Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Cell ; 185(14): 2523-2541.e30, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35738284

RESUMEN

Stem cell research endeavors to generate specific subtypes of classically defined "cell types." Here, we generate >90% pure human artery or vein endothelial cells from pluripotent stem cells within 3-4 days. We specified artery cells by inhibiting vein-specifying signals and vice versa. These cells modeled viral infection of human vasculature by Nipah and Hendra viruses, which are extraordinarily deadly (∼57%-59% fatality rate) and require biosafety-level-4 containment. Generating pure populations of artery and vein cells highlighted that Nipah and Hendra viruses preferentially infected arteries; arteries expressed higher levels of their viral-entry receptor. Virally infected artery cells fused into syncytia containing up to 23 nuclei, which rapidly died. Despite infecting arteries and occupying ∼6%-17% of their transcriptome, Nipah and Hendra largely eluded innate immune detection, minimally eliciting interferon signaling. We thus efficiently generate artery and vein cells, introduce stem-cell-based toolkits for biosafety-level-4 virology, and explore the arterial tropism and cellular effects of Nipah and Hendra viruses.


Asunto(s)
Virus Hendra , Virus Nipah , Células Madre Pluripotentes , Arterias , Células Endoteliales , Virus Hendra/genética , Humanos , Tropismo
2.
Emerg Infect Dis ; 29(10): 2159-2163, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37735788

RESUMEN

Several occurrences of human-to-human transmission of Andes virus, an etiological agent of hantavirus cardiopulmonary syndrome, are documented. Syrian hamsters consistently model human hantavirus cardiopulmonary syndrome, yet neither transmission nor shedding has been investigated. We demonstrate horizontal virus transmission and show that Andes virus is shed efficiently from both inoculated and contact-infected hamsters.


Asunto(s)
Orthohantavirus , Animales , Cricetinae , Humanos , Mesocricetus , Síndrome
3.
Clin Infect Dis ; 63(8): 1026-33, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27531847

RESUMEN

BACKGROUND: The ongoing Ebola outbreak in West Africa has resulted in 28 646 suspected, probable, and confirmed Ebola virus infections. Nevertheless, malaria remains a large public health burden in the region affected by the outbreak. A joint Centers for Disease Control and Prevention/National Institutes of Health diagnostic laboratory was established in Monrovia, Liberia, in August 2014, to provide laboratory diagnostics for Ebola virus. METHODS: All blood samples from suspected Ebola virus-infected patients admitted to the Médecins Sans Frontières ELWA3 Ebola treatment unit in Monrovia were tested by quantitative real-time polymerase chain reaction for the presence of Ebola virus and Plasmodium species RNA. Clinical outcome in laboratory-confirmed Ebola virus-infected patients was analyzed as a function of age, sex, Ebola viremia, and Plasmodium species parasitemia. RESULTS: The case fatality rate of 1182 patients with laboratory-confirmed Ebola virus infections was 52%. The probability of surviving decreased with increasing age and decreased with increasing Ebola viral load. Ebola virus-infected patients were 20% more likely to survive when Plasmodium species parasitemia was detected, even after controlling for Ebola viral load and age; those with the highest levels of parasitemia had a survival rate of 83%. This effect was independent of treatment with antimalarials, as this was provided to all patients. Moreover, treatment with antimalarials did not affect survival in the Ebola virus mouse model. CONCLUSIONS: Plasmodium species parasitemia is associated with an increase in the probability of surviving Ebola virus infection. More research is needed to understand the molecular mechanism underlying this remarkable phenomenon and translate it into treatment options for Ebola virus infection.


Asunto(s)
Coinfección , Ebolavirus , Fiebre Hemorrágica Ebola/complicaciones , Fiebre Hemorrágica Ebola/mortalidad , Malaria/complicaciones , Malaria/parasitología , Parasitemia , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Niño , Preescolar , Modelos Animales de Enfermedad , Ebolavirus/genética , Femenino , Fiebre Hemorrágica Ebola/diagnóstico , Fiebre Hemorrágica Ebola/epidemiología , Humanos , Lactante , Recién Nacido , Malaria/diagnóstico , Malaria/epidemiología , Masculino , Ratones , Persona de Mediana Edad , Carga de Parásitos , Plasmodium/genética , Tasa de Supervivencia , Carga Viral , Adulto Joven
4.
Emerg Infect Dis ; 22(2): 323-6, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26814608

RESUMEN

Malaria is a major public health concern in the countries affected by the Ebola virus disease epidemic in West Africa. We determined the feasibility of using molecular malaria diagnostics during an Ebola virus disease outbreak and report the incidence of Plasmodium spp. parasitemia in persons with suspected Ebola virus infection.


Asunto(s)
Coinfección , Brotes de Enfermedades , Ebolavirus , Fiebre Hemorrágica Ebola/epidemiología , Malaria/diagnóstico , Malaria/parasitología , Humanos , Malaria Falciparum/diagnóstico , Malaria Falciparum/parasitología , Carga de Parásitos , Plasmodium falciparum/clasificación , Plasmodium falciparum/genética , Prevalencia
5.
Nat Commun ; 15(1): 1826, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38418477

RESUMEN

Bats are increasingly recognized as reservoirs of emerging zoonotic pathogens. Egyptian rousette bats (ERBs) are the known reservoir of Marburg virus (MARV), a filovirus that causes deadly Marburg virus disease (MVD) in humans. However, ERBs harbor MARV asymptomatically, likely due to a coadapted and specific host immunity-pathogen relationship. Recently, we measured transcriptional responses in MARV-infected ERB whole tissues, showing that these bats possess a disease tolerant strategy that limits pro-inflammatory gene induction, presumably averting MVD-linked immunopathology. However, the host resistant strategy by which ERBs actively limit MARV burden remains elusive, which we hypothesize requires localized inflammatory responses unresolvable at bulk-tissue scale. Here, we use dexamethasone to attenuate ERB pro-inflammatory responses and assess MARV replication, shedding and disease. We show that MARV-infected ERBs naturally mount coordinated pro-inflammatory responses at liver foci of infection, comprised of recruited mononuclear phagocytes and T cells, the latter of which proliferate with likely MARV-specificity. When pro-inflammatory responses are diminished, ERBs display heightened MARV replication, oral/rectal shedding and severe MVD-like liver pathology, demonstrating that ERBs balance immunoprotective tolerance with discreet MARV-resistant pro-inflammatory responses. These data further suggest that natural ERB immunomodulatory stressors like food scarcity and habitat disruption may potentiate viral shedding, transmission and therefore outbreak risk.


Asunto(s)
Quirópteros , Filoviridae , Enfermedad del Virus de Marburg , Marburgvirus , Animales , Humanos , Marburgvirus/genética , Inmunidad
6.
Front Immunol ; 14: 1306501, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38259437

RESUMEN

Several filoviruses, including Marburg virus (MARV), cause severe disease in humans and nonhuman primates (NHPs). However, the Egyptian rousette bat (ERB, Rousettus aegyptiacus), the only known MARV reservoir, shows no overt illness upon natural or experimental infection, which, like other bat hosts of zoonoses, is due to well-adapted, likely species-specific immune features. Despite advances in understanding reservoir immune responses to filoviruses, ERB peripheral blood responses to MARV and how they compare to those of diseased filovirus-infected spillover hosts remain ill-defined. We thus conducted a longitudinal analysis of ERB blood gene responses during acute MARV infection. These data were then contrasted with a compilation of published primate blood response studies to elucidate gene correlates of filovirus protection versus disease. Our work expands on previous findings in MARV-infected ERBs by supporting both host resistance and disease tolerance mechanisms, offers insight into the peripheral immunocellular repertoire during infection, and provides the most direct known cross-examination between reservoir and spillover hosts of the most prevalently-regulated response genes, pathways and activities associated with differences in filovirus pathogenesis and pathogenicity.


Asunto(s)
Quirópteros , Filoviridae , Marburgvirus , Humanos , Animales , Filoviridae/genética , Tolerancia Inmunológica , Inmunidad
7.
Curr Biol ; 31(2): 257-270.e5, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33157026

RESUMEN

Marburg virus (MARV) is among the most virulent pathogens of primates, including humans. Contributors to severe MARV disease include immune response suppression and inflammatory gene dysregulation ("cytokine storm"), leading to systemic damage and often death. Conversely, MARV causes little to no clinical disease in its reservoir host, the Egyptian rousette bat (ERB). Previous genomic and in vitro data suggest that a tolerant ERB immune response may underlie MARV avirulence, but no significant examination of this response in vivo yet exists. Here, using colony-bred ERBs inoculated with a bat isolate of MARV, we use species-specific antibodies and an immune gene probe array (NanoString) to temporally characterize the transcriptional host response at sites of MARV replication relevant to primate pathogenesis and immunity, including CD14+ monocytes/macrophages, critical immune response mediators, primary MARV targets, and skin at the inoculation site, where highest viral loads and initial engagement of antiviral defenses are expected. Our analysis shows that ERBs upregulate canonical antiviral genes typical of mammalian systems, such as ISG15, IFIT1, and OAS3, yet demonstrate a remarkable lack of significant induction of proinflammatory genes classically implicated in primate filoviral pathogenesis, including CCL8, FAS, and IL6. Together, these findings offer the first in vivo functional evidence for disease tolerance as an immunological mechanism by which the bat reservoir asymptomatically hosts MARV. More broadly, these data highlight factors determining disparate outcomes between reservoir and spillover hosts and defensive strategies likely utilized by bat hosts of other emerging pathogens, knowledge that may guide development of effective antiviral therapies.


Asunto(s)
Quirópteros/inmunología , Reservorios de Enfermedades/virología , Tolerancia Inmunológica/inmunología , Enfermedad del Virus de Marburg/inmunología , Marburgvirus/inmunología , Animales , Infecciones Asintomáticas , Quirópteros/sangre , Quirópteros/genética , Quirópteros/virología , Femenino , Regulación de la Expresión Génica/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Tolerancia Inmunológica/genética , Masculino , Enfermedad del Virus de Marburg/virología , Monocitos/inmunología
8.
Nat Rev Immunol ; 17(3): 195-207, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28111475

RESUMEN

Two of the most important contemporary emerging viruses that affect human health in Africa are Ebola virus (EBOV) and Lassa virus (LASV). The 2013-2016 West African outbreak of EBOV was responsible for more than 11,000 deaths, primarily in Guinea, Sierra Leone and Liberia. LASV is constantly emerging in these and surrounding West African countries, with an estimate of more than 500,000 cases of Lassa fever, and approximately 5,000 deaths, annually. Both EBOV and LASV are zoonotic, and human infection often results in a severe haemorrhagic fever in both cases. However, the contribution of specific immune responses to disease differs between EBOV and LASV. This Review examines innate and adaptive immune responses to these viruses with the goal of delineating responses that are associated with protective versus pathogenic outcomes.


Asunto(s)
Fiebre Hemorrágica Ebola/inmunología , Fiebre de Lassa/inmunología , Ebolavirus/inmunología , Humanos , Virus Lassa/inmunología
9.
Sci Rep ; 2: 736, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23071900

RESUMEN

Encephalitis is a hallmark of Nipah virus (NiV) infection in humans. The exact route of entry of NiV into the central nervous system (CNS) is unknown. Here, we performed a spatio-temporal analysis of NiV entry into the CNS of hamsters. NiV initially predominantly targeted the olfactory epithelium in the nasal turbinates. From there, NiV infected neurons were visible extending through the cribriform plate into the olfactory bulb, providing direct evidence of rapid CNS entry. Subsequently, NiV disseminated to the olfactory tubercle and throughout the ventral cortex. Transmission electron microscopy on brain tissue showed extravasation of plasma cells, neuronal degeneration and nucleocapsid inclusions in affected tissue and axons, providing further evidence for axonal transport of NiV. NiV entry into the CNS coincided with the occurrence of respiratory disease, suggesting that the initial entry of NiV into the CNS occurs simultaneously with, rather than as a result of, systemic virus replication.


Asunto(s)
Sistema Nervioso Central/virología , Virus Nipah/fisiología , Mucosa Olfatoria/virología , Animales , Cricetinae , Infecciones por Henipavirus/virología , Inmunohistoquímica , Microscopía Electrónica de Transmisión , Neuronas/patología , Neuronas/virología , Bulbo Olfatorio/patología , Bulbo Olfatorio/virología , Mucosa Olfatoria/patología , Internalización del Virus
10.
J Immunol ; 179(3): 1796-802, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17641046

RESUMEN

Sin Nombre virus (SNV) is a highly pathogenic New World virus and etiologic agent of hantavirus cardiopulmonary syndrome. We have previously shown that replication-defective virus particles are able to induce a strong IFN-stimulated gene (ISG) response in human primary cells. RNA viruses often stimulate the innate immune response by interactions between viral nucleic acids, acting as a pathogen-associated molecular pattern, and cellular pattern-recognition receptors (PRRs). Ligand binding to PRRs activates transcription factors which regulate the expression of antiviral genes, and in all systems examined thus far, IFN regulatory factor 3 (IRF3) has been described as an essential intermediate for induction of ISG expression. However, we now describe a model in which IRF3 is dispensable for the induction of ISG transcription in response to viral particles. IRF3-independent ISG transcription in human hepatoma cell lines is initiated early after exposure to SNV virus particles in an entry- and replication-independent fashion. Furthermore, using gene knockdown, we discovered that this activation is independent of the best-characterized RNA- and protein-sensing PRRs including the cytoplasmic caspase recruitment domain-containing RNA helicases and the TLRs. SNV particles engage a heretofore unrecognized PRR, likely located at the cell surface, and engage a novel IRF3-independent pathway that activates the innate immune response.


Asunto(s)
Inmunidad Innata , Factor 3 Regulador del Interferón/fisiología , Virus Sin Nombre/inmunología , Virus Sin Nombre/metabolismo , Receptores Toll-Like/fisiología , Internalización del Virus , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/virología , Línea Celular Tumoral , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , ARN Helicasas DEAD-box/fisiología , Regulación Viral de la Expresión Génica/inmunología , Regulación Viral de la Expresión Génica/efectos de la radiación , Humanos , Factor 7 Regulador del Interferón/fisiología , Interferones/fisiología , Receptores Inmunológicos , Receptores Virales/fisiología , Virus Sin Nombre/patogenicidad , Virus Sin Nombre/efectos de la radiación , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 3/fisiología , Rayos Ultravioleta , Virión/inmunología , Replicación Viral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA