Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2823: 269-289, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39052226

RESUMO

In healthy cells, membrane-anchored wild-type RAS proteins (i.e., HRAS, KRAS4A, KRAS4B, and NRAS) regulate critical cellular processes (e.g., proliferation, differentiation, survival). When mutated, RAS proteins are principal oncogenic drivers in approximately 30% of all human cancers. Among them, KRAS mutants are found in nearly 80% of all patients diagnosed with RAS-driven malignancies and are regarded as high-priority anti-cancer drug targets. Due to the lack of highly qualified/specific RAS isoform and mutant RAS monoclonal antibodies, there is a vital need for an effective antibody-free approach capable of identifying and quantifying membrane-bound RAS proteins in isoform- and mutation-specific manner. Here, we describe the development of a simple antibody-free protocol that relies on ultracentrifugation to isolate the membrane fraction coupled with single-dimensional (1D) sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) to fractionate and enrich membrane-bound endogenous RAS isoforms. Next, bottom-up proteomics that utilizes in-gel digestion followed by reversed-phase high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS2) is used for detection and relative quantitation of all wild-type RAS proteins (i.e., HRAS, KRAS4A, KRAS4B, and NRAS) and corresponding RAS mutants (e.g., G12D, G13D, G12S, G12V). Notably, this simple 1D-SDS-PAGE-HPLC-MS2-based protocol can be automated and widely applied to multiple cancer cell lines to investigate concentration changes in membrane-bound endogenous RAS proteins and corresponding mutants in the context of drug discovery.


Assuntos
Eletroforese em Gel de Poliacrilamida , Mutação , Proteínas Proto-Oncogênicas p21(ras) , Espectrometria de Massas em Tandem , Humanos , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Linhagem Celular Tumoral , Cromatografia Líquida/métodos , Eletroforese em Gel de Poliacrilamida/métodos , Espectrometria de Massas em Tandem/métodos , Membrana Celular/metabolismo , Proteômica/métodos , Neoplasias/genética , Neoplasias/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas ras/metabolismo , Proteínas ras/genética
2.
Oncotarget ; 12(20): 2022-2038, 2021 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-34611477

RESUMO

Lung cancer is the leading cause of cancer-related deaths in the USA and worldwide. Yet, about 95% of new drug candidates validated in preclinical phase eventually fail in clinical trials. Such a high attrition rate is attributed mostly to the inability of conventional two-dimensionally (2D) cultured cancer cells to mimic native three-dimensional (3D) growth of malignant cells in human tumors. To ascertain phenotypical differences between these two distinct culture conditions, we carried out a comparative proteomic analysis of a membrane fraction obtained from 3D- and 2D-cultured NSCLC model cell line NCI-H23. This analysis revealed a map of 1,166 (24%) protein species regulated in culture dependent manner, including differential regulation of a subset of cell surface-based CD molecules. We confirmed exclusive expression of CD99, CD146 and CD239 in 3D culture. Furthermore, label-free quantitation, targeting KRas proteoform-specific peptides, revealed upregulation of both wild type and monoallelic KRas4BG12C mutant at the surface of 3D cultured cells. In order to reduce the high attrition rate of new drug candidates, the results of this study strongly suggests exploiting base-line molecular profiling of a large number of patient-derived NSCLC cell lines grown in 2D and 3D culture, prior to actual drug candidate testing.

3.
Cancers (Basel) ; 13(15)2021 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-34359745

RESUMO

The ATM serine/threonine kinase (HGNC: ATM) is involved in initiation of repair of DNA double-stranded breaks, and ATM inhibitors are currently being tested as anti-cancer agents in clinical trials, where pharmacodynamic (PD) assays are crucial to help guide dose and scheduling and support mechanism of action studies. To identify and quantify PD biomarkers of ATM inhibition, we developed and analytically validated a 51-plex assay (DDR-2) quantifying protein expression and DNA damage-responsive phosphorylation. The median lower limit of quantification was 1.28 fmol, the linear range was over 3 orders of magnitude, the median inter-assay variability was 11% CV, and 86% of peptides were stable for storage prior to analysis. Use of the assay was demonstrated to quantify signaling following ionizing radiation-induced DNA damage in both immortalized lymphoblast cell lines and primary human peripheral blood mononuclear cells, identifying PD biomarkers for ATM inhibition to support preclinical and clinical studies.

4.
Endocrinology ; 161(2)2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31875919

RESUMO

Testosterone production occurs in the Leydig cells of the testes and is essential for virilization, development, reproduction, and quality of life. Although the steroidogenic proteins involved in cholesterol conversion to testosterone (T) are well characterized, the causes of reduced T during fetal, neonatal, and adult life remain uncertain. It is well established that normal cellular function is achieved through fine-tuning of multiple rather than single protein networks. Our objective was to use mass spectrometry (MS)-based proteomics to identify which cellular pathways, other than the steroidogenic machinery, influence testosterone production in MA-10 mouse tumor Leydig cells. The 14-3-3 family of scaffolds mediate protein-protein interactions facilitating the crosstalk between protein networks. We previously showed that in MA-10 cells, 14-3-3γ is a critical regulator of steroidogenesis. Therefore, identifying proteins that interact with 14-3-3γ during steroidogenesis could provide clues into the other networks involved. Using liquid chromatography (LC)-MS, we identified 688 proteins that interact with 14-3-3γ and thus potentially impact MA-10 cell steroidogenesis. The identified proteins belong to multiple protein networks, including endoplasmic reticulum-Golgi cargo sorting and vesicle biogenesis, micro ribonucleic acid-induced gene silencing, inflammation, and vesicle trafficking, to name a few. We found that silencing one of the candidates, Sec23ip, a protein known to be involved in vesicle trafficking, resulted in decreased steroidogenesis. We further showed that in Sec23ip-silenced MA-10 cells, cholesterol mobilization from the cytoplasmic membrane to mitochondria is impaired. Taken together these data suggest that Sec23ip is involved in cholesterol trafficking to supply cholesterol for acute steroidogenesis through its interactions with 14-3-3γ.


Assuntos
Proteínas 14-3-3/metabolismo , Proteínas de Transporte/metabolismo , Células Intersticiais do Testículo/metabolismo , Testosterona/biossíntese , Animais , Linhagem Celular Tumoral , Colesterol/metabolismo , Masculino , Camundongos
5.
Artigo em Inglês | MEDLINE | ID: mdl-31274965

RESUMO

Herein we introduce the Visual Mass-Spec Share (vMS-Share), a new public mass spectrometric (MS) repository and data mining website/resource freely accessible at https://vmsshare.nist.gov. vMS-Share is a web-based application developed for instant visualization of raw MS data with integrated display of metadata optimized for the sharing of proteomics and metabolomics experimental results. Each MS-based identification is linked to a given experiment and the entire experimental data can then be viewed using the link associated with a given peptide and/or small molecule. Interactive and user-friendly visualizations are provided to the user via variety of easily accessible search filters.

6.
Nanomedicine ; 17: 266-275, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30794962

RESUMO

The complement system plays an essential role in both innate and adaptive immunity. The traditional understanding of this system comes from studies investigating complement proteins produced by the liver and present in plasma to "complement" the immune cell-mediated response to invading pathogens. Recently, it has been reported that immune cells including, but not limited to, T-cells and monocytes, express complement proteins. This complement is referred to as intracellular (IC) and implicated in the regulation of T-cell activation. The mechanisms and the structure-activity relationship between nanomaterials and IC, however, are currently unknown. Herein, we describe a structure-activity relationship study demonstrating that under in vitro conditions, only polymeric materials with cationic surfaces activate IC in T-cells. The effect also depends on particle size and occurs through a mechanism involving membrane damage, thereby IC on the cell surface serves as a self-opsonization marker in response to the nanoparticle-triggered danger affecting the cell integrity.


Assuntos
Ativação do Complemento , Ativação Linfocitária , Nanopartículas/efeitos adversos , Polímeros/efeitos adversos , Linfócitos T/imunologia , Cátions/efeitos adversos , Cátions/química , Células Cultivadas , Ativação do Complemento/efeitos dos fármacos , Humanos , Células Jurkat , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Nanopartículas/química , Polímeros/química , Linfócitos T/efeitos dos fármacos
7.
Oncotarget ; 9(41): 26431-26452, 2018 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-29899869

RESUMO

The most widely used cancer animal model is the human-murine tumor xenograft. Unbiased molecular dissection of tumor parenchyma versus stroma in human-murine xenografts is critical for elucidating dysregulated protein networks/pathways and developing therapeutics that may target these two functionally codependent compartments. Although antibody-reliant technologies (e.g., immunohistochemistry, imaging mass cytometry) are capable of distinguishing tumor-proper versus stromal proteins, the breadth or extent of targets is limited. Here, we report an antibody-free targeted cross-species glycoproteomic (TCSG) approach that enables direct dissection of human tumor parenchyma from murine tumor stroma at the molecular/protein level in tumor xenografts at a selectivity rate presently unattainable by other means. This approach was used to segment/dissect and obtain the protein complement phenotype of the tumor stroma and parenchyma of the metastatic human lung adenocarcinoma A549 xenograft, with no need for tissue microdissection prior to mass-spectrometry analysis. An extensive molecular map of the tumor proper and the associated microenvironment was generated along with the top functional N-glycosylated protein networks enriched in each compartment. Importantly, immunohistochemistry-based cross-validation of selected parenchymal and stromal targets applied on human tissue samples of lung adenocarcinoma and normal adjacent tissue is indicative of a noteworthy translational capacity for this unique approach that may facilitate identifications of novel targets for next generation antibody therapies and development of real time preclinical tumor models.

8.
Elife ; 72018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29359686

RESUMO

While there have been tremendous efforts to target oncogenic RAS signaling from inside the cell, little effort has focused on the cell-surface. Here, we used quantitative surface proteomics to reveal a signature of proteins that are upregulated on cells transformed with KRASG12V, and driven by MAPK pathway signaling. We next generated a toolkit of recombinant antibodies to seven of these RAS-induced proteins. We found that five of these proteins are broadly distributed on cancer cell lines harboring RAS mutations. In parallel, a cell-surface CRISPRi screen identified integrin and Wnt signaling proteins as critical to RAS-transformed cells. We show that antibodies targeting CDCP1, a protein common to our proteomics and CRISPRi datasets, can be leveraged to deliver cytotoxic and immunotherapeutic payloads to RAS-transformed cancer cells and report for RAS signaling status in vivo. Taken together, this work presents a technological platform for attacking RAS from outside the cell.


Assuntos
Anticorpos/metabolismo , Antineoplásicos/metabolismo , Portadores de Fármacos/metabolismo , Fatores Imunológicos/metabolismo , Proteínas de Membrana/metabolismo , Terapia de Alvo Molecular/métodos , Neoplasias/terapia , Anticorpos/imunologia , Linhagem Celular Tumoral , Humanos , Fatores Imunológicos/imunologia , Proteínas de Membrana/imunologia , Proteínas ras/metabolismo
9.
Methods Mol Biol ; 1723: 95-110, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29344855

RESUMO

The heterogeneity present in solid tumors adds significant difficulty to scientific analysis and improved understanding. Fundamentally, solid tumor formation consists of cancer cells proper along with stromal elements. The burgeoning malignant process is dependent upon modified stromal elements. Collectively, the stroma forms an essential microenvironment, which is indispensable for the survival and growth of the malignant neoplasm. This cellular heterogeneity makes molecular profiling of solid tumors via mass spectrometry (MS)-based proteomics a daunting task. Laser capture microdissection (LCM) is commonly used to obtain distinct histological cell types (e.g., tumor parenchymal cells, stromal cells) from tumor tissue and attempt to address the tumor heterogeneity interference with downstream liquid chromatography (LC) MS analysis. To provide optimal LC-MS analysis of micro-scale and/or nano-scale tissue sections, we modified and optimized a silver-stained one-dimensional sodium dodecyl sulfate polyacrylamide gel electrophoresis (1D-SDS-PAGE) protocol for the LC-MS analysis of LCM-procured fresh-frozen tissue specimens. Presented is a detailed in-gel digestion protocol adjusted specifically to maximize the proteome coverage of amount-limited LCM samples, and facilitate in-depth molecular profiling. Following LCM, targeted tissue sections are further fractionated using silver-stained 1D-SDS-PAGE to resolve and visualize tissue proteins prior to in-gel digestion and subsequent LC-MS analysis.


Assuntos
Cromatografia Líquida/métodos , Eletroforese em Gel de Poliacrilamida/métodos , Secções Congeladas/métodos , Microdissecção e Captura a Laser/métodos , Neoplasias/metabolismo , Proteínas/análise , Espectrometria de Massas em Tandem/métodos , Separação Celular/métodos , Humanos , Proteínas/isolamento & purificação , Proteômica/métodos , Prata/química
10.
Methods Mol Biol ; 1647: 71-90, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28808996

RESUMO

The discovery of novel drug targets and biomarkers via mass spectrometry (MS)-based proteomic analysis of clinical specimens has proven to be challenging. The wide dynamic range of protein concentration in clinical specimens and the high background/noise originating from highly abundant proteins in tissue homogenates and serum/plasma encompass two major analytical obstacles. Immunoaffinity depletion of highly abundant blood-derived proteins from serum/plasma is a well-established approach adopted by numerous researchers; however, the utilization of this technique for immunodepletion of tissue homogenates obtained from fresh frozen clinical specimens is lacking. We first developed immunoaffinity depletion of highly abundant blood-derived proteins from tissue homogenates, using renal cell carcinoma as a model disease, and followed this study by applying it to different tissue types. Tissue homogenate immunoaffinity depletion of highly abundant proteins may be equally important as is the recognized need for depletion of serum/plasma, enabling more sensitive MS-based discovery of novel drug targets, and/or clinical biomarkers from complex clinical samples. Provided is a detailed protocol designed to guide the researcher through the preparation and immunoaffinity depletion of fresh frozen tissue homogenates for two-dimensional liquid chromatography, tandem mass spectrometry (2D-LC-MS/MS)-based molecular profiling of tissue specimens in the context of drug target and/or biomarker discovery.


Assuntos
Métodos Analíticos de Preparação de Amostras , Biomarcadores Farmacológicos/análise , Proteínas Sanguíneas/isolamento & purificação , Terapia de Alvo Molecular , Proteômica , Manejo de Espécimes/métodos , Espectrometria de Massas em Tandem/métodos , Carcinoma de Células Renais/metabolismo , Cromatografia Líquida/métodos , Humanos , Neoplasias Renais/metabolismo
11.
J Pharm Sci ; 106(7): 1760-1763, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28285019

RESUMO

Vaccine delivery systems play pivotal role in effective antigen delivery. These systems often contain adjuvants that stimulate specific immune response and are important for vaccines' efficacy and safety. Oil-in-water vaccine delivery lipid emulsion systems containing monophosphoryl lipid A (MPLA) as immune modulator have been extensively investigated in vaccine trials. Herein, we describe a simple orthogonal method, for quantitative measurement of MPLA in an oil-in-water lipid delivery system using direct transesterification reaction followed by gas-chromatography-mass spectrometry analysis. In this protocol, the transesterification reaction results in the release of fatty acid methyl esters followed by gas-chromatography-mass spectrometry-based targeted quantification of the specific 3-hydroxytetradecanoate fatty acid methyl ester to measure the concentration of MPLA in an oil-in-water lipid emulsion system.


Assuntos
Adjuvantes Imunológicos/análise , Emulsões/química , Lipídeo A/análogos & derivados , Óleos/química , Veículos Farmacêuticos/química , Vacinas/análise , Adjuvantes Imunológicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Esterificação , Cromatografia Gasosa-Espectrometria de Massas/métodos , Lipídeo A/administração & dosagem , Lipídeo A/análise , Vacinas/administração & dosagem , Água/química
12.
Oncotarget ; 7(52): 86948-86971, 2016 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-27894102

RESUMO

Oncogenic Ras mutants play a major role in the etiology of most aggressive and deadly carcinomas in humans. In spite of continuous efforts, effective pharmacological treatments targeting oncogenic Ras isoforms have not been developed. Cell-surface proteins represent top therapeutic targets primarily due to their accessibility and susceptibility to different modes of cancer therapy. To expand the treatment options of cancers driven by oncogenic Ras, new targets need to be identified and characterized at the surface of cancer cells expressing oncogenic Ras mutants. Here, we describe a mass spectrometry-based method for molecular profiling of the cell surface using KRasG12V transfected MCF10A (MCF10A-KRasG12V) as a model cell line of constitutively activated KRas and native MCF10A cells transduced with an empty vector (EV) as control. An extensive molecular map of the KRas surface was achieved by applying, in parallel, targeted hydrazide-based cell-surface capturing technology and global shotgun membrane proteomics to identify the proteins on the KRasG12V surface. This method allowed for integrated proteomic analysis that identified more than 500 cell-surface proteins found unique or upregulated on the surface of MCF10A-KRasG12V cells. Multistep bioinformatic processing was employed to elucidate and prioritize targets for cross-validation. Scanning electron microscopy and phenotypic cancer cell assays revealed changes at the cell surface consistent with malignant epithelial-to-mesenchymal transformation secondary to KRasG12V activation. Taken together, this dataset significantly expands the map of the KRasG12V surface and uncovers potential targets involved primarily in cell motility, cellular protrusion formation, and metastasis.


Assuntos
Proteínas de Membrana/análise , Proteínas Mutantes/análise , Proteômica/métodos , Proteínas Proto-Oncogênicas p21(ras)/análise , Antígenos CD/análise , Antígenos de Neoplasias , Basigina/análise , Moléculas de Adesão Celular/análise , Linhagem Celular Tumoral , Movimento Celular , Biologia Computacional , Transição Epitelial-Mesenquimal , Glicoproteínas/classificação , Glicoproteínas/fisiologia , Humanos , Espectrometria de Massas , Microscopia Eletrônica de Varredura , Proteínas de Neoplasias/análise
13.
Endocrinology ; 156(1): 346-59, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25380237

RESUMO

Neonatal gonocytes are direct precursors of spermatogonial stem cells, the cell pool that supports spermatogenesis. Although unipotent in vivo, gonocytes express pluripotency genes common with embryonic stem cells. Previously, we found that all-trans retinoic acid (RA) induced the expression of differentiation markers and a truncated form of platelet-derived growth factor receptor (PDGFR)ß in rat gonocytes, as well as in F9 mouse embryonal carcinoma cells, an embryonic stem cell-surrogate that expresses somatic lineage markers in response to RA. The present study is focused on identifying the signaling pathways involved in RA-induced gonocyte and F9 cell differentiation. Mitogen-activated protein kinase kinase (MEK) 1/2 activation was required during F9 cell differentiation towards somatic lineage, whereas its inhibition potentiated RA-induced Stra8 expression, suggesting that MEK1/2 acts as a lineage specification switch in F9 cells. In both cell types, RA increased the expression of the spermatogonial/premeiotic marker Stra8, which is in line with F9 cells being at a stage before somatic-germline lineage specification. Inhibiting PDGFR kinase activity reduced RA-induced Stra8 expression. Interestingly, RA increased the expression of PDGFRα variant forms in both cell types. Together, these results suggest a potential cross talk between RA and PDGFR signaling pathways in cell differentiation. RA receptor-α inhibition partially reduced RA effects on Stra8 in gonocytes, indicating that RA acts in part via RA receptor-α. RA-induced gonocyte differentiation was significantly reduced by inhibiting SRC (v-src avian sarcoma [Schmidt-Ruppin A-2] viral oncogene) and JAK2/STAT5 (Janus kinase 2/signal transducer and activator of transcription 5) activities, implying that these signaling molecules play a role in gonocyte differentiation. These results suggest that gonocyte and F9 cell differentiation is regulated via cross talk between RA and PDGFRs using different downstream pathways.


Assuntos
Células-Tronco de Carcinoma Embrionário/citologia , Células-Tronco de Carcinoma Embrionário/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Tretinoína/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/fisiologia , Animais , Animais Recém-Nascidos , Diferenciação Celular , Linhagem Celular , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica , Ceratolíticos/farmacologia , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Masculino , Camundongos , Fator de Crescimento Derivado de Plaquetas/genética , Ratos , Ratos Sprague-Dawley , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Testículo/citologia , Testículo/crescimento & desenvolvimento
14.
Biol Reprod ; 91(4): 96, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25210128

RESUMO

Mitochondria are home to many cellular processes, including oxidative phosphorylation and fatty acid metabolism, and in steroid-synthesizing cells, they are involved in cholesterol import and metabolism, which is the initiating step in steroidogenesis. The formation of macromolecular protein complexes aids in the regulation and efficiency of these mitochondrial functions, though because of their dynamic nature, they are hard to identify. To overcome this problem, we used Blue-Native PAGE with whole-gel mass spectrometry on isolated mitochondria from control and hormone-treated MA-10 mouse tumor Leydig cells. The presence of multiple mitochondrial protein complexes was shown. Although these were qualitatively similar under control and human chorionic gonadotropin (hCG)-stimulated conditions, quantitative differences in the components of the complexes emerged after hCG treatment. A prominent decrease was observed with proteins involved in fatty acid import into the mitochondria, implying that mitochondrial beta-oxidation is not essential for steroidogenesis. To confirm this observation, we inhibited fatty acid import utilizing the CPT1a inhibitor etomoxir, resulting in increased steroid production. Conversely, stimulation of mitochondrial beta-oxidation with metformin resulted in a dose-dependent reduction in steroidogenesis. These changes were accompanied by changes in mitochondrial respiration and in the lactic acid formed during glycolysis. Taken together, these results suggest that upon hormonal stimulation, mitochondria efficiently import cholesterol for steroid production at the expense of other lipids necessary for energy production, specifically fatty acids required for beta-oxidation.


Assuntos
Ácidos Graxos/metabolismo , Células Intersticiais do Testículo/metabolismo , Mitocôndrias/metabolismo , Esteroides/biossíntese , Animais , Transporte Biológico , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Hipoglicemiantes/farmacologia , Masculino , Metformina/farmacologia , Camundongos , Oxirredução , Transcriptoma
15.
J Biol Chem ; 289(38): 26542-26553, 2014 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-25086053

RESUMO

The 14-3-3 protein family comprises adaptors and scaffolds that regulate intracellular signaling pathways. The 14-3-3γ isoform is a negative regulator of steroidogenesis that is hormonally induced and transiently functions at the initiation of steroidogenesis by delaying maximal steroidogenesis in MA-10 mouse tumor Leydig cells. Treatment of MA-10 cells with the cAMP analog 8-bromo-cAMP (8-Br-cAMP), which stimulates steroidogenesis, triggers the interaction of 14-3-3γ with the steroidogenic acute regulatory protein (STAR) in the cytosol, limiting STAR activity to basal levels. Over time, this interaction ceases, allowing for a 2-fold induction in STAR activity and maximal increase in the rate of steroid formation. The 14-3-3γ/STAR pattern of interaction was found to be opposite that of the 14-3-3γ homodimerization pattern. Phosphorylation and acetylation of 14-3-3γ showed similar patterns to homodimerization and STAR binding, respectively. 14-3-3γ Ser(58) phosphorylation and 14-3-3γ Lys(49) acetylation were blocked using trans-activator of HIV transcription factor 1 peptides coupled to 14-3-3γ sequences containing Ser(58) or Lys(49). Blocking either one of these modifications further induced 8-Br-cAMP-induced steroidogenesis while reducing lipid storage, suggesting that the stored cholesterol is used for steroid formation. Taken together, these results indicate that Ser(58) phosphorylation and Lys(49) acetylation of 14-3-3γ occur in a coordinated time-dependent manner to regulate 14-3-3γ homodimerization. 14-3-3γ Ser(58) phosphorylation is required for STAR interactions under control conditions, and 14-3-3γ Lys(49) acetylation is important for the cAMP-dependent induction of these interactions.


Assuntos
Proteínas 14-3-3/metabolismo , Colesterol/biossíntese , AMP Cíclico/fisiologia , Células Intersticiais do Testículo/metabolismo , Processamento de Proteína Pós-Traducional , Acetilação , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Masculino , Camundongos , Dados de Sequência Molecular , Fosfoproteínas/metabolismo , Fosforilação , Ligação Proteica , Multimerização Proteica
16.
Biomark Med ; 8(2): 269-86, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24521024

RESUMO

The discovery of clinically relevant cancer biomarkers using mass spectrometry (MS)-based proteomics has proven difficult, primarily because of the enormous dynamic range of blood-derived protein concentrations and the fact that the 22 most abundant blood-derived proteins constitute approximately 99% of the total plasma protein mass. Immunodepletion of clinical body fluid specimens (e.g., serum/plasma) for the removal of highly abundant proteins is a reasonable and reproducible solution. Often overlooked, clinical tissue specimens also contain a formidable amount of highly abundant blood-derived proteins present in tissue-embedded networks of blood/lymph capillaries and interstitial fluid. Hence, the dynamic range impediment to biomarker discovery remains a formidable obstacle, regardless of clinical sample type (solid tissue and/or body fluid). Thus, we optimized and applied simultaneous immunodepletion of blood-derived proteins from solid tissue and peripheral blood, using clear cell renal cell carcinoma as a model disease. Integrative analysis of data from this approach and genomic data obtained from the same type of tumor revealed concordant key pathways and protein targets germane to clear cell renal cell carcinoma. This includes the activation of the lipogenic pathway characterized by increased expression of adipophilin (PLIN2) along with 'cadherin switching', a phenomenon indicative of transcriptional reprogramming linked to renal epithelial dedifferentiation. We also applied immunodepletion of abundant blood-derived proteins to various tissue types (e.g., adipose tissue and breast tissue) showing unambiguously that the removal of abundant blood-derived proteins represents a powerful tool for the reproducible profiling of tissue proteomes. Herein, we show that the removal of abundant blood-derived proteins from solid tissue specimens is of equal importance to depletion of body fluids and recommend its routine use in the context of biological discovery and/or cancer biomarker research. Finally, this perspective presents the background, rationale and strategy for using tissue-directed high-resolution/accuracy MS-based shotgun proteomics to detect genuine tumor proteins in the peripheral blood of a patient diagnosed with nonmetastatic cancer, employing concurrent liquid chromatography-MS analysis of immunodepleted clinical tissue and blood specimens.


Assuntos
Biomarcadores Tumorais/análise , Espectrometria de Massas , Anticorpos/imunologia , Biomarcadores Tumorais/sangue , Proteínas Sanguíneas/imunologia , Proteínas Sanguíneas/isolamento & purificação , Carcinoma de Células Renais/sangue , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Cromatografia Líquida de Alta Pressão , Perfilação da Expressão Gênica , Humanos , Neoplasias Renais/sangue , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Proteômica
17.
Mol Cell Proteomics ; 13(3): 907-17, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24443746

RESUMO

Adoption of targeted mass spectrometry (MS) approaches such as multiple reaction monitoring (MRM) to study biological and biomedical questions is well underway in the proteomics community. Successful application depends on the ability to generate reliable assays that uniquely and confidently identify target peptides in a sample. Unfortunately, there is a wide range of criteria being applied to say that an assay has been successfully developed. There is no consensus on what criteria are acceptable and little understanding of the impact of variable criteria on the quality of the results generated. Publications describing targeted MS assays for peptides frequently do not contain sufficient information for readers to establish confidence that the tests work as intended or to be able to apply the tests described in their own labs. Guidance must be developed so that targeted MS assays with established performance can be made widely distributed and applied by many labs worldwide. To begin to address the problems and their solutions, a workshop was held at the National Institutes of Health with representatives from the multiple communities developing and employing targeted MS assays. Participants discussed the analytical goals of their experiments and the experimental evidence needed to establish that the assays they develop work as intended and are achieving the required levels of performance. Using this "fit-for-purpose" approach, the group defined three tiers of assays distinguished by their performance and extent of analytical characterization. Computational and statistical tools useful for the analysis of targeted MS results were described. Participants also detailed the information that authors need to provide in their manuscripts to enable reviewers and readers to clearly understand what procedures were performed and to evaluate the reliability of the peptide or protein quantification measurements reported. This paper presents a summary of the meeting and recommendations.


Assuntos
Bioensaio/métodos , Biologia , Espectrometria de Massas/métodos , Medicina , Peptídeos/metabolismo , Animais , Guias como Assunto , Humanos , Marcação por Isótopo , Proteômica/normas , Padrões de Referência , Software
18.
J Immunol ; 191(2): 892-901, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23776175

RESUMO

Recognition of microbial products by TLRs is critical for mediating innate immune responses to invading pathogens. In this study, we identify a novel scaffold protein in TLR4 signaling called SAM and SH3 domain containing protein 1 (SASH1). Sash1 is expressed across all microvascular beds and functions as a scaffold molecule to independently bind TRAF6, TAK1, IκB kinase α, and IκB kinase ß. This interaction fosters ubiquitination of TRAF6 and TAK1 and promotes LPS-induced NF-κB, JNK, and p38 activation, culminating in increased production of proinflammatory cytokines and increased LPS-induced endothelial migration. Our findings suggest that SASH1 acts to assemble a signaling complex downstream of TLR4 to activate early endothelial responses to receptor activation.


Assuntos
Células Endoteliais/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Receptor 4 Toll-Like/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Movimento Celular , Ativação Enzimática , Quinase I-kappa B/metabolismo , Imunidade Inata , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lipopolissacarídeos/imunologia , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Interferência de RNA , Transdução de Sinais , Proteínas Supressoras de Tumor/genética , Ubiquitinação , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Methods Mol Biol ; 1002: 71-83, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23625395

RESUMO

The discovery of effective cancer biomarkers is essential for the development of both advanced molecular diagnostics and new therapies/medications. Finding and exploiting useful clinical biomarkers for cancer patients is fundamentally linked to improving outcomes. Towards these aims, the heterogeneous nature of tumors represents a significant problem. Thus, methods establishing an effective functional linkage between laser capture microdissection (LCM) and mass spectrometry (MS) provides for an enhanced molecular profiling of homogenous, specifically targeted cell populations from solid tumors. Utilizing frozen tissue avoids molecular degradation and bias that can be induced by other preservation techniques. Since clinical samples are often of a small quantity, tissue losses must be minimized. Therefore, all steps are carried out in the same single tube. Proteins are identified through peptide sequencing and subsequent matching against a specific proteomic database. Using such an approach enhances clinical biomarker discovery in the following ways. First, LCM allows for the complexity of a solid tumor to be reduced. Second, MS provides for the profiling of proteins, which are the ultimate bio-effectors. Third, by selecting for tumor proper or microenvironment-specific cells from clinical samples, the heterogeneity of individual solid tumors is directly addressed. Finally, since proteins are the targets of most pharmaceuticals, the enriched protein data streams can then be further analyzed for potential biomarkers, drug targets, pathway elucidation, as well as an enhanced understanding of the various pathologic processes under study. Within this context, the following method illustrates in detail a synergy between LCM and MS for an enhanced molecular profiling of solid tumors and clinical biomarker discovery.


Assuntos
Secções Congeladas , Microdissecção e Captura a Laser/métodos , Proteínas/análise , Proteoma/análise , Proteômica/métodos , Biomarcadores Tumorais/análise , Separação Celular , Humanos , Espectrometria de Massas , Neoplasias/química , Proteínas/química
20.
Methods Mol Biol ; 1002: 133-49, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23625401

RESUMO

Differential (18)O/(16)O stable isotopic labeling that relies on post-digestion (18)O exchange is a simple and efficient method for the relative quantitation of proteins in complex mixtures. This method incorporates two (18)O atoms onto the C-termini of proteolytic peptides resulting in a 4 Da mass-tag difference between (18)O- and (16)O-labeled peptides. This allows for wide-range relative quantitation of proteins in complex mixtures using shotgun proteomics. Because of minimal sample consumption and unrestricted peptide tagging, the post-digestion (18)O exchange is suitable for labeling of low-abundance membrane proteins enriched from cancer cell lines or clinical specimens, including tissues and body fluids. This chapter describes a protocol that applies post-digestion (18)O labeling to elucidate putative endogenous tumor hypoxia markers in the plasma membrane fraction enriched from a hypoxia-adapted malignant melanoma cell line. Plasma membrane proteins from hypoxic and normoxic cells were differentially tagged using (18)O/(16)O stable isotopic labeling. The initial tryptic digestion and solubilization of membrane proteins were carried out in a buffer containing 60 % methanol followed by post-digestion (18)O exchange/labeling in buffered 20 % methanol. The differentially labeled peptides were mixed in a 1:1 ratio and fractionated using off-line strong cation exchange (SCX) liquid chromatography followed by on-line reversed-phase nano-flow RPLC-MS identification and quantitation of peptides/proteins in respective SCX fractions. The present protocol illustrates the utility of (18)O/(16)O stable isotope labeling in the context of quantitative shotgun proteomics that provides a basis for the discovery of hypoxia-induced membrane protein markers in malignant melanoma cell lines.


Assuntos
Biomarcadores Tumorais/análise , Marcação por Isótopo , Melanoma/metabolismo , Isótopos de Oxigênio/química , Proteínas/análise , Tripsina/metabolismo , Hipóxia Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Cromatografia Líquida , Humanos , Espectrometria de Massas , Proteínas/química , Proteômica/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA