Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Am J Obstet Gynecol ; 212(5): 611.e1-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25524398

RESUMEN

OBJECTIVE: The objective of the study was to evaluate the upper genital tract (UGT) presence of vaginal bacterial species using sensitive molecular methods capable of detecting fastidious bacterial vaginosis (BV)-associated bacteria. STUDY DESIGN: Vaginal swabs were collected prior to hysterectomy. The excised uterus was sterilely opened and swabs collected from the endometrium and upper endocervix. DNA was tested in 11 quantitative polymerase chain reaction (PCR) assays for 12 bacterial species: Lactobacillus iners, L crispatus, L jensenii, Gardnerella vaginalis, Atopobium vaginae, Megasphaera spp, Prevotella spp, Leptotrichia/Sneathia, BVAB1, BVAB2, BVAB3, and a broad-range16S ribosomal ribonucleic acid gene assay. Endometrial fluid was tested with Luminex and an enzyme-linked immunosorbent assay for cytokines and defensins and tissue for gene expression of defensins and cathelicidin. RESULTS: We enrolled 58 women: mean aged 43±7 years, mostly white (n=46; 79%) and BV negative (n=43; 74%). By species-specific quantitative PCR, 55 (95%) had UGT colonization with at least 1 species (n=52) or were positive by 16S PCR (n=3). The most common species were L iners (45% UGT, 61% vagina), Prevotella spp (33% UGT, 76% vagina) and L crispatus (33% UGT, 56% vagina). Median quantities of bacteria in the UGT were lower than vaginal levels by 2-4 log10 ribosomal ribonucleic acid gene copies per swab. There were no differences in the endometrial inflammatory markers between women with no bacteria, Lactobacillus only, or any BV-associated species in the UGT. CONCLUSION: Our data suggest that the endometrial cavity is not sterile in most women undergoing hysterectomy and that the presence of low levels of bacteria in the uterus is not associated with significant inflammation.


Asunto(s)
Portador Sano/epidemiología , Cuello del Útero/microbiología , Endometrio/microbiología , Vagina/microbiología , Actinobacteria/genética , Actinobacteria/aislamiento & purificación , Adulto , Portador Sano/microbiología , Femenino , Gardnerella vaginalis/genética , Gardnerella vaginalis/aislamiento & purificación , Humanos , Histerectomía , Lactobacillus/genética , Lactobacillus/aislamiento & purificación , Leptotrichia/genética , Leptotrichia/aislamiento & purificación , Megasphaera/genética , Megasphaera/aislamiento & purificación , Microbiota , Persona de Mediana Edad , Tipificación Molecular , Prevotella/genética , Prevotella/aislamiento & purificación , ARN Ribosómico 16S/genética , Enfermedades Uterinas/cirugía
2.
J Gen Virol ; 95(Pt 3): 578-590, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24323639

RESUMEN

Polymorphonuclear neutrophils (PMN) infiltrate the respiratory tract early after viral infection and can contribute to both host defence and pathology. Coronaviruses are important causes of respiratory tract infections, ranging from mild to severe depending on the viral strain. This study evaluated the role of PMN during a non-fatal pulmonary coronavirus infection in the natural host. Rat coronavirus (RCoV) causes respiratory disease in adult rats, characterized by an early PMN response, viral replication and inflammatory lesions in the lungs, mild weight loss and effective resolution of infection. To determine their role during RCoV infection, PMN were depleted and the effects on disease progression, viral replication, inflammatory response and lung pathology were analysed. Compared with RCoV infection in control animals, PMN-depleted rats had worsened disease with weight loss, clinical signs, mortality and prolonged pulmonary viral replication. PMN-depleted animals had fewer macrophages and lymphocytes in the respiratory tract, corresponding to lower chemokine levels. Combined with in vitro experiments showing that PMN express cytokines and chemokines in response to RCoV-infected alveolar epithelial cells, these findings support a role for PMN in eliciting an inflammatory response to RCoV infection. Despite their critical role in the protection from severe disease, the presence of PMN was correlated with haemorrhagic lesions, epithelial barrier permeability and cellular inflammation in the lungs. This study demonstrated that while PMN are required for an effective antiviral response, they also contribute to lung pathology during RCoV infection.


Asunto(s)
Infecciones por Coronavirus/veterinaria , Coronavirus de la Rata/inmunología , Neutrófilos/inmunología , Alveolos Pulmonares/inmunología , Enfermedades de los Roedores/inmunología , Animales , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Coronavirus de la Rata/fisiología , Citocinas/inmunología , Masculino , Alveolos Pulmonares/patología , Alveolos Pulmonares/virología , Ratas , Ratas Endogámicas F344 , Enfermedades de los Roedores/patología , Enfermedades de los Roedores/virología
3.
Nat Commun ; 15(1): 8161, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39289368

RESUMEN

Gene drives are genetic modifications designed to propagate efficiently through a population. Most applications rely on homologous recombination during sexual reproduction in diploid organisms such as insects, but we recently developed a gene drive in herpesviruses that relies on co-infection of cells by wild-type and engineered viruses. Here, we report on a viral gene drive against human herpes simplex virus 1 (HSV-1) and show that it propagates efficiently in cell culture and during HSV-1 infection in mice. We describe high levels of co-infection and gene drive-mediated recombination in neuronal tissues during herpes encephalitis as the infection progresses from the site of inoculation to the peripheral and central nervous systems. In addition, we show evidence that a superinfecting gene drive virus could recombine with wild-type viruses during latent infection. These findings indicate that HSV-1 achieves high rates of co-infection and recombination during viral infection, a phenomenon that is currently underappreciated. Overall, this study shows that a viral gene drive could spread in vivo during HSV-1 infection, paving the way toward therapeutic applications.


Asunto(s)
Herpes Simple , Herpesvirus Humano 1 , Animales , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Ratones , Herpes Simple/virología , Herpes Simple/genética , Humanos , Coinfección/virología , Tecnología de Genética Dirigida/métodos , Femenino , Células Vero , Chlorocebus aethiops , Encefalitis por Herpes Simple/genética , Encefalitis por Herpes Simple/virología , Ratones Endogámicos C57BL , Recombinación Genética/genética , Genes Virales/genética
4.
Nat Commun ; 15(1): 4018, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38740820

RESUMEN

Anti-HSV therapies are only suppressive because they do not eliminate latent HSV present in ganglionic neurons, the source of recurrent disease. We have developed a potentially curative approach against HSV infection, based on gene editing using HSV-specific meganucleases delivered by adeno-associated virus (AAV) vectors. Gene editing performed with two anti-HSV-1 meganucleases delivered by a combination of AAV9, AAV-Dj/8, and AAV-Rh10 can eliminate 90% or more of latent HSV DNA in mouse models of orofacial infection, and up to 97% of latent HSV DNA in mouse models of genital infection. Using a pharmacological approach to reactivate latent HSV-1, we demonstrate that ganglionic viral load reduction leads to a significant decrease of viral shedding in treated female mice. While therapy is well tolerated, in some instances, we observe hepatotoxicity at high doses and subtle histological evidence of neuronal injury without observable neurological signs or deficits. Simplification of the regimen through use of a single serotype (AAV9) delivering single meganuclease targeting a duplicated region of the HSV genome, dose reduction, and use of a neuron-specific promoter each results in improved tolerability while retaining efficacy. These results reinforce the curative potential of gene editing for HSV disease.


Asunto(s)
Dependovirus , Edición Génica , Herpes Simple , Herpesvirus Humano 1 , Carga Viral , Esparcimiento de Virus , Animales , Edición Génica/métodos , Femenino , Dependovirus/genética , Ratones , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Herpes Simple/genética , Herpes Simple/virología , Herpes Simple/terapia , Modelos Animales de Enfermedad , Latencia del Virus/genética , Humanos , Vectores Genéticos/genética , Células Vero , Terapia Genética/métodos , Herpes Genital/terapia , Herpes Genital/virología , ADN Viral/genética
5.
Hepatol Commun ; 7(2): e0009, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-37074875

RESUMEN

BACKGROUND AND AIMS: Adeno-associated virus (AAV) vectors are widely used to deliver therapeutic transgenes to distinct tissues, including the liver. Vectors based on naturally occurring AAV serotypes as well as vectors using engineered capsids have shown variations in tissue tropism and level of transduction between different mouse models. Moreover, results obtained in rodents frequently lack translatability into large animal studies. In light of the increasing interest in AAV vectors for human gene therapy, an increasing number of studies are being performed in nonhuman primates. To keep animal numbers to a minimum and thus optimize the process of AAV capsid selection, we developed a multiplex barcoding approach to simultaneously evaluate the in vivo vector performance for a set of serotypes and capsid-engineered AAV vectors across multiple organs. APPROACH AND RESULTS: Vector biodistribution and transgene expression were assessed by quantitative PCR, quantitative reverse transcription PCR, vector DNA amplicon Illumina sequencing and vRNAseq in male and female rhesus macaques simultaneously dosed with a mixture of barcoded naturally occurring or engineered AAV vectors encoding the same transgene. As expected, our findings show animal-to-animal variation in both the biodistribution and tissue transduction pattern, which was partly influenced by each animal's distinctive serological status. CONCLUSIONS: This method offers a robust approach to AAV vector optimization that can be used to identify and validate AAV vectors for gene delivery to potentially any anatomical site or cell type.


Asunto(s)
Cápside , Dependovirus , Animales , Ratones , Femenino , Masculino , Humanos , Cápside/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Distribución Tisular , Macaca mulatta/genética , Macaca mulatta/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Terapia Genética/métodos
6.
Am J Respir Cell Mol Biol ; 46(6): 833-41, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22312020

RESUMEN

The alveolar epithelium is a critical target for pulmonary viruses and can produce proinflammatory cytokines and chemokines upon viral infection. However, the molecular interactions between virus-infected alveolar epithelial cells and inflammatory cells, including polymorphonuclear leukocytes (PMNs), have not been thoroughly characterized. Rat coronavirus (RCoV) is used as a model to study the immune response to viral infection in the lung of the natural host. We have developed an in vitro model to characterize the response of PMNs to RCoV-infected type I-like alveolar epithelial (AT1) cells, the primary target for RCoV infection in the alveoli. Multiple CXC chemokines that signal through CXCR2 were required for PMN chemotaxis toward medium from RCoV-infected AT1-like cells (RCoV-AT1). Furthermore, RCoV-AT1 inhibited spontaneous PMN apoptosis, including activation of effector caspase 3 and initiator caspases 8 and 9. Use of a selective inhibitor of CXCR2, SB265610, demonstrated that CXCR2 signaling was required for RCoV-AT1-mediated inhibition of PMN apoptosis. These data suggest that CXC chemokines produced by RCoV-infected AT1-like cells inhibit PMN apoptosis during infection. These studies provide new insight into the molecular mechanisms whereby alveolar epithelial cells direct the functions of PMNs during viral infection of the lung.


Asunto(s)
Apoptosis/fisiología , Quimiotaxis/fisiología , Neutrófilos/fisiología , Alveolos Pulmonares/fisiología , Animales , Células Epiteliales/fisiología , Ratas
7.
Nanotoxicology ; 14(7): 908-928, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32574512

RESUMEN

Silver nanoparticles (AgNP) are used in multiple applications but primarily in the manufacturing of antimicrobial products. Previous studies have identified AgNP toxicity in airway epithelial cells, but no in vitro studies to date have used organotypic cultures as a high-content in vitro model of the conducting airway to characterize the effects of interactions between host genetic and acquired factors, or gene × phenotype interactions (G × P), on AgNP toxicity. In the present study, we derived organotypic cultures from primary murine tracheal epithelial cells (MTEC) to characterize nominal and dosimetric dose-response relationships for AgNPs with a gold core on barrier dysfunction, glutathione (GSH) depletion, reactive oxygen species (ROS) production, lipid peroxidation, and cytotoxicity across two genotypes (A/J and C57BL/6J mice), two phenotypes ('Normal' and 'Type 2 [T2]-Skewed'), and two exposures (an acute exposure of 24 h and a subacute exposure of 4 h, every other day, over 5 days [5 × 4 h]). We characterized the 'T2-Skewed' phenotype as an in vitro model of chronic respiratory diseases, which was marked by increased sensitivity to AgNP-induced barrier dysfunction, GSH depletion, ROS production, lipid peroxidation, and cytotoxicity, suggesting that asthmatics are a sensitive population to AgNP exposures in occupational settings. This also suggests that exposure limits, which should be based upon the most sensitive population, should be derived using in vitro and in vivo models of chronic respiratory diseases. This study highlights the importance of considering dosimetry as well as G × P effects when screening and prioritizing potential respiratory toxicants. Such in vitro studies can be used to inform regulatory policy aimed at special protections for all populations.


Asunto(s)
Antibacterianos/toxicidad , Células Epiteliales/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Plata/toxicidad , Tráquea/efectos de los fármacos , Animales , Antibacterianos/química , Técnicas de Cultivo de Célula , Relación Dosis-Respuesta a Droga , Células Epiteliales/metabolismo , Células Epiteliales/patología , Genotipo , Glutatión/metabolismo , Oro/química , Oro/toxicidad , Peroxidación de Lípido/efectos de los fármacos , Nanopartículas del Metal/química , Ratones , Ratones Endogámicos C57BL , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Plata/química , Propiedades de Superficie , Tráquea/metabolismo , Tráquea/patología
8.
Toxicol Sci ; 173(1): 131-143, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31562762

RESUMEN

The airway epithelium is critical for maintaining innate and adaptive immune responses, and occupational exposures that disrupt its immune homeostasis may initiate and amplify airway inflammation. In our previous study, we demonstrated that silver nanoparticles (AgNP), which are engineered nanomaterials used in multiple applications but primarily in the manufacturing of many antimicrobial products, induce toxicity in organotypic cultures derived from murine tracheal epithelial cells (MTEC), and those differentiated toward a "Type 2 [T2]-Skewed" phenotype experienced an increased sensitivity to AgNP toxicity, suggesting that asthmatics could be a sensitive population to AgNP exposures in occupational settings. However, the mechanistic basis for this genotype × phenotype (G × P) interaction has yet to be defined. In this study, we conducted transcriptional profiling using RNA-sequencing to predict the enrichment of specific canonical pathways and upstream transcriptional regulators to assist in defining a mechanistic basis for G × P effects on AgNP toxicity. Organotypic cultures were derived from MTEC across 2 genetically inbred mouse strains (A/J and C57BL/6J mice), 2 phenotypes ("Normal" and "T2-Skewed"), and 1 AgNP exposure (an acute 24 h exposure) to characterize G × P effects on transcriptional response to AgNP toxicity. The "T2-Skewed" phenotype was marked by increased pro-inflammatory T17 responses to AgNP toxicity, which are significant predictors of neutrophilic/difficult-to-control asthma and suggests that asthmatics could be a sensitive population to AgNP exposures in occupational settings. This study highlights the importance of considering G × P effects when identifying these sensitive populations, whose underlying genetics or diseases could directly modify their response to AgNP exposures.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Plata/toxicidad , Animales , Antibacterianos , Recuento de Células , Epitelio , Genotipo , Ratones , Ratones Endogámicos C57BL , Fenotipo , Sistema Respiratorio , Pruebas de Toxicidad
9.
Nat Commun ; 11(1): 4148, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32811834

RESUMEN

We evaluate gene editing of HSV in a well-established mouse model, using adeno-associated virus (AAV)-delivered meganucleases, as a potentially curative approach to treat latent HSV infection. Here we show that AAV-delivered meganucleases, but not CRISPR/Cas9, mediate highly efficient gene editing of HSV, eliminating over 90% of latent virus from superior cervical ganglia. Single-cell RNA sequencing demonstrates that both HSV and individual AAV serotypes are non-randomly distributed among neuronal subsets in ganglia, implying that improved delivery to all neuronal subsets may lead to even more complete elimination of HSV. As predicted, delivery of meganucleases using a triple AAV serotype combination results in the greatest decrease in ganglionic HSV loads. The levels of HSV elimination observed in these studies, if translated to humans, would likely significantly reduce HSV reactivation, shedding, and lesions. Further optimization of meganuclease delivery and activity is likely possible, and may offer a pathway to a cure for HSV infection.


Asunto(s)
Desoxirribonucleasas/genética , Dependovirus/genética , Infecciones del Ojo/terapia , Edición Génica/métodos , Herpes Simple/terapia , Herpesvirus Humano 1/genética , Latencia del Virus/genética , Animales , Sistemas CRISPR-Cas/genética , Células Cultivadas , Chlorocebus aethiops , Infecciones del Ojo/genética , Infecciones del Ojo/virología , Femenino , Células HEK293 , Herpes Simple/genética , Herpesvirus Humano 1/patogenicidad , Humanos , Ratones , Neuronas/metabolismo , Neuronas/virología , RNA-Seq , Análisis de la Célula Individual , Ganglio Cervical Superior/metabolismo , Ganglio Cervical Superior/virología , Células Vero
10.
Biochem Biophys Res Commun ; 370(2): 338-43, 2008 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-18373977

RESUMEN

Our previous studies indicate that hMRE11 plays a role in MMR, and this function of hMRE11 is most likely mediated by the hMLH1-hMRE11 interaction. Here, we explored the functional implications of the hMLH1-hMRE11 interaction in MMR and the effects of hMLH1 mutations on their interaction. Our in vitro MMR assay demonstrated that the dominant-negative hMRE11(452-634) mutant peptide (i.e., harboring only the hMLH1-interacting domain) imparted a significant reduction in both 3' excision and 3'-directed MMR activities. Furthermore, the expression of hMRE11(452-634), and to a lesser extent hMRE11(1-634) (ATLD1), impaired G2/M checkpoint control in response to MNU and cisplatin treatments, rendering cells resistant to killings by these two anticancer drugs. Analysis of 38 hMLH1 missense mutations showed that the majority of mutations caused significant (>50%) reductions in their interaction with hMRE11, suggesting a potential link between aberrant protein interaction and the pathogenic effects of hMLH1 variants.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Disparidad de Par Base , Reparación del ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , División Celular/genética , Línea Celular , Proteínas de Unión al ADN/genética , Fase G2/genética , Humanos , Proteína Homóloga de MRE11 , Homólogo 1 de la Proteína MutL , Mutación Missense , Proteínas Nucleares/genética , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Técnicas del Sistema de Dos Híbridos
11.
Virology ; 483: 96-107, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25965799

RESUMEN

Infection of type II alveolar epithelial (ATII) cells by influenza A viruses (IAV) correlates with severe respiratory disease in humans and mice. To understand pathogenic mechanisms during IAV infection of ATII cells, murine ATII cells were cultured to maintain a differentiated phenotype, infected with IAV-PR8, which causes severe lung pathology in mice, and proteomics analyses were performed using liquid chromatography-mass spectrometry. PR8 infection increased levels of proteins involved in interferon signaling, antigen presentation, and cytoskeleton regulation. Proteins involved in mitochondrial membrane permeability, energy metabolism, and chromatin formation had reduced levels in PR8-infected cells. Phenotypic markers of ATII cells in vivo were identified, confirming the differentiation status of the cultures. Surfactant protein B had decreased levels in PR8-infected cells, which was confirmed by immunoblotting and immunofluorescence assays. Analysis of ATII cell protein profiles will elucidate cellular processes in IAV pathogenesis, which may provide insight into potential therapies to modulate disease severity.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/virología , Regulación hacia Abajo , Virus de la Influenza A/crecimiento & desarrollo , Proteína B Asociada a Surfactante Pulmonar/metabolismo , Animales , Células Cultivadas , Cromatografía Liquida , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Immunoblotting , Espectrometría de Masas , Ratones Endogámicos C57BL , Proteómica
12.
Virus Res ; 175(2): 110-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23639425

RESUMEN

Severe respiratory viral infections are associated with spread to the alveoli of the lungs. There are multiple murine models of severe respiratory viral infections that have been used to identify viral and host factors that contribute to disease severity. Primary cultures of murine alveolar epithelial cells provide a robust in vitro model to perform mechanistic studies that can be correlated with in vivo studies to identify cell type-specific factors that contribute to pathology within the alveoli of the lung during viral infection. In this study, we established an in vitro model to compare the responses of type I (ATI) and type II (ATII) alveolar epithelial cells to infection by respiratory viruses used in murine models: mouse-adapted severe acute respiratory syndrome-associated coronavirus (SARS-CoV, v2163), murine coronavirus MHV-1, and influenza A (H1N1) virus, strain PR8. Murine alveolar cells cultured to maintain an ATII cell phenotype, determined by expression of LBP180, were susceptible to infection by all three viruses. In contrast, ATII cells that were cultured to trans-differentiate into an ATI-like cell phenotype were susceptible to MHV-1 and PR8, but not mouse-adapted SARS-CoV. Epithelial cells produce cytokines in response to viral infections, thereby activating immune responses. Thus, virus-induced cytokine expression was quantified in ATI and ATII cells. Both cell types had increased expression of IL-1ß mRNA upon viral infection, though at different levels. While MHV-1 and PR8 induced expression of a number of shared cytokines in ATI cells, there were several cytokines whose expression was induced uniquely by MHV-1 infection. In summary, ATI and ATII cells exhibited differential susceptibilities and cytokine responses to infection by respiratory viruses. This in vitro model will be critical for future studies to determine the roles of these specialized cell types in the pathogenesis of respiratory viral infection.


Asunto(s)
Diferenciación Celular , Células Epiteliales/fisiología , Células Epiteliales/virología , Subtipo H1N1 del Virus de la Influenza A/fisiología , Virus de la Hepatitis Murina/fisiología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Animales , Células Cultivadas , Citocinas/biosíntesis , Femenino , Expresión Génica , Interacciones Huésped-Patógeno , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Virus de la Hepatitis Murina/crecimiento & desarrollo , Fenotipo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/crecimiento & desarrollo , Tropismo Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA