Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Natl Compr Canc Netw ; 20(2): 160-166, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35130494

RESUMEN

BACKGROUND: Most safety and efficacy trials of the SARS-CoV-2 vaccines excluded patients with cancer, yet these patients are more likely than healthy individuals to contract SARS-CoV-2 and more likely to become seriously ill after infection. Our objective was to record short-term adverse reactions to the COVID-19 vaccine in patients with cancer, to compare the magnitude and duration of these reactions with those of patients without cancer, and to determine whether adverse reactions are related to active cancer therapy. PATIENTS AND METHODS: A prospective, single-institution observational study was performed at an NCI-designated Comprehensive Cancer Center. All study participants received 2 doses of the Pfizer BNT162b2 vaccine separated by approximately 3 weeks. A report of adverse reactions to dose 1 of the vaccine was completed upon return to the clinic for dose 2. Participants completed an identical survey either online or by telephone 2 weeks after the second vaccine dose. RESULTS: The cohort of 1,753 patients included 67.5% who had a history of cancer and 12.0% who were receiving active cancer treatment. Local pain at the injection site was the most frequently reported symptom for all respondents and did not distinguish patients with cancer from those without cancer after either dose 1 (39.3% vs 43.9%; P=.07) or dose 2 (42.5% vs 40.3%; P=.45). Among patients with cancer, those receiving active treatment were less likely to report pain at the injection site after dose 1 compared with those not receiving active treatment (30.0% vs 41.4%; P=.002). The onset and duration of adverse events was otherwise unrelated to active cancer treatment. CONCLUSIONS: When patients with cancer were compared with those without cancer, few differences in reported adverse events were noted. Active cancer treatment had little impact on adverse event profiles.


Asunto(s)
COVID-19 , Neoplasias , Vacuna BNT162 , Vacunas contra la COVID-19 , Humanos , Neoplasias/tratamiento farmacológico , Estudios Prospectivos , ARN Mensajero , SARS-CoV-2
2.
Virology ; 563: 38-43, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34416448

RESUMEN

BST2/tetherin is a transmembrane protein with antiviral activity; it is synthesized following exposure to interferons, and restricts the release of budding virus particles by tethering them to the host cell membrane. We previously showed that BST2 is induced in primary neurons following measles virus (MV) infection or type I interferon; however, BST2 was dispensable for protection against challenge with neuron-restricted MV. Here, we define the contribution of BST-2 in neuronal MV infection. Surprisingly, and in contrast to its antiviral role in non-neuronal cells, murine BST2 promotes MV infection in brains of permissive mice and in primary neuron cultures. Moreover, BST2 expression was predominantly observed in the non-synaptic fraction of purified neurons. These studies highlight a cell-type dependent role of a well-characterized antiviral protein in enhancing neuronal infection.


Asunto(s)
Antígenos CD/metabolismo , Virus del Sarampión/fisiología , Glicoproteínas de Membrana/metabolismo , Neuronas/virología , Animales , Antígenos CD/genética , Encéfalo/metabolismo , Encéfalo/virología , Regulación de la Expresión Génica , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Neuronas/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Sinapsis
3.
mBio ; 12(2)2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33758092

RESUMEN

Viruses, including members of the herpes-, entero-, and morbillivirus families, are the most common cause of infectious encephalitis in mammals worldwide. During most instances of acute viral encephalitis, neurons are typically the initial cell type that is infected. However, as replication and spread ensue, other parenchymal cells can become viral targets, especially in chronic infections. Consequently, to ascertain how neurotropic viruses trigger neuropathology, it is crucial to identify which central nervous system (CNS) cell populations are susceptible and permissive throughout the course of infection, and to define how viruses spread between distinct cell types. Using a measles virus (MV) transgenic mouse model that expresses human CD46 (hCD46), the MV vaccine strain receptor, under the control of a neuron-specific enolase promoter (NSE-hCD46+ mice), a novel mode of viral spread between neurons and astrocytes was identified. Although hCD46 is required for initial neuronal infection, it is dispensable for heterotypic spread to astrocytes, which instead depends on glutamate transporters and direct neuron-astrocyte contact. Moreover, in the presence of RNase A, astrocyte infection is reduced, suggesting that nonenveloped ribonucleoproteins (RNP) may cross the neuron-astrocyte synaptic cleft. The characterization of this novel mode of intercellular transport offers insights into the unique interaction of neurons and glia and may reveal therapeutic targets to mitigate the life-threatening consequences of measles encephalitis.IMPORTANCE Viruses are the most important cause of infectious encephalitis in mammals worldwide; several thousand people, primarily the very young and the elderly, are impacted annually, and few therapies are reliably successful once neuroinvasion has occurred. To understand how viruses contribute to neuropathology, and to develop tools to prevent or ameliorate such infections, it is crucial to define if and how viruses disseminate among the different cell populations within the highly complex central nervous system. This study defines a noncanonical mode of viral transmission between neurons and astrocytes within the brain.


Asunto(s)
Astrocitos/virología , Vacuna Antisarampión/análisis , Virus del Sarampión/fisiología , Neuronas/virología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Encefalitis Viral/virología , Femenino , Humanos , Masculino , Proteína Cofactora de Membrana/genética , Ratones , Ratones Transgénicos
4.
PLoS One ; 16(2): e0244334, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33544724

RESUMEN

Wild elephant populations are declining rapidly due to rampant killing for ivory and body parts, range fragmentation, and human-elephant conflict. Wild and captive elephants are further impacted by viruses, including highly pathogenic elephant endotheliotropic herpesviruses. Moreover, while the rich genetic diversity of the ancient elephant lineage is disappearing, elephants, with their low incidence of cancer, have emerged as a surprising resource in human cancer research for understanding the intrinsic cellular response to DNA damage. However, studies on cellular resistance to transformation and herpesvirus reproduction have been severely limited, in part due to the lack of established elephant cell lines to enable in vitro experiments. This report describes creation of a recombinant plasmid, pAelPyV-1-Tag, derived from a wild isolate of African Elephant Polyomavirus (AelPyV-1), that can be used to create immortalized lines of elephant cells. This isolate was extracted from a trunk nodule biopsy isolated from a wild African elephant, Loxodonta africana, in Botswana. The AelPyV-1 genome contains open-reading frames encoding the canonical large (LTag) and small (STag) tumor antigens. We cloned the entire early region spanning the LTag and overlapping STag genes from this isolate into a high-copy vector to construct a recombinant plasmid, pAelPyV-1-Tag, which effectively transformed primary elephant endothelial cells. We expect that the potential of this reagent to transform elephant primary cells will, at a minimum, facilitate study of elephant-specific herpesviruses.


Asunto(s)
Antígenos Virales de Tumores/genética , Genoma Viral , Infecciones por Polyomavirus/veterinaria , Poliomavirus/aislamiento & purificación , Infecciones Tumorales por Virus/veterinaria , Animales , Animales Salvajes , Elefantes , Células Endoteliales/virología , Infecciones por Polyomavirus/diagnóstico , Infecciones Tumorales por Virus/diagnóstico
5.
J Virol ; 94(9)2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32051270

RESUMEN

Influenza A viruses (IAV) are lytic viruses that have recently been found to activate necroptosis in many of the cell types they infect. Necroptotic cell death is potently immunogenic and limits IAV spread by directly eliminating infected cells and by mobilizing both innate and adaptive immune responses. The benefits of necroptosis to the host, however, may sometimes be outweighed by the potentially deleterious hyperinflammatory consequences of activating this death modality in pulmonary and other tissues.


Asunto(s)
Gripe Humana/metabolismo , Necroptosis/fisiología , Orthomyxoviridae/metabolismo , Animales , Apoptosis , Caspasa 8/metabolismo , Muerte Celular , Humanos , Virus de la Influenza A/metabolismo , Gripe Humana/virología , Necrosis , Orthomyxoviridae/patogenicidad , Infecciones por Orthomyxoviridae/metabolismo , Fosforilación , Proteínas Quinasas/metabolismo
6.
Trends Immunol ; 40(9): 842-856, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31439415

RESUMEN

Interferons (IFNs) comprise a pleiotropic family of signaling molecules that are often the first line of defense against viral infection. Inflammatory responses induced by IFN are generally well tolerated during peripheral infections; yet, the same degree of inflammation during infection of the central nervous system (CNS) could be catastrophic. Thus, IFN responses must be modified within the CNS to ensure host survival. In this review, we discuss emerging principles highlighting unique aspects of antiviral effects of IFN protection following neurotropic viral infection, chiefly using new techniques in rodent models. Evaluation of these unique responses provides insights into how the immune system eradicates or controls pathogens, while avoiding host damage. Defining these principles may have direct impact on the development of novel clinical approaches.


Asunto(s)
Antivirales/inmunología , Interferones/inmunología , Virosis/inmunología , Animales , Humanos
7.
J Immunol ; 203(5): 1348-1355, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31358656

RESUMEN

Receptor-interacting protein kinase 1 (RIPK1) regulates cell fate and proinflammatory signaling downstream of multiple innate immune pathways, including those initiated by TNF-α, TLR ligands, and IFNs. Genetic ablation of Ripk1 results in perinatal lethality arising from both RIPK3-mediated necroptosis and FADD/caspase-8-driven apoptosis. IFNs are thought to contribute to the lethality of Ripk1-deficient mice by activating inopportune cell death during parturition, but how IFNs activate cell death in the absence of RIPK1 is not understood. In this study, we show that Z-form nucleic acid binding protein 1 (ZBP1; also known as DAI) drives IFN-stimulated cell death in settings of RIPK1 deficiency. IFN-activated Jak/STAT signaling induces robust expression of ZBP1, which complexes with RIPK3 in the absence of RIPK1 to trigger RIPK3-driven pathways of caspase-8-mediated apoptosis and MLKL-driven necroptosis. In vivo, deletion of either Zbp1 or core IFN signaling components prolong viability of Ripk1-/- mice for up to 3 mo beyond parturition. Together, these studies implicate ZBP1 as the dominant activator of IFN-driven RIPK3 activation and perinatal lethality in the absence of RIPK1.


Asunto(s)
Muerte Celular/fisiología , Proteínas de Unión al ARN/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis/fisiología , Caspasa 8/metabolismo , Línea Celular , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología
8.
J Virol ; 93(18)2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31270232

RESUMEN

Genomic material from many neurotropic RNA viruses (e.g., measles virus [MV], West Nile virus [WNV], Sindbis virus [SV], rabies virus [RV], and influenza A virus [IAV]) remains detectable in the mouse brain parenchyma long after resolution of the acute infection. The presence of these RNAs in the absence of overt central nervous system (CNS) disease has led to the suggestion that they are viral remnants, with little or no potential to reactivate. Here we show that MV RNA remains detectable in permissive mouse neurons long after challenge with MV and, moreover, that immunosuppression can cause RNA and protein synthesis to rebound, triggering neuropathogenesis months after acute viral control. Robust recrudescence of viral transcription and protein synthesis occurs after experimental depletion of cells of the adaptive immune response and is associated with a loss of T resident memory (Trm) lymphocytes within the brain. The disease associated with loss of immune control is distinct from that seen during the acute infection: immune cell-depleted, long-term-infected mice display severe gait and motor problems, in contrast to the wasting and lethal disease that occur during acute infection of immunodeficient hosts. These results illuminate the potential consequences of noncytolytic, immune-mediated viral control in the CNS and demonstrate that what were once considered "resolved" RNA viral infections may, in fact, induce diseases later in life that are distinct from those caused by acute infection.IMPORTANCE Viral infections of neurons are often not cytopathic; thus, once-infected neurons survive, and viral RNAs can be detected long after apparent viral control. These RNAs are generally considered viral fossils, unlikely to contribute to central nervous system (CNS) disease. Using a mouse model of measles virus (MV) neuronal infection, we show that MV RNA is maintained in the CNS of infected mice long after acute control and in the absence of overt disease. Viral replication is suppressed by the adaptive immune response; when these immune cells are depleted, viral protein synthesis recurs, inducing a CNS disease that is distinct from that observed during acute infection. The studies presented here provide the basis for understanding how persistent RNA infections in the CNS are controlled by the host immune response, as well as the pathogenic consequences of noncytolytic viral control.


Asunto(s)
Virus del Sarampión/genética , Neuronas/virología , Infecciones por Virus ARN/virología , Animales , Encéfalo/virología , Sistema Nervioso Central/virología , Modelos Animales de Enfermedad , Femenino , Masculino , Sarampión/virología , Virus del Sarampión/metabolismo , Ratones , Ratones Transgénicos , Neuronas/metabolismo , ARN/genética , ARN/metabolismo , Infecciones por Virus ARN/genética , Infecciones por Virus ARN/metabolismo , Virus ARN/genética , Virus ARN/metabolismo
9.
J Cell Sci ; 131(17)2018 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-30154209

RESUMEN

T cell-mediated adaptive immunity requires naïve, unstimulated T cells to transition from a quiescent metabolic state into a highly proliferative state upon T cell receptor engagement. This complex process depends on transcriptional changes mediated by Ca2+-dependent NFAT signaling, mTOR-mediated signaling and increased activity of the guanine nucleotide biosynthetic inosine-5'-monophosphate (IMP) dehydrogenase 1 and 2 enzymes (IMPDH1 and IMPDH2, hereafter IMPDH). Inhibitors of these pathways serve as potent immunosuppressants. Unexpectedly, we discovered that all three pathways converge to promote the assembly of IMPDH protein into micron-scale macromolecular filamentous structures in response to T cell activation. Assembly is post-transcriptionally controlled by mTOR and the Ca2+ influx regulator STIM1. Furthermore, IMPDH assembly and catalytic activity were negatively regulated by guanine nucleotide levels, suggesting a negative feedback loop that limits biosynthesis of guanine nucleotides. Filamentous IMPDH may be more resistant to this inhibition, facilitating accumulation of the higher GTP levels required for T cell proliferation.


Asunto(s)
IMP Deshidrogenasa/metabolismo , Linfocitos T/enzimología , Animales , Células Cultivadas , Nucleótidos de Guanina/metabolismo , IMP Deshidrogenasa/genética , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Bazo/enzimología , Bazo/inmunología , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo , Linfocitos T/inmunología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
10.
J Neuroimmunol ; 308: 25-29, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28187911

RESUMEN

Central nervous system consequences of viral infections are rare, but when they do occur, they are often serious and clinically challenging to manage. Our awareness of the perils of neuroinvasion by viruses is growing: the recently appreciated impact of Ebola and Zika virus infections on CNS integrity, decreases in vaccination coverage for potentially neurotropic viruses such as measles, and increased neurovirulence of some influenza strains collectively highlight the need for a better understanding of the viral-neural interaction. Defining these interactions and how they result in neuropathogenesis is paramount for the development of better clinical strategies, especially given the limited treatment options that are available due to the unique physiology of the brain that limits migration of blood-borne molecules into the CNS parenchyma. In this perspective, we discuss some unique aspects of neuronal viral infections and immune-mediated control that impact the pathogenic outcomes of these infections. Further, we draw attention to an often overlooked aspect of neuropathogenesis research: that lack of overt disease, which is often equated with survival post-infection, likely only scratches the surface of the myriad ways by which neurotropic infections can impair CNS function.


Asunto(s)
Enfermedades Virales del Sistema Nervioso Central/mortalidad , Sistema Nervioso Central/patología , Estimación de Kaplan-Meier , Animales , Sistema Nervioso Central/virología , Enfermedades Virales del Sistema Nervioso Central/genética , Modelos Animales de Enfermedad , Humanos , Interferón gamma/deficiencia , Interferón gamma/genética , Proteína Cofactora de Membrana/deficiencia , Proteína Cofactora de Membrana/genética , Ratones , Ratones Transgénicos , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Factor de Transcripción STAT1/deficiencia , Factor de Transcripción STAT1/genética
11.
Nat Rev Neurosci ; 17(12): 766-776, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27811921

RESUMEN

It is becoming clear that the manner by which the immune response resolves or contains infection by a pathogen varies according to the tissue that is affected. Unlike many peripheral cell types, CNS neurons are generally non-renewable. Thus, the cytolytic and inflammatory strategies that are effective in controlling infections in the periphery could be damaging if deployed in the CNS. Perhaps for this reason, the immune response to some CNS viral infections favours maintenance of neuronal integrity and non-neurolytic viral control. This modified immune response - when combined with the unique anatomy and physiology of the CNS - provides an ideal environment for the maintenance of viral genomes, including those of RNA viruses. Therefore, it is possible that such viruses can reactivate long after initial viral exposure, contributing to CNS disease.


Asunto(s)
Encéfalo/inmunología , Inmunidad Innata/fisiología , Neuronas/inmunología , Infecciones por Virus ARN/inmunología , Virus ARN/inmunología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Enfermedades del Sistema Nervioso Central/inmunología , Enfermedades del Sistema Nervioso Central/metabolismo , Enfermedades del Sistema Nervioso Central/patología , Humanos , Neuronas/metabolismo , Neuronas/patología , Infecciones por Virus ARN/metabolismo , Infecciones por Virus ARN/patología , Virus ARN/metabolismo
12.
Virology ; 499: 196-202, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27677156

RESUMEN

Immunity within the brain, specifically to virus-infected neurons, must be controlled to prevent neuron loss and impairment, though the process by which this occurs remains unclear. Here, we use a mouse model of neuron-restricted measles virus infection, in which immunocompetent adults survive challenge, whereas T and B cell-deficient mice succumb. This model allowed us to more precisely define the contributions of CD4+ T cells, CD8+ T cells, and B cells in neuroprotection. Both B cell knockout mice and mice depleted of CD8+ T cells survive challenge and show no signs of illness, though are less able to control viral replication than immunocompetent mice. In contrast, depletion of CD4+ T cells results in disease and death in all infected mice, though the kinetics of illness are delayed compared to RAG knockout mice. Our data suggest a coordinated interplay of adaptive immune components, which collectively controls viral spread and limits neuropathogenesis.


Asunto(s)
Linfocitos B/inmunología , Encéfalo/virología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Virus del Sarampión/fisiología , Virus del Sarampión/patogenicidad , Sarampión/inmunología , Animales , Encéfalo/inmunología , Femenino , Humanos , Masculino , Sarampión/virología , Ratones , Ratones Endogámicos C57BL , Tropismo Viral , Virulencia
13.
ACS Chem Neurosci ; 7(4): 435-41, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-26854733

RESUMEN

The central nervous system (CNS) coordinates all aspects of life, autonomic and sentient, though how it has evolved to contend with pathogenic infections remains, to a great degree, a mystery. The skull and cerebrospinal fluid (CSF) provide protection from blunt force contacts, and it was once thought that the blood-brain barrier (BBB) was a fortress that restricted pathogen entry and limited inflammation. Recent studies, however, have caused a revision of this viewpoint: the CNS is monitored by blood-borne lymphocytes, but can use alternative strategies to prevent or resolve many pathogenic challenges. In this Review, we discuss emerging principles that indicate how the CNS is immunologically unique from peripheral tissues. We focus on developments that include glymphatics, recently characterized brain lymphatic vessels, distinctions in innate and adaptive immune strategies, novel points of entry for neurotropic viruses, and, finally, how the periphery can influence CNS homeostasis and immune responses within the brain. Collectively, these attributes demand a re-evaluation of immunity in the brain: not privileged, but distinct.


Asunto(s)
Encéfalo , Enfermedades Virales del Sistema Nervioso Central , Neuroinmunomodulación/fisiología , Animales , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/fisiopatología , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/virología , Enfermedades Virales del Sistema Nervioso Central/inmunología , Enfermedades Virales del Sistema Nervioso Central/patología , Humanos , Inflamación , Sistema Linfático/inmunología , Sistema Linfático/metabolismo , Sistema Linfático/virología , Neuronas/clasificación , Neuronas/inmunología , Neuronas/virología
14.
J Virol ; 89(21): 11011-8, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26311886

RESUMEN

UNLABELLED: In permissive mouse central nervous system (CNS) neurons, measles virus (MV) spreads in the absence of hallmark viral budding or neuronal death, with transmission occurring efficiently and exclusively via the synapse. MV infection also initiates a robust type I interferon (IFN) response, resulting in the synthesis of a large number of genes, including bone marrow stromal antigen 2 (Bst2)/tetherin/CD317. Bst2 restricts the release of some enveloped viruses, but to date, its role in viral infection of neurons has not been assessed. Consequently, we investigated how Bst2 was induced and what role it played in MV neuronal infection. The magnitude of induction of neuronal Bst2 RNA and protein following IFN exposure and viral infection was notably higher than in similarly treated mouse embryo fibroblasts (MEFs). Bst2 synthesis was both IFN and Stat1 dependent. Although Bst2 prevented MV release from nonneuronal cells, its deletion had no effect on viral pathogenesis in MV-challenged mice. Our findings underscore how cell-type-specific differences impact viral infection and pathogenesis. IMPORTANCE: Viral infections of the central nervous system can lead to debilitating disease and death. Moreover, it is becoming increasingly clear that nonrenewable cells, including most central nervous system neurons, combat neurotropic viral infections in fundamentally different ways than other rapidly dividing and renewable cell populations. Here we identify type I interferon signaling as a key inducer of a known antiviral protein (Bst2) in neurons. Unexpectedly, the gene is dispensable for clearance of neurotropic viral infection despite its well-defined contribution to limiting the spread of enveloped viruses in proliferating cells. A deeper appreciation of the importance of cell type heterogeneity in antiviral immunity will aid in the identification of unique therapeutic targets for life-threatening viral infections.


Asunto(s)
Antígenos CD/metabolismo , Regulación Viral de la Expresión Génica/fisiología , Interferón Tipo I/metabolismo , Virus del Sarampión/fisiología , Sarampión/inmunología , Glicoproteínas de Membrana/metabolismo , Neuronas/metabolismo , Análisis de Varianza , Animales , Western Blotting , Células Cultivadas , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Hipocampo/citología , Ratones , Neuronas/virología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Neurochem ; 135(2): 309-22, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26190522

RESUMEN

The signal transduction molecule, Stat1, is critical for the expression of type I and II interferon (IFN)-responsive genes in most cells; however, we previously showed that primary hippocampal mouse neurons express low basal Stat1, with delayed and attenuated expression of IFN-responsive genes. Moreover, IFNγ-dependent resolution of a neurotropic viral challenge in permissive mice is Stat1-independent. Here, we show that exogenous IFNγ has no deleterious impact on neuronal viability, and staurosporine-induced apoptosis in neurons is significantly blunted by the addition of IFNγ, suggesting that IFNγ confers a pro-survival signal in neurons. To identify the pathways induced by IFNγ in neurons, the activation of alternative signal transducers associated with IFNγ signaling was assessed. Rapid and pronounced activation of extracellular signal regulated kinase (Erk1/2) was observed in neurons, compared to a modest response in fibroblasts. Moreover, the absence of Stat1 in primary fibroblasts led to enhanced Erk activation following IFNγ addition, implying that the cell-specific availability of signal transducers can diversify the cellular response following IFN engagement.


Asunto(s)
Interferón gamma/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Antimetabolitos/farmacología , Bromodesoxiuridina/farmacología , Supervivencia Celular/efectos de los fármacos , Citosol/metabolismo , Femenino , Hipocampo/citología , Hipocampo/embriología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Transgénicos , Neuroglía/efectos de los fármacos , Embarazo , Cultivo Primario de Células , Inhibidores de Proteínas Quinasas/farmacología , Estaurosporina/farmacología
16.
J Neuroimmunol ; 279: 11-9, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25669994

RESUMEN

The mechanisms by which neurons respond to inflammatory mediators such as interferons (IFNs) remain largely undefined. We previously showed that the activation and nuclear localization of the core IFN signaling molecule, Stat1, are muted and delayed in primary mouse hippocampal neurons treated with IFN gamma as compared to control mouse embryonic fibroblasts (MEFs). Here, we show that the kinetics of Stat1 and Stat2 activation following type I IFN exposure are also unique in neurons, affecting gene expression and neuronal response. Specifically, despite lower basal expression of many IFN stimulated genes in neurons, basal expression of the type I IFN themselves is significantly higher in primary hippocampal neurons compared to MEF. Elevated homeostatic IFN in neurons is critical and sufficient for early control of viral infection. These data provide further evidence that neurons exploit unique signaling responses to IFNs, and define an important contribution of homeostatic IFN within the CNS. Such differences are likely critical for the ability of neurons to survive a viral challenge.


Asunto(s)
Interferones/metabolismo , Virus del Sarampión/patogenicidad , Sarampión/metabolismo , Neuronas/metabolismo , 2',5'-Oligoadenilato Sintetasa/genética , 2',5'-Oligoadenilato Sintetasa/metabolismo , Animales , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Hipocampo/citología , Interferones/genética , Interferones/farmacología , Sarampión/patología , Proteína Cofactora de Membrana/genética , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/virología , Fosfopiruvato Hidratasa/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Factores de Tiempo , Ubiquitinas/genética , Ubiquitinas/metabolismo
17.
Annu Rev Virol ; 2(1): 451-71, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26958924

RESUMEN

The cultural impact of rabies, the fatal neurological disease caused by infection with rabies virus, registers throughout recorded history. Although rabies has been the subject of large-scale public health interventions, chiefly through vaccination efforts, the disease continues to take the lives of about 40,000-70,000 people per year, roughly 40% of whom are children. Most of these deaths occur in resource-poor countries, where lack of infrastructure prevents timely reporting and postexposure prophylaxis and the ubiquity of domestic and wild animal hosts makes eradication unlikely. Moreover, although the disease is rarer than other human infections such as influenza, the prognosis following a bite from a rabid animal is poor: There is currently no effective treatment that will save the life of a symptomatic rabies patient. This review focuses on the major unanswered research questions related to rabies virus pathogenesis, especially those connecting the disease progression of rabies with the complex dysfunction caused by the virus in infected cells. The recent applications of cutting-edge research strategies to this question are described in detail.


Asunto(s)
Virus de la Rabia/fisiología , Rabia/virología , Animales , Humanos , Conocimiento , Rabia/psicología , Virus de la Rabia/genética , Virus de la Rabia/patogenicidad , Virulencia
18.
Proc Natl Acad Sci U S A ; 110(33): E3109-18, 2013 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-23898178

RESUMEN

Interferons (IFNs) are cytokines with powerful immunomodulatory and antiviral properties, but less is known about how they induce cell death. Here, we show that both type I (α/ß) and type II (γ) IFNs induce precipitous receptor-interacting protein (RIP)1/RIP3 kinase-mediated necrosis when the adaptor protein Fas-associated death domain (FADD) is lost or disabled by phosphorylation, or when caspases (e.g., caspase 8) are inactivated. IFN-induced necrosis proceeds via progressive assembly of a RIP1-RIP3 "necrosome" complex that requires Jak1/STAT1-dependent transcription, but does not need the kinase activity of RIP1. Instead, IFNs transcriptionally activate the RNA-responsive protein kinase PKR, which then interacts with RIP1 to initiate necrosome formation and trigger necrosis. Although IFNs are powerful activators of necrosis when FADD is absent, these cytokines are likely not the dominant inducers of RIP kinase-driven embryonic lethality in FADD-deficient mice. We also identify phosphorylation on serine 191 as a mechanism that disables FADD and collaborates with caspase inactivation to allow IFN-activated necrosis. Collectively, these findings outline a mechanism of IFN-induced RIP kinase-dependent necrotic cell death and identify FADD and caspases as negative regulators of this process.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Interferón gamma/metabolismo , Modelos Moleculares , Necrosis/metabolismo , Transducción de Señal/fisiología , Animales , Células Cultivadas , Electroforesis en Gel de Poliacrilamida , Proteína de Dominio de Muerte Asociada a Fas/química , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteínas Activadoras de GTPasa/metabolismo , Inmunoprecipitación , Ratones , Ratones Noqueados , Fosforilación , Interferencia de ARN , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Factor de Transcripción STAT1/metabolismo , eIF-2 Quinasa/metabolismo
19.
PLoS Pathog ; 8(10): e1002971, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23071441

RESUMEN

Rabies virus (RABV) is a highly neurotropic pathogen that typically leads to mortality of infected animals and humans. The precise etiology of rabies neuropathogenesis is unknown, though it is hypothesized to be due either to neuronal death or dysfunction. Analysis of human brains post-mortem reveals surprisingly little tissue damage and neuropathology considering the dramatic clinical symptomology, supporting the neuronal dysfunction model. However, whether or not neurons survive infection and clearance and, provided they do, whether they are functionally restored to their pre-infection phenotype has not been determined in vivo for RABV, or any neurotropic virus. This is due, in part, to the absence of a permanent "mark" on once-infected cells that allow their identification long after viral clearance. Our approach to study the survival and integrity of RABV-infected neurons was to infect Cre reporter mice with recombinant RABV expressing Cre-recombinase (RABV-Cre) to switch neurons constitutively expressing tdTomato (red) to expression of a Cre-inducible EGFP (green), permanently marking neurons that had been infected in vivo. We used fluorescence microscopy and quantitative real-time PCR to measure the survival of neurons after viral clearance; we found that the vast majority of RABV-infected neurons survive both infection and immunological clearance. We were able to isolate these previously infected neurons by flow cytometry and assay their gene expression profiles compared to uninfected cells. We observed transcriptional changes in these "cured" neurons, predictive of decreased neurite growth and dysregulated microtubule dynamics. This suggests that viral clearance, though allowing for survival of neurons, may not restore them to their pre-infection functionality. Our data provide a proof-of-principle foundation to re-evaluate the etiology of human central nervous system diseases of unknown etiology: viruses may trigger permanent neuronal damage that can persist or progress in the absence of sustained viral antigen.


Asunto(s)
Encéfalo/virología , Neuronas/fisiología , Virus de la Rabia/inmunología , Rabia/inmunología , Animales , Encéfalo/inmunología , Encéfalo/patología , Supervivencia Celular , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Integrasas/genética , Ratones , Neuronas/inmunología , Neuronas/virología , Rabia/genética , Rabia/patología , Rabia/virología , Virus de la Rabia/patogenicidad , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcriptoma
20.
J Neuroimmunol ; 251(1-2): 33-8, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22769061

RESUMEN

Although interferon-gamma (IFN-γ) plays a critical role in the noncytolytic elimination of many neurotropic viral infections, the signaling response to this cytokine has not been extensively characterized in primary CNS neurons. We previously demonstrated that the IFN-γ response at the signaling and gene expression levels is temporally extended in primary mouse hippocampal neurons, as compared to the transient response of primary mouse embryonic fibroblasts (MEF). We hypothesize that the protracted kinetics of STAT1 phosphorylation in IFN-γ-treated neurons are due to extended receptor activation and/or delayed STAT1 dephosphorylation in the nucleus. Here, we show that in response to IFN-γ, the Janus kinases (JAK1/JAK2) associated with the neuronal IFN-γ receptor complex remain active for an extended period as compared to MEF. Experimental inactivation of JAK1/JAK2 in neurons after IFN-γ treatment did not reverse the extended STAT1 phosphorylation phenotype. These results suggest that the extended kinetics of neuronal IFN-γ signaling are a product of distinct negative feedback mechanisms operating at both the receptor and within the nucleus.


Asunto(s)
Hipocampo/metabolismo , Interferón gamma/metabolismo , Janus Quinasa 1/metabolismo , Janus Quinasa 2/metabolismo , Neuronas/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Interferón gamma/farmacología , Ratones , Neuronas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...