Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Traffic ; 18(7): 465-484, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28382714

RESUMEN

The covalent attachment of ubiquitin onto proteins can elicit a variety of downstream consequences. Attachment is mediated by a large array of E3 ubiquitin ligases, each thought be subject to regulatory control and to have a specific repertoire of substrates. Assessing the biological roles of ligases, and in particular, identifying their biologically relevant substrates has been a persistent yet challenging question. In this study, we describe tools that may help achieve both of these goals. We describe a strategy whereby the activity of a ubiquitin ligase has been enzymatically reversed, accomplished by fusing it to a catalytic domain of an exogenous deubiquitinating enzyme. We present a library of 72 "anti-ligases" that appear to work in a dominant-negative fashion to stabilize their cognate substrates against ubiquitin-dependent proteasomal and lysosomal degradation. We then used the ligase-deubiquitinating enzyme (DUb) library to screen for E3 ligases involved in post-Golgi/endosomal trafficking. We identify ligases previously implicated in these pathways (Rsp5 and Tul1), in addition to ligases previously localized to endosomes (Pib1 and Vps8). We also document an optimized workflow for isolating and analyzing the "ubiquitome" of yeast, which can be used with mass spectrometry to identify substrates perturbed by expression of particular ligase-DUb fusions.


Asunto(s)
Enzimas Desubicuitinizantes/metabolismo , Degradación Asociada con el Retículo Endoplásmico , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Membrana Celular/metabolismo , Enzimas Desubicuitinizantes/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Endosomas/enzimología , Aparato de Golgi/enzimología , Lisosomas/enzimología , Plásmidos , Transporte de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas de Saccharomyces cerevisiae/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
2.
Proc Natl Acad Sci U S A ; 113(16): E2335-44, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27044074

RESUMEN

The large-conductance, voltage-gated, calcium (Ca(2+))-activated potassium channel (BKCa) plays an important role in regulating Ca(2+)signaling and is implicated in the maintenance of uterine quiescence during pregnancy. We used immunopurification and mass spectrometry to identify proteins that interact with BKCain myometrium samples from term pregnant (≥37 wk gestation) women. From this screen, we identified alpha-2-macroglobulin (α2M). We then used immunoprecipitation followed by immunoblot and the proximity ligation assay to confirm the interaction between BKCaand both α2M and its receptor, low-density lipoprotein receptor-related protein 1 (LRP1), in cultured primary human myometrial smooth muscle cells (hMSMCs). Single-channel electrophysiological recordings in the cell-attached configuration demonstrated that activated α2M (α2M*) increased the open probability of BKCain an oscillatory pattern in hMSMCs. Furthermore, α2M* caused intracellular levels of Ca(2+)to oscillate in oxytocin-primed hMSMCs. The initiation of oscillations required an interaction between α2M* and LRP1. By using Ca(2+)-free medium and inhibitors of various Ca(2+)signaling pathways, we demonstrated that the oscillations required entry of extracellular Ca(2+)through store-operated Ca(2+)channels. Finally, we found that the specific BKCablocker paxilline inhibited the oscillations, whereas the channel opener NS11021 increased the rate of these oscillations. These data demonstrate that α2M* and LRP1 modulate the BKCachannel in human myometrium and that BKCaand its immunomodulatory interacting partners regulate Ca(2+)dynamics in hMSMCs during pregnancy.


Asunto(s)
Señalización del Calcio/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Miocitos del Músculo Liso/metabolismo , Miometrio/metabolismo , Embarazo/metabolismo , alfa-Macroglobulinas/metabolismo , Adulto , Células Cultivadas , Femenino , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Miometrio/citología , Tercer Trimestre del Embarazo/metabolismo
3.
Mol Cell Proteomics ; 15(6): 2169-85, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27099343

RESUMEN

Mutations in genes encoding components of the sarcolemmal dystrophin-glycoprotein complex (DGC) are responsible for a large number of muscular dystrophies. As such, molecular dissection of the DGC is expected to both reveal pathological mechanisms, and provides a biological framework for validating new DGC components. Establishment of the molecular composition of plasma-membrane protein complexes has been hampered by a lack of suitable biochemical approaches. Here we present an analytical workflow based upon the principles of protein correlation profiling that has enabled us to model the molecular composition of the DGC in mouse skeletal muscle. We also report our analysis of protein complexes in mice harboring mutations in DGC components. Bioinformatic analyses suggested that cell-adhesion pathways were under the transcriptional control of NFκB in DGC mutant mice, which is a finding that is supported by previous studies that showed NFκB-regulated pathways underlie the pathophysiology of DGC-related muscular dystrophies. Moreover, the bioinformatic analyses suggested that inflammatory and compensatory mechanisms were activated in skeletal muscle of DGC mutant mice. Additionally, this proteomic study provides a molecular framework to refine our understanding of the DGC, identification of protein biomarkers of neuromuscular disease, and pharmacological interrogation of the DGC in adult skeletal muscle https://www.mda.org/disease/congenital-muscular-dystrophy/research.


Asunto(s)
Distrofina/metabolismo , Glicoproteínas/metabolismo , Distrofias Musculares/metabolismo , Proteómica/métodos , Animales , Biología Computacional , Distrofina/genética , Humanos , Ratones , Músculo Esquelético/metabolismo , Mutación , Mapas de Interacción de Proteínas
4.
J Biol Chem ; 291(36): 18818-42, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27365400

RESUMEN

Aberrant androgen receptor (AR)-dependent transcription is a hallmark of human prostate cancers. At the molecular level, ligand-mediated AR activation is coordinated through spatial and temporal protein-protein interactions involving AR-interacting proteins, which we designate the "AR-interactome." Despite many years of research, the ligand-sensitive protein complexes involved in ligand-mediated AR activation in prostate tumor cells have not been clearly defined. Here, we describe the development, characterization, and utilization of a novel human LNCaP prostate tumor cell line, N-AR, which stably expresses wild-type AR tagged at its N terminus with the streptavidin-binding peptide epitope (streptavidin-binding peptide-tagged wild-type androgen receptor; SBP-AR). A bioanalytical workflow involving streptavidin chromatography and label-free quantitative mass spectrometry was used to identify SBP-AR and associated ligand-sensitive cytosolic proteins/protein complexes linked to AR activation in prostate tumor cells. Functional studies verified that ligand-sensitive proteins identified in the proteomic screen encoded modulators of AR-mediated transcription, suggesting that these novel proteins were putative SBP-AR-interacting proteins in N-AR cells. This was supported by biochemical associations between recombinant SBP-AR and the ligand-sensitive coatomer protein complex I (COPI) retrograde trafficking complex in vitro Extensive biochemical and molecular experiments showed that the COPI retrograde complex regulates ligand-mediated AR transcriptional activation, which correlated with the mobilization of the Golgi-localized ARA160 coactivator to the nuclear compartment of prostate tumor cells. Collectively, this study provides a bioanalytical strategy to validate the AR-interactome and define novel AR-interacting proteins involved in ligand-mediated AR activation in prostate tumor cells. Moreover, we describe a cellular system to study how compartment-specific AR-interacting proteins influence AR activation and contribute to aberrant AR-dependent transcription that underlies the majority of human prostate cancers.


Asunto(s)
Núcleo Celular/metabolismo , Proteína Coat de Complejo I/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Transcripción Genética , Transporte Activo de Núcleo Celular , Línea Celular Tumoral , Núcleo Celular/genética , Proteína Coat de Complejo I/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Aparato de Golgi/genética , Aparato de Golgi/metabolismo , Humanos , Masculino , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/genética , Proteómica , Receptores Androgénicos/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Am J Respir Cell Mol Biol ; 54(4): 469-81, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26334941

RESUMEN

Defects in the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel lead to viscous secretions from submucosal glands that cannot be properly hydrated and cleared by beating cilia in cystic fibrosis (CF) airways. The mechanisms by which CFTR, and the predominant epithelial sodium channel (ENaC), control the hydration and clearance of glandular secretions remain unclear. We used a proteomics approach to characterize the proteins contained in CF and non-CF submucosal gland fluid droplets and found that differentially regulated proteases (cathepsin S and H) and their antiprotease (cystatin C) influenced the equilibration of fluid on the airway surface and tracheal mucociliary clearance (MCC). Contrary to prevailing models of airway hydration and clearance, cystatin C, or raising the airway surface liquid (ASL) pH, inhibited cathepsin-dependent ENaC-mediated fluid absorption and raised the height of ASL, and yet decreased MCC velocity. Importantly, coupling of both CFTR and ENaC activities were required for effective MCC and for effective ASL height equilibration after volume challenge. Cystatin C-inhibitable cathepsins controlled initial phases of ENaC-mediated fluid absorption, whereas CFTR activity was required to prevent ASL dehydration. Interestingly, CF airway epithelia absorbed fluid more slowly owing to reduced cysteine protease activity in the ASL but became abnormally dehydrated with time. Our findings demonstrate that, after volume challenge, pH-dependent protease-mediated coupling of CFTR and ENaC activities are required for rapid fluid equilibration at the airway surface and for effective MCC. These findings provide new insights into how glandular fluid secretions may be equilibrated at the airway surface and how this process may be impaired in CF.


Asunto(s)
Bronquios/fisiopatología , Cistatina C/fisiología , Fibrosis Quística/fisiopatología , Proteoma , Tráquea/fisiopatología , Animales , Bronquios/metabolismo , Hurones , Células HEK293 , Humanos , Tráquea/metabolismo
6.
Glycobiology ; 26(12): 1284-1296, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27496765

RESUMEN

Both LARGE1 (formerly LARGE) and its paralog LARGE2 are bifunctional glycosyltransferases with xylosy- and glucuronyltransferase activities, and are capable of synthesizing polymers composed of a repeating disaccharide [-3Xylα1,3GlcAß1-]. Post-translational modification of the O-mannosyl glycan of α-dystroglycan (α-DG) with the polysaccharide is essential for it to act as a receptor for ligands in the extracellular matrix (ECM), and both LARGE paralogs contribute to the modification in vivo. LARGE1 and LARGE2 have different tissue distribution profiles and enzymatic properties; however, the functional difference of the homologs remains to be determined, and α-DG is the only known substrate for the modification by LARGE1 or LARGE2. Here we show that LARGE2 can modify proteoglycans (PGs) with the laminin-binding glycan. We found that overexpression of LARGE2, but not LARGE1, mediates the functional modification on the surface of DG-/-, Pomt1-/- and Fktn-/- embryonic stem cells. We identified a heparan sulfate-PG glypican-4 as a substrate for the LARGE2-dependent modification by affinity purification and subsequent mass spectrometric analysis. Furthermore, we showed that LARGE2 could modify several additional PGs with the laminin-binding glycan, most likely within the glycosaminoglycan (GAG)-protein linkage region. Our results indicate that LARGE2 can modify PGs with the GAG-like polysaccharide composed of xylose and glucuronic acid to confer laminin binding. Thus, LARGE2 may play a differential role in stabilizing the basement membrane and modifying its functions by augmenting the interactions between laminin globular domain-containing ECM proteins and PGs.


Asunto(s)
Glicosiltransferasas/metabolismo , Laminina/metabolismo , Proteoglicanos/metabolismo , Animales , Sitios de Unión , Células Madre Embrionarias/metabolismo , Glicosilación , Glicosiltransferasas/química , Laminina/química , Ratones , Ratones Noqueados , Proteoglicanos/química
7.
J Biol Chem ; 289(14): 10095-103, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24569989

RESUMEN

The large-conductance Ca(2+)-activated K(+) (BK(Ca)) channel is essential for maintaining the membrane in a hyperpolarized state, thereby regulating neuronal excitability, smooth muscle contraction, and secretion. The BK(Ca) α-subunit has three predicted initiation codons that generate proteins with N-terminal ends starting with the amino acid sequences MANG, MSSN, or MDAL. Because the N-terminal region and first transmembrane domain of the α-subunit are required for modulation by auxiliary ß1-subunits, we examined whether ß1 differentially modulates the N-terminal BK(Ca) α-subunit isoforms. In the absence of ß1, all isoforms had similar single-channel conductances and voltage-dependent activation. However, whereas ß1 did not modulate the voltage-activation curve of MSSN, ß1 induced a significant leftward shift of the voltage activation curves of both the MDAL and MANG isoforms. These shifts, of which the MDAL was larger, occurred at both 10 µM and 100 µM Ca(2+). The ß1-subunit increased the open dwell times of all three isoforms and decreased the closed dwell times of MANG and MDAL but increased the closed dwell times of MSSN. The distinct modulation of voltage activation by the ß1-subunit may be due to the differential effect of ß1 on burst duration and interburst intervals observed among these isoforms. Additionally, we observed that the related ß2-subunit induced comparable leftward shifts in the voltage-activation curves of all three isoforms, indicating that the differential modulation of these isoforms was specific to ß1. These findings suggest that the relative expression of the N-terminal isoforms can fine-tune BK(Ca) channel activity in cells, highlighting a novel mechanism of BK(Ca) channel regulation.


Asunto(s)
Calcio/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Femenino , Células HEK293 , Humanos , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
8.
BMC Cancer ; 15: 204, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25884570

RESUMEN

BACKGROUND: Identifying cellular signaling pathways that become corrupted in the presence of androgens that increase the metastatic potential of organ-confined tumor cells is critical to devising strategies capable of attenuating the metastatic progression of hormone-naïve, organ-confined tumors. In localized prostate cancers, gene fusions that place ETS-family transcription factors under the control of androgens drive gene expression programs that increase the invasiveness of organ-confined tumor cells. C-X-C chemokine receptor type 4 (CXCR4) is a downstream target of ERG, whose upregulation in prostate-tumor cells contributes to their migration from the prostate gland. Recent evidence suggests that CXCR4-mediated proliferation and metastasis of tumor cells is regulated by CXCR7 through its scavenging of chemokine CXCL12. However, the role of androgens in regulating CXCR4-mediated motility with respect to CXCR7 function in prostate-cancer cells remains unclear. METHODS: Immunocytochemistry, western blot, and affinity-purification analyses were used to study how androgens influenced the expression, subcellular localization, and function of CXCR7, CXCR4, and androgen receptor (AR) in LNCaP prostate-tumor cells. Moreover, luciferase assays and quantitative polymerase chain reaction (qPCR) were used to study how chemokines CXCL11 and CXCL12 regulate androgen-regulated genes (ARGs) in LNCaP prostate-tumor cells. Lastly, cell motility assays were carried out to determine how androgens influenced CXCR4-dependent motility through CXCL12. RESULTS: Here we show that, in the LNCaP prostate-tumor cell line, androgens coordinate the expression of CXCR4 and CXCR7, thereby promoting CXCL12/CXCR4-mediated cell motility. RNA interference experiments revealed functional interactions between AR and CXCR7 in these cells. Co-localization and affinity-purification experiments support a physical interaction between AR and CXCR7 in LNCaP cells. Unexpectedly, CXCR7 resided in the nuclear compartment and modulated AR-mediated transcription. Moreover, androgen-mediated cell motility correlated positively with the co-localization of CXCR4 and CXCR7 receptors, suggesting that cell migration may be linked to functional CXCR4/CXCR7 heterodimers. Lastly, CXCL12-mediated cell motility was CXCR7-dependent, with CXCR7 expression required for optimal expression of CXCR4 protein. CONCLUSIONS: Overall, our results suggest that inhibition of CXCR7 function might decrease the metastatic potential of organ-confined prostate cancers.


Asunto(s)
Movimiento Celular/genética , Quimiocina CXCL12/biosíntesis , Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Receptores CXCR4/biosíntesis , Receptores CXCR/biosíntesis , Andrógenos/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Quimiocina CXCL11/biosíntesis , Quimiocina CXCL11/genética , Quimiocina CXCL12/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Interferencia de ARN , Receptores Androgénicos/metabolismo , Receptores CXCR/genética , Receptores CXCR4/genética , Transducción de Señal/genética
9.
Appl Opt ; 54(9): 2413-23, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25968530

RESUMEN

Liquid scintillation counters measure charged particle-emitting radioactive isotopes and are used for environmental studies, nuclear chemistry, and life science. Alpha and beta emissions arising from the material under study interact with the scintillation cocktail to produce light. The prototypical liquid scintillation counter employs low-level photon-counting detectors to measure the arrival of the scintillation. For reliable operation, the counting instrument must convey the scintillation light to the detectors efficiently and predictably. Current best practices employ the use of two or more detectors for coincidence processing to discriminate true scintillation events from background events due to instrumental effects such as photomultiplier tube dark rates, tube flashing, or other light emission not generated in the scintillation cocktail vial. In low-background liquid scintillation counters, additional attention is paid to shielding the scintillation cocktail from naturally occurring radioactive material present in the laboratory and within the instrument's construction materials. Low-background design is generally at odds with optimal light collection. This study presents the evolution of a light collection design for liquid scintillation counting (LSC) in a low-background shield. The basic approach to achieve both good light collection and a low-background measurement is described. The baseline signals arising from the scintillation vial are modeled and methods to efficiently collect scintillation light are presented as part of the development of a customized low-background, high-sensitivity LSC system.

10.
Proc Natl Acad Sci U S A ; 107(21): 9671-6, 2010 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-20457893

RESUMEN

Localized chromatin modifications of histone tails play an important role in regulating gene transcription, and aberration of these processes leads to carcinogenesis. Methylated histone lysine residues, a key player in chromatin remodeling, are demethylated by the JmjC class of enzymes. Here we show that JMJD5 (now renamed KDM8), a JmjC family member, demethylates H3K36me2 and is required for cell cycle progression. Chromatin immunoprecipitation assays applied to human genome tiling arrays in conjunction with RNA microarray revealed that KDM8 occupies the coding region of cyclin A1 and directly regulates transcription. Mechanistic analyses showed that KDM8 functioned as a transcriptional activator by inhibiting HDAC recruitment via demethylation of H3K36me2, an epigenetic repressive mark. Tumor array experiments revealed KDM8 is overexpressed in several types of cancer. In addition, loss-of-function studies in MCF7 cells leads to cell cycle arrest. These studies identified KDM8 as an important cell cycle regulator.


Asunto(s)
Proliferación Celular , Ciclina A1/metabolismo , Histona Demetilasas/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Sistemas de Lectura Abierta , Acetilación , Línea Celular Tumoral , Ciclina A1/genética , Regulación Neoplásica de la Expresión Génica , Histona Demetilasas/genética , Histonas/metabolismo , Humanos , Neoplasias/genética , Interferencia de ARN , Transcripción Genética
11.
Retina ; 32(10): 2141-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23095728

RESUMEN

PURPOSE: To compare vitreous biopsy methods using analysis platforms used in proteomics biomarker discovery. METHODS: Vitreous biopsies from 10 eyes were collected sequentially using a 23-gauge needle and a 23-gauge vitreous cutter instrument. Paired specimens were evaluated by UV absorbance spectroscopy, sodium dodecyl sulfate-polyacrylamide gel electrophoresis, and liquid chromatography tandem mass spectrometry (LC-MS/MS). RESULTS: The total protein concentration obtained with a needle and vitrectomy instrument biopsy averaged 1.10 mg/mL (standard error of the mean = 0.35) and 1.13 mg/mL (standard error of the mean = 0.25), respectively. In eight eyes with low or medium viscidity, there was a very high correlation (R = 0.934) between the biopsy methods. When data from 2 eyes with high viscidity vitreous were included, the correlation was reduced (R = 0.704). The molecular weight protein sodium dodecyl sulfate-polyacrylamide gel electrophoresis profiles of paired needle and vitreous cutter samples were similar, except for a minority of pairs with single band intensity variance. Using LC-MS/MS, equivalent peptides were identified with similar frequencies (R ≥ 0.90) in paired samples. CONCLUSION: Proteins and peptides collected from vitreous needle biopsies are nearly equivalent to those obtained from a vitreous cutter instrument. This study suggests both techniques may be used for most proteomic and biomarker discovery studies of vitreoretinal diseases, although a minority of proteins and peptides may differ in concentration.


Asunto(s)
Biomarcadores/análisis , Biopsia/métodos , Proteínas del Ojo/análisis , Cuerpo Vítreo/química , Adolescente , Anciano , Biopsia/instrumentación , Cromatografía Liquida , Electroforesis en Gel de Poliacrilamida , Oftalmopatías/diagnóstico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteómica , Espectrofotometría Ultravioleta , Espectrometría de Masas en Tándem , Vitrectomía/instrumentación , Adulto Joven
12.
Sci Rep ; 11(1): 22208, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34782677

RESUMEN

Supraphysiologic androgen (SPA) inhibits cell proliferation in prostate cancer (PCa) cells by transcriptional repression of DNA replication and cell-cycle genes. In this study, quantitative glycoprotein profiling identified androgen-regulated glycoprotein networks associated with SPA-mediated inhibition of PCa cell proliferation, and androgen-regulated glycoproteins in clinical prostate tissues. SPA-regulated glycoprotein networks were enriched for translation factors and ribosomal proteins, proteins that are known to be O-GlcNAcylated in response to various cellular stresses. Thus, androgen-regulated glycoproteins are likely to be targeted for O-GlcNAcylation. Comparative analysis of glycosylated proteins in PCa cells and clinical prostate tissue identified androgen-regulated glycoproteins that are differentially expressed prostate tissues at various stages of cancer. Notably, the enzyme ectonucleoside triphosphate diphosphohydrolase 5 was found to be an androgen-regulated glycoprotein in PCa cells, with higher expression in cancerous versus non-cancerous prostate tissue. Our glycoproteomics study provides an experimental framework for characterizing androgen-regulated proteins and glycoprotein networks, toward better understanding how this subproteome leads to physiologic and supraphysiologic proliferation responses in PCa cells, and their potential use as druggable biomarkers of dysregulated AR-dependent signaling in PCa cells.


Asunto(s)
Andrógenos/metabolismo , Glicoproteínas/metabolismo , Enfermedades de la Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Proteoma , Proteómica , Biomarcadores , Línea Celular Tumoral , Biología Computacional/métodos , Regulación Neoplásica de la Expresión Génica , Glicoproteínas/genética , Humanos , Masculino , Espectrometría de Masas , Enfermedades de la Próstata/etiología , Neoplasias de la Próstata/etiología , Proteómica/métodos , Transducción de Señal
13.
Pharmacy (Basel) ; 9(4)2021 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-34941623

RESUMEN

PURPOSE: Staphylococcus aureus is a leading cause of bacteremia with a 30-day mortality of 20%. This study evaluated outcomes after implementation of a pharmacist-driven Staphylococcus aureus bacteremia (SAB) initiative in a community hospital. METHODS: This retrospective cohort analysis compared patients admitted with SAB between May 2015 and April 2018 (intervention group) to those admitted between May 2012 and April 2015 (historical control group). Pharmacists were notified of and responded to blood cultures positive for Staphylococcus aureus by contacting provider(s) with a bundle of recommendations. Components of the SAB bundle included prompt source control, selection of appropriate intravenous antibiotics, appropriate duration of therapy, repeat blood cultures, echocardiography, and infectious diseases consult. Demographics (age, gender, and race) were collected at baseline. Primary outcome was in-hospital mortality. Compliance with bundle components was also assessed. RESULTS: Eighty-three patients in the control group and 110 patients in the intervention group were included in this study. Demographics were similar at baseline. In-hospital mortality was lower in the intervention group (3.6% vs. 15.7%; p = 0.0033). Bundle compliance was greater in the intervention group (69.1% vs. 39.8%; p < 0.0001). CONCLUSIONS: We observed a significant reduction in in-hospital mortality and increased treatment bundle compliance in the intervention cohort with implementation of a pharmacist-driven SAB initiative. Pharmacists' participation in the care of SAB patients in the form of recommending adherence to treatment bundle components drastically improved clinical outcomes. Widespread adoption and implementation of similar practice models at other institutions may reduce in-hospital mortality for this relatively common and life-threatening infection.

14.
Cancer Res ; 64(1): 347-55, 2004 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-14729644

RESUMEN

Prostate cancer is unusual among neoplasms in that it may be diagnosed at a curable stage through detection of a protein in serum, the serine protease prostate-specific antigen (PSA). PSA is secreted by both normal and neoplastic prostate epithelial cells in response to androgenic hormones and has found widespread use in cancer screening. Because PSA screening is controversial due to sensitivity and specificity issues, efforts continue to focus on the identification and characterization of additional markers that may be used for diagnostic and therapeutic purposes. In this study, we report the application of quantitative proteomic techniques that incorporate isotope coded affinity tag reagents and tandem mass spectrometry to comprehensively identify secreted and cell surface proteins from neoplastic prostate epithelium. LNCaP cells, a prostate tumor-derived cell line that secretes PSA in response to androgen exposure, were grown in a low protein-defined media under androgen-stimulated (A+) and -starved (A-) conditions. Proteomic analysis of the media identified in excess of 600 proteins, 524 of which could be quantified. Nine percent of the proteins had A+/A- ratios > 2.0, including PSA, and 2.5% had ratios < 0.5. A subset of these androgen-regulated proteins appeared to be expressed in abundance. Of these, selected mass spectrometry observations were confirmed by Western analysis. The findings suggest that androgen-mediated release of proteins may occur through the activation of proteolytic enzymes rather than exclusively through transcriptional or translational control mechanisms. On the basis of their known functional roles, several of the abundant androgen-regulated proteins may participate in the progression of neoplastic epithelial cell growth and should be considered as potential serum markers of neoplastic prostate diseases.


Asunto(s)
Proteínas de Neoplasias/análisis , Neoplasias de la Próstata/química , Proteoma/química , Biomarcadores de Tumor/análisis , Línea Celular Tumoral , Medios de Cultivo Condicionados , Enzimas/análisis , Células Epiteliales/química , Humanos , Masculino , Orgánulos/química , Antígeno Prostático Específico/análisis , Fracciones Subcelulares/química
15.
J Phys Chem B ; 109(46): 21496-8, 2005 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-16853790

RESUMEN

A series of oligomers consisting of ethynyl-linked azobenzene units was prepared using Pd-catalyzed cross coupling. The linear and nonlinear optical properties of the oligomers were investigated. The molecular second hyperpolarizability, gamma, followed the power law gamma proportional, variant n(2.12+/-0.05) (n is the number of repeat units) for unusually large molecular lengths exceeding 360 conjugated bonds (>49 nm). The exceptional exciton delocalization length is attributed to the rigidity of the conjugated backbone.

16.
Mol Endocrinol ; 17(9): 1726-37, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12775765

RESUMEN

Androgen-ablation therapy is an effective method for treating prostate cancer. However, prostate tumors that survive long-term androgen-ablation therapy are classified as androgen-independent as they proliferate in the absence of androgens, and they tend to be enriched for neuroendocrine (NE) cells. Androgen withdrawal causes androgen-dependent prostate cancer cells to adopt a pronounced NE phenotype, suggesting that androgen receptor (AR) represses an intrinsic NE transdifferentiation process in prostate cancer cells. In this report we show that short interfering RNA-induced AR silencing induced a NE phenotype that manifested itself in the growth of dendritic-like processes in both the androgen-dependent LNCaP and androgen-independent LNCaP-AI human prostate cancer cells. Western blot analysis revealed that neuronal-specific enolase, a marker of the neuronal lineage, was increased by AR knockdown in LNCaP cells. The expression levels of the neuronal-specific cytoskeletal proteins beta-tubulin III, nestin, and glial acidic fibrillary protein were also characterized in AR knockdown cells. Most interestingly, AR silencing induced beta-tubulin III expression in LNCaP cells, while AR knockdown increased glial acidic fibrillary protein levels in both LNCaP and LNCaP-AI cells. Lastly, AR silencing reduced the proliferative capacity of LNCaP and LNCaP-AI cells. Our data demonstrate that AR actively represses an intrinsic NE transdifferentiation process in androgen-responsive prostate cancer cells and suggest a potential link between AR inactivation and the increased frequency of NE cells in androgen-independent tumors.


Asunto(s)
Diferenciación Celular/fisiología , Sistemas Neurosecretores/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Biomarcadores , Carcinoma/metabolismo , Silenciador del Gen/fisiología , Humanos , Masculino , Fenotipo , Interferencia de ARN/fisiología , ARN Interferente Pequeño , Receptores Androgénicos/genética , Células Tumorales Cultivadas
17.
Perm J ; 19(1): 34-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25432002

RESUMEN

CONTEXT: The role at admission of nasal polymerase chain reaction (PCR) for patients with methicillin-resistant Staphylococcus aureus (MRSA) in guiding antibiotic therapy for lower respiratory tract infection is unknown. OBJECTIVE: To determine whether nasal MRSA PCR at admission can predict the absence of MRSA in lower respiratory tract secretions. DESIGN: We performed a retrospective study of adult patients admitted to a large urban hospital. Patients had a nasal MRSA PCR test and a lower respiratory tract culture obtained within 48 hours of admission and the culture yielded S aureus. MAIN OUTCOME MEASURES: Sensitivity, specificity, and positive and negative predictive values. RESULTS: Our results showed high sensitivity (93.3%) and negative predictive value (95.2%) of nasal PCR for MRSA in the lower respiratory tract. CONCLUSION: With its high sensitivity and negative predictive value, a nasal MRSA PCR test performed within 48 hours of hospital admission could help guide the discontinuation of MRSA-directed empiric antibiotic therapy in patients who are unlikely to be infected with this organism. A prospective study is needed to confirm these findings.


Asunto(s)
Portador Sano/diagnóstico , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Cavidad Nasal/microbiología , Neumonía Estafilocócica/diagnóstico , Reacción en Cadena de la Polimerasa/estadística & datos numéricos , Adulto , Anciano , Antibacterianos/uso terapéutico , Portador Sano/tratamiento farmacológico , Portador Sano/microbiología , Femenino , Hospitales Urbanos/estadística & datos numéricos , Humanos , Masculino , Persona de Mediana Edad , Neumonía Estafilocócica/tratamiento farmacológico , Neumonía Estafilocócica/microbiología , Valor Predictivo de las Pruebas , Estudios Prospectivos , Estudios Retrospectivos , Sensibilidad y Especificidad
18.
Mol Endocrinol ; 29(8): 1195-218, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26181434

RESUMEN

The aberrant expression of androgen receptor (AR)-dependent transcriptional programs is a defining pathology of the development and progression of prostate cancers. Transcriptional cofactors that bind AR are critical determinants of prostate tumorigenesis. To gain a deeper understanding of the proteins linked to AR-dependent gene transcription, we performed a DNA-affinity chromatography-based proteomic screen designed to identify proteins involved in AR-mediated gene transcription in prostate tumor cells. Functional experiments validated the coregulator roles of known AR-binding proteins in AR-mediated transcription in prostate tumor cells. More importantly, novel coregulatory functions were detected in components of well-established cell surface receptor-dependent signal transduction pathways. Further experimentation demonstrated that components of the TNF, TGF-ß, IL receptor, and epidermal growth factor signaling pathways modulated AR-dependent gene transcription and androgen-dependent proliferation in prostate tumor cells. Collectively, our proteomic dataset demonstrates that the cell surface receptor- and AR-dependent pathways are highly integrated, and provides a molecular framework for understanding how disparate signal-transduction pathways can influence AR-dependent transcriptional programs linked to the development and progression of human prostate cancers.


Asunto(s)
Membrana Celular/metabolismo , Receptores Androgénicos/metabolismo , Receptores de Superficie Celular/metabolismo , Andrógenos/química , Sitios de Unión , Línea Celular Tumoral , Biología Computacional , Citocinas/metabolismo , ADN/química , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Espectrometría de Masas , Mutación , Neoplasias de la Próstata/metabolismo , Proteómica , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Factores de Transcripción/metabolismo , Transcripción Genética
19.
ACS Appl Mater Interfaces ; 4(2): 492-502, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22188314

RESUMEN

Kapton polyimde is extensively used in solar arrays, spacecraft thermal blankets, and space inflatable structures. Upon exposure to atomic oxygen in low Earth orbit (LEO), Kapton is severely eroded. An effective approach to prevent this erosion is to incorporate polyhedral oligomeric silsesquioxane (POSS) into the polyimide matrix by copolymerizing POSS monomers with the polyimide precursor. The copolymerization of POSS provides Si and O in the polymer matrix on the nano level. During exposure of POSS polyimide to atomic oxygen, organic material is degraded, and a silica passivation layer is formed. This silica layer protects the underlying polymer from further degradation. Laboratory and space-flight experiments have shown that POSS polyimides are highly resistant to atomic-oxygen attack, with erosion yields that may be as little as 1% those of Kapton. The results of all the studies indicate that POSS polyimide would be a space-survivable replacement for Kapton on spacecraft that operate in the LEO environment.

20.
Genes Cancer ; 2(10): 956-78, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22701762

RESUMEN

Increasing evidence suggests that the disruption of androgen-mediated cellular processes, such as cell proliferation and cell differentiation, contributes to the development of early-stage androgen-dependent prostate cancers. Large-scale mRNA profiling experiments have paved the way in identifying androgen-regulated gene networks that control the proliferation, survival, and differentiation of prostate cancer cells. Despite these extensive research efforts, it remains to be determined whether all androgen-mediated mRNA changes faithfully translate into changes in protein abundance that influence prostate tumorigenesis. Here, we report on a mass spectrometry-based quantitative proteomics analysis that identified known androgen signaling pathways and also novel, androgen-sensitive microsome-associated proteins and protein networks that had not been discovered by gene network studies in human LNCaP prostate cancer cells. Androgen-sensitive microsome-associated proteins encoded components of the insulin growth factor-1 (IGF-1), phosphoinositide 3-kinase (PI3K)/AKT, and extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) signaling pathways. Further bioinformatic analyses showed most of the androgen-sensitive microsome-associated protein networks play roles in cell proliferation and differentiation. Functional validation experiments showed that the androgen-sensitive microsome-associated proteins Janus kinase 2 (JAK2) and I-kappa B kinase complex-associated protein (IKAP) modulated the expression of prostate epithelial and neuronal markers, attenuated proliferation through an androgen receptor-dependent mechanism, and co-regulated androgen receptor-mediated transcription in LNCaP cells. Further biochemical analyses showed that the increased proliferation in JAK2 knockdown cells was mediated by activation of the mammalian target of rapamycin (mTOR), as determined by increased phosphorylation of several downstream targets (p70 S6 kinase, translational repressor 4E-BP1, and 40S ribosomal S6 protein). We conclude that the expression of microsome-associated proteins that were previously implicated in the tumorigenesis of prostate epithelial cells is strongly influenced by androgens. These findings provide a molecular framework for exploring the mechanisms underlying prostate tumorigenesis and how these protein networks might be attenuated or potentiated in disrupting the growth and survival of human prostate cancers.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA