Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Nature ; 615(7953): 678-686, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36922586

RESUMEN

Dengue is a major health threat and the number of symptomatic infections caused by the four dengue serotypes is estimated to be 96 million1 with annually around 10,000 deaths2. However, no antiviral drugs are available for the treatment or prophylaxis of dengue. We recently described the interaction between non-structural proteins NS3 and NS4B as a promising target for the development of pan-serotype dengue virus (DENV) inhibitors3. Here we present JNJ-1802-a highly potent DENV inhibitor that blocks the NS3-NS4B interaction within the viral replication complex. JNJ-1802 exerts picomolar to low nanomolar in vitro antiviral activity, a high barrier to resistance and potent in vivo efficacy in mice against infection with any of the four DENV serotypes. Finally, we demonstrate that the small-molecule inhibitor JNJ-1802 is highly effective against viral infection with DENV-1 or DENV-2 in non-human primates. JNJ-1802 has successfully completed a phase I first-in-human clinical study in healthy volunteers and was found to be safe and well tolerated4. These findings support the further clinical development of JNJ-1802, a first-in-class antiviral agent against dengue, which is now progressing in clinical studies for the prevention and treatment of dengue.


Asunto(s)
Antivirales , Virus del Dengue , Dengue , Primates , Proteínas no Estructurales Virales , Animales , Humanos , Ratones , Antivirales/efectos adversos , Antivirales/farmacología , Antivirales/uso terapéutico , Ensayos Clínicos Fase I como Asunto , Dengue/tratamiento farmacológico , Dengue/prevención & control , Dengue/virología , Virus del Dengue/clasificación , Virus del Dengue/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Farmacorresistencia Viral , Técnicas In Vitro , Terapia Molecular Dirigida , Primates/virología , Unión Proteica/efectos de los fármacos , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
2.
J Virol ; 97(9): e0066923, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37655939

RESUMEN

Boosting herpes simplex virus (HSV)-specific immunity in the genital tissues of HSV-positive individuals to increase control of HSV-2 recurrent disease and virus shedding is an important goal of therapeutic immunization and would impact HSV-2 transmission. Experimental therapeutic HSV-2 vaccines delivered by a parenteral route have resulted in decreased recurrent disease in experimental animals. We used a guinea pig model of HSV-2 infection to test if HSV-specific antibody and cell-mediated responses in the vaginal mucosa would be more effectively increased by intravaginal (Ivag) therapeutic immunization compared to parenteral immunization. Therapeutic immunization with HSV glycoproteins and CpG adjuvant increased glycoprotein-specific IgG titers in vaginal secretions and serum to comparable levels in Ivag- and intramuscular (IM)-immunized animals. However, the mean numbers of HSV glycoprotein-specific antibody secreting cells (ASCs) and IFN-γ SCs were greater in Ivag-immunized animals demonstrating superior boosting of immunity in the vaginal mucosa compared to parenteral immunization. Therapeutic Ivag immunization also resulted in a significant decrease in the cumulative mean lesion days compared to IM immunization. There was no difference in the incidence or magnitude of HSV-2 shedding in either therapeutic immunization group compared to control-treated animals. Collectively, these data demonstrated that Ivag therapeutic immunization was superior compared to parenteral immunization to boost HSV-2 antigen-specific ASC and IFN-γ SC responses in the vagina and control recurrent HSV-2 disease. These results suggest that novel antigen delivery methods providing controlled release of optimized antigen/adjuvant combinations in the vaginal mucosa would be an effective approach for therapeutic HSV vaccines. IMPORTANCE HSV-2 replicates in skin cells before it infects sensory nerve cells where it establishes a lifelong but mostly silent infection. HSV-2 occasionally reactivates, producing new virus which is released back at the skin surface and may be transmitted to new individuals. Some HSV-specific immune cells reside at the skin site of the HSV-2 infection that can quickly activate and clear new virus. Immunizing people already infected with HSV-2 to boost their skin-resident immune cells and rapidly control the new HSV-2 infection is logical, but we do not know the best way to administer the vaccine to achieve this goal. In this study, a therapeutic vaccine given intravaginally resulted in significantly better protection against HSV-2 disease than immunization with the same vaccine by a conventional route. Immunization by the intravaginal route resulted in greater stimulation of vaginal-resident, virus-specific cells that produced antibody and produced immune molecules to rapidly clear virus.


Asunto(s)
Herpes Genital , Herpes Simple , Herpesvirus Humano 2 , Animales , Femenino , Cobayas , Humanos , Adyuvantes Inmunológicos , Anticuerpos Antivirales , Glicoproteínas/metabolismo , Herpes Genital/prevención & control , Herpes Simple/metabolismo , Herpesvirus Cercopitecino 1 , Herpesvirus Humano 2/fisiología , Inmunización , Linfocitos T , Vagina/inmunología , Vagina/virología
3.
J Virol ; 93(4)2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30463981

RESUMEN

Reactivation of herpes simplex virus 2 (HSV-2) results in infection of epithelial cells at the neuro-epithelial junction and shedding of virus at the epithelial surface. Virus shedding can occur in either the presence or absence of clinical disease and is usually of short duration, although the shedding frequency varies among individuals. The basis for host control of virus shedding is not well understood, although adaptive immune mechanisms are thought to play a central role. To determine the importance of CD4+ T cells in control of HSV-2 shedding, this subset of immune cells was depleted from HSV-2-infected guinea pigs by injection of an anti-CD4 monoclonal antibody (MAb). Guinea pigs were treated with the depleting MAb after establishment of a latent infection, and vaginal swabs were taken daily to monitor shedding by quantitative PCR. The cumulative number of HSV-2 shedding days and the mean number of days virus was shed were significantly increased in CD4-depleted compared to control-treated animals. However, there was no difference in the incidence of recurrent disease between the two treatment groups. Serum antibody levels and the number of HSV-specific antibody-secreting cells in secondary lymphoid tissues were unaffected by depletion of CD4+ T cells; however, the frequency of functional HSV-specific, CD8+ gamma interferon-secreting cells was significantly decreased. Together, these results demonstrate an important role for CD4+ T lymphocytes in control of virus shedding that may be mediated in part by maintenance of HSV-specific CD8+ T cell populations. These results have important implications for development of therapeutic vaccines designed to control HSV-2 shedding.IMPORTANCE Sexual transmission of HSV-2 results from viral shedding following reactivation from latency. The immune cell populations and mechanisms that control HSV-2 shedding are not well understood. This study examined the role of CD4+ T cells in control of virus shedding using a guinea pig model of genital HSV-2 infection that recapitulates the shedding of virus experienced by humans. We found that the frequency of virus-shedding episodes, but not the incidence of clinical disease, was increased by depletion of CD4+ T cells. The HSV-specific antibody response was not diminished, but frequency of functional HSV-reactive CD8+ T cells was significantly diminished by CD4 depletion. These results confirm the role of cell-mediated immunity and highlight the importance of CD4+ T cells in controlling HSV shedding, suggesting that therapeutic vaccines designed to reduce transmission by controlling HSV shedding should include specific enhancement of HSV-specific CD4+ T cell responses.


Asunto(s)
Herpesvirus Humano 2/fisiología , Esparcimiento de Virus/inmunología , Esparcimiento de Virus/fisiología , Animales , Anticuerpos Antivirales/inmunología , Células Productoras de Anticuerpos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Femenino , Cobayas/virología , Herpes Simple/inmunología , Herpesvirus Humano 2/metabolismo , Herpesvirus Humano 2/patogenicidad , Inmunidad Celular/inmunología , Proteínas del Envoltorio Viral/inmunología
4.
J Gen Virol ; 98(10): 2507-2519, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28949904

RESUMEN

The mosquito-borne disease dengue is caused by four serologically and genetically related flaviviruses termed DENV-1 to DENV-4. Dengue is a global public health concern, with both the geographical range and burden of disease increasing rapidly. Clinically, dengue ranges from a relatively mild self-limiting illness to a severe life-threatening and sometimes fatal disease. Infection with one DENV serotype produces life-long homotypic immunity, but incomplete and short-term heterotypic protection. The development of small-animal models that recapitulate the characteristics of the disseminated disease seen clinically has been difficult, slowing the development of vaccines and therapeutics. The AG129 mouse (deficient in interferon alpha/beta and gamma receptor signalling) has proven to be valuable for this purpose, with the development of models of disseminated DENV-2,-3 and -4 disease. Recently, a DENV-1 AG129 model was described, but it requires antibody-dependent enhancement (ADE) to produce lethality. Here we describe a new AG129 model utilizing a non-mouse-adapted DENV-1 strain, West Pacific 74, that does not require ADE to induce lethal disease. Following high-titre intraperitoneal challenge, animals experience a virus infection with dissemination to multiple visceral tissues, including the liver, spleen and intestine. The animals also become thrombocytopenic, but vascular leakage is less prominent than in AG129 models with other DENV serotypes. Taken together, our studies demonstrate that this model is an important addition to dengue research, particularly for understanding the pathological basis of the disease between DENV serotypes and allowing the full spectrum of activity to test comparisons for putative vaccines and antivirals.


Asunto(s)
Virus del Dengue/crecimiento & desarrollo , Dengue/patología , Modelos Animales de Enfermedad , Aedes , Animales , Anticuerpos Antivirales/inmunología , Acrecentamiento Dependiente de Anticuerpo , Línea Celular , Chlorocebus aethiops , Citocinas/biosíntesis , Dengue/virología , Virus del Dengue/clasificación , Recuento de Eritrocitos , Intestinos/patología , Intestinos/virología , Hígado/patología , Hígado/virología , Ratones , Ratones Noqueados , Bazo/patología , Bazo/virología , Trombocitopenia/virología , Células Vero
5.
Antimicrob Agents Chemother ; 60(8): 4600-9, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27185807

RESUMEN

Injury occurring on the surface of the rectal mucosal lining that causes defects in barrier function may result in increased risk for transmission of infection by HIV and other pathogens. Such injury could occur from microbicidal or other topical agents, mechanical trauma during consensual or nonconsensual intercourse, or inflammatory conditions. Tools for evaluation of rectal mucosal barrier function for assessing the mucosa under these conditions are lacking, particularly those that can provide in vivo structural and functional barrier integrity assessment and are adaptable to longitudinal imaging. We investigated confocal endomicroscopy (CE) as a means for in vivo imaging of the rectal epithelial barrier in the ovine model following spatially confined injury to the surface at a controlled site using a topical application of the microbicide test agent benzalkonium chloride. Topical and intravenous (i.v.) fluorescent probes were used with CE to provide subcellular resolution imaging of the mucosal surface and assessment of barrier function loss. A 3-point CE grading system based on cellular structure integrity and leakage of dye through the mucosa showed significant differences in score between untreated (1.19 ± 0.53) and treated (2.55 ± 0.75) tissue (P < 0.0001). Histological grading confirmed findings of barrier compromise. The results indicate that CE is an effective means for detecting epithelial injury and barrier loss following localized trauma in a large-animal model. CE is promising for real-time rectal mucosal evaluation after injury or trauma or topical application of emerging biomedical prevention strategies designed to combat HIV.


Asunto(s)
Infecciones por VIH/prevención & control , Mucosa Intestinal/citología , Microscopía Confocal/métodos , Recto/citología , Animales , Modelos Animales de Enfermedad , Mucosa Intestinal/metabolismo , Ovinos
6.
J Virol ; 89(2): 1254-66, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25392217

RESUMEN

UNLABELLED: The mosquito-borne disease dengue (DEN) is caused by four serologically and genetically related viruses, termed DENV-1 to DENV-4. Infection with one DENV usually leads to acute illness and results in lifelong homotypic immunity, but individuals remain susceptible to infection by the other three DENVs. The lack of a small-animal model that mimics systemic DEN disease without neurovirulence has been an obstacle, but DENV-2 models that resemble human disease have been recently developed in AG129 mice (deficient in interferon alpha/beta and interferon gamma receptor signaling). However, comparable DENV-1, -3, and -4 models have not been developed. We utilized a non-mouse-adapted DENV-3 Thai human isolate to develop a lethal infection model in AG129 mice. Intraperitoneal inoculation of six to eight-week-old animals with strain C0360/94 led to rapid, fatal disease. Lethal C0360/94 infection resulted in physical signs of illness, high viral loads in the spleen, liver, and large intestine, histological changes in the liver and spleen tissues, and increased serum cytokine levels. Importantly, the animals developed vascular leakage, thrombocytopenia, and leukopenia. Overall, we have developed a lethal DENV-3 murine infection model, with no evidence of neurotropic disease based on a non-mouse-adapted human isolate, which can be used to investigate DEN pathogenesis and to evaluate candidate vaccines and antivirals. This suggests that murine models utilizing non-mouse-adapted isolates can be obtained for all four DENVs. IMPORTANCE: Dengue (DEN) is a mosquito-borne disease caused by four DENV serotypes (DENV-1, -2, -3, and -4) that have no treatments or vaccines. Primary infection with one DENV usually leads to acute illness followed by lifelong homotypic immunity, but susceptibility to infection by the other three DENVs remains. Therefore, a vaccine needs to protect from all four DENVs simultaneously. To date a suitable animal model to mimic systemic human illness exists only for DENV-2 in immunocompromised mice using passaged viruses; however, models are still needed for the remaining serotypes. This study describes establishment of a lethal systemic DENV-3 infection model with a human isolate in immunocompromised mice and is the first report of lethal infection by a nonadapted clinical DENV isolate without evidence of neurological disease. Our DENV-3 model provides a relevant platform to test DEN vaccines and antivirals.


Asunto(s)
Virus del Dengue/crecimiento & desarrollo , Dengue/patología , Dengue/virología , Modelos Animales de Enfermedad , Estructuras Animales/patología , Estructuras Animales/virología , Animales , Dengue/inmunología , Virus del Dengue/inmunología , Ratones Noqueados , Receptores de Interferón/deficiencia , Análisis de Supervivencia
7.
J Microsc ; 264(2): 227-237, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27351717

RESUMEN

BACKGROUND: Concern regarding the effect of epithelial damage to the colorectal surface and possible impact on sexually transmitted infection transmission prompts the need for methods to evaluate the mucosal microscopic surface in preclinical studies examining such injury. This includes determining the effect of topical HIV prevention products on mucosal barrier integrity. In vivo imaging with high-resolution endomicroscopy could reveal defects in the mucosal barrier resulting from injury/surface trauma. METHODS: Confocal endomicroscopy was investigated to assess the ability to image surface injury resulting from topical application of a chemical used in lubricants and microbial products. Mice treated with a 50 µL rectal dose of 0.2% benzalkonium chloride solution, 1% benzalkonium chloride or phosphate-buffered saline control for 20 min were imaged in vivo using confocal endomicroscopy for assessment of epithelial disruption. Following imaging, mice were sacrificed and rectal tissue evaluated by histology. Confocal images were graded based on degree of disruption to crypt and epithelial microstructure. Histology was graded based on percent of epithelial disruption observed in stained sections. Confocal image features were confirmed by high-resolution two-photon microscopy. RESULTS: Based on quantitative grading of in vivo confocal endomicroscopy images, disruption at the microscopic scale was observed following treatment with benzalkonium chloride, with increased injury occurring with higher dose. Epithelial disruption at the lumen surface, evident between crypts and alteration in crypt structure on the luminal side were observed in confocal endomicroscopy and confirmed by histology. CONCLUSIONS: High-resolution imaging by confocal endomicroscopy can be used as a noninvasive tool for rapid visual assessment of rectal epithelial integrity following surface injury, potentially providing valuable indication of epithelial injury or trauma.


Asunto(s)
Mucosa Intestinal/patología , Microscopía Confocal/métodos , Recto/patología , Animales , Compuestos de Benzalconio/toxicidad , Femenino , Infecciones por VIH/prevención & control , Mucosa Intestinal/diagnóstico por imagen , Mucosa Intestinal/efectos de los fármacos , Ratones , Recto/diagnóstico por imagen
8.
J Gen Virol ; 96(10): 3035-3048, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26296350

RESUMEN

Dengue is a mosquito-borne disease caused by four related but distinct dengue viruses, DENV-1 to DENV-4. Dengue is endemic in most tropical countries, and over a third of the world's population is at risk of being infected. Although the global burden is high, no vaccine or antiviral is licensed to combat this disease. An obstacle complicating dengue research is the lack of animal challenge models that mimic human disease. Advances in immunocompromised murine infection models resulted in development of lethal DENV-2, DENV-3 and DENV-4 models in AG129 mice, which are deficient in both the IFN-α/ß receptor (IFN-α/ßR) and the IFN-γ receptor (IFN-γR). These models mimic features of dengue disease in humans. Here, we characterized lethal infection of AG129 mice by DENV-4 strain TVP-376 and found that AG129 mice developed clinical signs of illness and high viral loads in multiple tissues and succumbed 5 days after infection. Moreover, the splenic and hepatic histopathology of TVP-376-infected mice demonstrated the presence of cell activation and destruction of tissue architecture. Furthermore, infected mice had heightened levels of circulating cytokines. Comparison of the virulence phenotypes of DENV-4 strain TVP-376 and DENV-2 strain D2S10 revealed that TVP-376-induced mortality occurred in the absence of both IFN-α/ßR and IFN-γR signalling, but not with intact signalling from the IFN-γR, whereas D2S10 required the absence of IFN-α/ßR signalling only, indicating that it is more virulent than TVP-376. In conclusion, TVP-376 is lethal in AG129 mice, and this model provides a useful platform to investigate vaccine candidates and antivirals against DENV-4.


Asunto(s)
Dengue/patología , Dengue/virología , Modelos Animales de Enfermedad , Receptor de Interferón alfa y beta/deficiencia , Receptores de Interferón/deficiencia , Estructuras Animales/virología , Animales , Citocinas/sangre , Dengue/inmunología , Hígado/patología , Ratones de la Cepa 129 , Ratones Noqueados , Bazo/patología , Carga Viral , Receptor de Interferón gamma
9.
J Virol ; 88(7): 3678-94, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24429362

RESUMEN

UNLABELLED: Persistent infection is a key feature of hepatitis C virus (HCV). However, chimpanzee infections with cell culture-derived viruses (JFH1 or related chimeric viruses that replicate efficiently in cell culture) have been limited to acute-transient infections with no pathogenicity. Here, we report persistent infection with chronic hepatitis in a chimpanzee challenged with cell culture-derived genotype 1a virus (H77S.2) containing 6 cell culture-adaptive mutations. Following acute-transient infection with a chimeric H77/JFH1 virus (HJ3-5), intravenous (i.v.) challenge with 10(6) FFU H77S.2 virus resulted in immediate seroconversion and, following an unusual 4- to 6-week delay, persistent viremia accompanied by alanine aminotransferase (ALT) elevation, intrahepatic innate immune responses, and diffuse hepatopathy. This first persistent infection with cell culture-produced HCV provided a unique opportunity to assess evolution of cell culture-adapted virus in vivo. Synonymous and nonsynonymous nucleotide substitution rates were greatest during the first 8 weeks of infection. Of 6 cell culture-adaptive mutations in H77S.2, Q1067R (NS3) had reverted to Q1067 and S2204I (NS5A) was replaced by T2204 within 8 weeks of infection. By 62 weeks, 4 of 6 mutations had reverted to the wild-type sequence, and all reverted to the wild-type sequence by 194 weeks. The data suggest H77S.2 virus has greater potential for persistence and pathogenicity than JFH1 and demonstrate both the capacity of a nonfit virus to persist for weeks in the liver in the absence of detectable viremia as well as strong selective pressure against cell culture-adaptive mutations in vivo. IMPORTANCE: This study shows that mutations promoting the production of infectious genotype 1a HCV in cell culture have the opposite effect and attenuate replication in the liver of the only fully permissive animal species other than humans. It provides the only example to date of persistent infection in a chimpanzee challenged with cell culture-produced virus and provides novel insight into the forces shaping molecular evolution of that virus during 5 years of persistent infection. It demonstrates that a poorly fit virus can replicate for weeks within the liver in the absence of detectable viremia, an observation that expands current concepts of HCV pathogenesis and that is relevant to relapses observed with direct-acting antiviral therapies.


Asunto(s)
Evolución Molecular , Hepacivirus/genética , Hepacivirus/aislamiento & purificación , Hepatitis C Crónica/virología , Mutación , Cultivo de Virus , Alanina Transaminasa/sangre , Animales , Modelos Animales de Enfermedad , Genotipo , Hepacivirus/clasificación , Hígado/patología , Pan troglodytes , Viremia
10.
J Biol Chem ; 288(27): 19370-85, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23640885

RESUMEN

The FGF14 protein controls biophysical properties and subcellular distribution of neuronal voltage-gated Na(+) (Nav) channels through direct binding to the channel C terminus. To gain insights into the dynamic regulation of this protein/protein interaction complex, we employed the split luciferase complementation assay to screen a small molecule library of kinase inhibitors against the FGF14·Nav1.6 channel complex and identified inhibitors of GSK3 as hits. Through a combination of a luminescence-based counter-screening, co-immunoprecipitation, patch clamp electrophysiology, and quantitative confocal immunofluorescence, we demonstrate that inhibition of GSK3 reduces the assembly of the FGF14·Nav channel complex, modifies FGF14-dependent regulation of Na(+) currents, and induces dissociation and subcellular redistribution of the native FGF14·Nav channel complex in hippocampal neurons. These results further emphasize the role of FGF14 as a critical component of the Nav channel macromolecular complex, providing evidence for a novel GSK3-dependent signaling pathway that might control excitability through specific protein/protein interactions.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Hipocampo/metabolismo , Complejos Multiproteicos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Neuronas/metabolismo , Animales , Inhibidores Enzimáticos/farmacología , Factores de Crecimiento de Fibroblastos/genética , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Células HEK293 , Hipocampo/citología , Humanos , Ratones , Ratones Noqueados , Complejos Multiproteicos/genética , Canal de Sodio Activado por Voltaje NAV1.6/genética , Neuronas/citología , Ratas , Transducción de Señal/efectos de los fármacos
11.
Sex Transm Dis ; 40(11): 854-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24113407

RESUMEN

BACKGROUND: Successful development of topical rectal microbicides requires preclinical evaluation in suitable large animal models. Our previous studies have demonstrated the benefits of high-resolution optical coherence tomography (OCT) to visualize subclinical microbicide toxicity in the sheep vagina. In the current study, we evaluated the potential application of colonoscopy and OCT to visualize and quantify the effects of topical products on sheep colorectal tissue, as assessed by advanced imaging techniques. METHODS: Yearling virginal female sheep were treated rectally with a single 8-mL dose of 0.2% benzalkonium chloride (BZK) solution or phosphate-buffered saline control. Imaging was performed before and 30 minutes after treatment. Colonoscopy findings were evaluated based on mucosal disruption. Optical coherence tomography images were graded based on the integrity of the mucosal layer. Biopsies collected after treatment were evaluated by histology for validation of OCT scoring. RESULTS: Mucosal disruption was observed by colonoscopy in BZK-treated animals, whereas none was present in controls. In contrast to colonoscopy, high-resolution in-depth OCT imaging provided visualization of the morphology of the mucosal layer and underlying muscularis, thus enabling detection of microscopic abnormalities. Noninvasive quantification of drug-induced injury after validation of the scoring system (categories 1, 2, 3) showed increased scores after treatment with BZK (P < 0.001), indicating mucosal injury. CONCLUSIONS: High-resolution OCT can be used as highly sensitive tool to evaluate rectal microbicide effects. Because the sheep rectum has both gross and microscopic similarities to the human, this model is a useful addition to current methods of rectal product toxicity.


Asunto(s)
Canal Anal/patología , Antiinfecciosos Locales/farmacología , Compuestos de Benzalconio/farmacología , Mucosa Intestinal/patología , Tomografía de Coherencia Óptica , Animales , Colonoscopía , Modelos Animales de Enfermedad , Femenino , Ovinos , Vagina/patología
12.
Am J Obstet Gynecol ; 208(4): 282.e1-7, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23333551

RESUMEN

OBJECTIVE: High-resolution optical coherence tomography can be used noninvasively to evaluate vaginal morphologic features, including epithelial thickness, to assess this protective barrier in transmission of sexually transmitted infections and to monitor tissue response to topical medications and hormonal fluctuations. We examined the use of optical coherence tomography to measure epithelial thickness noninvasively before and after topical treatment with a drug that causes epithelial thinning. STUDY DESIGN: Twelve female sheep were treated with intravaginal placebo (n = 4) or nonoxynol-9 (n = 8). Vaginal optical coherence tomography images were obtained before and 24 hours after treatment. Four sheep in the nonoxynol-9 group were also examined on days 3 and 7. Vaginal biopsies were obtained on the last examination day. Epithelial thickness was measured in optical coherence tomography images and in hematoxylin and eosin-stained histologic sections from biopsies. Statistical analysis was performed using analyses of variance (significance P < .05). RESULTS: Baseline optical coherence tomography epithelial thickness measurements were similar (85 ± 19 µm placebo, 78 ± 20 µm nonoxynol-9; P = .52). Epithelial thinning was significant after nonoxynol-9 (32 ± 22 µm) compared with placebo (80 ± 15 µm) 24 hours after treatment (P < .0001). In the 4 nonoxynol-9-treated sheep followed for 7 days, epithelial thickness returned to baseline by day 3, and increased significantly on day 7. Epithelial thickness measurements from histology were not significantly different than optical coherence tomography (P = .98 nonoxynol-9, P = .93 hydroxyethyl cellulose). CONCLUSION: Drug-induced changes in the epithelium were clearly detectable using optical coherence tomography imaging. Optical coherence tomography and histology epithelial thickness measurements were similar, validating optical coherence tomography as a noninvasive method for epithelial thickness measurement, providing an important tool for quantitative and longitudinal monitoring of vaginal epithelial changes.


Asunto(s)
Epitelio/efectos de los fármacos , Nonoxinol/farmacología , Espermicidas/farmacología , Vagina/efectos de los fármacos , Administración Intravaginal , Animales , Epitelio/patología , Femenino , Nonoxinol/administración & dosificación , Ovinos , Espermicidas/administración & dosificación , Tomografía de Coherencia Óptica , Vagina/patología
13.
Viruses ; 15(7)2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37515275

RESUMEN

Ebola virus (EBOV) causes Ebola virus disease (EVD), which is characterized by hemorrhagic fever with high mortality rates in humans. EBOV sexual transmission has been a concern since the 2014-2016 outbreak in Africa, as persistent infection in the testis and transmission to women was demonstrated. The only study related to establishing an intravaginal small animal infection model was recently documented in IFNAR-/- mice using wild-type and mouse-adapted EBOV (maEBOV), and resulted in 80% mortality, supporting epidemiological data. However, this route of transmission is still poorly understood in women, and the resulting EVD from it is understudied. Here, we contribute to this field of research by providing data from immunocompetent BALB/c mice. We demonstrate that progesterone priming increased the likelihood of maEBOV vaginal infection and of exhibiting the symptoms of disease and seroconversion. However, our data suggest subclinical infection, regardless of the infective dose. We conclude that maEBOV can infect BALB/c mice through vaginal inoculation, but that this route of infection causes significantly less disease compared to intraperitoneal injection at a similar dose, which is consistent with previous studies using other peripheral routes of inoculation in that animal model. Our data are inconsistent with the disease severity described in female patients, therefore suggesting that BALB/c mice are unsuitable for modeling typical EVD following vaginal challenge with maEBOV. Further studies are required to determine the mechanisms by which EVD is attenuated in BALB/c mice, using maEBOV via the vaginal route, as in our experimental set-up.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Humanos , Masculino , Animales , Femenino , Ratones , Ratones Endogámicos BALB C , Vagina , Modelos Animales
14.
Vaccine ; 2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-38407992

RESUMEN

Chikungunya virus (CHIKV) a mosquito-borne alphavirus is the causative agent of Chikungunya (CHIK), a disease with low mortality but high acute and chronic morbidity resulting in a high overall burden of disease. After the acute disease phase, chronic disease including persistent arthralgia is very common, and can cause fatigue and pain that is severe enough to limit normal activities. On average, around 40% of people infected with CHIKV will develop chronic arthritis, which may last for months or years. Recommendations for protection from CHIKV focus on infection control through preventing mosquito proliferation. There is currently no licensed antiviral drug or vaccine against CHIKV. Therefore, one of the most important public health impacts of vaccination would be to decrease burden of disease and economic losses in areas impacted by the virus, and prevent or reduce chronic morbidity associated with CHIK. This benefit would particularly be seen in Low and Middle Income Countries (LMIC) and socio-economically deprived areas, as they are more likely to have more infections and more severe outcomes. This 'Vaccine Value Profile' (VVP) for CHIK is intended to provide a high-level, holistic assessment of the information and data that are currently available to inform the potential public health, economic and societal value of vaccines in the development pipeline and vaccine-like products.This VVP was developed by a working group of subject matter experts from academia, non-profit organizations, public private partnerships, and multi-lateral organizations. All contributors have extensive expertise on various elements of the CHIK VVP and collectively aimed to identify current research and knowledge gaps.The VVP was developed using only existing and publicly available information.

15.
Sex Transm Dis ; 39(9): 720-5, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22902670

RESUMEN

BACKGROUND: Use of the stimulant methamphetamine (METH) is increasingly common, with >35 million users worldwide. There is a known association between stimulant use and an increased incidence of HIV and other sexually transmitted infections (STIs). METH is known to have immune modulatory properties. However, the impact of METH on normal immune responses and disease pathogenesis with STIs has not been fully examined. METHODS: We used a well-characterized murine model to investigate the impact of METH use on genital herpes simplex virus type 2 infection. Plaque assay and quantitative real-time polymerase chain reaction were used to measure viral replication. Cytokine bead array and enzyme-linked immunosorbent assay were used to determine levels of cytokines during host innate immune response. RESULTS: METH treatment altered behavior, onset of clinical signs, and disease progression. METH-treated mice also had a thinned vaginal epithelium and an increase in virus present in the sensory ganglia. In addition, METH produced a local dysregulation of cytokine secretion that contrasts with its minimal impact on systemic cytokine secretion. CONCLUSIONS: Results suggest that the METH alterations of the host immune response partially contribute to enhanced genital herpes disease progression. These findings will improve understanding of METH use on host immune responses and susceptibility to disease.


Asunto(s)
Citocinas/inmunología , Herpes Genital/inmunología , Herpesvirus Humano 2/inmunología , Metanfetamina/farmacología , Vagina/inmunología , Animales , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Ensayo de Inmunoadsorción Enzimática , Femenino , Herpesvirus Humano 2/patogenicidad , Inmunidad Innata , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Vagina/virología
16.
BMC Infect Dis ; 12: 48, 2012 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-22375797

RESUMEN

BACKGROUND: The development of safe topical microbicides that can preserve the integrity of cervicovaginal tract epithelial barrier is of great interest as this may minimize the potential for increased susceptibility to STI infections. High resolution imaging to assess epithelial integrity in a noninvasive manner could be a valuable tool for preclinical testing of candidate topical agents. METHODS: A quantitative approach using confocal fluorescence microendoscopy (CFM) for assessment of microbicide-induced injury to the vaginal epithelium was developed. Sheep were treated intravaginally with one of five agents in solution (PBS; 0.02% benzalkonium chloride (BZK); 0.2% BZK) or gel formulation (hydroxyethyl cellulose (HEC); Gynol II nonoxynol-9 gel (N-9)). After 24 hours the vaginal tract was removed, labeled with propidium iodide (PI), imaged, then fixed for histology. An automated image scoring algorithm was developed for quantitative assessment of injury and applied to the data set. Image-based findings were validated with histological visual gradings that describe degree of injury and measurement of epithelial thickness. RESULTS: Distinct differences in PI staining were detected following BZK and N-9 treatment. Images from controls had uniformly distributed nuclei with defined borders, while those after BZK or N-9 showed heavily stained and disrupted nuclei, which increased in proportion to injury detected on histology. The confocal scoring system revealed statistically significant scores for each agent versus PBS controls with the exception of HEC and were consistent with histology scores of injury. CONCLUSIONS: Confocal microendoscopy provides a sensitive, objective, and quantitative approach for non-invasive assessment of vaginal epithelial integrity and could serve as a tool for real-time safety evaluation of emerging intravaginal topical agents.


Asunto(s)
Antiinfecciosos/efectos adversos , Endoscopía/métodos , Epitelio/efectos de los fármacos , Epitelio/patología , Vagina/efectos de los fármacos , Vagina/patología , Administración Tópica , Animales , Antiinfecciosos/administración & dosificación , Femenino , Histocitoquímica , Índice de Severidad de la Enfermedad , Ovinos
17.
NPJ Vaccines ; 7(1): 39, 2022 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-35322047

RESUMEN

The envelope (E) protein of flaviviruses is functionally associated with viral tissue tropism and pathogenicity. For yellow fever virus (YFV), viscerotropic disease primarily involving the liver is pathognomonic for wild-type (WT) infection. In contrast, the live-attenuated vaccine (LAV) strain 17D does not cause viscerotropic disease and reversion to virulence is associated with neurotropic disease. The relationship between structure-function of the E protein for WT strain Asibi and its LAV derivative 17D strain is poorly understood; however, changes to WT and vaccine epitopes have been associated with changes in virulence. Here, a panel of Asibi and 17D infectious clone mutants were generated with single-site mutations at the one membrane residue and each of the eight E protein amino acid substitutions that distinguish the two strains. The mutants were characterized with respect to WT-specific and vaccine-specific monoclonal antibodies (mAbs) binding to virus plus binding of virus to brain, liver, and lung membrane receptor preparations (MRPs) generated from AG129 mice. This approach shows that amino acids in the YFV E protein domains (ED) I and II contain the WT E protein epitope, which overlap with those that mediate YFV binding to mouse liver. Furthermore, amino acids in EDIII associated with the vaccine epitope overlap with those that facilitate YFV binding mouse brain MRPs. Taken together, these data suggest that the YFV E protein is a key determinant in the phenotype of WT and 17D vaccine strains of YFV.

18.
NPJ Vaccines ; 7(1): 38, 2022 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-35301331

RESUMEN

Oropouche virus (OROV) is an arthropod-borne orthobunyavirus found in South America and causes Oropouche fever, a febrile infection similar to dengue. It is the second most prevalent arthropod-borne viral disease in South America after dengue. Over 500,000 cases have been diagnosed since the virus was first discovered in 1955; however, this is likely a significant underestimate given the limited availability of diagnostics. No fatalities have been reported to date, however, up to 60% of cases have a recurrent phase of disease within one month of recovery from the primary disease course. The main arthropod vector is the biting midge Culicoides paraensis, which has a geographic range as far north as the United States and demonstrates the potential for OROV to geographically expand. The transmission cycle is incompletely understood and vertebrate hosts include both non-human primates and birds further supporting the potential ability of the virus to spread. A number of candidate antivirals have been evaluated against OROV in vitro but none showed antiviral activity. Surprisingly, there is only one report in the literature on candidate vaccines. We suggest that OROV is an undervalued pathogen much like chikungunya, Schmallenberg, and Zika viruses were before they emerged. Overall, OROV is an important emerging disease that has been under-investigated and has the potential to cause large epidemics in the future. Further research, in particular candidate vaccines, is needed for this important pathogen.

19.
Antimicrob Agents Chemother ; 55(6): 2566-75, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21444700

RESUMEN

PSI-352938 is a novel cyclic phosphate prodrug of ß-D-2'-deoxy-2'-α-fluoro-2'-ß-C-methylguanosine 5'-monophosphate that has potent activity against hepatitis C virus (HCV) in vitro. The studies described here characterize the in vitro anti-HCV activity of PSI-352938, alone and in combination with other inhibitors of HCV, and the cross-resistance profile of PSI-352938. The effective concentration required to achieve 50% inhibition for PSI-352938, determined using genotype 1a-, 1b-, and 2a-derived replicons stably expressed in the Lunet cell line, were 0.20, 0.13, and 0.14 µM, respectively. The active 5'-triphosphate metabolite, PSI-352666, inhibited recombinant NS5B polymerase from genotypes 1 to 4 with comparable 50% inhibitory concentrations. In contrast, PSI-352938 did not inhibit the replication of hepatitis B virus or human immunodeficiency virus in vitro. PSI-352666 did not significantly affect the activity of human DNA and RNA polymerases. PSI-352938 and its cyclic phosphate metabolites did not affect the cyclic GMP-mediated activation of protein kinase G. Clearance studies using replicon cells demonstrated that PSI-352938 cleared cells of HCV replicon RNA and prevented replicon rebound. An additive to synergistic effect was observed when PSI-352938 was combined with other classes of HCV inhibitors, including alpha interferon, ribavirin, NS3/4A inhibitors, an NS5A inhibitor, and nucleoside/nucleotide and nonnucleoside inhibitors. Cross-resistance studies showed that PSI-352938 remained fully active against replicons containing the S282T or the S96T/N142T amino acid alteration. Replicons that contain mutations conferring resistance to various classes of nonnucleoside inhibitors also remained sensitive to inhibition by PSI-352938. PSI-352938 is currently being evaluated in a phase I clinical study in genotype 1-infected individuals.


Asunto(s)
Antivirales/farmacología , Óxidos P-Cíclicos/farmacología , Desoxiguanosina/análogos & derivados , Hepacivirus/efectos de los fármacos , Nucleósidos/farmacología , Profármacos/farmacología , ARN Viral/biosíntesis , Replicón/efectos de los fármacos , Desoxiguanosina/farmacología , Farmacorresistencia Viral , Humanos
20.
Hepatology ; 51(6): 1912-21, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20512985

RESUMEN

UNLABELLED: Silymarin, an extract from milk thistle (Silybum marianum), and its purified flavonolignans have been recently shown to inhibit hepatitis C virus (HCV) infection, both in vitro and in vivo. In the current study, we further characterized silymarin's antiviral actions. Silymarin had antiviral effects against hepatitis C virus cell culture (HCVcc) infection that included inhibition of virus entry, RNA and protein expression, and infectious virus production. Silymarin did not block HCVcc binding to cells but inhibited the entry of several viral pseudoparticles (pp), and fusion of HCVpp with liposomes. Silymarin but not silibinin inhibited genotype 2a NS5B RNA-dependent RNA polymerase (RdRp) activity at concentrations 5 to 10 times higher than required for anti-HCVcc effects. Furthermore, silymarin had inefficient activity on the genotype 1b BK and four 1b RDRPs derived from HCV-infected patients. Moreover, silymarin did not inhibit HCV replication in five independent genotype 1a, 1b, and 2a replicon cell lines that did not produce infectious virus. Silymarin inhibited microsomal triglyceride transfer protein activity, apolipoprotein B secretion, and infectious virion production into culture supernatants. Silymarin also blocked cell-to-cell spread of virus. CONCLUSION: Although inhibition of in vitro NS5B polymerase activity is demonstrable, the mechanisms of silymarin's antiviral action appear to include blocking of virus entry and transmission, possibly by targeting the host cell.


Asunto(s)
Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Silimarina/farmacología , Línea Celular Tumoral , Humanos , Proteínas no Estructurales Virales/antagonistas & inhibidores , Internalización del Virus/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda