Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Intervalo de año de publicación
1.
Science ; 385(6704): eadk4898, 2024 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-38781354

RESUMEN

After infection of B cells, Epstein-Barr virus (EBV) engages host pathways that mediate cell proliferation and transformation, contributing to the propensity of the virus to drive immune dysregulation and lymphomagenesis. We found that the EBV protein EBNA2 initiates nicotinamide adenine dinucleotide (NAD) de novo biosynthesis by driving expression of the metabolic enzyme indoleamine 2,3-dioxygenase 1 (IDO1) in infected B cells. Virus-enforced NAD production sustained mitochondrial complex I activity, to match adenosine triphosphate (ATP) production with bioenergetic requirements of proliferation and transformation. In transplant patients, IDO1 expression in EBV-infected B cells, and a serum signature of increased IDO1 activity, preceded development of lymphoma. In humanized mice infected with EBV, IDO1 inhibition reduced both viremia and lymphomagenesis. Virus-orchestrated NAD biosynthesis is therefore a druggable metabolic vulnerability of EBV-driven B cell transformation, opening therapeutic possibilities for EBV-related diseases.


Asunto(s)
Adenosina Trifosfato , Linfocitos B , Transformación Celular Viral , Infecciones por Virus de Epstein-Barr , Antígenos Nucleares del Virus de Epstein-Barr , Herpesvirus Humano 4 , Indolamina-Pirrol 2,3,-Dioxigenasa , NAD , Animales , Humanos , Ratones , Adenosina Trifosfato/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Proliferación Celular , Complejo I de Transporte de Electrón/metabolismo , Infecciones por Virus de Epstein-Barr/virología , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/fisiología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Linfoma/virología , NAD/metabolismo , Proteínas Virales , Viremia
2.
Nat Immunol ; 24(12): 2008-2020, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38012409

RESUMEN

Our increased understanding of how key metabolic pathways are activated and regulated in malignant cells has identified metabolic vulnerabilities of cancers. Translating this insight to the clinics, however, has proved challenging. Roadblocks limiting efficacy of drugs targeting cancer metabolism may lie in the nature of the metabolic ecosystem of tumors. The exchange of metabolites and growth factors between cancer cells and nonmalignant tumor-resident cells is essential for tumor growth and evolution, as well as the development of an immunosuppressive microenvironment. In this Review, we will examine the metabolic interplay between tumor-resident cells and how targeted inhibition of specific metabolic enzymes in malignant cells could elicit pro-tumorigenic effects in non-transformed tumor-resident cells and inhibit the function of tumor-specific T cells. To improve the efficacy of metabolism-targeted anticancer strategies, a holistic approach that considers the effect of metabolic inhibitors on major tumor-resident cell populations is needed.


Asunto(s)
Ecosistema , Neoplasias , Humanos , Neoplasias/metabolismo , Carcinogénesis , Linfocitos T/metabolismo , Microambiente Tumoral
3.
J Autoimmun ; 124: 102714, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34403915

RESUMEN

BACKGROUND: Viral infections may trigger autoimmunity in genetically predisposed individuals. Immunizations mimic viral infections immunologically, but only in rare instances vaccinations coincide with the onset of autoimmunity. Inadvertent vaccine injection into periarticular shoulder tissue can cause inflammatory tissue damage ('shoulder injury related to vaccine administration, SIRVA). Thus, this accident provides a model to study if vaccine-induced pathogen-specific immunity accompanied by a robust inflammatory insult may trigger autoimmunity in specific genetic backgrounds. METHODS: We studied 16 otherwise healthy adults with suspected SIRVA occurring following a single work-related influenza immunization campaign in 2017. We performed ultrasound, immunophenotypic analyses, HLA typing, and influenza- and self-reactivity functional immunoassays. Vaccine-related bone toxicity and T cell/osteoclast interactions were assessed in vitro. FINDINGS: Twelve of the 16 subjects had evidence of inflammatory tissue damage on imaging, including bone erosions in six. Tissue damage was associated with a robust peripheral blood T and B cell activation signature and extracellular matrix-reactive autoantibodies. All subjects with erosions were HLA-DRB1*04 positive and showed extracellular matrix-reactive HLA-DRB1*04 restricted T cell responses targeting heparan sulfate proteoglycan (HSPG). Antigen-specific T cells potently activated osteoclasts via RANK/RANK-L, and the osteoclast activation marker Trap5b was high in sera of patients with an erosive shoulder injury. In vitro, the vaccine component alpha-tocopheryl succinate recapitulated bone toxicity and stimulated osteoclasts. Auto-reactivity was transient, with no evidence of progression to rheumatoid arthritis or overt autoimmune disease. CONCLUSION: Vaccine misapplication, potentially a genetic predisposition, and vaccine components contribute to SIRVA. The association with autoimmunity risk allele HLA-DRB1*04 needs to be further investigated. Despite transient autoimmunity, SIRVA was not associated with progression to autoimmune disease during two years of follow-up.


Asunto(s)
Inflamación/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Cápsula Articular/inmunología , Orthomyxoviridae/fisiología , Osteoclastos/inmunología , Linfocitos T/inmunología , Adulto , Autoinmunidad , Enfermedad Crónica , Matriz Extracelular/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Cadenas HLA-DRB1/genética , Proteoglicanos de Heparán Sulfato/inmunología , Prueba de Histocompatibilidad , Humanos , Masculino , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Fosfatasa Ácida Tartratorresistente/sangre , Vacunación/efectos adversos , Adulto Joven
4.
Theranostics ; 11(9): 4011-4029, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33754045

RESUMEN

Rationale: Adenylosuccinate lyase (ADSL) is an essential enzyme for de novo purine biosynthesis. Here we sought to investigate the putative role of ADSL in colorectal carcinoma (CRC) carcinogenesis and response to antimetabolites. Methods: ADSL expression levels were assessed by immunohistochemistry or retrieved from The Cancer Genome Atlas (TCGA) dataset. The effects of ADSL silencing or overexpression were evaluated on CRC cell proliferation, cell migration and cell-cycle. In vivo tumor growth was assessed by the chicken chorioallantoic membrane (CAM). Transfected cell lines or patient-derived organoids (PDO) were treated with 5-fluorouracil (5-FU) and 6-mercaptopurine (6-MP) and drug response was correlated with ADSL expression levels. Metabolomic and transcriptomic profiling were performed to identify dysregulated pathways and ADSL downstream effectors. Mitochondrial respiration and glycolytic capacity were measured using Seahorse; mitochondrial membrane potential and the accumulation of ROS were measured by FACS using MitoTracker Red and MitoSOX staining, respectively. Activation of canonical pathways was assessed by immunohistochemistry and immunoblotting. Results: ADSL expression is significantly increased in CRC tumors compared to non-tumor tissue. ADSL-high CRCs show upregulation of genes involved in DNA synthesis, DNA repair and cell cycle. Accordingly, ADSL overexpression accelerated progression through the cell cycle and significantly increased proliferation and migration in CRC cell lines. Additionally, ADSL expression increased tumor growth in vivo and sensitized CRCs to 6-MP in vitro, ex vivo (PDOs) and in vivo (CAM model). ADSL exerts its oncogenic function by affecting mitochondrial function via alteration of the TCA cycle and impairment of mitochondrial respiration. The KEAP1-NRF2 and mTORC1-cMyc axis are independently activated upon ADSL overexpression and may favor the survival and proliferation of ROS-accumulating cells, favoring DNA damage and tumorigenesis. Conclusions: Our results suggest that ADSL is a novel oncogene in CRC, modulating mitochondrial function, metabolism and oxidative stress, thus promoting cell cycle progression, proliferation and migration. Our results also suggest that ADSL is a predictive biomarker of response to 6-mercaptopurine in the pre-clinical setting.


Asunto(s)
Adenilosuccinato Liasa/genética , Neoplasias Colorrectales/genética , Mitocondrias/genética , Factor 2 Relacionado con NF-E2/genética , Oncogenes/genética , Proteínas Proto-Oncogénicas c-myc/genética , Serina-Treonina Quinasas TOR/genética , Células CACO-2 , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Respiración de la Célula/genética , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica/genética , Células HT29 , Humanos , Mitocondrias/patología
5.
Nat Commun ; 12(1): 1460, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33674584

RESUMEN

Mitochondria are important regulators of macrophage polarisation. Here, we show that arginase-2 (Arg2) is a microRNA-155 (miR-155) and interleukin-10 (IL-10) regulated protein localized at the mitochondria in inflammatory macrophages, and is critical for IL-10-induced modulation of mitochondrial dynamics and oxidative respiration. Mechanistically, the catalytic activity and presence of Arg2 at the mitochondria is crucial for oxidative phosphorylation. We further show that Arg2 mediates this process by increasing the activity of complex II (succinate dehydrogenase). Moreover, Arg2 is essential for IL-10-mediated downregulation of the inflammatory mediators succinate, hypoxia inducible factor 1α (HIF-1α) and IL-1ß in vitro. Accordingly, HIF-1α and IL-1ß are highly expressed in an LPS-induced in vivo model of acute inflammation using Arg2-/- mice. These findings shed light on a new arm of IL-10-mediated metabolic regulation, working to resolve the inflammatory status of the cell.


Asunto(s)
Arginasa/metabolismo , Interleucina-10/metabolismo , Macrófagos/metabolismo , Mitocondrias/metabolismo , Animales , Arginasa/genética , Regulación hacia Abajo , Femenino , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados/genética , Mitocondrias/enzimología , Succinato Deshidrogenasa/metabolismo
6.
Sci Signal ; 12(599)2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31530731

RESUMEN

Transforming growth factor-ß (TGF-ß) is produced by tumors, and increased amounts of this cytokine in the tumor microenvironment and serum are associated with poor patient survival. TGF-ß-mediated suppression of antitumor T cell responses contributes to tumor growth and survival. However, TGF-ß also has tumor-suppressive activity; thus, dissecting cell type-specific molecular effects may inform therapeutic strategies targeting this cytokine. Here, using human peripheral and tumor-associated lymphocytes, we investigated how tumor-derived TGF-ß suppresses a key antitumor function of CD4+ T cells, interferon-γ (IFN-γ) production. Suppression required the expression and phosphorylation of Smad proteins in the TGF-ß signaling pathway, but not their nuclear translocation, and depended on oxygen availability, suggesting a metabolic basis for these effects. Smad proteins were detected in the mitochondria of CD4+ T cells, where they were phosphorylated upon treatment with TGF-ß. Phosphorylated Smad proteins were also detected in the mitochondria of isolated tumor-associated lymphocytes. TGF-ß substantially impaired the ATP-coupled respiration of CD4+ T cells and specifically inhibited mitochondrial complex V (ATP synthase) activity. Last, inhibition of ATP synthase alone was sufficient to impair IFN-γ production by CD4+ T cells. These results, which have implications for human antitumor immunity, suggest that TGF-ß targets T cell metabolism directly, thus diminishing T cell function through metabolic paralysis.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Interferón gamma/inmunología , Mitocondrias/inmunología , Neoplasias/inmunología , Consumo de Oxígeno/inmunología , Factor de Crecimiento Transformador beta/inmunología , Adenosina Trifosfato/inmunología , Adenosina Trifosfato/metabolismo , Animales , Linfocitos T CD4-Positivos/metabolismo , Humanos , Interferón gamma/metabolismo , Mitocondrias/metabolismo , ATPasas de Translocación de Protón Mitocondriales/inmunología , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Fosforilación/inmunología , Transducción de Señal/inmunología , Proteínas Smad/inmunología , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/sangre , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral/inmunología
7.
Nat Immunol ; 20(10): 1311-1321, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31527833

RESUMEN

Whether screening the metabolic activity of immune cells facilitates discovery of molecular pathology remains unknown. Here we prospectively screened the extracellular acidification rate as a measure of glycolysis and the oxygen consumption rate as a measure of mitochondrial respiration in B cells from patients with primary antibody deficiency. The highest oxygen consumption rate values were detected in three study participants with persistent polyclonal B cell lymphocytosis (PPBL). Exome sequencing identified germline mutations in SDHA, which encodes succinate dehydrogenase subunit A, in all three patients with PPBL. SDHA gain-of-function led to an accumulation of fumarate in PPBL B cells, which engaged the KEAP1-Nrf2 system to drive the transcription of genes encoding inflammatory cytokines. In a single patient trial, blocking the activity of the cytokine interleukin-6 in vivo prevented systemic inflammation and ameliorated clinical disease. Overall, our study has identified pathological mitochondrial retrograde signaling as a disease modifier in primary antibody deficiency.


Asunto(s)
Linfocitos B/inmunología , Complejo II de Transporte de Electrones/genética , Inflamación/metabolismo , Linfocitosis/inmunología , Mitocondrias/metabolismo , Mutación/genética , Antiinflamatorios/farmacología , Respiración de la Célula , Células Cultivadas , Fumaratos/metabolismo , Glucólisis , Humanos , Inflamación/genética , Interleucina-6/antagonistas & inhibidores , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Consumo de Oxígeno , Estudios Prospectivos , Transducción de Señal , Secuenciación del Exoma
8.
Immunity ; 48(3): 542-555.e6, 2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29523440

RESUMEN

Glycolysis is linked to the rapid response of memory CD8+ T cells, but the molecular and subcellular structural elements enabling enhanced glucose metabolism in nascent activated memory CD8+ T cells are unknown. We found that rapid activation of protein kinase B (PKB or AKT) by mammalian target of rapamycin complex 2 (mTORC2) led to inhibition of glycogen synthase kinase 3ß (GSK3ß) at mitochondria-endoplasmic reticulum (ER) junctions. This enabled recruitment of hexokinase I (HK-I) to the voltage-dependent anion channel (VDAC) on mitochondria. Binding of HK-I to VDAC promoted respiration by facilitating metabolite flux into mitochondria. Glucose tracing pinpointed pyruvate oxidation in mitochondria, which was the metabolic requirement for rapid generation of interferon-γ (IFN-γ) in memory T cells. Subcellular organization of mTORC2-AKT-GSK3ß at mitochondria-ER contact sites, promoting HK-I recruitment to VDAC, thus underpins the metabolic reprogramming needed for memory CD8+ T cells to rapidly acquire effector function.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Retículo Endoplásmico/metabolismo , Metabolismo Energético , Memoria Inmunológica , Mitocondrias/metabolismo , Transducción de Señal , Respiración de la Célula , Retículo Endoplásmico/ultraestructura , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucólisis , Membranas Intracelulares/metabolismo , Activación de Linfocitos , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Mitocondrias/ultraestructura , Modelos Biológicos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina/deficiencia
9.
Nat Rev Immunol ; 18(1): 19-34, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28944771

RESUMEN

In healthy individuals, metabolically quiescent T cells survey lymph nodes and peripheral tissues in search of cognate antigens. During infection, T cells that encounter cognate antigens are activated and - in a context-specific manner - proliferate and/or differentiate to become effector T cells. This process is accompanied by important changes in cellular metabolism (known as metabolic reprogramming). The magnitude and spectrum of metabolic reprogramming as it occurs in T cells in the context of acute infection ensure host survival. By contrast, altered T cell metabolism, and hence function, is also observed in various disease states, in which T cells actively contribute to pathology. In this Review, we introduce the idea that the spectrum of immune cell metabolic states can provide a basis for categorizing human diseases. Specifically, we first summarize the metabolic and interlinked signalling requirements of T cells responding to acute infection. We then discuss how metabolic reprogramming of T cells is linked to disease.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Infecciones/metabolismo , Inflamación/metabolismo , Síndrome Metabólico/metabolismo , Neoplasias/metabolismo , Linfocitos T/metabolismo , Aminoácidos/metabolismo , Enfermedades Autoinmunes/inmunología , Glucosa/metabolismo , Humanos , Infecciones/inmunología , Inflamación/inmunología , Metabolismo de los Lípidos , Ganglios Linfáticos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Síndrome Metabólico/inmunología , Neoplasias/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Escape del Tumor/inmunología , Microambiente Tumoral/inmunología
11.
Eur J Immunol ; 44(12): 3614-20, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25251877

RESUMEN

ATP-binding cassette (ABC) transporters, including ABC-transporter B1 (ABCB1), extrude drugs, metabolites, and other compounds (such as mitotracker green (MTG)) from cells. Susceptibility of CD4(+) regulatory T (Treg) cells to the ABCB1-substrate cyclophosphamide (CPA) has been reported. Here, we characterized ABCB1 expression and function in human CD4(+) T-cell subsets. Naïve, central memory, and effector-memory CD4(+) T cells, but not Treg cells, effluxed MTG in an ABCB1-dependent manner. In line with this, ABCB1 mRNA and protein was expressed by nonregulatory CD4(+) T-cell subsets, but not Treg cells. In vitro, the ABCB1-substrate CPA was cytotoxic for Treg cells at a 100-fold lower dose than for nonregulatory counterparts, and, inversely, verapamil, an inhibitor of ABC transporters, increased CPA-toxicity in nonregulatory CD4(+) T cells but not Treg cells. Thus, Treg cells lack expression of ABCB1, rendering them selectively susceptible to CPA. Our findings provide mechanistic support for therapeutic strategies using CPA to boost anti-tumor immunity by selectively depleting Treg cells.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Ciclofosfamida/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Subfamilia B de Transportador de Casetes de Unión a ATP/inmunología , Antineoplásicos Alquilantes/farmacocinética , Apoptosis/inmunología , Ciclofosfamida/farmacocinética , Citotoxinas/farmacocinética , Citotoxinas/farmacología , Femenino , Regulación de la Expresión Génica/inmunología , Humanos , Depleción Linfocítica/métodos , Masculino , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Linfocitos T Reguladores/citología
12.
Nat Immunol ; 14(10): 1064-72, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23955661

RESUMEN

Antigen-experienced memory T cells acquire effector function with innate-like kinetics; however, the metabolic requirements of these cells are unknown. Here we show that rapid interferon-γ (IFN-γ) production of effector memory (EM) CD8(+) T cells, activated through stimulation mediated by the T cell antigen receptor (TCR) and the costimulatory receptor CD28 or through cognate interactions, was linked to increased glycolytic flux. EM CD8(+) T cells exhibited more glyceraldehyde-3-phosphate dehydrogenase (GAPDH) activity at early time points, before proliferation commenced, than did naive cells activated under similar conditions. CD28 signaling via the serine-threonine kinase Akt and the metabolic-checkpoint kinase mTORC2 was needed to sustain TCR-mediated immediate-early glycolysis. Unlike glycolysis in proliferating cells, immediate-early glycolysis in memory CD8(+) T cells was rapamycin insensitive. Thus, CD8(+) memory T cells have an Akt-dependent 'imprinted' glycolytic potential that is required for efficient immediate-early IFN-γ recall responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Memoria Inmunológica , Ensamble y Desensamble de Cromatina , Epítopos de Linfocito T/inmunología , Glucólisis , Herpesvirus Humano 4/inmunología , Humanos , Interferón gamma/biosíntesis , Interferón gamma/genética , Activación de Linfocitos , Diana Mecanicista del Complejo 2 de la Rapamicina , Metaboloma , Metabolómica , Complejos Multiproteicos/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
13.
PLoS Pathog ; 9(3): e1003200, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23505367

RESUMEN

Infection of the developing fetus with human cytomegalovirus (HCMV) is a major cause of central nervous system disease in infants and children; however, mechanism(s) of disease associated with this intrauterine infection remain poorly understood. Utilizing a mouse model of HCMV infection of the developing CNS, we have shown that peripheral inoculation of newborn mice with murine CMV (MCMV) results in CNS infection and developmental abnormalities that recapitulate key features of the human infection. In this model, animals exhibit decreased granule neuron precursor cell (GNPC) proliferation and altered morphogenesis of the cerebellar cortex. Deficits in cerebellar cortical development are symmetric and global even though infection of the CNS results in a non-necrotizing encephalitis characterized by widely scattered foci of virus-infected cells with mononuclear cell infiltrates. These findings suggested that inflammation induced by MCMV infection could underlie deficits in CNS development. We investigated the contribution of host inflammatory responses to abnormal cerebellar development by modulating inflammatory responses in infected mice with glucocorticoids. Treatment of infected animals with glucocorticoids decreased activation of CNS mononuclear cells and expression of inflammatory cytokines (TNF-α, IFN-ß and IFNγ) in the CNS while minimally impacting CNS virus replication. Glucocorticoid treatment also limited morphogenic abnormalities and normalized the expression of developmentally regulated genes within the cerebellum. Importantly, GNPC proliferation deficits were normalized in MCMV infected mice following glucocorticoid treatment. Our findings argue that host inflammatory responses to MCMV infection contribute to deficits in CNS development in MCMV infected mice and suggest that similar mechanisms of disease could be responsible for the abnormal CNS development in human infants infected in-utero with HCMV.


Asunto(s)
Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Encefalitis/tratamiento farmacológico , Glucocorticoides/uso terapéutico , Infecciones por Herpesviridae/tratamiento farmacológico , Muromegalovirus , Animales , Animales Recién Nacidos/virología , Encéfalo/virología , Proliferación Celular , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/virología , Enfermedades del Sistema Nervioso Central/virología , Cerebelo/efectos de los fármacos , Cerebelo/embriología , Cerebelo/virología , Dexametasona/uso terapéutico , Modelos Animales de Enfermedad , Encefalitis/virología , Infecciones por Herpesviridae/virología , Interferón beta/biosíntesis , Interferón gamma/biosíntesis , Ratones , Ratones Endogámicos BALB C , Células-Madre Neurales/metabolismo , Células-Madre Neurales/virología , Prednisolona/uso terapéutico , Factor de Necrosis Tumoral alfa/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA