Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Infect Dis ; 213(5): 703-11, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26582961

RESUMEN

Animal models recapitulating human Ebola virus disease (EVD) are critical for insights into virus pathogenesis. Ebola virus (EBOV) isolates derived directly from human specimens do not, without adaptation, cause disease in immunocompetent adult rodents. Here, we describe EVD in mice engrafted with human immune cells (hu-BLT). hu-BLT mice developed EVD following wild-type EBOV infection. Infection with high-dose EBOV resulted in rapid, lethal EVD with high viral loads, alterations in key human antiviral immune cytokines and chemokines, and severe histopathologic findings similar to those shown in the limited human postmortem data available. A dose- and donor-dependent clinical course was observed in hu-BLT mice infected with lower doses of either Mayinga (1976) or Makona (2014) isolates derived from human EBOV cases. Engraftment of the human cellular immune system appeared to be essential for the observed virulence, as nonengrafted mice did not support productive EBOV replication or develop lethal disease. hu-BLT mice offer a unique model for investigating the human immune response in EVD and an alternative animal model for EVD pathogenesis studies and therapeutic screening.


Asunto(s)
Ebolavirus/fisiología , Regulación de la Expresión Génica/inmunología , Fiebre Hemorrágica Ebola/inmunología , Animales , Encéfalo/virología , Citocinas/genética , Citocinas/metabolismo , Fiebre Hemorrágica Ebola/orina , Fiebre Hemorrágica Ebola/virología , Humanos , Riñón/virología , Hígado/virología , Pulmón/virología , Masculino , Ratones , Ratones Transgénicos , ARN Viral/aislamiento & purificación , Bazo/virología , Testículo/virología , Replicación Viral
2.
Emerg Infect Dis ; 21(10): 1816-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26401603

RESUMEN

To determine the utility of oral swabs for diagnosing infection with Ebola virus, we used a guinea pig model and obtained daily antemortem and postmortem swab samples. According to quantitative reverse transcription PCR analysis, the diagnostic value was poor for antemortem swab samples but excellent for postmortem samples.


Asunto(s)
Diagnóstico Precoz , Ebolavirus , Fiebre Hemorrágica Ebola/diagnóstico , Boca/virología , Manejo de Especímenes/métodos , Animales , Cobayas , Fiebre Hemorrágica Ebola/virología , Humanos
3.
Sci Rep ; 12(1): 9403, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672343

RESUMEN

Human rabies remains a globally significant public health problem. Replacement of polyclonal anti-rabies immunoglobulin (RIG), a passive component of rabies post-exposure prophylaxis (PEP), with a monoclonal antibody (MAb), would eliminate the cost and availability constraints associated with RIG. Our team has developed and licensed a human monoclonal antibody RAB1 (Rabishield©), as the replacement for RIG where canine rabies is enzootic. However, for the highly diverse rabies viruses of North America, a cocktail containing two or more MAbs targeting different antigenic sites of the rabies glycoprotein should be included to ensure neutralization of all variants of the virus. In this study, two MAb cocktails, R172 (RAB1-RAB2) and R173 (RAB1-CR57), were identified and evaluated against a broad range of rabies variants from North America. R173 was found to be the most potent cocktail, as it neutralized all the tested North American RABV isolates and demonstrated broad coverage of isolates from both terrestrial and bat species. R173 could be a promising candidate as an alternative or replacement for RIG PEP in North America.


Asunto(s)
Antineoplásicos Inmunológicos , Vacunas Antirrábicas , Virus de la Rabia , Rabia , Animales , Anticuerpos Monoclonales , Anticuerpos Antivirales , Perros , Humanos , Profilaxis Posexposición
4.
mSphere ; 6(1)2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33536322

RESUMEN

Smallpox, caused by Variola virus (VARV), was eradicated in 1980; however, VARV bioterrorist threats still exist, necessitating readily available therapeutics. Current preparedness activities recognize the importance of oral antivirals and recommend therapeutics with different mechanisms of action. Monkeypox virus (MPXV) is closely related to VARV, causing a highly similar clinical human disease, and can be used as a surrogate for smallpox antiviral testing. The prairie dog MPXV model has been characterized and used to study the efficacy of antipoxvirus therapeutics, including recently approved TPOXX (tecovirimat). Brincidofovir (BCV; CMX001) has shown antiviral activity against double-stranded DNA viruses, including poxviruses. To determine the exposure of BCV following oral administration to prairie dogs, a pharmacokinetics (PK) study was performed. Analysis of BCV plasma concentrations indicated variability, conceivably due to the outbred nature of the animals. To determine BCV efficacy in the MPXV prairie dog model, groups of animals were intranasally challenged with 9 × 105 plaque-forming units (PFU; 90% lethal dose [LD90]) of MPXV on inoculation day 0 (ID0). Animals were divided into groups based on the first day of BCV treatment relative to inoculation day (ID-1, ID0, or ID1). A trend in efficacy was noted dependent upon treatment initiation (57% on ID-1, 43% on ID0, and 29% on ID1) but was lower than demonstrated in other animal models. Analysis of the PK data indicated that BCV plasma exposure (maximum concentration [Cmax]) and the time of the last quantifiable concentration (AUClast) were lower than in other animal models administered the same doses, indicating that suboptimal BCV exposure may explain the lower protective effect on survival.IMPORTANCE Preparedness activities against highly transmissible viruses with high mortality rates have been highlighted during the ongoing coronavirus disease 2019 (COVID-19) pandemic. Smallpox, caused by variola virus (VARV) infection, is highly transmissible, with an estimated 30% mortality. Through an intensive vaccination campaign, smallpox was declared eradicated in 1980, and routine smallpox vaccination of individuals ceased. Today's current population has little/no immunity against VARV. If smallpox were to reemerge, the worldwide results would be devastating. Recent FDA approval of one smallpox antiviral (tecovirimat) was a successful step in biothreat preparedness; however, orthopoxviruses can become resistant to treatment, suggesting the need for multiple therapeutics. Our paper details the efficacy of the investigational smallpox drug brincidofovir in a monkeypox virus (MPXV) animal model. Since brincidofovir has not been tested in vivo against smallpox, studies with the related virus MPXV are critical in understanding whether it would be protective in the event of a smallpox outbreak.


Asunto(s)
Citosina/análogos & derivados , Monkeypox virus/efectos de los fármacos , Organofosfonatos/farmacología , Organofosfonatos/farmacocinética , Viruela/tratamiento farmacológico , Animales , Antivirales/farmacocinética , Antivirales/farmacología , Benzamidas/farmacocinética , Benzamidas/farmacología , Citosina/farmacocinética , Citosina/farmacología , Modelos Animales de Enfermedad , Perros , Femenino , Isoindoles/farmacocinética , Isoindoles/farmacología , Masculino , Virus de la Viruela/efectos de los fármacos
5.
Viruses ; 12(2)2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-32033253

RESUMEN

Currently, no rabies virus-specific antiviral drugs are available. Ranpirnase has strong antitumor and antiviral properties associated with its ribonuclease activity. TMR-001, a proprietary bulk drug substance solution of ranpirnase, was evaluated against rabies virus in three cell types: mouse neuroblastoma, BSR (baby hamster kidney cells), and bat primary fibroblast cells. When TMR-001 was added to cell monolayers 24 h preinfection, rabies virus release was inhibited for all cell types at three time points postinfection. TMR-001 treatment simultaneous with infection and 24 h postinfection effectively inhibited rabies virus release in the supernatant and cell-to-cell spread with 50% inhibitory concentrations of 0.2-2 nM and 20-600 nM, respectively. TMR-001 was administered at 0.1 mg/kg via intraperitoneal, intramuscular, or intravenous routes to Syrian hamsters beginning 24 h before a lethal rabies virus challenge and continuing once per day for up to 10 days. TMR-001 at this dose, formulation, and route of delivery did not prevent rabies virus transit from the periphery to the central nervous system in this model (n = 32). Further aspects of local controlled delivery of other active formulations or dose concentrations of TMR-001 or ribonuclease analogues should be investigated for this class of drugs as a rabies antiviral therapeutic.


Asunto(s)
Antivirales/farmacología , Virus de la Rabia/efectos de los fármacos , Ribonucleasas/farmacología , Liberación del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Línea Celular , Células Cultivadas , Quirópteros , Cricetinae , Femenino , Fibroblastos/virología , Mesocricetus , Ratones , Rabia/prevención & control , Virus de la Rabia/fisiología , Ribonucleasas/administración & dosificación
6.
Vaccines (Basel) ; 7(3)2019 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-31349649

RESUMEN

Rabies is preventable through vaccination, but the need to mount annual canine vaccination campaigns presents major challenges in rabies control and prevention. The development of a rabies vaccine that ensures lifelong immunity and animal population management in one dose could be extremely advantageous. A nonsurgical alternative to spay/neuter is a high priority for animal welfare, but irreversible infertility in one dose has not been achieved. Towards this goal, we developed a rabies virus-vectored immunocontraceptive vaccine ERA-2GnRH, which protected against rabies virus challenge and induced >80% infertility in mice after three doses in a live, liquid-vaccine formulation (Wu et al., 2014). To improve safety and use, we formulated an inactivated vaccine in a thermo-responsive chitosan hydrogel for one-dose delivery and studied the immune responses in mice. The hydrogel did not cause any injection site reactions, and the killed ERA-2GnRH vaccine induced high and persistent rabies virus neutralizing antibodies (rVNA) in mice. The rVNA in the hydrogel group reached an average of 327.40 IU/mL, more than 200 times higher than the liquid vaccine alone. The Gonadotropin-releasing hormone (GnRH) antibodies were also present and lasted longer in the hydrogel group, but did not prevent fertility in mice, reflecting a possible threshold level of GnRH antibodies for contraception. In conclusion, the hydrogel facilitated a high and long-lasting immunity, and ERA-2GnRH is a promising dual vaccine candidate. Future studies will focus on rabies protection in target species and improving the anti-GnRH response.

7.
PLoS One ; 14(9): e0222612, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31557167

RESUMEN

Monkeypox virus (MPXV) is a member of the genus Orthopoxvirus, endemic in Central and West Africa. This viral zoonosis was introduced into the United States in 2003 via African rodents imported for the pet trade and caused 37 human cases, all linked to exposure to MPXV-infected black-tailed prairie dogs (Cynomys ludovicianus). Prairie dogs have since become a useful model of MPXV disease, utilized for testing of potential medical countermeasures. In this study, we used recombinant MPXV containing the firefly luciferase gene (luc) and in vivo imaging technology to characterize MPXV pathogenesis in the black-tailed prairie dog in real time. West African (WA) MPXV could be visualized using in vivo imaging in the nose, lymph nodes, intestines, heart, lung, kidneys, and liver as early as day 6 post infection (p.i.). By day 9 p.i., lesions became visible on the skin and in some cases in the spleen. After day 9 p.i., luminescent signal representing MPXV replication either increased, indicating a progression to what would be a fatal infection, or decreased as infection was resolved. Use of recombinant luc+ MPXV allowed for a greater understanding of how MPXV disseminates throughout the body in prairie dogs during the course of infection. This technology will be used to reduce the number of animals required in future pathogenesis studies as well as aid in determining the effectiveness of potential medical countermeasures.


Asunto(s)
Monkeypox virus , Mpox/veterinaria , Sciuridae/virología , Animales , Modelos Animales de Enfermedad , Femenino , Corazón/virología , Intestinos/virología , Riñón/virología , Hígado/virología , Mediciones Luminiscentes/veterinaria , Pulmón/virología , Ganglios Linfáticos/virología , Masculino , Mpox/patología , Mpox/virología , Nariz/virología
8.
Nat Commun ; 8: 14446, 2017 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-28194016

RESUMEN

The Egyptian rousette bat (ERB) is a natural reservoir host for Marburg virus (MARV); however, the mechanisms by which MARV is transmitted bat-to-bat and to other animals are unclear. Here we co-house MARV-inoculated donor ERBs with naive contact ERBs. MARV shedding is detected in oral, rectal and urine specimens from inoculated bats from 5-19 days post infection. Simultaneously, MARV is detected in oral specimens from contact bats, indicating oral exposure to the virus. In the late study phase, we provide evidence that MARV can be horizontally transmitted from inoculated to contact ERBs by finding MARV RNA in blood and oral specimens from contact bats, followed by MARV IgG antibodies in these same bats. This study demonstrates that MARV can be horizontally transmitted from inoculated to contact ERBs, thereby providing a model for filovirus maintenance in its natural reservoir host and a potential mechanism for virus spillover to other animals.


Asunto(s)
Quirópteros/virología , Reservorios de Enfermedades/virología , Enfermedad del Virus de Marburg/virología , Marburgvirus/fisiología , Animales , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Egipto , Femenino , Filoviridae/fisiología , Inmunoglobulina G/inmunología , Masculino , Enfermedad del Virus de Marburg/sangre , Enfermedad del Virus de Marburg/transmisión , Marburgvirus/genética , Marburgvirus/inmunología , ARN Viral/sangre , ARN Viral/genética , Esparcimiento de Virus
10.
J Wildl Dis ; 51(1): 113-24, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25375951

RESUMEN

Marburg virus (Marburg marburgvirus; MARV) causes sporadic outbreaks of Marburg hemorrhagic fever (MHF) in Africa. The Egyptian fruit bat (Rousettus aegyptiacus) has been identified as a natural reservoir based most-recently on the repeated isolation of MARV directly from bats caught at two locations in southwestern Uganda where miners and tourists separately contracted MHF from 2007-08. Despite learning much about the ecology of MARV through extensive field investigations, there remained unanswered questions such as determining the primary routes of virus shedding and the severity of disease, if any, caused by MARV in infected bats. To answer these questions and others, we experimentally infected captive-bred R. aegyptiacus with MARV under high (biosafety level 4) containment. These experiments have shown infection profiles consistent with R. aegyptiacus being a bona fide natural reservoir host for MARV and demonstrated routes of viral shedding capable of infecting humans and other animals.


Asunto(s)
Quirópteros/virología , Enfermedad del Virus de Marburg/virología , Marburgvirus/aislamiento & purificación , Boca/virología , Esparcimiento de Virus , Animales , Masculino , ARN Viral/aislamiento & purificación , Carga Viral
11.
Antiviral Res ; 101: 26-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24184127

RESUMEN

Nipah virus (NiV) continues to cause outbreaks of fatal human encephalitis due to spillover from its bat reservoir. We determined that a single dose of replication-defective vesicular stomatitis virus (VSV)-based vaccine vectors expressing either the NiV fusion (F) or attachment (G) glycoproteins protected hamsters from over 1000 times LD50 NiV challenge. This highly effective single-dose protection coupled with an enhanced safety profile makes these candidates ideal for potential use in livestock and humans.


Asunto(s)
Portadores de Fármacos , Infecciones por Henipavirus/prevención & control , Virus Nipah/inmunología , Vesiculovirus/genética , Vacunas Virales/inmunología , Animales , Cricetinae , Modelos Animales de Enfermedad , Mesocricetus , Virus Nipah/genética , Análisis de Supervivencia , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas Estructurales Virales/genética , Proteínas Estructurales Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA