Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Intervalo de año de publicación
1.
Proteomes ; 6(4)2018 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-30301203

RESUMEN

It is well accepted that treatment of chronic pain with morphine leads to µ opioid receptor (MOR) desensitization and the development of morphine tolerance. MOR activation by the selective peptide agonist, D-Ala2, N-MePhe4, Gly-ol]-enkephalin(DAMGO), leads to robust G protein receptor kinase activation, ß-arrestin recruitment, and subsequent receptor endocytosis, which does not occur in an activation by morphine. However, MOR activation by morphine induces receptor desensitization, in a Protein kinase C (PKC) dependent manner. PKC inhibitors have been reported to decrease receptor desensitization, reduce opiate tolerance, and increase analgesia. However, the exact role of PKC in these processes is not clearly delineated. The difficulties in establishing a particular role for PKC have been, in part, due to the lack of reagents that allow the selective identification of PKC targets. Recently, we generated a conformation state-specific anti-PKC antibody that preferentially recognizes the active state of this kinase. Using this antibody to selectively isolate PKC substrates and a proteomics strategy to establish the identity of the proteins, we examined the effect of morphine treatment on the PKC targets. We found an enhanced interaction of a number of proteins with active PKC, in the presence of morphine. In this article, we discuss the role of these proteins in PKC-mediated MOR desensitization and analgesia. In addition, we posit a role for some of these proteins in mediating pain by TrKA activation, via the activation of transient receptor potential cation channel subfamily V member 1 (TRPV1). Finally, we discuss how these new PKC interacting proteins and pathways could be targeted for the treatment of pain.

2.
Curr Protoc Chem Biol ; 10(2): e42, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29927112

RESUMEN

The protein kinase C (PKC) family of serine/ threonine kinases has been shown to play active roles as either suppressors or promoters of carcinogenesis in different types of tumors. Using antibodies that preferentially recognize the active conformation of classical PKCs (cPKCs), we have previously shown that in breast cancer samples the expression levels of cPKCs were similar in estrogen receptor-positive (ER+ ) as compared to triple-negative tumors; however, the levels of active cPKCs were different. Determining the activation status of PKCs and other kinases in tumors may thus aid therapeutic decisions. Further, in basic science these tools may be used to understand the spatial and temporal dynamics of PKC signaling under different stimuli and for co-immunoprecipitation studies to detect binding partners and substrates of active cPKCs. In this article, we describe how monoclonal and polyclonal anti-active state PKC antibodies can be obtained using rational approaches to select bona fide epitopes through inspection of the crystal structure of classical PKCs coupled to molecular modeling studies. We believe that this methodology can be used for other kinases and multi-domain enzymes that undergo changes in their conformation upon activation. © 2018 by John Wiley & Sons, Inc.


Asunto(s)
Anticuerpos/química , Anticuerpos/inmunología , Proteína Quinasa C/química , Proteína Quinasa C/inmunología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Dominio Catalítico , Humanos , Conformación Proteica , Proteína Quinasa C/metabolismo
3.
Neuropsychopharmacology ; 42(13): 2527-2536, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28425495

RESUMEN

Studies show that neuropeptide-receptor systems in the basolateral amygdala (BLA) play an important role in the pathology of anxiety and other mood disorders. Since GPR171, a recently deorphanized receptor for the abundant neuropeptide BigLEN, is expressed in the BLA, we investigated its role in fear and anxiety-like behaviors. To carry out these studies we identified small molecule ligands using a homology model of GPR171 to virtually screen a library of compounds. One of the hits, MS0021570_1, was identified as a GPR171 antagonist based on its ability to block (i) BigLEN-mediated activation of GPR171 in heterologous cells, (ii) BigLEN-mediated hyperpolarization of BLA pyramidal neurons, and (iii) feeding induced by DREADD-mediated activation of BigLEN containing AgRP neurons in the arcuate nucleus. The role of GPR171 in anxiety-like behavior or fear conditioning was evaluated following systemic or intra-BLA administration of MS0021570_1, as well as following lentiviral-mediated knockdown of GPR171 in the BLA. We find that systemic administration of MS0021570_1 attenuates anxiety-like behavior while intra-BLA administration or knockdown of GPR171 in the BLA reduces anxiety-like behavior and fear conditioning. These results indicate that the BigLEN-GPR171 system plays an important role in these behaviors and could be a novel target to develop therapeutics to treat psychiatric disorders.


Asunto(s)
Ansiedad/metabolismo , Complejo Nuclear Basolateral/metabolismo , Condicionamiento Psicológico/fisiología , Miedo/fisiología , Neuropéptidos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Complejo Nuclear Basolateral/citología , Complejo Nuclear Basolateral/efectos de los fármacos , Células CHO , Condicionamiento Psicológico/efectos de los fármacos , Cricetulus , Miedo/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Moleculares , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Técnicas de Cultivo de Tejidos
4.
Int J Parasitol Drugs Drug Resist ; 6(1): 74-84, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27054066

RESUMEN

Parasitic diseases cause ∼ 500,000 deaths annually and remain a major challenge for therapeutic development. Using a rational design based approach, we developed peptide inhibitors with anti-parasitic activity that were derived from the sequences of parasite scaffold proteins LACK (Leishmania's receptor for activated C-kinase) and TRACK (Trypanosoma receptor for activated C-kinase). We hypothesized that sequences in LACK and TRACK that are conserved in the parasites, but not in the mammalian ortholog, RACK (Receptor for activated C-kinase), may be interaction sites for signaling proteins that are critical for the parasites' viability. One of these peptides exhibited leishmanicidal and trypanocidal activity in culture. Moreover, in infected mice, this peptide was also effective in reducing parasitemia and increasing survival without toxic effects. The identified peptide is a promising new anti-parasitic drug lead, as its unique features may limit toxicity and drug-resistance, thus overcoming central limitations of most anti-parasitic drugs.


Asunto(s)
Leishmania/efectos de los fármacos , Péptidos/síntesis química , Péptidos/farmacología , Proteínas Protozoarias/antagonistas & inhibidores , Receptores de Superficie Celular/antagonistas & inhibidores , Tripanocidas/farmacología , Trypanosoma/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Antígenos de Protozoos/química , Diseño de Fármacos , Leishmania/química , Leishmania/genética , Leishmaniasis/tratamiento farmacológico , Leishmaniasis/parasitología , Ratones , Parasitemia/tratamiento farmacológico , Péptidos/administración & dosificación , Proteínas Protozoarias/química , Receptores de Cinasa C Activada , Receptores de Superficie Celular/química , Alineación de Secuencia , Tripanocidas/administración & dosificación , Tripanocidas/química , Trypanosoma/genética , Tripanosomiasis/tratamiento farmacológico , Tripanosomiasis/parasitología
5.
Sci Signal ; 9(420): re3, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-27016527

RESUMEN

Despite the efforts of pharmaceutical companies to develop specific kinase modulators, few drugs targeting kinases have been completely successful in the clinic. This is primarily due to the conserved nature of kinases, especially in the catalytic domains. Consequently, many currently available inhibitors lack sufficient selectivity for effective clinical application. Kinases phosphorylate their substrates to modulate their activity. One of the important steps in the catalytic reaction of protein phosphorylation is the correct positioning of the target residue within the catalytic site. This positioning is mediated by several regions in the substrate binding site, which is typically a shallow crevice that has critical subpockets that anchor and orient the substrate. The structural characterization of this protein-protein interaction can aid in the elucidation of the roles of distinct kinases in different cellular processes, the identification of substrates, and the development of specific inhibitors. Because the region of the substrate that is recognized by the kinase can be part of a linear consensus motif or a nonlinear motif, advances in technology beyond simple linear sequence scanning for consensus motifs were needed. Cost-effective bioinformatics tools are already frequently used to predict kinase-substrate interactions for linear consensus motifs, and new tools based on the structural data of these interactions improve the accuracy of these predictions and enable the identification of phosphorylation sites within nonlinear motifs. In this Review, we revisit kinase-substrate interactions and discuss the various approaches that can be used to identify them and analyze their binding structures for targeted drug development.


Asunto(s)
Biología Computacional/métodos , Sistemas de Liberación de Medicamentos , Inhibidores de Proteínas Quinasas , Proteínas Quinasas , Secuencias de Aminoácidos , Animales , Biología Computacional/tendencias , Sistemas de Liberación de Medicamentos/métodos , Sistemas de Liberación de Medicamentos/tendencias , Humanos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato
6.
Sci Rep ; 6: 22114, 2016 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-26911897

RESUMEN

Protein kinase C (PKC) plays a regulatory role in key pathways in cancer. However, since phosphorylation is a step for classical PKC (cPKC) maturation and does not correlate with activation, there is a lack of tools to detect active PKC in tissue samples. Here, a structure-based rational approach was used to select a peptide to generate an antibody that distinguishes active from inactive cPKC. A peptide conserved in all cPKCs, C2Cat, was chosen since modeling studies based on a crystal structure of PKCß showed that it is localized at the interface between the C2 and catalytic domains of cPKCs in an inactive kinase. Anti-C2Cat recognizes active cPKCs at least two-fold better than inactive kinase in ELISA and immunoprecipitation assays, and detects the temporal dynamics of cPKC activation upon receptor or phorbol stimulation. Furthermore, the antibody is able to detect active PKC in human tissue. Higher levels of active cPKC were observed in the more aggressive triple negative breast cancer tumors as compared to the less aggressive estrogen receptor positive tumors. Thus, this antibody represents a reliable, hitherto unavailable and a valuable tool to study PKC activation in cells and tissues. Similar structure-based rational design strategies can be broadly applied to obtain active-state specific antibodies for other signal transduction molecules.


Asunto(s)
Anticuerpos/metabolismo , Neoplasias de la Mama/metabolismo , Neuroblastoma/metabolismo , Proteína Quinasa C beta/metabolismo , Sitios de Unión/inmunología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Carcinogénesis , Línea Celular Tumoral , Activación Enzimática , Femenino , Humanos , Isoenzimas/inmunología , Estadificación de Neoplasias , Neuroblastoma/inmunología , Neuroblastoma/patología , Fragmentos de Péptidos/inmunología , Conformación Proteica , Dominios Proteicos/genética , Proteína Quinasa C beta/genética , Proteína Quinasa C beta/inmunología , Receptores de Estrógenos/metabolismo , Transducción de Señal , Relación Estructura-Actividad
7.
São Paulo; s.n; s.n; 2016. 145 p. tab, graf, ilus.
Tesis en Portugués | LILACS | ID: biblio-847514

RESUMEN

A família proteína quinases C (PKC) é composta por dez isoenzimas, as quais são capazes de fosforilar resíduos de serina e treonina. A ativação dessas quinases envolve mudanças conformacionais, como a remoção do pseudo-substrato do sítio ativo e associação dessas enzimas com lipídeos em membranas biológicas. Além disso, três fosforilações são importantes para a maturação/ enovelamento da enzima e não estão associadas com o estado de ativação das cPKCs. Apesar dessas quinases estarem envolvidas em vários processos patológicos, como carcinogênese e doenças cardiovasculares, ainda não se estabeleceu a relação entre estado de ativação das PKCs com essas doenças. Isso se deve, em parte, à ausência de ferramentas que possibilitam a distinção das formas ativas e inativas das PKCs. Na presente tese, baseando-se em mudanças conformacionais sofridas pelas PKCs durante o processo de ativação, dois anticorpos contra cPKCs ativas foram racionalmente desenvolvidos, sendo um anticorpo policlonal (anti-C2Cat) e outro monoclonal (4.8E). O anticorpo anti-C2Cat foi desenvolvido a partir de imunização de coelhos com um peptídeo localizado na região de interação entre os domínios C2 e catalítico na PKC inativa. Já o anticorpo monoclonal 4.8E foi produzido após a imunização de camundongos Balb/ C com extrato de proteínas proveniente de células HEK293T superexpressando formas constitutivamente ativas da PKCßI. A seletividade de anti-C2Cat e 4.8E por cPKCs ativas foi demonstrada por ensaios de ELISA e de imunoprecipitação, sendo que os anticorpos sempre apresentaram maior afinidade por cPKCs ativas purificadas, superexpressas ou mesmo as endógenas. O anticorpo anti-C2Cat foi capaz de monitorar a dinâmica espaço-temporal da ativação das cPKCs em linhagens de neuroblastoma (Neuro-2A e SK-N-SH) estimuladas com PMA, morfina, ATP ou glutamato por diferentes tempos. Ainda, um maior conteúdo de cPKCs ativas foi detectado por anti-C2Cat na linhagem de câncer de mama MDA-MB-231 (triplo- negativa) do que em células MCF-7 (ER+). Em acordo com esses dados, anti-C2Cat identificou uma maior ativação de cPKCs em tumores mais agressivos de câncer de mama (subtipo triplo-negativo) do que em tumores menos agressivos (ER+, subtipo luminal). Os anticorpos conformacionais anti-C2Cat e 4.8E foram aplicados para elucidar vias de sinalização que levam à carcinogênese em células MDA-MB-231, por meio da realização de ensaios de co-imunoprecipitação, seguida pela identificação das proteínas por espectrometria de massas. Usando essa abordagem, os resultados sugerem que as cPKCs ativas possam estar envolvidas com a tradução de proteínas envolvidas na migração celular, como actina. Em conjunto, os resultado obtidos na presente tese demonstram duas formas racionais de desenvolver anticorpos contra cPKCs ativas, sendo que algumas aplicações para estas ferramentas foram demonstradas. Estratégias baseadas em mudanças conformacionais, similares às apresentadas aqui, poderão ser utilizadas para a produção racional de anticorpos contra outras quinases ou proteínas


The protein kinase C family (PKC) is composed of ten isoenzymes, which are capable of phosphorylating serine and threonine amino acid residues. PKC activation involves conformational changes, such as removing the pseudo-substrate from the active site and binding of the enzyme to lipids in biological membranes. In addition, PKC undergoes three phosphorylations that are important for the maturation/ folding of the enzyme and are not linked with activation status. Despite the fact that these kinases are involved in various pathological processes, such as carcinogenesis and cardiovascular disease, a relationship between PKC activation status with these diseases has not yet been established. This is partly due to the lack of tools to detect active PKC in tissue samples. In this thesis, based on conformational changes suffered by PKC during its activation, two antibodies against active cPKCs were rationally developed; a polyclonal antibody (anti-C2Cat) and a monoclonal (4.8E). Anti-C2Cat was produced after immunization of rabbits with a peptide located at the interface between the C2 and catalytic domains of cPKCs in an inactive PKC. The monoclonal antibody 4.8E was produced after immunization of Balb/C mice with total lysates from HEK293T cells overexpressing constitutively active forms of PKCßI. The anti-C2Cat and 4.8E specificity by active cPKCs was demonstrated by ELISA and immunoprecipitation assays, where the antibodies always showed higher affinity to active cPKCs. Anti-C2Cat was able to detect the temporal and spatial dynamics of cPKC activation upon receptor (morphine, ATP or glutamate) or phorbol ester stimulation in neuroblastoma lines (Neuro-2A and SK-N-SH). Futhermore, anti-C2Cat is able to detect active PKC in human tissues. Higher levels of active cPKC were observed in the more aggressive triple negative breast cancer tumors as compared to the less aggressive estrogen receptor positive tumors. Also, both antibodies were applied to study signaling pathways that lead to carcinogenesis in MDA-MB-231 cells by performing co-immunoprecipitation and mass spectrometry. Using this approach, the results suggest that active cPKCs may be involved in translation of proteins involved in cell migration, such as actin. Taken together, the results obtained in this thesis showed two rational ways to develop antibodies against active cPKCs and some applications for these tools were demonstrated. Strategies based on conformational changes, similar to those presented herein may be used for rational production of antibodies against other kinases and proteins


Asunto(s)
Animales , Masculino , Femenino , Ratones , Conejos , Anticuerpos Monoclonales/análisis , Anticuerpos/análisis , Proteína Quinasa C/efectos adversos , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Neoplasias de la Mama/complicaciones , Línea Celular Tumoral/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , Técnica del Anticuerpo Fluorescente/métodos , Hibridomas , Inmunoprecipitación/métodos , Espectrometría de Masas/métodos
8.
Sci Signal ; 7(350): ra105, 2014 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-25372052

RESUMEN

Linear consensus motifs are short contiguous sequences of residues within a protein that can form recognition modules for protein interaction or catalytic modification. Protein kinase specificity and the matching of kinases to substrates have been mostly defined by phosphorylation sites that occur in linear consensus motifs. However, phosphorylation can also occur within sequences that do not match known linear consensus motifs recognized by kinases and within flexible loops. We report the identification of Thr(253) in α-tubulin as a site that is phosphorylated by protein kinase C ßI (PKCßI). Thr(253) is not part of a linear PKC consensus motif. Instead, Thr(253) occurs within a region on the surface of α-tubulin that resembles a PKC phosphorylation site consensus motif formed by basic residues in different parts of the protein, which come together in the folded protein to form the recognition motif for PKCßI. Mutations of these basic residues decreased substrate phosphorylation, confirming the presence of this "structurally formed" consensus motif and its importance for the protein kinase-substrate interaction. Analysis of previously reported protein kinase A (PKA) and PKC substrates identified sites within structurally formed consensus motifs in many substrates of these two kinase families. Thus, the concept of consensus phosphorylation site motif needs to be expanded to include sites within these structurally formed consensus motifs.


Asunto(s)
Fosfotransferasas/química , Secuencias de Aminoácidos , Animales , Catálisis , Bovinos , Proteínas Quinasas Dependientes de AMP Cíclico/química , Proteínas Fluorescentes Verdes/química , Células HEK293 , Células HeLa , Humanos , Lisina/química , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Mutación , Fosforilación , Pliegue de Proteína , Proteína Quinasa C/química , Treonina/química , Tubulina (Proteína)/química
9.
J Proteomics ; 94: 497-512, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24332066

RESUMEN

The protein kinase C (PKC) family of serine/threonine kinases participate in embryonic stem cell (ESC) proliferation/self-renewal. A few stimuli that induce ESC proliferation activate several PKC isoenzymes including δPKC, however, the role of this isoenzyme under basal conditions that maintain undifferentiated ESCs remains to be determined. Herewith, we aimed to characterize signaling events that occur in undifferentiated ESCs upon δPKC activation. Using phosphoproteomics and a δPKC specific activator peptide, ψδRACK, it was seen that the majority of proteins whose phosphorylation increased upon δPKC activation participate in cell proliferation. Network analysis of these proteins directly connected δPKC to Raf1 and 14-3-3. Experimental validation studies showed that activation of δPKC increased its binding to 14-3-3, transiently activated ERK1/2 and increased ESC proliferation. Independently inhibiting MEK or PI3 kinase both led to a decrease in proliferation of approximately 50%, but δPKC activation only recovered the effect of PI3 kinase inhibition suggesting that ERK1/2 activation via δPKC is probably a parallel pathway to PI3 kinase and that both pathways are necessary for undifferentiated ESC proliferation. BIOLOGICAL SIGNIFICANCE: The use of embryonic stem cells and induced pluripotent stem cells for regenerative therapies is still a challenge. Understanding the underlying mechanisms that keep these cells proliferating with the ability to differentiate in more than 200 cell types (self-renewal) will aid in the future use of these cells therapeutically. Using a targeted phosphoproteomics study, insights into signaling pathways involved in ESC proliferation can be obtained. Modulating these pathways will aid the obtention of a larger number of self-renewing stem cells and induced pluripotent stem cells that can be used therapeutically.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Células Madre Embrionarias/enzimología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Péptidos/farmacología , Proteína Quinasa C-delta/metabolismo , Proteínas 14-3-3/metabolismo , Línea Celular , Células Madre Embrionarias/citología , Activación Enzimática/efectos de los fármacos , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo
10.
J Infect Dis ; 204(3): 478-86, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21742848

RESUMEN

BACKGROUND: Chagas disease is caused by the protozoan parasite Trypanosoma cruzi, which exhibits a high genetic variability. TcI, TcII, or mixed TcI/TcII strains may be found during acute human infection while mainly TcII parasites are present at the chronic stage of disease. In a previously studied Chagas disease outbreak, we identified mixed TcI/TcII strains in the vector Triatoma tibiamaculata and only TcII strains in infected humans, indicating that T. cruzi populations may be selected within the human host. METHODS: Utilizing molecular typing and cell biology techniques, we investigated the interaction of TcI, TcII, and mixed TcI/TcII strains with macrophages, an important cell population implicated in controlling protozoan infection. RESULTS: TcII but not TcI strains were selected by both human and murine macrophages in vitro and by peritoneal cavity cells in vivo. Biological analysis revealed that, compared with TcI, TcII strains display higher infective and multiplicative ability as well as lower doubling time inside macrophages. However, TcI and TcII strains present similar susceptibility to interferon-γ-activated macrophages in vitro. CONCLUSIONS: Taken together, our results reveal the existence of an intracellular selection process in macrophages that favors TcII, but not TcI, when infection occurs with vector-derived mixed TcI/TcII strains.


Asunto(s)
Enfermedad de Chagas/parasitología , Macrófagos/parasitología , Trypanosoma cruzi/clasificación , Enfermedad Aguda , Animales , Anticuerpos Antiprotozoarios/inmunología , Humanos , Interferón gamma/farmacología , Activación de Macrófagos , Masculino , Ratones , Ratones Endogámicos BALB C , Filogenia
11.
Braz. j. vet. res. anim. sci ; 43(4): 537-541, 2006. ilus, tab
Artículo en Portugués | LILACS | ID: lil-461493

RESUMEN

Os vírus da anemia infecciosa eqüina (EIAV), da influenza eqüina tipo 2 (EIV-2) e o herpesvírus eqüino tipo 1 (EHV-1) são agentes causadores de enfermidades que podem causar graves prejuízos econômicos. O objetivo deste presente estudo foi estimar a freqüência de anticorpos contra o EIAV, EIV-2 e o EHV-1 em rebanhos do sul do Estado do Pará, Brasil. Os anticorpos contra EIAV, EIV-2 e EHV- 1 foram detectados pelo teste de IDGA, pelo método de inibição da hemaglutinação e pela técnica de soroneutralização (TCID50 =100), respectivamente. Amostras de sangue de 672, 514 e de 506 equídeos saudáveis e sem histórico de vacinação contra nenhum dos três vírus foram testadas, respectivamente, para EIAV, EIV-2, EHV-1. A seguinte freqüência de soro reativos foi observada: 1,34% para o EIAV; 35,79% para o EIV-2; 45,45% para o EHV-1. Estes resultados indicam que estes agentes estão presentes no rebanho paraense e a adoção de medidas de manejo e profilaxia devem ser priorizadas, garantindo deste modo, a prosperidade da eqüideocultura brasileira.


Equine infectious anemia virus (EIA V), equine influenza virus type 2 (EIV-2) and equine herpesvirus type 1 (EHV-1) are the causal agents of diseases that may bring economical Iosses. The aim of this present study was to estimate the frequency of antibodies against EIAV, EIV-2 and EHV-1 in herds of south Pará State, Brazil. Antibodies against EIAV, EIV-2 and EHV-1 were detected by AGID, hemagglutination inhibition method and serum neutralization technique (TCID50 =100), respectively. Blood samples of 572, 514, and 506 healthy equine unvaccinated against any of the three viruses were tested, respectively, for EIAV, EIV-2 and EHV-1. The following frequencies of serum reactors animals were observed: EIAV,1,34%; EIV-2, 35,79%; EHV-1, 45,45%. These results show that the agents are present in herds from Pará herds and the adoption of measures of management and prophylaxis should be prioritized, ensuring, thereby, the prosperity of brazilian's breeding equine.


Asunto(s)
Anticuerpos Antivirales/aislamiento & purificación , Epidemiología , Herpesvirus Équido 1 , Caballos , Pruebas de Hemaglutinación/métodos , Pruebas de Neutralización/métodos , Virus de la Anemia Infecciosa Equina/aislamiento & purificación , /aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...