Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
1.
Sci Transl Med ; 16(754): eadi6887, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38959328

RESUMEN

Virulent infectious agents such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and methicillin-resistant Staphylococcus aureus (MRSA) induce tissue damage that recruits neutrophils, monocyte, and macrophages, leading to T cell exhaustion, fibrosis, vascular leak, epithelial cell depletion, and fatal organ damage. Neutrophils, monocytes, and macrophages recruited to pathogen-infected lungs, including SARS-CoV-2-infected lungs, express phosphatidylinositol 3-kinase gamma (PI3Kγ), a signaling protein that coordinates both granulocyte and monocyte trafficking to diseased tissues and immune-suppressive, profibrotic transcription in myeloid cells. PI3Kγ deletion and inhibition with the clinical PI3Kγ inhibitor eganelisib promoted survival in models of infectious diseases, including SARS-CoV-2 and MRSA, by suppressing inflammation, vascular leak, organ damage, and cytokine storm. These results demonstrate essential roles for PI3Kγ in inflammatory lung disease and support the potential use of PI3Kγ inhibitors to suppress inflammation in severe infectious diseases.


Asunto(s)
COVID-19 , Fosfatidilinositol 3-Quinasa Clase Ib , Inflamación , SARS-CoV-2 , COVID-19/patología , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Animales , Inflamación/patología , Humanos , Tratamiento Farmacológico de COVID-19 , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Pulmón/patología , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Permeabilidad Capilar/efectos de los fármacos , Ratones Endogámicos C57BL , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/patología
2.
Antivir Chem Chemother ; 28: 2040206620950143, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-34161179

RESUMEN

Clinical evidence suggests that Zika virus contributes to Guillain-Barré syndrome that causes temporary paralysis. We utilized a recently described Zika virus mouse model of temporary flaccid paralysis to address the hypothesis that treatment with an N-methyl-D-aspartate receptor antagonist, memantine, can reduce the incidence of paralysis. Aged interferon alpha/beta-receptor knockout mice were used because of their sublethal susceptibility to Zika virus infection. Fifteen to twenty-five percent of mice infected with a Puerto Rico strain of Zika virus develop acute flaccid paralysis beginning at days 8-9 and peaked at days 10-12. Mice recover from paralysis within a week of onset. In two independent studies, twice daily oral administration of memantine at 60 mg/kg/day on days 4 through 9 after viral challenge significantly reduced the incidence of paralysis. No efficacy was observed with treatments from days 9 through 12. Memantine treatment in cell culture or mice did not affect viral titers. These data indicate that early treatment of memantine before onset of paralysis is efficacious, but treatments beyond the onset of paralysis were not efficacious. The effect of this N-methyl-D-aspartate receptor antagonist on the incidence of Zika virus-induced paralysis may provide guidance for investigations on the mechanism of paralysis.


Asunto(s)
Antivirales/farmacología , Modelos Animales de Enfermedad , Síndrome de Guillain-Barré/tratamiento farmacológico , Memantina/farmacología , Parálisis/tratamiento farmacológico , Infección por el Virus Zika/tratamiento farmacológico , Animales , Células Cultivadas , Femenino , Masculino , Ratones , Ratones Noqueados , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Virus Zika/efectos de los fármacos
3.
Sci Rep ; 9(1): 19531, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31862897

RESUMEN

Clinical evidence is mounting that Zika virus can contribute to Guillain-Barré syndrome which causes temporary paralysis, yet the mechanism is unknown. We investigated the mechanism of temporary acute flaccid paralysis caused by Zika virus infection in aged interferon αß-receptor knockout mice used for their susceptibility to infection. Twenty-five to thirty-five percent of mice infected subcutaneously with Zika virus developed motor deficits including acute flaccid paralysis that peaked 8-10 days after viral challenge. These mice recovered within a week. Despite Zika virus infection in the spinal cord, motor neurons were not destroyed. We examined ultrastructures of motor neurons and synapses by transmission electron microscopy. The percent coverage of motor neurons by boutons was reduced by 20%; more specifically, flattened-vesicle boutons were reduced by 46%, and were normalized in recovering mice. Using electromyographic procedures employed in people to help diagnose Guillain-Barré syndrome, we determined that nerve conduction velocities between the sciatic notch and the gastrocnemius muscle were unchanged in paralyzed mice. However, F-wave latencies were increased in paralyzed mice, which suggests that neuropathy may exist between the sciatic notch to the nerve rootlets. Reversible synaptic retraction may be a previously unrecognized cofactor along with peripheral neuropathy for the development of Guillain-Barré syndrome during Zika virus outbreaks.


Asunto(s)
Neuronas Motoras/fisiología , Parálisis/etiología , Infección por el Virus Zika/complicaciones , Virus Zika/patogenicidad , Animales , Electrofisiología , Femenino , Síndrome de Guillain-Barré/virología , Masculino , Ratones , Parálisis/virología , Enfermedades del Sistema Nervioso Periférico/etiología , Enfermedades del Sistema Nervioso Periférico/virología , ARN Viral/genética , Receptor de Interferón alfa y beta/metabolismo
4.
Sci Rep ; 9(1): 8325, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31171800

RESUMEN

Zika virus (ZIKV) is an arboviral infection that has been shown to be sexually transmitted. The study outlined herein aims to determine if accessory sex glands and epididymal epithelial cells are sources of viral persistence in subacute and chronic ZIKV infection, and if infection of these organs is important in sexual transmission during long-term (chronic) infection. Male interferon type I receptor knockout (Ifnar-/-) mice were challenged with ZIKV and reproductive tissues were harvested 14 and 35 days post infection (DPI) for inoculation studies and 14, 35 and 70 DPI for histopathology. Artificial insemination fluid derived from epididymal flush and seminal plasma from the prostate and seminal vesicle was obtained from ZIKV inoculated and sham-infected males. Naïve interferon type I and II receptor knockout (AG129) female mice were pre-treated with progesterone and inoculated intravaginally with artificial insemination fluid from ZIKV-infected males. ZIKV RNA was detected in the artificial insemination fluid generated from epididymal flush or seminal plasma from ZIKV infected males at 14 and 35 DPI. ZIKV antigens were only detected in seminiferous tubules at 14 DPI. Epididymal epithelial cells did not show ZIKV antigen immunoreactivity at 14, 35 or 70 DPI. Severe fibrosing orchitis (end stage orchitis) was observed at 35 and 70 DPI. Mild inflammation and peri-tubular fibrosis were observed in the epididymis following clearance of virus. Viral RNA was not detected by PCR in whole blood samples of females from any intravaginal experimental group and only detected in 20% of subcutaneously inoculated animals (derived from 1 experimentally infected male) at 35 DPI. While ZIKV RNA and antigens can be detected in the male reproductive tract at 14 DPI and RNA can also be detected at 35 DPI, intravaginal inoculation of artificial insemination fluid from these time-points failed to result in viremia in naïve females inoculated intravaginally. These studies support the hypothesis that epididymal epithelial cells are critical to sexual transmission in immunocompromised mice. Additionally, acute but not chronic male reproductive tract infection with ZIKV results in infectious virus capable of being sexually transmitted in mice.


Asunto(s)
Antígenos Virales/química , Testículo/virología , Infección por el Virus Zika/complicaciones , Infección por el Virus Zika/transmisión , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Epidídimo , Femenino , Masculino , Ratones , Ratones Noqueados , Progesterona/uso terapéutico , ARN Viral , Receptor de Interferón alfa y beta/genética , Semen/virología , Testículo/patología , Resultado del Tratamiento , Virus Zika
5.
Viruses ; 11(2)2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30678320

RESUMEN

Zika virus (ZIKV) infection can cause severe congenital diseases, such as microcephaly, ocular defects and arthrogryposis in fetuses, and Guillain⁻Barré syndrome in adults. Efficacious therapeutic treatments for infected patients, as well as prophylactic treatments to prevent new infections are needed for combating ZIKV infection. Here, we report that ZIKV-specific human polyclonal antibodies (SAB-155), elicited in transchromosomal bovine (TcB), provide significant protection from infection by ZIKV in STAT2 knockout (KO) golden Syrian hamsters both prophylactically and therapeutically. These antibodies also prevent testicular lesions in this hamster model. Our data indicate that antibody-mediated immunotherapy is effective in treating ZIKV infection. Because suitable quantities of highly potent human polyclonal antibodies can be quickly produced from the TcB system against ZIKV and have demonstrated therapeutic efficacy in a small animal model, they have the potential as an effective countermeasure against ZIKV infection.


Asunto(s)
Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Inmunización Pasiva , Factor de Transcripción STAT2/genética , Infección por el Virus Zika/prevención & control , Infección por el Virus Zika/terapia , Animales , Animales Modificados Genéticamente , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Bovinos , Cricetinae , Modelos Animales de Enfermedad , Femenino , Técnicas de Inactivación de Genes , Humanos , Masculino , Testículo/patología , Testículo/virología , Virus Zika
6.
Sci Rep ; 8(1): 15379, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30337585

RESUMEN

Zika virus (ZIKV) is an arboviral infection that may be sexually transmitted. The present study aims to determine if accessory sex glands are a potential source of infectious virus and important in sexual transmission. Male interferon type I receptor knockout (Ifnar-/-) mice were challenged subcutaneously with a Puerto Rican ZIKV isolate. Reproductive tissues were harvested seven days after viral challenge and artificial insemination fluid derived from epididymis or homogenized accessory sex glands (seminal plasma) was obtained. Naïve interferon type I and II receptor knockout (AG129) females were pre-treated with progesterone, and inoculated intravaginally with either epididymal flush or seminal plasma from ZIKV-infected males. ZIKV RNA was demonstrated in the artificial insemination fluid and ZIKV antigen was detected in epididymal epithelial cells but not within seminiferous tubules at the time of artificial insemination fluid collection. Peripheral viremia, demonstrated by ZIKV RNA in whole blood samples of females from each challenge group was observed. Infectious virus was present in both epididymal fluid and seminal plasma. These studies provide evidence of passage of virus from epididymal flush and seminal plasma to naïve females via artificial insemination and provides a model for the study of sexual transmission of ZIKV.


Asunto(s)
Coito , Semen/virología , Infección por el Virus Zika/transmisión , Virus Zika/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Noqueados
7.
Sci Rep ; 8(1): 9384, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29925850

RESUMEN

Zika virus (ZIKV) can cause various diseases in offspring after congenital infection. The purpose of this study was to identify disease phenotypes in pups exposed to ZIKV in utero. Female interferon-α/ß, -γ receptor knockout mice (AG129) were infected intraperitoneally with ZIKV 7.5 days' post coitus (dpc). Viral RNA, antigen and infectious virus were detected in some, but not all, maternal and fetal tissues at various times during gestation. Fetuses of infected dams had significant intrauterine growth restriction (IUGR), which was more pronounced as females neared parturition. Pups born to infected dams were significantly smaller and had significantly shortened skull lengths, as determined by measurement with a caliper and by micro-CT analysis, as compared with age-matched controls. Growth rates of exposed pups after birth, however, was similar to sham-exposed offspring. Viral RNA was detected in pups of infected dams after birth. A lower survival rate was observed in neonates exposed to ZIKV in utero. A mortality rate of over 50%, attributed to consequences of ZIKV infection, occurred after birth in pups born to infected dams. A transient hearing loss was observed in some animals exposed to virus in utero. No motor deficits or cognitive deficits were detected using running wheel or viral paresis scoring assays. Abnormalities in offspring included smaller size, shorter skull length and increased neonatal mortality, while the only functional deficit we could detect was a low incidence of transient hearing loss.


Asunto(s)
Infección por el Virus Zika/complicaciones , Virus Zika/patogenicidad , Animales , Modelos Animales de Enfermedad , Femenino , Retardo del Crecimiento Fetal/virología , Inmunohistoquímica , Ratones , Ratones Noqueados , Microscopía Confocal , Tomografía Computarizada por Tomografía de Emisión de Positrones , Embarazo , Complicaciones Infecciosas del Embarazo/virología , ARN Viral/genética , Virus Zika/genética
8.
J Neurovirol ; 24(3): 273-290, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29476408

RESUMEN

Zika virus (ZIKV) has received widespread attention because of its effect on the developing fetus. It is becoming apparent, however, that severe neurological sequelae, such as Guillian-Barrë syndrome (GBS), myelitis, encephalitis, and seizures can occur after infection of adults. This study demonstrates that a contemporary strain of ZIKV can widely infect astrocytes and neurons in the brain and spinal cord of adult, interferon α/ß receptor knockout mice (AG129 strain) and cause progressive hindlimb paralysis, as well as severe seizure-like activity during the acute phase of disease. The severity of hindlimb motor deficits correlated with increased numbers of ZIKV-infected lumbosacral spinal motor neurons and decreased numbers of spinal motor neurons. Electrophysiological compound muscle action potential (CMAP) amplitudes in response to stimulation of the lumbosacral spinal cord were reduced when obvious motor deficits were present. ZIKV immunoreactivity was high, intense, and obvious in tissue sections of the brain and spinal cord. Infection in the brain and spinal cord was also associated with astrogliosis as well as T cell and neutrophil infiltration. CMAP and histological analysis indicated that peripheral nerve and muscle functions were intact. Consequently, motor deficits in these circumstances appear to be primarily due to myelitis and possibly encephalitis as opposed to a peripheral neuropathy or a GBS-like syndrome. Thus, acute ZIKV infection of adult AG129 mice may be a useful model for ZIKV-induced myelitis, encephalitis, and seizure activity.


Asunto(s)
Encefalitis/fisiopatología , Trastornos Motores/fisiopatología , Mielitis/fisiopatología , Convulsiones/fisiopatología , Infección por el Virus Zika/fisiopatología , Virus Zika/patogenicidad , Potenciales de Acción/fisiología , Animales , Astrocitos/inmunología , Astrocitos/patología , Astrocitos/virología , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/virología , Modelos Animales de Enfermedad , Encefalitis/inmunología , Encefalitis/virología , Femenino , Humanos , Interferón-alfa/deficiencia , Interferón-alfa/genética , Interferón-alfa/inmunología , Interferón beta/deficiencia , Interferón beta/genética , Interferón beta/inmunología , Interferón gamma/deficiencia , Interferón gamma/genética , Interferón gamma/inmunología , Masculino , Ratones , Ratones Noqueados , Trastornos Motores/inmunología , Trastornos Motores/virología , Neuronas Motoras/inmunología , Neuronas Motoras/patología , Neuronas Motoras/virología , Músculo Esquelético/fisiología , Mielitis/inmunología , Mielitis/virología , Neutrófilos/inmunología , Neutrófilos/patología , Neutrófilos/virología , Convulsiones/inmunología , Convulsiones/virología , Médula Espinal/inmunología , Médula Espinal/patología , Médula Espinal/virología , Linfocitos T/inmunología , Linfocitos T/patología , Linfocitos T/virología , Virus Zika/crecimiento & desarrollo , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología
9.
Am J Pathol ; 188(4): 904-915, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29378173

RESUMEN

An understanding of the pathogenesis of infection with the Zika virus in the male reproductive tract is vital for the development of vaccines and antivirals that will limit or prevent sexual transmission. Two common immunocompromised mouse strains used in transmission studies-male with genes encoding interferon types I and II receptor gene knockout (IFNAR/IFNGR; AG129) and with interferon type 1 receptor knockout (Ifnar-/-) were infected with a Puerto Rican Zika virus isolate (PRVABC59), and pathology was assessed 5 to 11 days after infection. Virus was detected by immunohistochemistry and quantitative RT-PCR in the testicle and epididymis of AG129 and Ifnar-/- mice, and by immunohistochemistry in the prostate and seminal vesicle of infected AG129 mice. Severe disease manifestations initiating as epididymitis and progressing to orchitis were observed in both models, with more severe inflammation noted in the AG129 mouse strain. Significant inflammation was not observed in any evaluated accessory sex gland at any point during infection. Time-course analysis of infection revealed an increase in the severity of disease within the epididymis of both strains, indicating a potential route of sexual transmission. Male mice with Ifnar-/- may better recapitulate Zika virus in humans and provide insight into the mechanism of sexual transmission, due to milder histopathologic lesions, the presence of histologically normal sperm in epididymal tubules, and an ability to survive the acute phase of disease.


Asunto(s)
Genitales Masculinos/patología , Genitales Masculinos/virología , Receptor de Interferón alfa y beta/deficiencia , Infección por el Virus Zika/patología , Infección por el Virus Zika/virología , Virus Zika/fisiología , Enfermedad Aguda , Animales , Epidídimo/patología , Inflamación/patología , Masculino , Ratones Noqueados , ARN Viral/análisis , Receptor de Interferón alfa y beta/metabolismo , Testículo/patología
10.
Viruses ; 10(1)2018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-29329211

RESUMEN

Enterovirus D68 (EV-D68) caused a large outbreak in the summer and fall of 2014 in the United States. It causes serious respiratory disease, but causation of associated paralysis is controversial, because the virus is not routinely identified in cerebrospinal fluid. To establish clinical correlates with human disease, we evaluated EV-D68 infection in non-lethal paralysis mouse models. Ten-day-old mice lacking interferon responses were injected intraperitoneally with the virus. Paralysis developed in hindlimbs. After six weeks of paralysis, the motor neurons were depleted due to viral infection. Hindlimb muscles were also infected and degenerating. Even at the earliest stage of paralysis, muscles were still infected and were degenerating, in addition to presence of virus in the spinal cord. To model natural respiratory infection, five-day-old mice were infected intranasally with EV-D68. Two of the four infected mice developed forelimb paralysis. The affected limbs had muscle disease, but no spinal cord infection was detected. The unique contributions of this study are that EV-D68 causes paralysis in mice, and that causation by muscle disease, with or without spinal cord disease, may help to resolve the controversy that the virus can cause paralysis, even if it cannot be identified in cerebrospinal fluid.


Asunto(s)
Enterovirus Humano D/patogenicidad , Infecciones por Enterovirus/fisiopatología , Mielitis/virología , Miositis/virología , Parálisis/etiología , Animales , Infecciones por Enterovirus/virología , Masculino , Ratones , Neuronas Motoras/virología , Atrofia Muscular/fisiopatología , Atrofia Muscular/virología , Mielitis/fisiopatología , Miositis/fisiopatología , Parálisis/virología , Receptor de Interferón alfa y beta/deficiencia , Receptores de Interferón/deficiencia , Médula Espinal/virología , Receptor de Interferón gamma
11.
Antiviral Res ; 149: 26-33, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29126900

RESUMEN

Nucleic acid polymers (NAPs) block the release of HBsAg from infected hepatocytes. These compounds have been previously shown to have the unique ability to eliminate serum surface antigen in DHBV-infected Pekin ducks and achieve multilog reduction of HBsAg or HBsAg loss in patients with chronic HBV infection and HBV/HDV coinfection. In ducks and humans, the blockage of HBsAg release by NAPs occurs by the selective targeting of the assembly and/or secretion of subviral particles (SVPs). The clinically active NAP species REP 2055 and REP 2139 were investigated in other relevant animal models of HBV infection including woodchucks chronically infected with WHV, HBV transgenic mice and HBV infected SCID-Hu mice. The liver accumulation of REP 2139 in woodchucks following subcutaneous administration was examined and was found to be similar to that observed in mice and ducks. However, in woodchucks, NAP treatment was associated with only mild (36-79% relative to baseline) reductions in WHsAg (4/10 animals) after 3-5 weeks of treatment without changes in serum WHV DNA. In HBV infected SCID-Hu mice, REP 2055 treatment was not associated with any reduction of HBsAg, HBeAg or HBV DNA in the serum after 28 days of treatment. In HBV transgenic mice, no reductions in serum HBsAg were observed with REP 2139 with up to 12 weeks of treatment. In conclusion, the antiviral effects of NAPs in DHBV infected ducks and patients with chronic HBV infection were weak or absent in woodchuck and mouse models despite similar liver accumulation of NAPs in all these species, suggesting that the mechanisms of SVP assembly and or secretion present in rodent models differs from that in DHBV and chronic HBV infections.


Asunto(s)
Virus de la Hepatitis B/efectos de los fármacos , Hepatitis B/virología , Ácidos Nucleicos/farmacología , Polímeros , Animales , Biomarcadores , Modelos Animales de Enfermedad , Hepatitis B/sangre , Hepatitis B/tratamiento farmacológico , Antígenos de Superficie de la Hepatitis B/sangre , Virus de la Hepatitis B de la Marmota/efectos de los fármacos , Humanos , Marmota , Ratones , Ratones Transgénicos , Ácidos Nucleicos/química , Polímeros/química , Roedores , Replicación Viral/efectos de los fármacos
12.
Virology ; 507: 89-95, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28431283

RESUMEN

Zika virus (ZIKV) infection was investigated in adult and fetal STAT2 knock-out (KO) hamsters. Subcutaneous injection of ZIKV of adults resulted in morbidity, mortality, and infection of the uterus, placenta, brain, spinal cord, and testicles, thus providing an opportunity to evaluate congenital ZIKV infection in a second rodent species besides mice. ZIKV-infected cells with morphologies of Sertoli cells and spermatogonia were observed in the testes, which may have implications for sexual transmission and male sterility. Neonates exposed as fetuses to ZIKV at 8 days post-coitus were not smaller than controls. Nevertheless, infectious virus and ZIKV RNA was detected in some, but not all, placentas and fetal brains of KO hamsters. STAT2 KO hamsters may be useful for addressing sexual transmission, pathogenesis, routes of fetal infection, and neurological disease outcomes, and may also be used in antiviral or vaccine studies to identify intervention strategies.


Asunto(s)
Factor de Transcripción STAT2/deficiencia , Infección por el Virus Zika/embriología , Virus Zika/fisiología , Animales , Encéfalo/patología , Encéfalo/virología , Cricetinae , Modelos Animales de Enfermedad , Femenino , Feto/patología , Feto/virología , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Masculino , Mesocricetus , Ratones Noqueados , Placenta/patología , Placenta/virología , Embarazo , Complicaciones Infecciosas del Embarazo/patología , Complicaciones Infecciosas del Embarazo/virología , Factor de Transcripción STAT2/genética , Virus Zika/genética , Infección por el Virus Zika/genética , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/virología
13.
Antiviral Res ; 137: 14-22, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27838352

RESUMEN

Zika virus (ZIKV) is currently undergoing pandemic emergence. While disease is typically subclinical, severe neurologic manifestations in fetuses and newborns after congenital infection underscore an urgent need for antiviral interventions. The adenosine analog BCX4430 has broad-spectrum activity against a wide range of RNA viruses, including potent in vivo activity against yellow fever, Marburg and Ebola viruses. We tested this compound against African and Asian lineage ZIKV in cytopathic effect inhibition and virus yield reduction assays in various cell lines. To further evaluate the efficacy in a relevant animal model, we developed a mouse model of severe ZIKV infection, which recapitulates various human disease manifestations including peripheral virus replication, conjunctivitis, encephalitis and myelitis. Time-course quantification of viral RNA accumulation demonstrated robust viral replication in several relevant tissues, including high and persistent viral loads observed in the brain and testis. The presence of viral RNA in various tissues was confirmed by an infectious culture assay as well as immunohistochemical staining of tissue sections. Treatment of ZIKV-infected mice with BCX4430 significantly improved outcome even when treatment was initiated during the peak of viremia. The demonstration of potent activity of BCX4430 against ZIKV in a lethal mouse model warrant its continued clinical development.


Asunto(s)
Antivirales/farmacología , Antivirales/uso terapéutico , Nucleósidos de Purina/farmacología , Nucleósidos de Purina/uso terapéutico , Infección por el Virus Zika/tratamiento farmacológico , Virus Zika/efectos de los fármacos , Adenina/análogos & derivados , Adenosina/análogos & derivados , Animales , Antivirales/administración & dosificación , Encéfalo/virología , Línea Celular , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Nucleósidos de Purina/administración & dosificación , Pirrolidinas , ARN Viral , Testículo/virología , Carga Viral/efectos de los fármacos , Viremia , Replicación Viral/efectos de los fármacos , Infección por el Virus Zika/virología
14.
J Neurovirol ; 23(2): 186-204, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27761801

RESUMEN

Neurological respiratory deficits are serious outcomes of West Nile virus (WNV) disease. WNV patients requiring intubation have a poor prognosis. We previously reported that WNV-infected rodents also appear to have respiratory deficits when assessed by whole-body plethysmography and diaphragmatic electromyography. The purpose of this study was to determine if the nature of the respiratory deficits in WNV-infected rodents is neurological and if deficits are due to a disorder of brainstem respiratory centers, cervical spinal cord (CSC) phrenic motor neuron (PMN) circuitry, or both. We recorded phrenic nerve (PN) activity and found that in WNV-infected mice, PN amplitude is reduced, corroborating a neurological basis for respiratory deficits. These results were associated with a reduction in CSC motor neuron number. We found no dramatic deficits, however, in brainstem-mediated breathing rhythm generation or responses to hypercapnia. PN frequency and pattern parameters were normal, and all PN parameters changed appropriately upon a CO2 challenge. Histological analysis revealed generalized microglia activation, astrocyte reactivity, T cell and neutrophil infiltration, and mild histopathologic lesions in both the brainstem and CSC, but none of these were tightly correlated with PN function. Similar results in PN activity, brainstem function, motor neuron number, and histopathology were seen in WNV-infected hamsters, except that histopathologic lesions were more severe. Taken together, the results suggest that respiratory deficits in acute WNV infection are primarily due to a lower motor neuron disorder affecting PMNs and the PN rather than a brainstem disorder. Future efforts should focus on markers of neuronal dysfunction, axonal degeneration, and myelination.


Asunto(s)
Tronco Encefálico/inmunología , Neuronas Motoras/inmunología , Nervio Frénico/inmunología , Médula Espinal/inmunología , Fiebre del Nilo Occidental/inmunología , Animales , Astrocitos/inmunología , Astrocitos/patología , Astrocitos/virología , Tronco Encefálico/patología , Tronco Encefálico/virología , Recuento de Células , Cricetulus , Electromiografía/métodos , Femenino , Humanos , Masculino , Ratones , Microglía/inmunología , Microglía/patología , Microglía/virología , Neuronas Motoras/patología , Neuronas Motoras/virología , Conducción Nerviosa , Infiltración Neutrófila , Nervio Frénico/patología , Nervio Frénico/virología , Médula Espinal/patología , Médula Espinal/virología , Linfocitos T/inmunología , Linfocitos T/patología , Linfocitos T/virología , Fiebre del Nilo Occidental/patología , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/patogenicidad , Virus del Nilo Occidental/fisiología
15.
J Virol ; 90(6): 3086-92, 2016 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-26739045

RESUMEN

UNLABELLED: Arbidol (ARB) is a synthetic antiviral originally developed to combat influenza viruses. ARB is currently used clinically in several countries but not in North America. We have previously shown that ARB inhibits in vitro hepatitis C virus (HCV) by blocking HCV entry and replication. In this report, we expand the list of viruses that are inhibited by ARB and demonstrate that ARB suppresses in vitro infection of mammalian cells with Ebola virus (EBOV), Tacaribe arenavirus, and human herpesvirus 8 (HHV-8). We also confirm suppression of hepatitis B virus and poliovirus by ARB. ARB inhibited EBOV Zaire Kikwit infection when added before or at the same time as virus infection and was less effective when added 24 h after EBOV infection. Experiments with recombinant vesicular stomatitis virus (VSV) expressing the EBOV Zaire glycoprotein showed that infection was inhibited by ARB at early stages, most likely at the level of viral entry into host cells. ARB inhibited HHV-8 replication to a similar degree as cidofovir. Our data broaden the spectrum of antiviral efficacy of ARB to include globally prevalent viruses that cause significant morbidity and mortality. IMPORTANCE: There are many globally prevalent viruses for which there are no licensed vaccines or antiviral medicines. Some of these viruses, such as Ebola virus or members of the arenavirus family, rapidly cause severe hemorrhagic diseases that can be fatal. Other viruses, such as hepatitis B virus or human herpesvirus 8 (HHV-8), establish persistent infections that cause chronic illnesses, including cancer. Thus, finding an affordable, effective, and safe drug that blocks many viruses remains an unmet medical need. The antiviral drug arbidol (ARB), already in clinical use in several countries as an anti-influenza treatment, has been previously shown to suppress the growth of many viruses. In this report, we expand the list of viruses that are blocked by ARB in a laboratory setting to include Ebola virus, Tacaribe arenavirus, and HHV-8, and we propose ARB as a broad-spectrum antiviral drug that may be useful against hemorrhagic viruses.


Asunto(s)
Antivirales/farmacología , Indoles/farmacología , Virus/efectos de los fármacos , Animales , Línea Celular , Humanos , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Estructura Molecular , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
16.
PLoS One ; 9(9): e106535, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25180779

RESUMEN

Evidence indicates that West Nile virus (WNV) employs Ca(2+) influx for its replication. Moreover, calcium buffer proteins, such as calbindin D28k (CB-D28k), may play an important role mitigating cellular destruction due to disease processes, and more specifically, in some neurological diseases. We addressed the hypothesis that CB-D28k inhibits WNV replication in cell culture and infected rodents. WNV envelope immunoreactivity (ir) was not readily co-localized with CB-D28k ir in WNV-infected Vero 76 or motor neuron-like NSC34 cells that were either stably or transiently transfected with plasmids coding for CB-D28k gene. This was confirmed in cultured cells fixed on glass coverslips and by flow cytometry. Moreover, WNV infectious titers were reduced in CB-D28k-transfected cells. As in cell culture studies, WNV env ir was not co-localized with CB-D28k ir in the cortex of an infected WNV hamster, or in the hippocampus of an infected mouse. Motor neurons in the spinal cord typically do not express CB-D28k and are susceptible to WNV infection. Yet, CB-D28k was detected in the surviving motor neurons after the initial phase of WNV infection in hamsters. These data suggested that induction of CB-D28k elicit a neuroprotective response to WNV infection.


Asunto(s)
Calbindina 1/fisiología , Virus del Nilo Occidental/fisiología , Animales , Calbindina 1/genética , Línea Celular , Chlorocebus aethiops , Cricetinae , Femenino , Interacciones Huésped-Patógeno/fisiología , Mesocricetus , Ratones , Ratones Endogámicos C57BL , Neuronas Motoras/virología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Médula Espinal/virología , Células Vero , Replicación Viral/fisiología , Fiebre del Nilo Occidental/fisiopatología , Fiebre del Nilo Occidental/prevención & control , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/patogenicidad
17.
PLoS Pathog ; 10(7): e1004290, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25077483

RESUMEN

Japanese encephalitis virus (JEV), a mosquito-borne flavivirus that causes fatal neurological disease in humans, is one of the most important emerging pathogens of public health significance. JEV represents the JE serogroup, which also includes West Nile, Murray Valley encephalitis, and St. Louis encephalitis viruses. Within this serogroup, JEV is a vaccine-preventable pathogen, but the molecular basis of its neurovirulence remains unknown. Here, we constructed an infectious cDNA of the most widely used live-attenuated JE vaccine, SA14-14-2, and rescued from the cDNA a molecularly cloned virus, SA14-14-2MCV, which displayed in vitro growth properties and in vivo attenuation phenotypes identical to those of its parent, SA14-14-2. To elucidate the molecular mechanism of neurovirulence, we selected three independent, highly neurovirulent variants (LD50, <1.5 PFU) from SA14-14-2MCV (LD50, >1.5×105 PFU) by serial intracerebral passage in mice. Complete genome sequence comparison revealed a total of eight point mutations, with a common single G1708→A substitution replacing a Gly with Glu at position 244 of the viral E glycoprotein. Using our infectious SA14-14-2 cDNA technology, we showed that this single Gly-to-Glu change at E-244 is sufficient to confer lethal neurovirulence in mice, including rapid development of viral spread and tissue inflammation in the central nervous system. Comprehensive site-directed mutagenesis of E-244, coupled with homology-based structure modeling, demonstrated a novel essential regulatory role in JEV neurovirulence for E-244, within the ij hairpin of the E dimerization domain. In both mouse and human neuronal cells, we further showed that the E-244 mutation altered JEV infectivity in vitro, in direct correlation with the level of neurovirulence in vivo, but had no significant impact on viral RNA replication. Our results provide a crucial step toward developing novel therapeutic and preventive strategies against JEV and possibly other encephalitic flaviviruses.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/genética , Encefalitis Japonesa/virología , Vacunas contra la Encefalitis Japonesa/genética , Glicoproteínas de Membrana/genética , Mutación/genética , Sistema Nervioso/virología , Proteínas del Envoltorio Viral/genética , Secuencia de Aminoácidos , Animales , Northern Blotting , Western Blotting , Clonación Molecular , Virus de la Encefalitis Japonesa (Especie)/inmunología , Encefalitis Japonesa/genética , Encefalitis Japonesa/inmunología , Femenino , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Vacunas contra la Encefalitis Japonesa/inmunología , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos ICR , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Conformación Proteica , Homología de Secuencia de Aminoácido , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Virulencia/genética , Replicación Viral
18.
Antimicrob Agents Chemother ; 58(11): 6607-14, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25155605

RESUMEN

No effective antiviral therapies are currently available to treat disease after infection with yellow fever virus (YFV). A Syrian golden hamster model of yellow fever (YF) was used to characterize the effect of treatment with BCX4430, a novel adenosine nucleoside analog. Significant improvement in survival was observed after treatment with BCX4430 at 4 mg/kg of body weight per day dosed intraperitoneally (i.p.) twice daily (BID). Treatment with BCX4430 at 12.5 mg/kg/day administered i.p. BID for 7 days offered complete protection from mortality and also resulted in significant improvement of other YF disease parameters, including weight loss, serum alanine aminotransferase levels (6 days postinfection [dpi]), and viremia (4 dpi). In uninfected hamsters, BCX4430 at 200 mg/kg/day administered i.p. BID for 7 days was well tolerated and did not result in mortality or weight loss, suggesting a potentially wide therapeutic index. Treatment with BCX4430 at 12 mg/kg/day i.p. remained effective when administered once daily and for only 4 days. Moreover, BCX4430 dosed at 200 mg/kg/day i.p. BID for 7 days effectively treated YF, even when treatment was delayed up to 4 days after virus challenge, corresponding with peak viral titers in the liver and serum. BCX4430 treatment did not preclude a protective antibody response, as higher neutralizing antibody (nAb) concentrations corresponded with increasing delays of treatment initiation, and greater nAb responses resulted in the protection of animals from a secondary challenge with YFV. In summary, BCX4430 is highly active in a hamster model of YF, even when treatment is initiated at the peak of viral replication.


Asunto(s)
Antivirales/uso terapéutico , Nucleósidos de Purina/uso terapéutico , Fiebre Amarilla/tratamiento farmacológico , Virus de la Fiebre Amarilla/efectos de los fármacos , Virus de la Fiebre Amarilla/inmunología , Adenina/análogos & derivados , Adenosina/análogos & derivados , Adenosina/uso terapéutico , Alanina Transaminasa/sangre , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Células Cultivadas , Cricetinae , Modelos Animales de Enfermedad , Femenino , Mesocricetus , Pirrolidinas , Resultado del Tratamiento , Ensayo de Placa Viral , Viremia/tratamiento farmacológico , Viremia/virología , Fiebre Amarilla/mortalidad , Fiebre Amarilla/virología
19.
Antiviral Res ; 108: 10-3, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24837607

RESUMEN

To characterize the impact on lung function, we assessed plethysmography parameters in a course of infection with mouse-adapted A/Pennsylvania/14/2010 (H3N2) influenza virus. Several parameters, represented by enhanced pause (penh) and ratio of inspiratory/expiratory time (Ti/Te), were observed that had early (1-7dpi) and robust changes regardless of virus challenge dose. Other parameters, characterized by tidal volume (TV), breathing frequency (freq) and end inspiratory pause (EIP), changed later (7-15dpi) during the course of infection and had a virus challenge dose effect. A third category of lung function parameters, such as peak inspiratory flow, had early, virus challenge-independent changes followed by later changes that were challenge dependent. These parameters changed in a similar manner after infection with a non-mouse adapted virus, although the time-course of many parameters was delayed somewhat when compared with mouse-adapted virus. Histopathological assessment of lung samples corresponded with changes in lung function parameters. This study demonstrates the utility of plethysmography in assessing disease in a mouse model of mild influenza virus infection.


Asunto(s)
Pulmón/patología , Infecciones por Orthomyxoviridae/diagnóstico , Infecciones por Orthomyxoviridae/patología , Pletismografía/métodos , Neumonía Viral/diagnóstico , Neumonía Viral/patología , Pruebas de Función Respiratoria/métodos , Animales , Femenino , Histocitoquímica , Subtipo H3N2 del Virus de la Influenza A/crecimiento & desarrollo , Ratones Endogámicos BALB C
20.
Antiviral Res ; 103: 32-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24393669

RESUMEN

Yellow fever virus (YFV) causes significant human disease and mortality in tropical regions of South and Central America and Africa, despite the availability of an effective vaccine. No specific therapy for YF is available. We previously showed that the humanized monoclonal antibody (MAb) 2C9-cIgG provided prophylactic and therapeutic protection from mortality in interferon receptor-deficient strain AG129 mice challenged with YF 17D-204 vaccine. In this study we tested the prophylactic and therapeutic efficacy of this MAb against virulent YFV infection in an immunocompetent hamster model. Intraperitoneal (ip) administration of a single dose of MAb 2C9-cIgG 24h prior to YFV challenge resulted in significantly improved survival rates in animals treated with 380 or 38 µg of MAb compared to untreated animals. Treatment with the higher dose also resulted in significantly improved weight gain and reductions in serum alanine aminotransferase (ALT) and virus titers in serum and liver. Prophylactic treatment with 2C9-cIgG 24h prior to virus challenge prevented the development of a virus-neutralizing antibody (vnAb) response in hamsters. Administration of a single ip dose of 380 µg of 2C9-cIgG as late as 72 h post-YFV challenge also resulted in significant improvement in survival rates. Hamsters treated at 4-72 h post-virus challenge developed a robust vnAb response. Enhanced survival and improvement of various disease parameters in the hamster model when MAb 2C9-cIgG is administered up to 3 days after virus challenge demonstrate the clinical potential of specific antibody therapy for YF.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Inmunización Pasiva/métodos , Fiebre Amarilla/tratamiento farmacológico , Fiebre Amarilla/prevención & control , Alanina Transaminasa/sangre , Animales , Peso Corporal , Cricetinae , Modelos Animales de Enfermedad , Hígado/virología , Mesocricetus , Suero/virología , Análisis de Supervivencia , Resultado del Tratamiento , Carga Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...