Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 18(7): e0288808, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37471346

RESUMO

SARS-CoV-2, the causative agent of Covid-19, is shed from infected persons in respiratory droplets, feces, and urine. Using quantitative PCR (qPCR), our group hypothesized that we could detect SARS-CoV-2 in wastewater samples collected on a university campus prior to the detection of the virus in individuals on campus. Wastewater samples were collected 3 times a week from 5 locations on the main campus of the University of North Carolina Wilmington (UNCW) from July 24, 2020 to December 21, 2020. Post-collection, total RNA was extracted and SARS-CoV-2 RNA in the samples was detected by qPCR. SARS-CoV-2 signal was detected on campus beginning on August 19 as classes began and the signal increased in both intensity and breadth as the Fall semester progressed. A comparison of two RNA extraction methods from wastewater showed that SARS-CoV-2 was detected more frequently on filter samples versus the direct extracts. Aligning our wastewater data with the reported SARS-CoV-2 cases on the campus Covid-19 dashboard showed the virus signal was routinely detected in the wastewater prior to clusters of individual cases being reported. These data support the testing of wastewater for the presence of SARS-CoV-2 and may be used as part of a surveillance program for detecting the virus in a community prior to an outbreak occurring and could ultimately be incorporated with other SARS-CoV-2 metrics to better inform public health enabling a quick response to contain or mitigating spread of the virus.


Assuntos
Vigilância em Saúde Pública , RNA Viral , SARS-CoV-2 , Águas Residuárias , Humanos , COVID-19/epidemiologia , RNA Viral/isolamento & purificação , SARS-CoV-2/genética , SARS-CoV-2/isolamento & purificação , Universidades , Águas Residuárias/virologia , Vigilância em Saúde Pública/métodos , North Carolina/epidemiologia
2.
Front Vet Sci ; 10: 1165917, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37275614

RESUMO

Equine herpesvirus type 1 (EHV-1) is a highly transmissible pathogen that leads to a variety of clinical disease outcomes in infected horses. A major sequela that can occur after an EHV-1 infection is a neurological disease termed equine herpesvirus myeloencephalopathy (EHM). Clinical manifestations of EHM include fever, ataxia, incontinence, and partial to full paralysis, which may ultimately lead to the euthanization of the infected horse. To develop an effective treatment strategy for EHM, it is critical that the specific virus-host interactions that lead to EHM be investigated so that safe and effective therapeutic interventions can be developed and delivered. In this study, we examined the ability of four non-steroidal anti-inflammatory drugs (NSAIDs), a steroidal anti-inflammatory drug (dexamethasone), a Rho-kinase (ROCK) inhibitor, and a JAK/STAT inhibitor (AG490) to reduce EHV-1 virus yields and cell-to-cell spread. We show that the NSAID, flunixin meglumine (FM), and the JAK/STAT inhibitor, AG490, significantly reduced virus yields in endothelial and epithelial cell lines, and this inhibition was similar for two neurologic and two non-neurologic EHV-1 strains. In addition to reducing virus yields, AG490 and FM also significantly reduced the ability of EHV-1 to spread laterally from cell to cell.

3.
J Med Chem ; 64(17): 12651-12669, 2021 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-34415160

RESUMO

We previously described the development of a DNA-alkylating compound that showed selective toxicity in breast cancer cells. This compound contained an estrogen receptor α (ERα)-binding ligand and a DNA-binding/methylating component that could selectively methylate the N3-position of adenines at adenine-thymine rich regions of DNA. Herein, we describe mechanistic investigations that demonstrate that this class of compounds facilitate the translocation of the ERα-compound complex to the nucleus and induce the expression of ERα target genes. We confirm that the compounds show selective toxicity in ERα-expressing cells, induce ERα localization in the nucleus, and verify the essential role of ERα in modulating the toxicity. Minor alterations in the compound structure significantly affects the DNA binding ability, which correlates to the DNA-methylating ability. These studies demonstrate the utility of DNA-alkylating compounds to accomplish targeted inhibition of the growth of specific cancer cells; an approach that may overcome shortcomings of currently used chemotherapy agents.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Metilação de DNA , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Estradiol/administração & dosagem , Estradiol/farmacologia , Feminino , Humanos , Células MCF-7 , Modelos Moleculares , Simulação de Dinâmica Molecular , Estrutura Molecular , Relação Estrutura-Atividade
4.
Biochemistry ; 56(2): 421-440, 2017 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-28000448

RESUMO

DNA-alkylating drugs continue to remain an important weapon in the arsenal against cancers. However, they typically suffer from several shortcomings because of the indiscriminate DNA damage that they cause and their inability to specifically target cancer cells. We have developed a strategy for overcoming the deficiencies in current DNA-alkylating chemotherapy drugs by designing a site-specific DNA-methylating agent that can target cancer cells because of its selective uptake via glucose transporters, which are overexpressed in most cancers. The design features of the molecule, its synthesis, its reactivity with DNA, and its toxicity in human glioblastoma cells are reported here. In this molecule, a glucosamine unit, which can facilitate uptake via glucose transporters, is conjugated to one end of a bispyrrole triamide unit, which is known to bind to the minor groove of DNA at A/T-rich regions. A methyl sulfonate moiety is tethered to the other end of the bispyrrole unit to serve as a DNA-methylating agent. This molecule produces exclusively N3-methyladenine adducts upon reaction with DNA and is an order of magnitude more toxic to treatment resistant human glioblastoma cells than streptozotocin is, a Food and Drug Administration-approved, glycoconjugated DNA-methylating drug. Cellular uptake studies using a fluorescent analogue of our molecule provide evidence of uptake via glucose transporters and localization within the nucleus of cells. These results demonstrate the feasibility of our strategy for developing more potent anticancer chemotherapeutics, while minimizing common side effects resulting from off-target damage.


Assuntos
Antineoplásicos Alquilantes/síntese química , Adutos de DNA/biossíntese , DNA de Neoplasias/antagonistas & inibidores , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Glicoconjugados/síntese química , Neuroglia/efeitos dos fármacos , Adenina/análogos & derivados , Adenina/química , Adenina/metabolismo , Alcanossulfonatos/química , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacologia , Transporte Biológico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Adutos de DNA/química , Dano ao DNA , Metilação de DNA , DNA de Neoplasias/química , DNA de Neoplasias/metabolismo , Expressão Gênica , Glucosamina/química , Proteínas Facilitadoras de Transporte de Glucose/genética , Glicoconjugados/metabolismo , Glicoconjugados/farmacologia , Humanos , Simulação de Dinâmica Molecular , Terapia de Alvo Molecular , Neuroglia/metabolismo , Neuroglia/patologia , Conformação de Ácido Nucleico , Pirróis/química , Estreptozocina/farmacologia
5.
Vet Microbiol ; 192: 52-59, 2016 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-27527764

RESUMO

Equine herpesvirus myeloencephalopathy (EHM), a disease caused by equine herpesvirus type 1 (EHV-1), is characterized by severe inflammation, thrombosis, and hypoxia in central nervous system (CNS) endothelial cells, which can result in a spectrum of clinical signs including urinary incontinence, ataxia, and paralysis. Strains of EHV-1 that contain a single point mutation within the viral DNA polymerase (nucleotide A2254>G2254: amino acid N752→D752) are isolated from EHM afflicted horses at higher frequencies than EHV-1 strains that do not harbor this mutation. Due to the correlation between the DNA Pol mutation and EHM disease, EHV-1 strains that contain the mutation have been designated as neurologic. In this study, we measured virus replication, cell to cell spread efficacy, and host inflammatory responses in equine endothelial cells infected with 12 different strains of EHV-1. Two strains, T953 (Ohio 2003) (neurologic) and Kentucky A (KyA) (non-neurologic), have well described disease phenotypes while the remaining strains used in this study are classified as neurologic or non-neurologic based solely on the presence or absence of the DNA pol mutation, respectively. Results show that the neurologic strains do not replicate better or spread more efficiently in endothelial cells. Also, the majority of the host inflammatory genes were modulated similarly regardless of EHV-1 genotype. Analyses of host gene expression showed that a subset of pro-inflammatory cytokines, including the CXCR3 ligands CXCL9, CXCL10, and CXCL11, as well as CCL5, IL-6 and TNF-α were consistently up-regulated in endothelial cells infected with each EHV-1 strain. The identification of specific pro-inflammatory cytokines in endothelial cells that are modulated by EHV-1 provides further insight into the factors that contribute to the immunopathology observed after infection and may also reveal new targets for disease intervention.


Assuntos
Células Endoteliais/virologia , Regulação da Expressão Gênica/fisiologia , Herpesvirus Equídeo 1/fisiologia , Cavalos , Inflamação/metabolismo , Animais , Linhagem Celular , Células Endoteliais/metabolismo , Coelhos , Ensaio de Placa Viral , Replicação Viral
6.
J Gen Virol ; 95(Pt 7): 1554-1563, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24722677

RESUMO

Equid herpesvirus type 1 (EHV-1) was shown to use an unusual receptor for cellular entry - MHC-I molecules. Here, we demonstrated that the closely related EHV, EHV-4, also uses this strategy for cellular invasion, both in equine cells in culture and in the heterologous, non-permissive murine mastocytoma cell line (P815) after stable transfection with horse MHC-I genes. Using a panel of P815 cell lines transfected with individual horse MHC-I genes, we provided support for the hypothesis that EHV-1 and EHV-4 target classical polymorphic MHC-I molecules as viral entry receptors. All known equine MHC-I molecules from the two principal classical polymorphic loci specify alanine at position 173 (A173), whilst other MHC-I loci encoded different amino acids at this position and did not permit viral entry. Site-directed mutagenesis of position 173 diminished or enhanced viral entry, depending upon the initial amino acid. However, there were other, as yet undefined, constraints to this process: MHC-I genes from two non-classical loci carried A173 but did not enable viral entry in P815 transfectants. Our study suggested that the capacity to bind MHC-I molecules arose in the common ancestor of EHV-1 and EHV-4. The widespread occurrence of A173 in classical polymorphic horse MHC-I molecules indicated that horses of most MHC haplotypes should be susceptible to infection via this entry portal.


Assuntos
Herpesvirus Equídeo 4/fisiologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Receptores Virais/metabolismo , Internalização do Vírus , Animais , Linhagem Celular , Análise Mutacional de DNA , Herpesvirus Equídeo 1/fisiologia , Antígenos de Histocompatibilidade Classe I/genética , Cavalos , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Ligação Viral
7.
J Virol ; 86(5): 2882-6, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22205738

RESUMO

The cytolytic animal virus equine herpesvirus type 1 (EHV-1) was evaluated for its oncolytic potential against five human glioblastoma cell lines. EHV-1 productively infected four of these cell lines, and the degree of infection was positively correlated with glioma cell death. No human major histocompatibility complex class 1 (MHC-I) was detected in the resistant glioma line, while infection of the susceptible glioma cell lines, which expressed human MHC-I, were blocked with antibody to MHC-I, indicating that human MHC-I acts as an EHV-1 entry receptor on glioma cells.


Assuntos
Neoplasias Encefálicas/virologia , Glioblastoma/virologia , Herpesvirus Equídeo 1/fisiologia , Terapia Viral Oncolítica/instrumentação , Vírus Oncolíticos/fisiologia , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Glioblastoma/terapia , Herpesvirus Equídeo 1/genética , Humanos , Vírus Oncolíticos/genética
8.
J Virol ; 84(18): 9027-34, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20610718

RESUMO

In this study, Equus caballus major histocompatibility complex class I (MHC-I) was identified as a cellular entry receptor for the alphaherpesvirus equine herpesvirus type 1 (EHV-1). This novel EHV-1 receptor was discovered using a cDNA library from equine macrophages. cDNAs from this EHV-1-susceptible cell type were inserted into EHV-1-resistant B78H1 murine melanoma cells, these cells were infected with an EHV-1 lacZ reporter virus, and cells that supported virus infection were identified by X-Gal (5-bromo-4-chloro-3-indolyl-beta-d-galactopyranoside) staining. Positive cells were subjected to several rounds of purification to obtain homogeneous cell populations that were shown to be uniformly infected with EHV-1. cDNAs from these cell populations were amplified by PCR and then sequenced. The sequence data revealed that the EHV-1-susceptible cells had acquired an E. caballus MHC-I cDNA. Cell surface expression of this receptor was verified by confocal immunofluorescence microscopy. The MHC-I cDNA was cloned into a mammalian expression vector, and stable B78H1 cell lines were generated that express this receptor. These cell lines were susceptible to EHV-1 infection while the parental B78H1 cells remained resistant to infection. In addition, EHV-1 infection of the B78H1 MHC-I-expressing cell lines was inhibited in a dose-dependent manner by an anti-MHC-I antibody.


Assuntos
Herpesvirus Equídeo 1/fisiologia , Antígenos de Histocompatibilidade Classe I/fisiologia , Cavalos/virologia , Receptores Virais/fisiologia , Internalização do Vírus , Animais , Linhagem Celular , Membrana Celular/química , Expressão Gênica , Biblioteca Gênica , Genes Reporter , Macrófagos , Camundongos , Microscopia Confocal , Microscopia de Fluorescência , Análise de Sequência de DNA , Coloração e Rotulagem/métodos , Transfecção , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
9.
Vet Microbiol ; 141(1-2): 12-21, 2010 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-19713056

RESUMO

To initiate infection, equine herpesvirus type 1 (EHV-1) attaches to heparan sulfate on cell surfaces and then interacts with a putative glycoprotein D receptor(s). After attachment, virus entry occurs either by direct fusion of the virus envelope with the plasma membrane or via endocytosis followed by fusion between the virus envelope and an endosomal membrane. Upon fusion, de-enveloped virus particles are deposited into the cytoplasm and travel to the nucleus for viral replication. In this report, we examined the mechanism of EHV-1 intracellular trafficking and investigated the ability of EHV-1 to utilize specific cellular components to efficiently travel to the nucleus post-entry. Using a panel of microtubule-depolymerizing drugs and inhibitors of microtubule motor proteins, we show that EHV-1 infection is dependent on both the integrity of the microtubule network and the minus-end microtubule motor protein, dynein. In addition, we show that EHV-1 actively induces the acetylation of tubulin, a marker of microtubule stabilization, as early as 15 min post-infection. Finally, our data support a role for the cellular kinase, ROCK1, in virus trafficking to the nucleus.


Assuntos
Dineínas/metabolismo , Herpesvirus Equídeo 1/patogenicidade , Microtúbulos/metabolismo , Quinases Associadas a rho/metabolismo , Acetilação , Animais , Western Blotting , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Herpesvirus Equídeo 1/genética , Herpesvirus Equídeo 1/metabolismo , Cavalos , Microscopia Confocal , Coelhos , Tubulina (Proteína)/metabolismo
10.
J Virol ; 83(7): 2951-61, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19129446

RESUMO

Both initial infection and cell-to-cell spread by herpes simplex virus type 1 (HSV-1) require the interaction of the viral glycoprotein D (gD) with an entry receptor on the cell surface. The two major HSV entry receptors, herpesvirus entry mediator (HVEM) and nectin-1, mediate infection independently but are coexpressed on a variety of cells. To determine if both receptors are active in these instances, we have established mutant viruses that are selectively impaired for recognition of one or the other receptor. In plaque assays, these viruses showed approximately 1,000-fold selectivity for the matched receptor over the mismatched receptor. Separate assays showed that each virus is impaired for both infection and spread through the mismatched receptor. We tested several human tumor cell lines for susceptibility to these viruses and observed that HT29 colon carcinoma cells are susceptible to infection by nectin-1-restricted virus but are highly resistant to HVEM-restricted virus infection, despite readily detectable HVEM expression on the cell surface. HVEM cDNA isolated from HT29 cells rendered HSV-resistant cells permissive for infection by the HVEM-restricted virus, suggesting that HT29 cells lack a cofactor for HVEM-mediated infection or express an HVEM-specific inhibitory factor. Passaging of HVEM-restricted virus on nectin-1-expressing cells yielded a set of gD missense mutations that each restored functional recognition of nectin-1. These mutations identify residues that likely play a role in shaping the nectin-1 binding site of gD. Our findings illustrate the utility of these receptor-restricted viruses in studying the early events in HSV infection.


Assuntos
Moléculas de Adesão Celular/metabolismo , Herpesvirus Humano 1/fisiologia , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Internalização do Vírus , Substituição de Aminoácidos/genética , Linhagem Celular Tumoral , Células Epiteliais/virologia , Herpesvirus Humano 1/genética , Humanos , Mutação de Sentido Incorreto , Nectinas , Membro 14 de Receptores do Fator de Necrose Tumoral/genética , Receptores Virais/genética , Receptores Virais/metabolismo , Proteínas do Envelope Viral/genética , Ensaio de Placa Viral
11.
J Virol ; 81(20): 10879-89, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17670830

RESUMO

Equine herpesvirus type 1 (EHV-1), a member of the Alphaherpesviridae, displays a broad host range in vitro, allowing for detailed study of the mechanisms of productive infection, including attachment and entry, in various cell culture systems. Previously, we showed that EHV-1 infects Chinese hamster ovary (CHO-K1) cells even though these cells do not express a known alphaherpesvirus entry receptor. In this report, we show by electron microscopy and an infectious recovery assay that entry into CHO-K1 cells occurs via an endocytic or phagocytic mechanism, while entry into equine dermal (ED) or rabbit kidney (RK13) cells occurs by direct fusion at the cell surface. In both cases (endocytic/phagocytic or direct fusion), entry leads to productive infection. Using drugs that inhibit clathrin-dependent or caveola-dependent endocytosis, we showed that EHV-1 entry into CHO-K1 cells does not require clathrin or caveolae. We also show that EHV-1 infection requires the activation of cell signaling molecules. In particular, we demonstrate that activation of the serine/threonine Rho kinase ROCK1 is critical for infection. Inhibition of this kinase by drugs or overexpression of a negative regulator of ROCK1 significantly blocked EHV-1 infection. These results show that EHV-1 can enter disparate cell types by at least two distinct mechanisms and that productive infection is dependent upon the activation of ROCK1.


Assuntos
Infecções por Herpesviridae/etiologia , Herpesvirus Equídeo 1/patogenicidade , Quinases Associadas a rho/metabolismo , Animais , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Endocitose , Ativação Enzimática , Cavalos , Microscopia Eletrônica , Fagocitose , Coelhos , Transdução de Sinais , Internalização do Vírus
12.
Virology ; 360(2): 477-91, 2007 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-17157347

RESUMO

Herpes simplex virus type 1 (HSV-1) entry into permissive cells involves attachment to cell-surface glycosaminoglycans (GAGs) and fusion of the virus envelope with the cell membrane triggered by the binding of glycoprotein D (gD) to cognate receptors. In this study, we characterized the observation that soluble forms of the gD ectodomain (sgD) can mediate entry of gD-deficient HSV-1. We examined the efficiency and receptor specificity of this activity and used sequential incubation protocols to determine the order and stability of the initial interactions required for entry. Surprisingly, virus binding to GAGs did not increase the efficiency of sgD-mediated entry and gD-deficient virus was capable of attaching to GAG-deficient cells in the absence of sgD. These observations suggested a novel binding interaction that may play a role in normal HSV infection.


Assuntos
Herpesvirus Humano 1/fisiologia , Receptores Virais/fisiologia , Proteínas do Envelope Viral/fisiologia , Ligação Viral , Internalização do Vírus , Animais , Células CHO , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Cricetulus , Deleção de Genes , Glicosaminoglicanos/deficiência , Herpesvirus Humano 1/genética , Humanos , Receptores Virais/genética , Células Vero , Proteínas do Envelope Viral/genética
13.
J Virol ; 79(5): 3169-73, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15709036

RESUMO

The well-described herpesvirus entry receptors HveA (TNFRSF14), HveB (nectin 2), and HveC (nectin 1) have been shown to mediate the entry of alphaherpesviruses. Our findings showed that the alphaherpesvirus equine herpesvirus 1 (EHV-1) efficiently entered and replicated in CHO-K1 cells that lack the entry receptors HveA, HveB, and HveC, demonstrating that EHV-1 utilizes a unique entry receptor. As with other alphaherpesviruses, efficient EHV-1 entry was dependent on glycoprotein D and cell surface glycosaminoglycans.


Assuntos
Herpesvirus Equídeo 1/fisiologia , Herpesvirus Equídeo 1/patogenicidade , Receptores Virais/fisiologia , Animais , Células CHO , Chlorocebus aethiops , Cricetinae , Glicosaminoglicanos/fisiologia , Infecções por Herpesviridae/etiologia , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/virologia , Doenças dos Cavalos/etiologia , Doenças dos Cavalos/virologia , Cavalos , Células Vero , Proteínas do Envelope Viral/fisiologia
14.
Virus Genes ; 29(1): 9-17, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15215680

RESUMO

One of the consequences of equine herpesvirus 1 (EHV-1) infection in the natural host is a neurological disease that can lead to paralysis. The pathology associated with EHV-1-induced neurological disease includes vasculitis of the small blood vessels within the central nervous system and subsequent damage to the surrounding neural tissue. In a previous study, an EHV-1 recombinant KyA virus (KgI/gE/75) was generated in which the sequences encoding glycoprotein I (gI) and glycoprotein E (gE) were repaired [Frampton et al. 2002 (Virus Research 90: 287-301)] using genes of the pathogenic EHV-1 strain 89c25. In contrast to the parental KyA virus that lacks gI and gE, the recombinant KgI/gE/75 was able to spread to the brains of CBA mice after intranasal infection. Infection resulted in a meningoencephalitis characterized by lymphocytic cuffing of small blood vessels within the brain, consistent with that observed in EHV-1-infected horses exhibiting neurological signs. KgI/gE/75 was able to elicit cytopathology in the lung prior to spread to the brain. However, like the attenuated KyA strain, KgI/gE/75 did not persist in the lung and was completely cleared from lung tissue by day 5 postinfection. We propose that gI and gE are neurovirulence factors for EHV-1, and that the CBA mouse model can be extended to study neurologic sequelae resulting after EHV-1 infection.


Assuntos
Encéfalo/patologia , Herpesvirus Equídeo 1/patogenicidade , Meningoencefalite/fisiopatologia , Recombinação Genética , Proteínas do Envelope Viral/metabolismo , Animais , Encéfalo/virologia , Feminino , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/fisiopatologia , Infecções por Herpesviridae/virologia , Herpesvirus Equídeo 1/genética , Herpesvirus Equídeo 1/metabolismo , Pulmão/patologia , Pulmão/virologia , Meningoencefalite/patologia , Meningoencefalite/virologia , Camundongos , Camundongos Endogâmicos CBA , Proteínas do Envelope Viral/genética , Virulência
15.
Viral Immunol ; 16(3): 307-20, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14583146

RESUMO

Equine herpesvirus 1 (EHV-1)-specific antibody-secreting cells (ASC) isolated from the lung and spleen of mice at 12 months after immunization with attenuated EHV-1 KyA, heat-killed KyA, or recombinant viral glycoprotein D (rgD) assessed by ELISPOT showed a three- to fivefold increase in three immunoglobulin isotypes at 3 days post-challenge with pathogenic EHV-1 RacL11 as compared to control mice. ELISPOT assays demonstrated a high frequency of cells secreting proinflammatory tumor necrosis factor-alpha (TNF-alpha), interferon gamma (IFN-gamma), and interleukin 4 (IL-4) in the lungs in response to infection with KyA or RacL11 or immunization with rgD. Cytokine production elicited by EHV-1 KyA or RacL11 infection revealed similar frequencies of EHV-1-specific IFN-gamma and IL-4 spot forming cells in the mediastinal lymph nodes and spleen. However, KyA induced significantly greater amounts of IFN-gamma producing cells in the lungs than did RacL11. Intranasal immunization with KyA or rgD induced long-term immunity that provided protection against pathogenic EHV-1 challenge infection at 12 months post-immunization. Overall, the data indicate that immunization with infectious KyA or rgD induces significant levels of cytokines, virus-specific ASC in the lungs and spleen, and long-term virus specific B-cell responses.


Assuntos
Anticorpos Antivirais/biossíntese , Citocinas/biossíntese , Herpesvirus Equídeo 1/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Células Produtoras de Anticorpos/imunologia , Linfócitos B/imunologia , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/genética , Feminino , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/prevenção & controle , Herpesvirus Equídeo 1/patogenicidade , Imunização , Memória Imunológica , Interferon gama/biossíntese , Interleucina-4/biossíntese , Camundongos , Camundongos Endogâmicos CBA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/imunologia , Fatores de Tempo , Fator de Necrose Tumoral alfa/biossíntese
16.
Virus Res ; 90(1-2): 287-301, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12457983

RESUMO

The pathogenesis of three equine herpesvirus 1 (EHV-1) recombinants was assessed in a CBA mouse model. Sequences encoding the majority of glycoproteins I (gI) and E (gE) were deleted from the pathogenic EHV-1 strain RacL11 (L11deltagIdeltagE), and sequences comprising the 3859 bp deletion within the strain KyA U(S) segment, which includes genes 73 (gI), 74 (gE), and 75 (putative 10 kDa protein 75), were re-inserted into attenuated KyA (KgI/gE/75). In addition, genes gE and 75 were inserted into KyA to generate the EHV-1 recombinant KgE/75. The insertion of the 3859 bp U(S) segment was sufficient to confer virulence to KyA, as indicated by pronounced signs of clinical disease including substantial weight loss. A large plaque morphology was observed in cells infected with KgI/gE/75 compared with KyA, and a small plaque phenotype was observed in cells infected with L11deltagIdeltagE compared with RacL11. These data indicate that gI and/or gI and gE contribute to the ability of EHV-1 to spread directly from cell-to-cell. The deletion of both gI and gE from the pathogenic RacL11 strain did not reduce clinical signs of disease in infected mice, but did decrease mortality compared with RacL11. Furthermore, the insertion of genes 74 (gE) and 75 into the vaccine strain KyA did not alter the attenuated phenotype of this virus. Finally, KgI/gE/75 and RacL11 elicited the production of the proinflammatory chemokines MIP-1alpha, MIP-1beta, and MIP-2 in the lungs of infected mice, while KyA did not, suggesting that gI and/or gI and gE contribute to the up-regulation of these mediators of inflammation. These findings show that gI, and/or gI and gE restore a virulent phenotype to the EHV-1 KyA strain, and indicate that virulence factors, in addition to gI and gE, contribute to the pathogenesis of the RacL11 strain.


Assuntos
Infecções por Herpesviridae/veterinária , Herpesvirus Equídeo 1/patogenicidade , Doenças dos Cavalos/virologia , Modelos Animais , Proteínas do Envelope Viral/genética , Animais , Linhagem Celular , Quimiocinas/metabolismo , Efeito Citopatogênico Viral , Feminino , Infecções por Herpesviridae/fisiopatologia , Infecções por Herpesviridae/virologia , Herpesvirus Equídeo 1/genética , Herpesvirus Equídeo 1/crescimento & desenvolvimento , Doenças dos Cavalos/fisiopatologia , Cavalos , Camundongos , Camundongos Endogâmicos CBA , Mutação , Ratos , Recombinação Genética , Proteínas do Envelope Viral/metabolismo , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...