Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Physiol Rep ; 8(19): e14610, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33038060

RESUMO

BACKGROUND: Resistant Starch (RS) improves CKD outcomes. In this report, we study how RS modulates host-microbiome interactions in CKD by measuring changes in the abundance of proteins and bacteria in the gut. In addition, we demonstrate RS-mediated reduction in CKD-induced kidney damage. METHODS: Eight mice underwent 5/6 nephrectomy to induce CKD and eight served as healthy controls. CKD and Healthy (H) groups were further split into those receiving RS (CKDRS, n = 4; HRS, n = 4) and those on normal diet (CKD, n = 4, H, n = 4). Kidney injury was evaluated by measuring BUN/creatinine and by histopathological evaluation. Cecal contents were analyzed using mass spectrometry-based metaproteomics and de novo sequencing using PEAKS. All the data were analyzed using R/Bioconductor packages. RESULTS: The 5/6 nephrectomy compromised kidney function as seen by an increase in BUN/creatinine compared to healthy groups. Histopathology of kidney sections showed reduced tubulointerstitial injury in the CKDRS versus CKD group; while no significant difference in BUN/creatinine was observed between the two CKD groups. Identified proteins point toward a higher population of butyrate-producing bacteria, reduced abundance of mucin-degrading bacteria in the RS fed groups, and to the downregulation of indole metabolism in CKD groups. CONCLUSION: RS slows the progression of chronic kidney disease. Resistant starch supplementation leads to active bacterial proliferation and the reduction of harmful bacterial metabolites.


Assuntos
Microbioma Gastrointestinal/fisiologia , Rim/metabolismo , Insuficiência Renal Crônica/metabolismo , Amido Resistente/metabolismo , Animais , Bactérias/metabolismo , Nitrogênio da Ureia Sanguínea , Modelos Animais de Doenças , Progressão da Doença , Rim/fisiopatologia , Masculino , Camundongos , Insuficiência Renal Crônica/fisiopatologia
2.
Mol Omics ; 16(4): 316-326, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32347222

RESUMO

Quantitative proteomics generates large datasets with increasing depth and quantitative information. With the advance of mass spectrometry and increasingly larger data sets, streamlined methodologies and tools for analysis and visualization of phosphoproteomics are needed both at the protein and modified peptide levels. To assist in addressing this need, we developed ProteoViz, which includes a set of R scripts that perform normalization and differential expression analysis of both the proteins and enriched phosphorylated peptides, and identify sequence motifs, kinases, and gene set enrichment pathways. The tool generates interactive visualization plots that allow users to interact with the phosphoproteomics results and quickly identify proteins and phosphorylated peptides of interest for their biological study. The tool also links significant phosphosites with sequence motifs and pathways that will help explain the experimental conditions and guide future experiments. Here, we present the workflow and demonstrate its functionality by analyzing a phosphoproteomic data set from two lymphoma cell lines treated with kinase inhibitors. The scripts and data are freely available at and via the ProteomeXchange with identifier PXD015606.


Assuntos
Biologia Computacional/métodos , Fosfoproteínas/metabolismo , Mapeamento de Interação de Proteínas/métodos , Proteômica , Software , Motivos de Aminoácidos , Linhagem Celular , Bases de Dados Genéticas , Feminino , Humanos , Masculino , Espectrometria de Massas/métodos , Ligação Proteica , Proteômica/métodos , Transdução de Sinais , Fluxo de Trabalho
3.
PLoS One ; 14(1): e0199274, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30699108

RESUMO

BACKGROUND: Resistant starch is a prebiotic metabolized by the gut bacteria. It has been shown to attenuate chronic kidney disease (CKD) progression in rats. Previous studies employed taxonomic analysis using 16S rRNA sequencing and untargeted metabolomics profiling. Here we expand these studies by metaproteomics, gaining new insight into the host-microbiome interaction. METHODS: Differences between cecum contents in CKD rats fed a diet containing resistant starch with those fed a diet containing digestible starch were examined by comparative metaproteomics analysis. Taxonomic information was obtained using unique protein sequences. Our methodology results in quantitative data covering both host and bacterial proteins. RESULTS: 5,834 proteins were quantified, with 947 proteins originating from the host organism. Taxonomic information derived from metaproteomics data surpassed previous 16S RNA analysis, and reached species resolutions for moderately abundant taxonomic groups. In particular, the Ruminococcaceae family becomes well resolved-with butyrate producers and amylolytic species such as R. bromii clearly visible and significantly higher while fibrolytic species such as R. flavefaciens are significantly lower with resistant starch feeding. The observed changes in protein patterns are consistent with fiber-associated improvement in CKD phenotype. Several known host CKD-associated proteins and biomarkers of impaired kidney function were significantly reduced with resistant starch supplementation. Data are available via ProteomeXchange with identifier PXD008845. CONCLUSIONS: Metaproteomics analysis of cecum contents of CKD rats with and without resistant starch supplementation reveals changes within gut microbiota at unprecedented resolution, providing both functional and taxonomic information. Proteins and organisms differentially abundant with RS supplementation point toward a shift from mucin degraders to butyrate producers.


Assuntos
Proteínas de Bactérias/análise , Ceco/microbiologia , Microbioma Gastrointestinal , Proteoma/análise , Proteômica , Insuficiência Renal Crônica/induzido quimicamente , Ruminococcus , Amido/efeitos adversos , Animais , Progressão da Doença , Masculino , Ratos , Ratos Sprague-Dawley , Insuficiência Renal Crônica/microbiologia , Ruminococcus/classificação , Ruminococcus/crescimento & desenvolvimento , Amido/farmacologia
4.
J Proteome Res ; 17(10): 3384-3395, 2018 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-30209945

RESUMO

The staphylococcal accessory regulator A ( sarA) impacts the extracellular accumulation of Staphylococcus aureus virulence factors at the level of intracellular production and extracellular protease-mediated degradation. We previously used a proteomics approach that measures protein abundance of all proteoforms to demonstrate that mutation of sarA results in increased levels of extracellular proteases and assesses the impact of this on the accumulation of S. aureus exoproteins. Our previous approach was limited as it did not take into account that large, stable proteolytic products from a given protein could result in false negatives when quantified by total proteoforms. Here, our goal was to use an expanded proteomics approach utilizing a dual quantitative method for measuring abundance at both the total proteoform and full-length exoprotein levels to alleviate these false negatives and thereby provide for characterization of protease-dependent and -independent effects of sarA mutation on the S. aureus exoproteome. Proteins present in conditioned medium from overnight, stationary phase cultures of the USA300 strain LAC, an isogenic sarA mutant, and a sarA mutant unable to produce any of the known extracellular proteases ( sarA/protease) were resolved using one-dimensional gel electrophoresis. Quantitative proteomic comparisons of sarA versus sarA/protease mutants identified proteins that were cleaved in a protease-dependent manner owing to mutation of sarA, and comparisons of sarA/protease mutant versus the LAC parent strain identified proteins in which abundance was altered in a sarA mutant in a protease-independent manner. Furthermore, the proteins uniquely identified by the full-length data analysis approach eliminated false negatives observed in the total proteoform analysis. This expanded approach provided for a more comprehensive analysis of the impact of mutating sarA on the S. aureus exoproteome.


Assuntos
Proteínas de Bactérias/metabolismo , Peptídeo Hidrolases/metabolismo , Proteoma/metabolismo , Proteômica/métodos , Staphylococcus aureus/metabolismo , Proteínas de Bactérias/genética , Biofilmes , Regulação Bacteriana da Expressão Gênica , Staphylococcus aureus Resistente à Meticilina/genética , Staphylococcus aureus Resistente à Meticilina/metabolismo , Mutação/genética , Proteoma/genética , Staphylococcus aureus/genética , Staphylococcus aureus/fisiologia , Espectrometria de Massas em Tandem , Virulência/genética
5.
PLoS One ; 12(3): e0174771, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28350824

RESUMO

Acute radiation syndrome (ARS) is a complex multi-organ disease resulting from total body exposure to high doses of radiation. Individuals can be exposed to total body irradiation (TBI) in a number of ways, including terrorist radiological weapons or nuclear accidents. In order to determine whether an individual has been exposed to high doses of radiation and needs countermeasure treatment, robust biomarkers are needed to estimate radiation exposure from biospecimens such as blood or urine. In order to identity such candidate biomarkers of radiation exposure, high-resolution proteomics was used to analyze plasma from non-human primates following whole body irradiation (Co-60 at 6.7 Gy and 7.4 Gy) with a twelve day observation period. A total of 663 proteins were evaluated from the plasma proteome analysis. A panel of plasma proteins with characteristic time- and dose-dependent changes was identified. In addition to the plasma proteomics study reported here, we recently identified candidate biomarkers using urine from these same non-human primates. From the proteomic analysis of both plasma and urine, we identified ten overlapping proteins that significantly differentiate both time and dose variables. These shared plasma and urine proteins represent optimal candidate biomarkers of radiation exposure.


Assuntos
Proteínas Sanguíneas/análise , Proteoma/efeitos da radiação , Proteômica/métodos , Irradiação Corporal Total/métodos , Animais , Biomarcadores/sangue , Proteínas Sanguíneas/classificação , Análise por Conglomerados , Relação Dose-Resposta à Radiação , Eletroforese em Gel de Poliacrilamida , Raios gama , Macaca mulatta , Espectrometria de Massas/métodos , Proteoma/classificação , Transdução de Sinais/efeitos da radiação , Fatores de Tempo
6.
Toxicology ; 378: 114-124, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28082109

RESUMO

Cytochrome P450 2E1 (CYP2E1) metabolizes low molecular weight hydrophobic compounds, including 1,3-butadiene, which is converted by CYP2E1 to electrophilic epoxide metabolites that covalently modify cellular proteins and DNA. Previous CYP2E1 studies have mainly focused on the enzyme localized in the endoplasmic reticulum (erCYP2E1); however, active CYP2E1 has also been found in mitochondria (mtCYP2E1) and the distribution of CYP2E1 between organelles can influence an individual's response to exposure. Relatively few studies have focused on the contribution of mtCYP2E1 to activation of chemical toxicants. We hypothesized that CYP2E1 bioactivation of 1,3-butadiene within mitochondria adversely affects mitochondrial respiratory complexes I-IV. A population of Collaborative Cross mice was exposed to air (control) or 200ppm 1,3-butadiene. Subcellular fractions (mitochondria, DNA, and microsomes) were collected from frozen livers and CYP2E1 activity was measured in microsomes and mitochondria. Individual activities of mitochondrial respiratory complexes I-IV were measured using in vitro assays and purified mitochondrial fractions. In air- and 1,3-butadiene-exposed mouse samples, mtDNA copy numbers were assessed by RT-PCR, and mtDNA integrity was assessed through a PCR-based assay. No significant changes in mtDNA copy number or integrity were observed; however, there was a decrease in overall activity of mitochondrial respiratory complexes I, II, and IV after 1,3-butadiene exposure. Additionally, higher mtCYP2E1 (but not erCYP2E1) activity was correlated with decreased mitochondrial respiratory complex activity (in complexes I-IV) in the 1,3-butadiene-exposed (not control) animals. Together, these results represent the first in vivo link between mitochondrial CYP2E1 activity and mitochondrial toxicity.


Assuntos
Butadienos/toxicidade , Carcinógenos/toxicidade , Citocromo P-450 CYP2E1/metabolismo , Mitocôndrias Hepáticas/efeitos dos fármacos , Animais , Variações do Número de Cópias de DNA , DNA Mitocondrial/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Camundongos , Mitocôndrias Hepáticas/metabolismo , Succinato Desidrogenase/metabolismo
7.
J Proteomics Bioinform ; 9(Suppl 10)2016.
Artigo em Inglês | MEDLINE | ID: mdl-26962295

RESUMO

The molecular effects of total body gamma-irradiation exposure are of critical importance as large populations of people could be exposed either by terrorists, nuclear blast, or medical therapy. In this study, we aimed to identify changes in the urine proteome using a non-human primate model system, Rhesus macaque, in order to characterize effects of acute radiation syndrome following whole body irradiation (Co-60) at 6.7 Gy and 7.4 Gy with a twelve day observation period. The urine proteome is potentially a valuable and non-invasive diagnostic for radiation exposure. Using high-resolution mass spectrometry, we identified 2346 proteins in the urine proteome. We show proteins involved in disease, cell adhesion, and metabolic pathway were significantly changed upon exposure to differing levels and durations of radiation exposure. Cell damage increased at a faster rate at 7.4 Gy compared with 6.7 Gy exposures. We report sets of proteins that are putative biomarkers of time- and dose-dependent radiation exposure. The proteomic study presented here is a comprehensive analysis of the urine proteome following radiation exposure.

8.
Mol Cell Proteomics ; 15(3): 765-75, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26621846

RESUMO

Normal cell growth is characterized by a regulated epigenetic program that drives cellular activities such as gene transcription, DNA replication, and DNA damage repair. Perturbation of this epigenetic program can lead to events such as mis-regulation of gene transcription and diseases such as cancer. To begin to understand the epigenetic program correlated to the development of melanoma, we performed a novel quantitative mass spectrometric analysis of histone post-translational modifications mis-regulated in melanoma cell culture as well as patient tumors. Aggressive melanoma cell lines as well as metastatic melanoma were found to have elevated histone H3 Lys(27) trimethylation (H3K27me3) accompanied by overexpressed methyltransferase EZH2 that adds the specific modification. The altered epigenetic program that led to elevated H3K27me3 in melanoma cell culture was found to directly silence transcription of the tumor suppressor genes RUNX3 and E-cadherin. The EZH2-mediated silencing of RUNX3 and E-cadherin transcription was also validated in advanced stage human melanoma tissues. This is the first study focusing on the detailed epigenetic mechanisms leading to EZH2-mediated silencing of RUNX3 and E-cadherin tumor suppressors in melanoma. This study underscores the utility of using high resolution mass spectrometry to identify mis-regulated epigenetic programs in diseases such as cancer, which could ultimately lead to the identification of biological markers for diagnostic and prognostic applications.


Assuntos
Histonas/metabolismo , Lisina/metabolismo , Espectrometria de Massas/métodos , Mieloma Múltiplo/metabolismo , Regulação para Cima , Caderinas/genética , Linhagem Celular Tumoral , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Humanos , Metilação , Mieloma Múltiplo/genética , Metástase Neoplásica , Processamento de Proteína Pós-Traducional
9.
PLoS One ; 10(6): e0127943, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26065416

RESUMO

Constitutive activation of the Rearranged during Transfection (RET) proto-oncogene leads to the development of MEN2A medullary thyroid cancer (MTC). The relatively clear genotype/phenotype relationship seen with RET mutations and the development of MEN2A is unusual in the fact that a single gene activity can drive the progression towards metastatic disease. Despite knowing the oncogene responsible for MEN2A, MTC, like most tumors of neural crest origin, remains largely resistant to chemotherapy. Constitutive activation of RET in a SK-N-MC cell line model reduces cell sensitivity to chemotherapy. In an attempt to identify components of the machinery responsible for the observed RET induced chemoresistance, we performed a proteomic screen of histones and associated proteins in cells with a constitutively active RET signaling pathway. The proteomic approach identified DNA-PKcs, a DNA damage response protein, as a target of the RET signaling pathway. Active DNA-PKcs, which is phosphorylated at site serine 2056 and localized to chromatin, was elevated within our model. Treatment with the RET inhibitor RPI-1 significantly reduced s2056 phosphorylation in RET cells as well as in a human medullary thyroid cancer cell line. Additionally, inhibition of DNA-PKcs activity diminished the chemoresistance observed in both cell lines. Importantly, we show that activated DNA-PKcs is elevated in medullary thyroid tumor samples and that expression correlates with expression of RET in thyroid tumors. These results highlight one mechanism by which RET signaling likely primes cells for rapid response to DNA damage and suggests DNA-PKcs as an additional target in MTC.


Assuntos
Carcinoma Neuroendócrino/metabolismo , Proteína Quinase Ativada por DNA/metabolismo , Proteínas Nucleares/metabolismo , Proteômica/métodos , Proteínas Proto-Oncogênicas c-ret/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Indóis/farmacologia , Fosforilação/efeitos dos fármacos , Proto-Oncogene Mas , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...