Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Tissue Res ; 392(1): 367-392, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36764940

RESUMEN

Prion diseases are fatal infectious neurodegenerative disorders and prototypic conformational diseases, caused by the conformational conversion of the normal cellular prion protein (PrPC) into the pathological PrPSc isoform. Examples are scrapie in sheep and goat, bovine spongiform encephalopathy (BSE) in cattle, chronic wasting disease (CWD) in cervids, and Creutzfeldt-Jacob disease (CJD) in humans. There are no therapies available, and animal prion diseases like BSE and CWD can negatively affect the economy, ecology, animal health, and possibly human health. BSE is a confirmed threat to human health, and mounting evidence supports the zoonotic potential of CWD. CWD is continuously expanding in North America in numbers and distribution and was recently identified in Scandinavian countries. CWD is the only prion disease occurring both in wild and farmed animals, which, together with extensive shedding of infectivity into the environment, impedes containment strategies. There is currently a strong push to develop vaccines against CWD, including ones that can be used in wildlife. The immune system does not develop a bona fide immune response against prion infection, as PrPC and PrPSc share an identical protein primary structure, and prions seem not to represent a trigger for immune responses. This asks for alternative vaccine strategies, which focus on PrPC-directed self-antibodies or exposure of disease-specific structures and epitopes. Several groups have established a proof-of-concept that such vaccine candidates can induce some levels of protective immunity in cervid and rodent models without inducing unwanted side effects. This review will highlight the most recent developments and discuss progress and challenges remaining.


Asunto(s)
Ciervos , Encefalopatía Espongiforme Bovina , Enfermedades por Prión , Priones , Vacunas , Enfermedad Debilitante Crónica , Animales , Bovinos , Humanos , Ovinos , Objetivos , Enfermedades por Prión/prevención & control , Enfermedades por Prión/metabolismo , Priones/metabolismo , Encefalopatía Espongiforme Bovina/metabolismo , Enfermedad Debilitante Crónica/prevención & control , Enfermedad Debilitante Crónica/metabolismo , Ciervos/metabolismo , Cabras
2.
Invest New Drugs ; 38(4): 909-921, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31375978

RESUMEN

Treatment response rates to current anticancer therapies for HER2 overexpressing breast cancer are limited and are associated with severe adverse drug reactions. Tyrosine kinases perform crucial roles in cellular processes by mediating cell signalling cascades. Ibrutinib is a recently approved Tyrosine Kinase Inhibitor (TKI) that has been shown be an effective therapeutic option for HER2 overexpressing breast cancer. The molecular mechanisms, pathways, or genes that are modulated by ibrutinib and the mechanism of action of ibrutinib in HER2 overexpressing breast cancer remain obscure. In this study, we have performed a kinome array analysis of ibrutinib treatment in two HER2 overexpressing breast cancer cell lines. Our analysis shows that ibrutinib induces changes in nuclear morphology and causes apoptosis via caspase-dependent extrinsic apoptosis pathway with the activation of caspases-8, caspase-3, and cleavage of PARP1. We further show that phosphorylated STAT3Y705 is upregulated and phosphorylated p21T145 is downregulated upon ibrutinib treatment. We propose that STAT3 upregulation is a passive response as a result of induction of DNA damage and downregulation of phosphorylated p21 is promoting cell cycle arrest and apoptosis in the two HER2 overexpressing cell lines. These results suggest that inhibitors of STAT3 phosphorylation may be potential options for combination therapy to help increase the efficacy of ibrutinib against HER2-overexpressing tumors.


Asunto(s)
Adenina/análogos & derivados , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Piperidinas/farmacología , Receptor ErbB-2 , Factor de Transcripción STAT3/metabolismo , Adenina/farmacología , Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Agammaglobulinemia Tirosina Quinasa/genética , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos
3.
Mol Cell Proteomics ; 17(5): 925-947, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29496907

RESUMEN

SRMS (Src-related kinase lacking C-terminal regulatory tyrosine and N-terminal myristoylation sites), also known as PTK 70 (Protein tyrosine kinase 70), is a non-receptor tyrosine kinase that belongs to the BRK family of kinases (BFKs). To date less is known about the cellular role of SRMS primarily because of the unidentified substrates or signaling intermediates regulated by the kinase. In this study, we used phosphotyrosine antibody-based immunoaffinity purification in large-scale label-free quantitative phosphoproteomics to identify novel candidate substrates of SRMS. Our analyses led to the identification of 1258 tyrosine-phosphorylated peptides which mapped to 663 phosphoproteins, exclusively from SRMS-expressing cells. DOK1, a previously characterized SRMS substrate, was also identified in our analyses. Functional enrichment analyses revealed that the candidate SRMS substrates were enriched in various biological processes including protein ubiquitination, mitotic cell cycle, energy metabolism and RNA processing, as well as Wnt and TNF signaling. Analyses of the sequence surrounding the phospho-sites in these proteins revealed novel candidate SRMS consensus substrate motifs. We utilized customized high-throughput peptide arrays to validate a subset of the candidate SRMS substrates identified in our MS-based analyses. Finally, we independently validated Vimentin and Sam68, as bona fide SRMS substrates through in vitro and in vivo assays. Overall, our study identified a number of novel and biologically relevant SRMS candidate substrates, which suggests the involvement of the kinase in a vast array of unexplored cellular functions.


Asunto(s)
Fosfoproteínas/metabolismo , Proteómica/métodos , Familia-src Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Línea Celular , Cromatografía de Afinidad , Simulación por Computador , Secuencia de Consenso , Proteínas de Unión al ADN/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Humanos , Espectrometría de Masas , Fosfopéptidos/química , Fosfopéptidos/metabolismo , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Análisis por Matrices de Proteínas , Proteoma/metabolismo , Proteínas de Unión al ARN/metabolismo , Reproducibilidad de los Resultados , Especificidad por Sustrato/efectos de los fármacos , Vimentina/metabolismo , Familia-src Quinasas/química
4.
Breast Cancer Res ; 19(1): 65, 2017 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-28583138

RESUMEN

BACKGROUND: Breast cancer cell lines are frequently used as model systems to study the cellular properties and biology of breast cancer. Our objective was to characterize a large, commonly employed panel of breast cancer cell lines obtained from the American Type Culture Collection (ATCC 30-4500 K) to enable researchers to make more informed decisions in selecting cell lines for specific studies. Information about these cell lines was obtained from a wide variety of sources. In addition, new information about cellular pathways that are activated within each cell line was generated. METHODS: We determined key protein expression data using immunoblot analyses. In addition, two analyses on serum-starved cells were carried out to identify cellular proteins and pathways that are activated in these cells. These analyses were performed using a commercial PathScan array and a novel and more extensive phosphopeptide-based kinome analysis that queries 1290 phosphorylation events in major signaling pathways. Data about this panel of breast cancer cell lines was also accessed from several online sources, compiled and summarized for the following areas: molecular classification, mRNA expression, mutational status of key proteins and other possible cancer-associated mutations, and the tumorigenic and metastatic capacity in mouse xenograft models of breast cancer. RESULTS: The cell lines that were characterized included 10 estrogen receptor (ER)-positive, 12 human epidermal growth factor receptor 2 (HER2)-amplified and 18 triple negative breast cancer cell lines, in addition to 4 non-tumorigenic breast cell lines. Within each subtype, there was significant genetic heterogeneity that could impact both the selection of model cell lines and the interpretation of the results obtained. To capture the net activation of key signaling pathways as a result of these mutational combinations, profiled pathway activation status was examined. This provided further clarity for which cell lines were particularly deregulated in common or unique ways. CONCLUSIONS: These two new kinase or "Kin-OMIC" analyses add another dimension of important data about these frequently used breast cancer cell lines. This will assist researchers in selecting the most appropriate cell lines to use for breast cancer studies and provide context for the interpretation of the emerging results.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Genómica , Proteómica , Animales , Biomarcadores , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Biología Computacional/métodos , Análisis Mutacional de ADN , Bases de Datos Genéticas , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genómica/métodos , Xenoinjertos , Humanos , Ratones , Proteoma , Proteómica/métodos , Transducción de Señal
5.
Brief Bioinform ; 16(5): 820-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25380664

RESUMEN

The majority of scientific resources are devoted to studying a relatively small number of model species, meaning that the ability to translate knowledge across species is of considerable importance. Obtaining species-specific knowledge enables targeted investigations of the biology and pathobiology of a particular species, and facilitates comparative analyses. Phosphorylation is the most widespread posttranslational modification in eukaryotes, and although many phosphorylation sites have been experimentally identified for some species, little or no data are available for others. Using the honeybee as a test organism, this case study illustrates the process of using protein sequence homology to identify putative phosphorylation sites in a species of interest using experimentally determined sites from other species. A number of issues associated with this process are examined and discussed. Several databases of experimentally determined phosphorylation sites exist; however, it can be difficult for the nonspecialist to ascertain how their contents compare. Thus, this case study assesses the content and comparability of several phosphorylation site databases. Additional issues examined include the efficacy of homology-based phosphorylation site prediction, the impact of the level of evolutionary relatedness between species in making these predictions, the ability to translate knowledge of phosphorylation sites across large evolutionary distances and the criteria that should be used in selecting probable phosphorylation sites in the species of interest. Although focusing on phosphorylation, the issues discussed here also apply to the homology-based cross-species prediction of other posttranslational modifications, as well as to sequence motifs in general.


Asunto(s)
Abejas/metabolismo , Evolución Biológica , Proteínas de Insectos/metabolismo , Animales , Humanos , Proteínas de Insectos/química , Funciones de Verosimilitud , Fosforilación , Homología de Secuencia de Aminoácido , Especificidad de la Especie
6.
Biochem Soc Trans ; 45(1): 65-77, 2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28202660

RESUMEN

There is increasing appreciation among researchers and clinicians of the value of investigating biology and pathobiology at the level of cellular kinase (kinome) activity. Kinome analysis provides valuable opportunity to gain insights into complex biology (including disease pathology), identify biomarkers of critical phenotypes (including disease prognosis and evaluation of therapeutic efficacy), and identify targets for therapeutic intervention through kinase inhibitors. The growing interest in kinome analysis has fueled efforts to develop and optimize technologies that enable characterization of phosphorylation-mediated signaling events in a cost-effective, high-throughput manner. In this review, we highlight recent advances to the central technologies currently available for kinome profiling and offer our perspectives on the key challenges remaining to be addressed.


Asunto(s)
Análisis por Matrices de Proteínas/métodos , Proteínas Quinasas/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Animales , Humanos , Fosforilación , Reproducibilidad de los Resultados , Transducción de Señal
7.
Mol Cell Proteomics ; 14(3): 646-57, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25573744

RESUMEN

The Syrian golden hamster has been increasingly used to study viral hemorrhagic fever (VHF) pathogenesis and countermeasure efficacy. As VHFs are a global health concern, well-characterized animal models are essential for both the development of therapeutics and vaccines as well as for increasing our understanding of the molecular events that underlie viral pathogenesis. However, the paucity of reagents or platforms that are available for studying hamsters at a molecular level limits the ability to extract biological information from this important animal model. As such, there is a need to develop platforms/technologies for characterizing host responses of hamsters at a molecular level. To this end, we developed hamster-specific kinome peptide arrays to characterize the molecular host response of the Syrian golden hamster. After validating the functionality of the arrays using immune agonists of defined signaling mechanisms (lipopolysaccharide (LPS) and tumor necrosis factor (TNF)-α), we characterized the host response in a hamster model of VHF based on Pichinde virus (PICV(1)) infection by performing temporal kinome analysis of lung tissue. Our analysis revealed key roles for vascular endothelial growth factor (VEGF), interleukin (IL) responses, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, and Toll-like receptor (TLR) signaling in the response to PICV infection. These findings were validated through phosphorylation-specific Western blot analysis. Overall, we have demonstrated that hamster-specific kinome arrays are a robust tool for characterizing the species-specific molecular host response in a VHF model. Further, our results provide key insights into the hamster host response to PICV infection and will inform future studies with high-consequence VHF pathogens.


Asunto(s)
Fiebre Hemorrágica Americana/virología , Pulmón/enzimología , Virus Pichinde/fisiología , Proteínas Quinasas/aislamiento & purificación , Proteoma/análisis , Animales , Modelos Animales de Enfermedad , Femenino , Fiebre Hemorrágica Americana/enzimología , Interleucinas/aislamiento & purificación , Pulmón/virología , Mesocricetus , FN-kappa B/aislamiento & purificación , Fosforilación , Transducción de Señal , Especificidad de la Especie , Receptores Toll-Like/aislamiento & purificación , Factor A de Crecimiento Endotelial Vascular/aislamiento & purificación
8.
J Proteome Res ; 15(8): 2760-7, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27367363

RESUMEN

The post-translational modification of proteins is critical for regulating their function. Although many post-translational modification sites have been experimentally determined, particularly in certain model organisms, experimental knowledge of these sites is severely lacking for many species. Thus, it is important to be able to predict sites of post-translational modification in such species. Previously, we described DAPPLE, a tool that facilitates the homology-based prediction of one particular post-translational modification, phosphorylation, in an organism of interest using known phosphorylation sites from other organisms. Here, we describe DAPPLE 2, which expands and improves upon DAPPLE in three major ways. First, it predicts sites for many post-translational modifications (20 different types) using data from several sources (15 online databases). Second, it has the ability to make predictions approximately 2-7 times faster than DAPPLE depending on the database size and the organism of interest. Third, it simplifies and accelerates the process of selecting predicted sites of interest by categorizing them based on gene ontology terms, keywords, and signaling pathways. We show that DAPPLE 2 can successfully predict known human post-translational modification sites using, as input, known sites from species that are either closely (e.g., mouse) or distantly (e.g., yeast) related to humans. DAPPLE 2 can be accessed at http://saphire.usask.ca/saphire/dapple2 .


Asunto(s)
Procesamiento Proteico-Postraduccional , Homología de Secuencia de Aminoácido , Programas Informáticos , Animales , Sitios de Unión , Bases de Datos de Proteínas , Humanos , Fosforilación
9.
Bioinformatics ; 31(14): 2388-90, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25750418

RESUMEN

UNLABELLED: Efforts to develop peptide-based vaccines, in particular those requiring site-specific targeting of self-proteins, rely on the ability to optimize the immunogenicity of the peptide epitopes. Currently, screening of candidate vaccines is typically performed through low-throughput, high-cost animal trials. To improve on this we present the program EpIC, which enables high-throughput prediction of peptide immunogenicity based on the endogenous occurrence of B-cell epitopes within native protein sequences. This information informs rational selection of immunogenicity-optimized epitopes for peptide vaccines. AVAILABILITY AND IMPLEMENTATION: EpIC is available as a web server at http://saphire.usask.ca/saphire/epic.


Asunto(s)
Epítopos de Linfocito B/inmunología , Programas Informáticos , Epítopos de Linfocito B/química , Péptidos/química , Péptidos/inmunología , Análisis de Secuencia de Proteína , Vacunas de Subunidad/inmunología
10.
J Virol ; 88(17): 9877-92, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24942569

RESUMEN

UNLABELLED: Ebola virus (EBOV) causes a severe hemorrhagic disease in humans and nonhuman primates, with a median case fatality rate of 78.4%. Although EBOV is considered a public health concern, there is a relative paucity of information regarding the modulation of the functional host response during infection. We employed temporal kinome analysis to investigate the relative early, intermediate, and late host kinome responses to EBOV infection in human hepatocytes. Pathway overrepresentation analysis and functional network analysis of kinome data revealed that transforming growth factor (TGF-ß)-mediated signaling responses were temporally modulated in response to EBOV infection. Upregulation of TGF-ß signaling in the kinome data sets correlated with the upregulation of TGF-ß secretion from EBOV-infected cells. Kinase inhibitors targeting TGF-ß signaling, or additional cell receptors and downstream signaling pathway intermediates identified from our kinome analysis, also inhibited EBOV replication. Further, the inhibition of select cell signaling intermediates identified from our kinome analysis provided partial protection in a lethal model of EBOV infection. To gain perspective on the cellular consequence of TGF-ß signaling modulation during EBOV infection, we assessed cellular markers associated with upregulation of TGF-ß signaling. We observed upregulation of matrix metalloproteinase 9, N-cadherin, and fibronectin expression with concomitant reductions in the expression of E-cadherin and claudin-1, responses that are standard characteristics of an epithelium-to-mesenchyme-like transition. Additionally, we identified phosphorylation events downstream of TGF-ß that may contribute to this process. From these observations, we propose a model for a broader role of TGF-ß-mediated signaling responses in the pathogenesis of Ebola virus disease. IMPORTANCE: Ebola virus (EBOV), formerly Zaire ebolavirus, causes a severe hemorrhagic disease in humans and nonhuman primates and is the most lethal Ebola virus species, with case fatality rates of up to 90%. Although EBOV is considered a worldwide concern, many questions remain regarding EBOV molecular pathogenesis. As it is appreciated that many cellular processes are regulated through kinase-mediated phosphorylation events, we employed temporal kinome analysis to investigate the functional responses of human hepatocytes to EBOV infection. Administration of kinase inhibitors targeting signaling pathway intermediates identified in our kinome analysis inhibited viral replication in vitro and reduced EBOV pathogenesis in vivo. Further analysis of our data also demonstrated that EBOV infection modulated TGF-ß-mediated signaling responses and promoted "mesenchyme-like" phenotypic changes. Taken together, these results demonstrated that EBOV infection specifically modulates TGF-ß-mediated signaling responses in epithelial cells and may have broader implications in EBOV pathogenesis.


Asunto(s)
Diferenciación Celular , Ebolavirus/fisiología , Hepatocitos/fisiología , Interacciones Huésped-Patógeno , Mesodermo/crecimiento & desarrollo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Fiebre Hemorrágica Ebola/patología , Humanos , Ratones Endogámicos BALB C
11.
Poult Sci ; 94(6): 1333-45, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25838314

RESUMEN

Defining cellular responses at the level of global cellular kinase (kinome) activity is a powerful approach to deciphering complex biology and identifying biomarkers. Here we report on the development of a chicken-specific peptide array and its application to characterizing kinome responses within the breast (pectoralis major) and thigh (iliotibialis) muscles of poultry subject to temperature stress to mimic conditions experienced by birds during commercial transport. Breast and thigh muscles exhibited unique kinome profiles, highlighting the distinct nature of these tissues. Against these distinct backgrounds, tissue- and temperature-specific kinome responses were observed. In breast, both cold and hot stresses activated calcium-dependent metabolic adaptations. Also within breast, but specific to cold stress, was the activation of ErbB signaling as well as dynamic patterns of phosphorylation of AMPK, a key regulatory enzyme of metabolism. In thigh, cold stress induced responses suggestive of the occurrence of tissue damage, including activation of innate immune signaling pathways and tissue repair pathways (TGF-ß). In contrast, heat stress in thigh activated pathways associated with protein and fat metabolism through adipocytokine and mammalian target of rapamycin (mTOR) signaling. Defining the responses of these tissues to these stresses through conventional markers of pH, glycolytic potential, and meat quality offered a similar conclusion of the tissue- and stressor-specific responses, validating the kinome results. Collectively, the results of this study highlight the unique cellular responses of breast and thigh tissues to heat and cold stresses and may offer insight into the unique susceptibilities, as well as functional consequences, of these tissues to thermal stress.


Asunto(s)
Proteínas Aviares/genética , Pollos/fisiología , Respuesta al Choque por Frío , Respuesta al Choque Térmico , Fosfotransferasas/genética , Proteoma/genética , Animales , Proteínas Aviares/metabolismo , Pollos/genética , Masculino , Músculo Esquelético/metabolismo , Especificidad de Órganos , Músculos Pectorales/metabolismo , Fosfotransferasas/metabolismo , Proteoma/metabolismo
12.
Infect Immun ; 82(1): 62-71, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24126524

RESUMEN

Mycoplasma bovis is one of the major causative pathogens of bovine respiratory complex disease (BRD), which is characterized by enzootic pneumonia, mastitis, pleuritis, and polyarthritis. M. bovis enters and colonizes bovine respiratory epithelial cells through inhalation of aerosol from contaminated air. The nature of the interaction between M. bovis and the bovine innate immune system is not well understood. We hypothesized that M. bovis invades blood monocytes and regulates cellular function to support its persistence and systemic dissemination. We used bovine-specific peptide kinome arrays to identify cellular signaling pathways that could be relevant to M. bovis-monocyte interactions in vitro. We validated these pathways using functional, protein, and gene expression assays. Here, we show that infection of bovine blood monocytes with M. bovis delays spontaneous or tumor necrosis factor alpha (TNF-α)/staurosporine-driven apoptosis, activates the NF-κB p65 subunit, and inhibits caspase-9 activity. We also report that M. bovis-infected bovine monocytes do not produce gamma interferon (IFN-γ) and TNF-α, although the level of production of interleukin-10 (IL-10) is elevated. Our findings suggest that M. bovis takes over the cellular machinery of bovine monocytes to prolong bacterial survival and to possibly facilitate subsequent systemic distribution.


Asunto(s)
Apoptosis/inmunología , Interferón gamma/biosíntesis , Interleucina-10/biosíntesis , Monocitos/microbiología , Infecciones por Mycoplasma/inmunología , Mycoplasma bovis/inmunología , Tuberculosis Bovina/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Apoptosis/fisiología , Caspasa 9/metabolismo , Bovinos , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Monocitos/inmunología , Monocitos/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Tuberculosis Bovina/metabolismo , Tuberculosis Bovina/microbiología
13.
Immunology ; 143(1): 68-80, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24666281

RESUMEN

Human cathelicidin LL-37 protects against infections and endotoxin-induced inflammation. In a recent study we have shown that IG-19, an LL-37-derived peptide, protects in a murine model of arthritis. Cytokine interleukin-32 (IL-32) is elevated and directly associated with the disease severity of inflammatory arthritis. Therefore, in this study we examined the effects of LL-37 and IG-19 on IL-32-induced responses in human peripheral blood-derived mononuclear cells (PBMC) and macrophages. We showed that CD14(+) monocytes are the primary cells that produce pro-inflammatory tumour necrosis factor-α (TNF-α) following stimulation of PBMC with IL-32. We demonstrated that LL-37 and IG-19 significantly suppress IL-32-induced production of pro-inflammatory cytokines, e.g. TNF-α and IL-1ß, without altering chemokine production. In contrast, LL-37 and IG-19 enhance the production of the anti-inflammatory cytokine IL-1RA. Further mechanistic studies revealed that LL-37 and IG-19 suppress IL-32-mediated phosphorylation of Fyn (Y420) Src kinase. In contrast, IL-32-mediated phosphorylation of AKT-1 (T308) and MKP-1 (S359) is not suppressed by the peptides. LL-37 and IG-19 alone induce the phosphorylation of MKP-1 (S359), which is a known negative regulator of inflammation. Furthermore, the peptides induce the activity of p44/42 mitogen-activated protein kinase, which is known to phosphorylate MKP-1 (S359). This is the first study to demonstrate the regulation of IL-32-induced inflammation by LL-37 and its derivative peptide IG-19. The mechanistic results from this study suggest that regulation of immune-mediated inflammation by these peptides may be controlled by the dual phosphatase MKP-1. We speculate that LL-37 and its derivatives may contribute to the control of immune-mediated inflammatory diseases.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/inmunología , Inflamación/inmunología , Interleucinas/inmunología , Leucocitos Mononucleares/inmunología , Macrófagos/inmunología , Transducción de Señal/inmunología , Western Blotting , Células Cultivadas , Citocinas/biosíntesis , Citocinas/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Catelicidinas
14.
Bioinformatics ; 29(13): 1693-5, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23658419

RESUMEN

SUMMARY: While many experimentally characterized phosphorylation sites exist for certain organisms, such as human, rat and mouse, few sites are known for other organisms, hampering related research efforts. We have developed a software pipeline called DAPPLE that automates the process of using known phosphorylation sites from other organisms to identify putative sites in an organism of interest. AVAILABILITY: DAPPLE is available as a web server at http://saphire.usask.ca. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Fosfoproteínas/química , Fosfoproteínas/metabolismo , Programas Informáticos , Animales , Bovinos , Humanos , Fosforilación , Análisis de Secuencia de Proteína , Homología de Secuencia de Aminoácido
15.
Vet Res ; 45: 54, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24885748

RESUMEN

Johne's disease (JD) is a chronic enteric infection of cattle caused by Mycobacterium avium subsp. paratuberculosis (MAP). The high economic cost and potential zoonotic threat of JD have driven efforts to develop tools and approaches to effectively manage this disease within livestock herds. Efforts to control JD through traditional animal management practices are complicated by MAP's ability to cause long-term environmental contamination as well as difficulties associated with diagnosis of JD in the pre-clinical stages. As such, there is particular emphasis on the development of an effective vaccine. This is a daunting challenge, in large part due to MAP's ability to subvert protective host immune responses. Accordingly, there is a priority to understand MAP's interaction with the bovine host: this may inform rational targets and approaches for therapeutic intervention. Here we review the early host defenses encountered by MAP and the strategies employed by the pathogen to avert or subvert these responses, during the critical period between ingestion and the establishment of persistent infection in macrophages.


Asunto(s)
Enfermedades de los Bovinos/inmunología , Inmunidad Innata , Mycobacterium avium subsp. paratuberculosis/inmunología , Paratuberculosis/inmunología , Animales , Bovinos , Enfermedades de los Bovinos/microbiología , Enfermedades de los Bovinos/transmisión , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Macrófagos/inmunología , Macrófagos/microbiología , Paratuberculosis/microbiología , Paratuberculosis/transmisión
16.
Mol Cell Proteomics ; 11(6): M111.015701, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22205724

RESUMEN

Monkeypox virus (MPXV) is comprised of two clades: Congo Basin MPXV, with an associated case fatality rate of 10%, and Western African MPXV, which is associated with less severe infection and minimal lethality. We thus postulated that Congo Basin and West African MPXV would differentially modulate host cell responses and, as many host responses are regulated through phosphorylation independent of transcription or translation, we employed systems kinomics with peptide arrays to investigate these functional host responses. Using this approach we have demonstrated that Congo Basin MPXV infection selectively down-regulates host responses as compared with West African MPXV, including growth factor- and apoptosis-related responses. These results were confirmed using fluorescence-activated cell sorting analysis demonstrating that West African MPXV infection resulted in a significant increase in apoptosis in human monocytes as compared with Congo Basin MPXV. Further, differentially phosphorylated kinases were identified through comparison of our MPXV data sets and validated as potential targets for pharmacological inhibition of Congo Basin MPXV infection, including increased Akt S473 phosphorylation and decreased p53 S15 phosphorylation. Inhibition of Akt S473 phosphorylation resulted in a significant decrease in Congo Basin MPXV virus yield (261-fold) but did not affect West African MPXV. In addition, treatment with staurosporine, an apoptosis activator resulted in a 49-fold greater decrease in Congo Basin MPXV yields as compared with West African MPXV. Thus, using a systems kinomics approach, our investigation demonstrates that West African and Congo Basin MPXV differentially modulate host cell responses and has identified potential host targets of therapeutic interest.


Asunto(s)
Monkeypox virus/fisiología , Mpox/metabolismo , Fosfoproteínas/metabolismo , Proteínas Quinasas/metabolismo , Proteoma/metabolismo , Animales , Apoptosis , Línea Celular , Chlorocebus aethiops , Análisis por Conglomerados , Interacciones Huésped-Patógeno , Humanos , Imidazoles/farmacología , Monocitos/enzimología , Monocitos/metabolismo , Monocitos/virología , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-met/metabolismo , Piridinas/farmacología , Transducción de Señal , Replicación Viral/efectos de los fármacos
17.
Infect Immun ; 81(8): 2861-72, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23716614

RESUMEN

Mycobacterium avium subsp. paratuberculosis is the causative agent of Johne's disease (JD) in cattle. M. avium subsp. paratuberculosis infects the gastrointestinal tract of calves, localizing and persisting primarily in the distal ileum. A high percentage of cattle exposed to M. avium subsp. paratuberculosis do not develop JD, but the mechanisms by which they resist infection are not understood. Here, we merge an established in vivo bovine intestinal segment model for M. avium subsp. paratuberculosis infection with bovine-specific peptide kinome arrays as a first step to understanding how infection influences host kinomic responses at the site of infection. Application of peptide arrays to in vivo tissue samples represents a critical and ambitious step in using this technology to understand host-pathogen interactions. Kinome analysis was performed on intestinal samples from 4 ileal segments subdivided into 10 separate compartments (6 M. avium subsp. paratuberculosis-infected compartments and 4 intra-animal controls) using bovine-specific peptide arrays. Kinome data sets clustered into two groups, suggesting unique binary responses to M. avium subsp. paratuberculosis. Similarly, two M. avium subsp. paratuberculosis-specific immune responses, characterized by different antibody, T cell proliferation, and gamma interferon (IFN-γ) responses, were also observed. Interestingly, the kinomic groupings segregated with the immune response groupings. Pathway and gene ontology analyses revealed that differences in innate immune and interleukin signaling and particular differences in the Wnt/ß-catenin pathway distinguished the kinomic groupings. Collectively, kinome analysis of tissue samples offers insight into the complex cellular responses induced by M. avium subsp. paratuberculosis in the ileum and provides a novel method to understand mechanisms that alter the balance between cell-mediated and antibody responses to M. avium subsp. paratuberculosis infection.


Asunto(s)
Mucosa Intestinal/microbiología , Mycobacterium avium subsp. paratuberculosis/inmunología , Paratuberculosis/inmunología , Paratuberculosis/microbiología , Transcriptoma , Animales , Bovinos , Modelos Animales de Enfermedad , Electroforesis en Gel de Poliacrilamida , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/inmunología , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Análisis por Micromatrices , Moco/metabolismo , Mycobacterium avium subsp. paratuberculosis/metabolismo , Mycobacterium avium subsp. paratuberculosis/patogenicidad , Paratuberculosis/metabolismo , Fosfotransferasas/biosíntesis
18.
Infect Immun ; 81(1): 226-37, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23115040

RESUMEN

Mycobacterium avium subsp. paratuberculosis is the causative agent of Johne's disease in cattle. The complex, multifaceted interaction of M. avium subsp. paratuberculosis with its host includes dampening the ability of infected cells to respond to stimuli that promote M. avium subsp. paratuberculosis clearance. By disrupting host defenses, M. avium subsp. paratuberculosis creates an intracellular environment that favors the establishment and maintenance of infection. Toll-like receptors (TLRs) are important sensors that initiate innate immune responses to microbial challenge and are also immunotherapeutic targets. For example, TLR9 contributes to host defense against M. avium subsp. paratuberculosis, and its agonists (CpG oligodeoxynucleotides [ODNs]) are under investigation for treatment of Johne's disease and other infections. Here we demonstrate that M. avium subsp. paratuberculosis infection changes the responsiveness of bovine monocytes to TLR9 stimulation. M. avium subsp. paratuberculosis inhibits classical TLR9-mediated responses despite a 10-fold increase in TLR9 expression and maintained uptake of CpG ODNs. Other TLR9-mediated responses, such as oxidative burst, which occur through noncanonical signaling, remain functional. Kinome analysis verifies that classic TLR9 signaling is blocked by M. avium subsp. paratuberculosis infection and that signaling instead proceeds through a Pyk2-mediated mechanism. Pyk2-mediated signaling does not hinder infection, as CpG ODNs fail to promote M. avium subsp. paratuberculosis clearance. Indeed, Pyk2 signaling appears to be an important aspect of M. avium subsp. paratuberculosis infection, as Pyk2 inhibitors significantly reduce the number of intracellular M. avium subsp. paratuberculosis bacteria. The actions of M. avium subsp. paratuberculosis on TLR9 signaling may represent a strategy to generate a host environment which is better suited for infection, revealing potential new targets for therapeutic intervention.


Asunto(s)
Monocitos/inmunología , Monocitos/microbiología , Mycobacterium avium subsp. paratuberculosis/metabolismo , Paratuberculosis/metabolismo , Receptor Toll-Like 9/metabolismo , Animales , Bovinos , Enfermedades de los Bovinos/inmunología , Enfermedades de los Bovinos/metabolismo , Enfermedades de los Bovinos/microbiología , Quinasa 2 de Adhesión Focal/inmunología , Quinasa 2 de Adhesión Focal/metabolismo , Interleucina-10/inmunología , Interleucina-10/metabolismo , Monocitos/metabolismo , Mycobacterium avium subsp. paratuberculosis/inmunología , Paratuberculosis/inmunología , Paratuberculosis/microbiología , Estallido Respiratorio/inmunología , Transducción de Señal/inmunología
19.
BMC Genomics ; 14: 854, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24314169

RESUMEN

BACKGROUND: Recently, questions have been raised regarding the ability of animal models to recapitulate human disease at the molecular level. It has also been demonstrated that cellular kinases, individually or as a collective unit (the kinome), play critical roles in regulating complex biology. Despite the intimate relationship between kinases and health, little is known about the variability, consistency and stability of kinome profiles across species and individuals. RESULTS: As a preliminary investigation of the existence of species- and individual-specific kinotypes (kinome signatures), peptide arrays were employed for the analysis of peripheral blood mononuclear cells collected weekly from human and porcine subjects (n = 6) over a one month period. The data revealed strong evidence for species-specific signalling profiles. Both humans and pigs also exhibited evidence for individual-specific kinome profiles that were independent of natural changes in blood cell populations. CONCLUSIONS: Species-specific kinotypes could have applications in disease research by facilitating the selection of appropriate animal models or by revealing a baseline kinomic signature to which treatment-induced profiles could be compared. Similarly, individual-specific kinotypes could have implications in personalized medicine, where the identification of molecular patterns or signatures within the kinome may depend on both the levels of kinome diversity and temporal stability across individuals.


Asunto(s)
Leucocitos Mononucleares/enzimología , Fosfotransferasas/metabolismo , Proteoma , Proteómica , Adulto , Animales , Análisis por Conglomerados , Activación Enzimática , Femenino , Redes Reguladoras de Genes , Humanos , Masculino , Persona de Mediana Edad , Péptidos/genética , Péptidos/metabolismo , Fosfotransferasas/genética , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Especificidad de la Especie , Porcinos , Adulto Joven
20.
Vet Res ; 44: 35, 2013 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-23682635

RESUMEN

Salmonella enterica serovar Typhimurium (Salmonella Typhimurium) infection of chickens that are more than a few days old results in asymptomatic cecal colonization with persistent shedding of bacteria. We hypothesized that while the bacterium colonizes and persists locally in the cecum it has systemic effects, including changes to metabolic pathways of skeletal muscle, influencing the physiology of the avian host. Using species-specific peptide arrays to perform kinome analysis on metabolic signaling pathways in skeletal muscle of Salmonella Typhimurium infected chickens, we have observed key metabolic changes that affected fatty acid and glucose metabolism through the 5'-adenosine monophosphate-activated protein kinase (AMPK) and the insulin/mammalian target of rapamycin (mTOR) signaling pathway. Over a three week time course of infection, we observed changes in the phosphorylation state of the AMPK protein, and proteins up and down the pathway. In addition, changes to a large subset of the protein intermediates of the insulin/mTOR pathway in the skeletal muscle were altered by infection. These changes occur in pathways with direct effects on fatty acid and glucose metabolism. This is the first report of significant cellular metabolic changes occurring systemically in chicken due to a Salmonella infection. These results have implications not only for animal production and health but also for the understanding of how Salmonella infection in the intestine can have widespread, systemic effects on the metabolism of chickens without disease-like symptoms.


Asunto(s)
Anticuerpos Antibacterianos/metabolismo , Pollos , Músculo Esquelético/metabolismo , Enfermedades de las Aves de Corral/microbiología , Salmonelosis Animal/microbiología , Salmonella typhimurium/fisiología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Ácidos Grasos/metabolismo , Insulina/metabolismo , Músculo Esquelético/microbiología , Fosforilación , Análisis por Matrices de Proteínas/veterinaria , Salmonella typhimurium/inmunología , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA