Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Vis Exp ; (197)2023 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-37522727

RESUMO

The use of viral vectors to treat genetic diseases has increased substantially in recent years, with over 2,000 studies registered to date. Adeno-associated viral (AAV) vectors have found particular success in the treatment of eye related diseases, as exemplified by the approval of voretigene neparvovec-rzyl. To bring new therapies to market, regulatory agencies typically request qualified or validated bioshedding studies to evaluate release of the vector into the environment. However, no official guidelines for the development of molecular based assays to support such shedding studies have been released by the United States Food and Drug Administration, leaving developers to determine best practices for themselves. The purpose of this protocol is to present a validatable protocol for the detection of AAV vectors in human tears by droplet digital polymerase chain reaction (ddPCR) in support of clinical bioshedding studies. This manuscript discusses current industry approaches to molecular assay validation and demonstrates that the method exceeds the target assay acceptance criteria currently proposed in white papers. Finally, steps critical in the performance of any ddPCR assay, regardless of application, are discussed.


Assuntos
Dependovirus , Oftalmopatias , Humanos , Dependovirus/genética , Reação em Cadeia da Polimerase/métodos , Terapia Genética/métodos , Vetores Genéticos/genética , Reação em Cadeia da Polimerase em Tempo Real/métodos
2.
Microorganisms ; 7(9)2019 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-31540200

RESUMO

Human adenovirus infection of the ocular surface is associated with severe keratoconjunctivitis and the formation of subepithelial corneal infiltrates, which may persist and impair vision for months to years following infection. Long term pathology persists well beyond the resolution of viral replication, indicating that the prolonged immune response is not virus-mediated. However, it is not clear how these responses are sustained or even initiated following infection. This review discusses recent work from our laboratory and others which demonstrates different entry pathways specific to both adenovirus and cell type. These findings suggest that adenoviruses may stimulate specific pattern recognition receptors in an entry/trafficking-dependent manner, leading to distinct immune responses dependent on the virus/cell type combination. Additional work is needed to understand the specific connections between adenoviral entry and the stimulation of innate immune responses by the various cell types present on the ocular surface.

3.
Am J Vet Res ; 80(5): 490-497, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31034270

RESUMO

OBJECTIVE: To determine the effects of orally administered raltegravir in cats with experimentally induced ocular and respiratory feline herpesvirus-1 (FHV-1) infection. ANIMALS: 14 healthy 6-month-old unvaccinated specific pathogen-free cats. PROCEDURES: On day 0, all cats were experimentally inoculated by topical application of 0.1 mL of a solution containing 106 plaque-forming units of FHV-1 strain FH2CS to the inferior conjunctival fornix of each eye. Cats were randomly assigned to receive either raltegravir (80 mg; n = 7) or lactose (250 mg; vehicle; 7), PO, every 12 hours for 14 days beginning on day 1. Cats were assigned clinical ocular and respiratory disease scores every other day from days 0 to 30. Conjunctival swab specimens were collected for detection of FHV-1 by virus isolation and real-time PCR assay at 3-day intervals from days 0 to 30. Confocal microscopy was performed on days 0 and 10 to assess corneal epithelial leukocyte infiltration. The assessed variables and duration of FHV-1 shedding were compared between the 2 treatment groups. RESULTS: Cats in both groups developed moderate to severe conjunctivitis and ulcerative keratitis characteristic of FHV-1 infection. Median duration of FHV-1 shedding was shorter and signs of ocular and respiratory disease were less severe for raltegravir-treated cats than for vehicle-treated cats. However, the mean conjunctival FHV-1 titer and corneal epithelial leukocyte count did not differ between the 2 groups. CONCLUSIONS AND CLINICAL RELEVANCE: Results suggested orally administered raltegravir might be effective for alleviation of ocular and respiratory signs of FHV-1 infection in cats. (Am J Vet Res 2019;80:490-497).


Assuntos
Antivirais/uso terapêutico , Doenças do Gato/tratamento farmacológico , Conjuntivite Viral/veterinária , Infecções por Herpesviridae/veterinária , Raltegravir Potássico/uso terapêutico , Infecções Respiratórias/veterinária , Varicellovirus , Animais , Doenças do Gato/virologia , Gatos , Conjuntivite Viral/tratamento farmacológico , Infecções por Herpesviridae/tratamento farmacológico , Distribuição Aleatória , Infecções Respiratórias/tratamento farmacológico , Infecções Respiratórias/virologia , Método Simples-Cego , Organismos Livres de Patógenos Específicos
4.
J Virol ; 92(20)2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30045987

RESUMO

Alphaherpesvirus-associated ocular infections in humans caused by human alphaherpesvirus 1 (HHV-1) remain challenging to treat due to the frequency of drug application required and the potential for the selection of drug-resistant viruses. Repurposing on-the-market drugs is a viable strategy to accelerate the pace of drug development. It has been reported that the human immunodeficiency virus (HIV) integrase inhibitor raltegravir inhibits HHV-1 replication by targeting the DNA polymerase accessory factor and limits terminase-mediated genome cleavage of human betaherpesvirus 5 (HHV-5). We have previously shown, both in vitro and in vivo, that raltegravir can also inhibit the replication of felid alphaherpesvirus 1 (FeHV-1), a common ocular pathogen of cats with a pathogenesis similar to that of HHV-1 ocular disease. In contrast to what was reported for HHV-1, we were unable to select for a raltegravir-resistant FeHV-1 strain in order to define any basis for drug action. A candidate-based approach to explore the mode of action of raltegravir against FeHV-1 showed that raltegravir did not impact FeHV-1 terminase function, as described for HHV-5. Instead, raltegravir inhibited DNA replication, similarly to HHV-1, but by targeting the initiation of viral DNA replication rather than elongation. In addition, we found that raltegravir specifically repressed late gene expression independently of DNA replication, and both activities are consistent with inhibition of ICP8. Taken together, these results suggest that raltegravir could be a valuable therapeutic agent against herpesviruses.IMPORTANCE The rise of drug-resistant herpesviruses is a longstanding concern, particularly among immunocompromised patients. Therefore, therapies targeting viral proteins other than the DNA polymerase that may be less likely to lead to drug-resistant viruses are urgently needed. Using FeHV-1, an alphaherpesvirus closely related to HHV-1 that similarly causes ocular herpes in its natural host, we found that the HIV integrase inhibitor raltegravir targets different stages of the virus life cycle beyond DNA replication and that it does so without developing drug resistance under the conditions tested. This shows that the drug could provide a viable strategy for the treatment of herpesvirus infections.


Assuntos
Inibidores de Integrase de HIV/farmacologia , Raltegravir Potássico/farmacologia , Varicellovirus/fisiologia , Replicação Viral/efeitos dos fármacos , Animais , Gatos , Linhagem Celular , DNA Viral/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Varicellovirus/efeitos dos fármacos , Proteínas Virais/metabolismo
5.
J Gen Virol ; 99(8): 1115-1128, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29916804

RESUMO

Anti-microbial compounds typically exert their action by directly interfering with one or more stages of the pathogen's life cycle. However, some compounds also have secondary effects on the host that aid in pathogen clearance. Raltegravir is a human immunodeficiency virus (HIV)-integrase inhibitor that has been shown to alter the host immune response to HIV in addition to its direct antiviral effect. Interestingly, raltegravir can also directly inhibit the replication of various herpesviruses. However, the host-targeted effects of this drug in the context of a herpesvirus infection have not been explored. Here, we used felid alphaherpesvirus 1 (FHV-1), a close relative of human alphaherpesvirus 1 (HHV-1) that similarly causes ocular herpes, to characterize the host-targeted effects of raltegravir on corneal epithelial cells during an alphaherpesvirus infection. Using RNA deep sequencing, we found that raltegravir specifically boosts the expression of anti-angiogenic factors and promotes metabolic homeostasis in FHV-1-infected cells. In contrast, few changes in host gene transcription were found in uninfected cells. Importantly, we were able to demonstrate that these effects were specific to raltegravir and independent of the direct-acting antiviral effect of the drug, since treatment with the DNA polymerase inhibitor phosphonoacetic acid did not induce these host-targeted effects. Taken together, these results indicate that raltegravir has profound and specific effects on the host transcription profile of herpesvirus-infected cells that may contribute to the overall antiviral activity of the drug and could provide therapeutic benefits in vivo. Furthermore, this study provides a framework for future efforts evaluating the host-targeted effects of anti-microbial compounds.


Assuntos
Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Inibidores de Integrase de HIV/farmacologia , Raltegravir Potássico/farmacologia , Transcriptoma/efeitos dos fármacos , Varicellovirus/efeitos dos fármacos , Animais , Gatos , Células Cultivadas , Epitélio Corneano/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Amplificação de Ácido Nucleico , Reprodutibilidade dos Testes , Replicação de Sequência Autossustentável , Organismos Livres de Patógenos Específicos , Varicellovirus/fisiologia
6.
Am J Vet Res ; 79(7): 762-769, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29943639

RESUMO

OBJECTIVE To determine the in vitro half maximal effective concentration (EC50) of ganciclovir for canine herpesvirus-1 (CHV-1) and to evaluate the efficacy of ganciclovir ophthalmic gel in dogs with experimentally induced ocular CHV-1 infection. ANIMALS 10 specific pathogen-free adult Beagles. PROCEDURES Cytotoxicity and EC50 of ganciclovir for CHV-1 were determined during in vitro experiments. During an in vivo experiment, dogs with experimentally induced ocular CHV-1 infections received 1 drop of 0.15% ganciclovir (ganciclovir group; n = 5) or artificial tear (control group; 5) ophthalmic gel in both eyes 5 times daily for 7 days, then 3 times daily for 7 days. For each dog, ophthalmic and confocal microscopic examinations were performed at predetermined times to determine severity of ocular disease and inflammation. Conjunctival swab specimens were collected at predetermined times for PCR assay analysis to determine CHV-1 shedding. RESULTS No in vitro cytotoxic effects were observed for ganciclovir concentrations ≤ 500µM. The EC50 of ganciclovir for CHV-1 was 37.7µM. No adverse effects associated with ganciclovir were observed during the in vivo experiment. Mean ocular disease and inflammation scores for the ganciclovir group were significantly lower than those for the control group. Mean duration of CHV-1 shedding for the ganciclovir group (0.4 days) was significantly shorter than that for the control group (6.2 days). CONCLUSIONS AND CLINICAL RELEVANCE Topical administration of 0.15% ganciclovir ophthalmic gel was well tolerated and effective in decreasing clinical disease scores, ocular tissue inflammation, and duration of viral shedding in dogs with experimentally induced ocular CHV-1 infection.


Assuntos
Administração Tópica , Antivirais/administração & dosagem , Doenças do Cão/tratamento farmacológico , Infecções Oculares Virais/veterinária , Ganciclovir/administração & dosagem , Infecções por Herpesviridae/veterinária , Animais , Doenças do Cão/virologia , Cães , Relação Dose-Resposta a Droga , Olho , Infecções Oculares Virais/tratamento farmacológico , Infecções por Herpesviridae/tratamento farmacológico , Herpesvirus Canídeo 1 , Células Madin Darby de Rim Canino , Microscopia Confocal , Reação em Cadeia da Polimerase em Tempo Real , Eliminação de Partículas Virais
7.
Viruses ; 9(11)2017 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-29156583

RESUMO

Ocular herpesviruses, most notably human alphaherpesvirus 1 (HSV-1), canid alphaherpesvirus 1 (CHV-1) and felid alphaherpesvirus 1 (FHV-1), infect and cause severe disease that may lead to blindness. CHV-1 and FHV-1 have a pathogenesis and induce clinical disease in their hosts that is similar to HSV-1 ocular infections in humans, suggesting that infection of dogs and cats with CHV-1 and FHV-1, respectively, can be used as a comparative natural host model of herpesvirus-induced ocular disease. In this review, we discuss both strengths and limitations of the various available model systems to study ocular herpesvirus infection, with a focus on the use of these non-traditional virus-natural host models. Recent work has demonstrated the robustness and reproducibility of experimental ocular herpesvirus infections in dogs and cats, and, therefore, these non-traditional models can provide additional insights into the pathogenesis of ocular herpesvirus infections.


Assuntos
Modelos Animais de Doenças , Doenças do Cão/virologia , Oftalmopatias/virologia , Infecções por Herpesviridae/fisiopatologia , Modelos Biológicos , Alphaherpesvirinae/patogenicidade , Animais , Gatos , Cães , Oftalmopatias/fisiopatologia , Infecções por Herpesviridae/virologia , Herpesvirus Canídeo 1/isolamento & purificação , Herpesvirus Canídeo 1/patogenicidade , Herpesvirus Canídeo 1/fisiologia
8.
Virus Res ; 242: 30-36, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28870469

RESUMO

Horses commonly develop gastric mucosal ulcers, similar to humans, a condition known as equine gastric ulcer syndrome (EGUS) that can lead to poor performance and lost training time and care expenses. Unlike humans, however, an infectious bacterial cause of ulcers has not been conclusively identified. Herpesviruses, while well-established causative agents of diseases such as cold sores, genital lesions, and certain types of cancer, have also been implicated in the development of a subset of gastric ulcers in humans. The presence of equid herpesviruses in the gastrointestinal tract and their potential contribution to EGUS has not been evaluated. Here, we provide the first evidence of equid gammaherpesviruses 2 and 5 (EHV-2 and -5) within the epithelium of the gastric mucosa of horses. These viruses were initially detected by a nested PCR screen of gastric tissue samples obtained from client- and university-owned horses with and without ulcers; however, no association with EGUS was found in this limited sample set. We then validated a highly sensitive in situ hybridization (ISH) assay and used this assay to characterize the distribution of these viruses in necropsy gastric tissue samples from five racehorses. Analyses revealed frequent EHV-2 and EHV-5 co-infections within the gastric mucosal epithelium, regardless of the ulcer status. These results are the first to demonstrate the presence of equid gammaherpesviruses in the gastric mucosa of horses and warrants further investigation to determine the contribution of these viruses to the development of EGUS and/or other gastrointestinal diseases.


Assuntos
Epitélio/virologia , Gammaherpesvirinae/isolamento & purificação , Mucosa Gástrica/virologia , Infecções por Herpesviridae/veterinária , Doenças dos Cavalos/virologia , Úlcera Gástrica/veterinária , Animais , Coinfecção/veterinária , Coinfecção/virologia , Gammaherpesvirinae/classificação , Gammaherpesvirinae/genética , Infecções por Herpesviridae/virologia , Cavalos , Hibridização de Ácido Nucleico , Reação em Cadeia da Polimerase , Úlcera Gástrica/virologia
9.
mSphere ; 2(2)2017.
Artigo em Inglês | MEDLINE | ID: mdl-28405631

RESUMO

Electric cell-substrate impedance sensing (ECIS) measures changes in an electrical circuit formed in a culture dish. As cells grow over a gold electrode, they block the flow of electricity and this is read as an increase in electrical impedance in the circuit. ECIS has previously been used in a variety of applications to study cell growth, migration, and behavior in response to stimuli in real time and without the need for cellular labels. Here, we demonstrate that ECIS is also a valuable tool with which to study infection by alphaherpesviruses. To this end, we used ECIS to study the kinetics of cells infected with felid herpesvirus type 1 (FHV-1), a close relative of the human alphaherpesviruses herpes simplex virus 1 (HSV-1) and HSV-2, and compared the results to those obtained with conventional infectivity assays. First, we demonstrated that ECIS can easily distinguish between wells of cells infected with different amounts of FHV-1 and provides information about the cellular response to infection. Second, we found ECIS useful in identifying differences between the replication kinetics of recombinant DsRed Express2-labeled FHV-1, created via CRISPR/Cas9 genome engineering, and wild-type FHV-1. Finally, we demonstrated that ECIS can accurately determine the half-maximal effective concentration of antivirals. Collectively, our data show that ECIS, in conjunction with current methodologies, is a powerful tool that can be used to monitor viral growth and study the cellular response to alphaherpesvirus infection. IMPORTANCE Alphaherpesviruses, including those that commonly infect humans, such as HSV-1 and HSV-2, typically infect and cause cellular damage to epithelial cells at mucosal surfaces, leading to disease. The development of novel technologies to study the cellular responses to infection may allow a more complete understanding of virus replication and the creation of novel antiviral therapies. This study demonstrates the use of ECIS to study various aspects of herpesvirus biology, with a specific focus on changes in cellular morphology as a result of infection. We conclude that ECIS represents a valuable new tool with which to study alphaherpesvirus infections in real time and in an objective and reproducible manner.

10.
Phys Rev Lett ; 117(5): 058101, 2016 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-27517794

RESUMO

We study the phase separation of binary lipid mixtures that form bicontinuous cubic phases. The competition between the nonuniform Gaussian membrane curvature and line tension leads to a very rich phase diagram, where we observe symmetry breaking of the membrane morphologies and reentrant phenomena due to the formation of bridges between segregated domains. Upon increasing the line tension contribution, we also find faceting of lipid domains that we explain using a simple argument based on the symmetry of the underlying surface and topology.

11.
Tissue Eng Part C Methods ; 22(6): 596-607, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27113698

RESUMO

Adult equine hepatocytes have proven challenging to culture long term in vitro as they rapidly lose their morphology and functionality, thus limiting studies on liver function and response to disease. In this study, we describe for the first time the differentiation of equine mesenchymal stromal cells (MSC) from a variety of sources into functional hepatocyte-like cells (HLC). First, we differentiated equine umbilical cord blood (UCB)-derived MSC into HLC and found that these cells exhibited a distinct polygonal morphology, stored glycogen as visualized by periodic acid Schiff's reagent staining, and were positive for albumin and other hepatocyte-specific genes. Second, we demonstrated that UCB-HLC could be revived following cryopreservation and retained their phenotype for at least 10 days. Third, we differentiated three sets of MSC from bone marrow (BM), adipose tissue (AT), and peripheral blood (PB), matched within the same horse. We achieved a 100% differentiation success rate with BM, 0% with AT, and 66% with PB. An additional set of nine PB-MSC samples resulted in an overall success rate of 42% (n = 12), and age or gender did not seem to have an effect on the success of hepatic differentiation from that source. In a final set of experiments, we evaluated the use of these HLC as tools in different fields of biomedical research like virology, to study viral growth, and toxicology, to study chemicals with hepatic toxicity. Equine HLC were found susceptible for infection with the equine herpesviruses type 1 (EHV-1), -2, and -5, and exhibited a more sensitive dose-dependent response to arsenic toxicity than the commonly used human hepatocellular cell line HepG2. Taken together, these data indicate that equine MSC can be efficiently differentiated into HLC and these equine HLC could be a useful tool for in vitro studies.


Assuntos
Tecido Adiposo/citologia , Células da Medula Óssea/citologia , Diferenciação Celular , Sangue Fetal/citologia , Hepatócitos/citologia , Células-Tronco Mesenquimais/citologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Feminino , Células Hep G2 , Cavalos , Humanos , Masculino , Fenótipo
12.
J Gen Virol ; 97(6): 1414-1425, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26959283

RESUMO

Feline herpesvirus type-1 (FHV-1) is the most common viral cause of ocular surface disease in cats. Many antiviral drugs are used to treat FHV-1, but require frequent topical application and most lack well-controlled in vivo studies to justify their clinical use. Therefore, better validation of current and novel treatment options are urgently needed. Here, we report on the development of a feline whole corneal explant model that supports FHV-1 replication and thus can be used as a novel model system to evaluate the efficacy of antiviral drugs. The anti-herpes nucleoside analogues cidofovir and acyclovir, which are used clinically to treat ocular herpesvirus infection in cats and have previously been evaluated in traditional two-dimensional feline cell cultures in vitro, were evaluated in this explant model. Both drugs suppressed FHV-1 replication when given every 12 h, with cidofovir showing greater efficacy. In addition, the potential efficacy of the retroviral integrase inhibitor raltegravir against FHV-1 was evaluated in cell culture as well as in the explant model. Raltegravir was not toxic to feline cells or corneas, and most significantly, inhibited FHV-1 replication at 500 µM in both systems. Importantly, this drug was effective when given only once every 24 h. Taken together, our data indicate that the feline whole corneal explant model is a useful tool for the evaluation of antiviral drugs and, furthermore, that raltegravir appears a promising novel antiviral drug to treat ocular herpesvirus infection in cats.


Assuntos
Antivirais/farmacologia , Córnea/virologia , Avaliação Pré-Clínica de Medicamentos/métodos , Técnicas de Cultura de Órgãos/métodos , Varicellovirus/efeitos dos fármacos , Cultura de Vírus/métodos , Aciclovir/farmacologia , Animais , Gatos , Cidofovir , Citosina/análogos & derivados , Citosina/farmacologia , Organofosfonatos/farmacologia
13.
J Ocul Pharmacol Ther ; 31(10): 642-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26418618

RESUMO

PURPOSE: The effects of cidofovir were investigated against canine herpesvirus-1 (CHV-1) in vitro and in dogs with experimentally induced recurrent ocular CHV-1 infection, a host-adapted pathogen animal model of ocular herpes simplex virus-1 (HSV-1) infection. METHODS: The cidofovir EC50 was determined for CHV-1 and HSV-1. A randomized, masked vehicle-controlled trial was performed using beagles with experimentally induced recurrent ocular CHV-1 infection. Dogs received 1 drop of 0.5% cidofovir solution or 0.9% sodium chloride solution (vehicle) in both eyes 2 times daily for 14 days. Dogs were monitored at intervals for 30 days by a clinical ophthalmic examination, in vivo confocal microscopy of the cornea and conjunctiva, ocular sample CHV-1 polymerase chain reaction assay, hemogram, and serum biochemistry panel. Clinical ocular disease scores were calculated and infiltrating leukocytes detected by in vivo confocal microscopy quantified. RESULTS: Cidofovir displayed similar in vitro antiviral activity against CHV-1 and HSV-1. Clinical ocular disease scores were significantly higher in the cidofovir group compared to the vehicle group. Marked conjunctival pigmentation and blepharitis were also detected in the cidofovir group, but not the vehicle group. Conjunctival and corneal leukocyte infiltration scores determined by in vivo confocal microscopy were significantly higher in the cidofovir group. Dogs administered cidofovir had significantly reduced durations of ocular viral shedding compared to the vehicle group. Hemogram and serum biochemistry panel values were unremarkable. CONCLUSIONS: Twice-daily application of topical 0.5% cidofovir ophthalmic solution reduced the duration of ocular viral shedding in dogs with experimentally induced recurrent ocular CHV-1 infection, but was associated with local ocular toxicity.


Assuntos
Citosina/análogos & derivados , Doenças do Cão/virologia , Infecções por Herpesviridae/tratamento farmacológico , Herpesvirus Canídeo 1/efeitos dos fármacos , Organofosfonatos/farmacologia , Administração Oftálmica , Animais , Antivirais/administração & dosagem , Antivirais/farmacologia , Antivirais/toxicidade , Chlorocebus aethiops , Cidofovir , Citosina/administração & dosagem , Citosina/farmacologia , Citosina/toxicidade , Modelos Animais de Doenças , Cães , Infecções por Herpesviridae/virologia , Células Madin Darby de Rim Canino , Microscopia Confocal , Organofosfonatos/administração & dosagem , Organofosfonatos/toxicidade , Distribuição Aleatória , Células Vero , Eliminação de Partículas Virais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...