Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 312
Filtrar
1.
Cell Rep ; 43(5): 114156, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38687642

RESUMEN

The maintenance of antigen-specific CD8+ T cells underlies the efficacy of vaccines and immunotherapies. Pathways contributing to CD8+ T cell loss are not completely understood. Uncovering the pathways underlying the limited persistence of CD8+ T cells would be of significant benefit for developing novel strategies of promoting T cell persistence. Here, we demonstrate that murine CD8+ T cells experience endoplasmic reticulum (ER) stress following activation and that the ER-associated degradation (ERAD) adapter Sel1L is induced in activated CD8+ T cells. Sel1L loss limits CD8+ T cell function and memory formation following acute viral infection. Mechanistically, Sel1L is required for optimal bioenergetics and c-Myc expression. Finally, we demonstrate that human CD8+ T cells experience ER stress upon activation and that ER stress is negatively associated with improved T cell functionality in T cell-redirecting therapies. Together, these results demonstrate that ER stress and ERAD are important regulators of T cell function and persistence.


Asunto(s)
Linfocitos T CD8-positivos , Estrés del Retículo Endoplásmico , Degradación Asociada con el Retículo Endoplásmico , Memoria Inmunológica , Animales , Humanos , Ratones , Enfermedad Aguda , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Coriomeningitis Linfocítica/patología , Ratones Endogámicos C57BL , Proteínas , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Masculino , Femenino
2.
Pediatr Res ; 95(2): 551-557, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38182822

RESUMEN

Lymphocytic choriomeningitis virus (LCMV) is a prevalent pathogen, whose natural host and reservoir is the wild mouse. Humans can be infected when they contact the secretions of mice. Most infections of postnatal humans result in mild illness. However, the consequences can be severe when the infection occurs during pregnancy, as the virus crosses the placenta to infect the fetus. LCMV infection of the human fetus can lead to severe neuropathologic effects, including microencephaly, hydrocephalus, focal destructive lesions, and cerebellar hypoplasia. Outcomes among children with congenital LCMV are variable, but most are permanently and severely disabled. The neonatal rat inoculated with LCMV models human prenatal infection. The rat model has demonstrated that effects of LCMV depend on host age at the time of infection. Some effects, including encephalomalacia and neuronal migration disturbances, are immune-mediated and depend on the actions of T-lymphocytes. Other effects, including cerebellar hypoplasia, are virus-mediated and do not depend on T-lymphocytes. Cerebellar neuronal migration disturbances are caused by immune-mediated corruption of Bergmann glia structure. The rat pup inoculated with LCMV is a superb animal model for human congenital infection. All neuropathologic effects observed in human congenital LCMV infection can be recapitulated in the rat model. IMPACT: Lymphocytic choriomeningitis virus (LCMV) is a prevalent human pathogen that can cause serious neurologic birth defects when the infection occurs during pregnancy. The effects of the virus on the developing brain depend strongly on the age of the host at the time of infection. Some of the pathologic effects of LCMV are immune-mediated and are driven by T-lymphocytes, while other pathologic effects are due to the virus itself.


Asunto(s)
Coriomeningitis Linfocítica , Virus de la Coriomeningitis Linfocítica , Malformaciones del Sistema Nervioso , Humanos , Embarazo , Femenino , Niño , Animales , Ratas , Ratones , Virus de la Coriomeningitis Linfocítica/fisiología , Encéfalo/patología , Coriomeningitis Linfocítica/congénito , Coriomeningitis Linfocítica/patología , Cerebelo/patología , Ratones Endogámicos C57BL , Discapacidades del Desarrollo
3.
Cell Death Dis ; 14(12): 838, 2023 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-38110339

RESUMEN

Acute infection and chronic infection are the two most common fates of pathogenic virus infections. While several factors that contribute to these fates are described, the critical control points and the mechanisms that underlie infection fate regulation are incompletely understood. Using the acute and chronic lymphocytic choriomeningitis virus (LCMV) infection model of mice, we find that the early dynamic pattern of the IFN-I response is a differentiating trait between both infection fates. Acute-infected mice generate a 2-wave IFN-I response while chronic-infected mice generate only a 1-wave response. The underlying cause is a temporal difference in CD8 T cell-mediated killing of splenic marginal zone CD169+ macrophages. It occurs later in acute infection and thus enables CD169+ marginal zone macrophages to produce the 2nd IFN-I wave. This is required for subsequent immune events including induction of inflammatory macrophages, generation of effector CD8+ T cells and virus clearance. Importantly, these benefits come at a cost for the host in the form of spleen fibrosis. Due to an earlier marginal zone destruction, these ordered immune events are deregulated in chronic infection. Our findings demonstrate the critical importance of kinetically well-coordinated sequential immune events for acute infection control and highlights that it may come at a cost for the host organism.


Asunto(s)
Coriomeningitis Linfocítica , Ratones , Animales , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/fisiología , Infección Persistente , Ratones Endogámicos C57BL , Linfocitos T CD8-positivos , Macrófagos/patología
4.
PLoS Pathog ; 19(11): e1011837, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38019895

RESUMEN

Neuropilin-1 (Nrp-1) expression on CD8+ T cells has been identified in tumor-infiltrating lymphocytes and in persistent murine gamma-herpes virus infections, where it interferes with the development of long-lived memory T cell responses. In parasitic and acute viral infections, the role of Nrp-1 expression on CD8+ T cells remains unclear. Here, we demonstrate a strong induction of Nrp-1 expression on CD8+ T cells in Plasmodium berghei ANKA (PbA)-infected mice that correlated with neurological deficits of experimental cerebral malaria (ECM). Likewise, the frequency of Nrp-1+CD8+ T cells was significantly elevated and correlated with liver damage in the acute phase of lymphocytic choriomeningitis virus (LCMV) infection. Transcriptomic and flow cytometric analyses revealed a highly activated phenotype of Nrp-1+CD8+ T cells from infected mice. Correspondingly, in vitro experiments showed rapid induction of Nrp-1 expression on CD8+ T cells after stimulation in conjunction with increased expression of activation-associated molecules. Strikingly, T cell-specific Nrp-1 ablation resulted in reduced numbers of activated T cells in the brain of PbA-infected mice as well as in spleen and liver of LCMV-infected mice and alleviated the severity of ECM and LCMV-induced liver pathology. Mechanistically, we identified reduced blood-brain barrier leakage associated with reduced parasite sequestration in the brain of PbA-infected mice with T cell-specific Nrp-1 deficiency. In conclusion, Nrp-1 expression on CD8+ T cells represents a very early activation marker that exacerbates deleterious CD8+ T cell responses during both, parasitic PbA and acute LCMV infections.


Asunto(s)
Coriomeningitis Linfocítica , Malaria Cerebral , Parásitos , Ratones , Animales , Neuropilina-1 , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica , Linfocitos T CD8-positivos/patología , Ratones Endogámicos C57BL
5.
Biochem Biophys Res Commun ; 661: 56-63, 2023 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-37087799

RESUMEN

T cell responses are regulated by co-stimulatory and inhibitory receptors along with T cell receptor- and cytokine-mediated signals. CD51 is a transmembrane glycoprotein of the integrin family that plays a role in cell adhesion, migration, tumorigenesis, and other cellular functions. In this study, we aimed to investigate the expression and function of CD51 on CD8 T cells. Upon in vitro T cell activation, CD51 expression was delayed but subsequently was upregulated in CD8 T cells upon cell division. Furthermore, CD51 was highly expressed in exhausted CD8 T cells in chronic LCMV infection, B16F10 melanoma, and CT26 colon carcinoma, and its expression level increased as cells became more differentiated. Using CRISPR-mediated knockdown, we found that the absence of CD51 led to a lower number of virus-specific CD8 T cells upon chronic lymphocytic choriomeningitis virus (LCMV) infection, although their granzyme B expression and cytokine production were maintained. Blocking CD51 also inhibited the in vitro proliferation of CD8 T cells. These results suggest that CD51 plays an important role in the early expansion of CD8 T cells and may have potential as an immunomodulatory target.


Asunto(s)
Coriomeningitis Linfocítica , Animales , Ratones , Linfocitos T CD8-positivos , Citocinas/metabolismo , Activación de Linfocitos , Coriomeningitis Linfocítica/metabolismo , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica , Ratones Endogámicos C57BL , Integrina alfaV/inmunología
6.
Methods Cell Biol ; 173: 121-131, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36653079

RESUMEN

Lymphocytic choriomeningitis virus (LCMV) is a frequently used animal model to study immune responses against acute and chronic viral infections. LCMV is a non-cytopathic virus, but destruction of infected cells by cytotoxic T lymphocytes (CTLs) can lead to severe damage of tissues. Virus-specific T cell responses have to be balanced. A low virus load leads to a strong T cell response and subsequently to viral control. In contrast, a high viral titer is associated with T cell exhaustion and chronic viral infections. During an intermediate LCMV viral load CD8+ T cells can cause immunopathology, which can have detrimental outcomes. The LCMV infection model offers the opportunity to study virus-specific CD4+ and CD8+ T cell responses during chronic and acute infections by quantifying LCMV-specific T cells by tetramer staining and measuring cytokine production and viral titers in different organs.


Asunto(s)
Coriomeningitis Linfocítica , Virus de la Coriomeningitis Linfocítica , Animales , Ratones , Linfocitos T CD8-positivos/patología , Coriomeningitis Linfocítica/patología , Linfocitos T Citotóxicos , Ratones Endogámicos C57BL
7.
Cell Physiol Biochem ; 56(4): 340-352, 2022 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-35926114

RESUMEN

In the context of chronic viral infections, the hepatic microenvironment dictates the outcome of the disease by influencing propagation of virus and regulation of cytotoxic CD8+ T cell response. Nevertheless, such regulation could be beneficial as it resolves the disease or could be detrimental as it causes liver pathological consequences. Liver pathology is a hallmark of chronic viral infection in both human and murine models. Such models show viral infection of hepatocytes and subsequent direct hepatic damage. Other compelling studies showed that liver injury was a consequence of overshooting CD8+ T cells response in experimental mice, so-called immune-mediated liver pathology. This review highlights the viral-induced immune mediated aspects of liver pathology based on the lymphocytic choriomeningitis virus (LCMV) and Hepatitis virus settings.


Asunto(s)
Coriomeningitis Linfocítica , Animales , Linfocitos T CD8-positivos , Humanos , Hígado/patología , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica , Ratones , Ratones Endogámicos C57BL , Linfocitos T Citotóxicos
8.
mBio ; 13(3): e0020522, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35502904

RESUMEN

Lymphocytic choriomeningitis virus (LCMV) is a well-studied mammarenavirus that can be fatal in congenital infections. However, our understanding of LCMV and its interactions with human host factors remains incomplete. Here, host determinants affecting LCMV infection were investigated through a genome-wide CRISPR knockout screen in A549 cells, a human lung adenocarcinoma line. We identified and validated a variety of novel host factors that play a functional role in LCMV infection. Among these, knockout of the sialomucin CD164, a heavily glycosylated transmembrane protein, was found to ablate infection with multiple LCMV strains but not other hemorrhagic mammarenaviruses in several cell types. Further characterization revealed a dependency of LCMV entry on the cysteine-rich domain of CD164, including an N-linked glycosylation site at residue 104 in that region. Given the documented role of LCMV with respect to transplacental human infections, CD164 expression was investigated in human placental tissue and placental cell lines. CD164 was found to be highly expressed in the cytotrophoblast cells, an initial contact site for pathogens within the placenta, and LCMV infection in placental cells was effectively blocked using a monoclonal antibody specific to the cysteine-rich domain of CD164. Together, this study identifies novel factors associated with LCMV infection of human tissues and highlights the importance of CD164, a sialomucin that previously had not been associated with viral infection. IMPORTANCE Lymphocytic choriomeningitis virus (LCMV) is a human-pathogenic mammarenavirus that can be fatal in congenital infections. Although frequently used in the study of persistent infections in the field of immunology, aspects of this virus's life cycle remain incomplete. For example, while viral entry has been shown to depend on a cell adhesion molecule, DAG1, genetic knockout of this gene allows for residual viral infection, implying that additional receptors can mediate cell entry. The significance of our study is the identification of host factors important for successful infection, including the sialomucin CD164, which had not been previously associated with viral infection. We demonstrated that CD164 is essential for LCMV entry into human cells and can serve as a possible therapeutic target for treatment of congenital infection.


Asunto(s)
Endolina , Coriomeningitis Linfocítica , Virus de la Coriomeningitis Linfocítica , Cisteína , Endolina/genética , Femenino , Humanos , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Placenta/virología , Embarazo , Sialomucinas
9.
Eur J Immunol ; 52(7): 1095-1111, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35389518

RESUMEN

Tissue-resident memory T cells (Trm) are retained in peripheral tissues after infection for enhanced protection against secondary encounter with the same pathogen. We have previously shown that the transcription factor Hobit and its homolog Blimp-1 drive Trm development after viral infection, but how and when these transcription factors mediate Trm formation remains poorly understood. In particular, the major impact of Blimp-1 in regulating several aspects of effector T-cell differentiation impairs study of its specific role in Trm development. Here, we used the restricted expression of Hobit in the Trm lineage to develop mice with a conditional deletion of Blimp-1 in Trm, allowing us to specifically investigate the role of both transcription factors in Trm differentiation. We found that Hobit and Blimp-1 were required for the upregulation of CD69 and suppression of CCR7 and S1PR1 on virus-specific Trm precursors after LCMV infection, underlining a role in their retention within tissues. The early impact of Hobit and Blimp-1 favored Trm formation and prevented the development of circulating memory T cells. Thus, our findings highlight a role of Hobit and Blimp-1 at the branching point of circulating and resident memory lineages by suppressing tissue egress of Trm precursors early during infection.


Asunto(s)
Linfocitos T CD8-positivos , Memoria Inmunológica , Coriomeningitis Linfocítica , Virus de la Coriomeningitis Linfocítica , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Factores de Transcripción , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Factor 1 de Unión al Dominio 1 de Regulación Positiva/inmunología , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo
10.
Virology ; 567: 77-86, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35032866

RESUMEN

Type-I interferon (IFN-I) signals exert a critical role in disease progression during viral infections. However, the immunomodulatory mechanisms by which IFN-I dictates disease outcomes remain to be fully defined. Here we report that IFN-I signals mediate thymic atrophy in viral infections, with more severe and prolonged loss of thymic output and unique kinetics and subtypes of IFN-α/ß expression in chronic infection compared to acute infection. Loss of thymic output was linked to inhibition of early stages of thymopoiesis (DN1-DN2 transition, and DN3 proliferation) and pronounced apoptosis during the late DP stage. Notably, infection-associated thymic defects were largely abrogated upon ablation of IFNαßR and partially mitigated in the absence of CD8 T cells, thus implicating direct as well as indirect effects of IFN-I on thymocytes. These findings provide mechanistic underpinnings for immunotherapeutic strategies targeting IFN-1 signals to manipulate disease outcomes during chronic infections and cancers.


Asunto(s)
Atrofia/virología , Interferón-alfa/inmunología , Interferón beta/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Timocitos/virología , Timo/virología , Animales , Atrofia/genética , Atrofia/inmunología , Atrofia/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Enfermedad Crónica , Femenino , Regulación de la Expresión Génica , Humanos , Memoria Inmunológica , Interferón-alfa/genética , Interferón beta/genética , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ganglios Linfáticos/virología , Depleción Linfocítica , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal/inmunología , Análisis de la Célula Individual , Timocitos/inmunología , Timocitos/patología , Timo/inmunología , Timo/patología
11.
J Exp Med ; 218(12)2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34709350

RESUMEN

Chronic viral infections are associated with hematopoietic suppression, bone marrow (BM) failure, and hematopoietic stem cell (HSC) exhaustion. However, how persistent viral challenge and inflammatory responses target BM tissues and perturb hematopoietic competence remains poorly understood. Here, we combine functional analyses with advanced 3D microscopy to demonstrate that chronic infection with lymphocytic choriomeningitis virus leads to (1) long-lasting decimation of the BM stromal network of mesenchymal CXCL12-abundant reticular cells, (2) proinflammatory transcriptional remodeling of remaining components of this key niche subset, and (3) durable functional defects and decreased competitive fitness in HSCs. Mechanistically, BM immunopathology is elicited by virus-specific, activated CD8 T cells, which accumulate in the BM via interferon-dependent mechanisms. Combined antibody-mediated inhibition of type I and II IFN pathways completely preempts degeneration of CARc and protects HSCs from chronic dysfunction. Hence, viral infections and ensuing immune reactions durably impact BM homeostasis by persistently decreasing the competitive fitness of HSCs and disrupting essential stromal-derived, hematopoietic-supporting cues.


Asunto(s)
Médula Ósea/virología , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/virología , Coriomeningitis Linfocítica/patología , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/virología , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Enfermedad Crónica , Regulación de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Interferones/metabolismo , Coriomeningitis Linfocítica/metabolismo , Coriomeningitis Linfocítica/virología , Ratones Endogámicos C57BL , Ratones Mutantes , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Receptor de Interferón alfa y beta/metabolismo
12.
Cells ; 10(8)2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34440912

RESUMEN

During acute infections, CD8+ T cells form various memory subpopulations to provide long-lasting protection against reinfection. T central memory (TCM), T effector memory (TEM), and long-lived effector (LLE) cells are circulating memory populations with distinct plasticity, migration patterns, and effector functions. Tissue-resident memory (TRM) cells permanently reside in the frontline sites of pathogen entry and provide tissue-specific protection upon reinfection. Here, using single-cell RNA-sequencing (scRNA-seq) and bulk RNA-seq, we examined the different and shared transcriptomes and regulators of TRM cells with other circulating memory populations. Furthermore, we identified heterogeneity within the TRM pool from small intestine and novel transcriptional regulators that may control the phenotypic and functional heterogeneity of TRM cells during acute infection. Our findings provide a resource for future studies to identify novel pathways for enhancing vaccination and immunotherapeutic approaches.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Perfilación de la Expresión Génica/métodos , Memoria Inmunológica/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Análisis de la Célula Individual/métodos , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Línea Celular , Células Cultivadas , Femenino , Intestino Delgado/citología , Intestino Delgado/inmunología , Intestino Delgado/virología , Coriomeningitis Linfocítica/patología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/fisiología , Ratones Endogámicos C57BL , RNA-Seq/métodos , Bazo/citología , Bazo/inmunología , Bazo/virología
13.
Cell Rep ; 35(2): 108966, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33852868

RESUMEN

Persistent virus infections can cause pathogenesis that is debilitating or lethal. During these infections, virus-specific T cells fail to protect due to weakened antiviral activity or failure to persist. These outcomes are governed by histone modifications, although it is unknown which enzymes contribute to T cell loss or impaired function over time. In this study, we show that T cell receptor-stimulated CD8+ T cells increase their expression of UTX (ubiquitously transcribed tetratricopeptide repeat, X chromosome) to enhance gene expression. During chronic lymphocytic choriomeningitis virus (LCMV) infection in mice, UTX binds to enhancers and transcription start sites of effector genes, allowing for improved cytotoxic T lymphocyte (CTL)-mediated protection, independent of its trimethylation of histone 3 lysine 27 (H3K27me3) demethylase activity. UTX also limits the frequency and durability of virus-specific CD8+ T cells, which correspond to increased expression of inhibitory receptors. Thus, UTX guides gene expression patterns in CD8+ T cells, advancing early antiviral defenses while reducing the longevity of CD8+ T cell responses.


Asunto(s)
Citotoxicidad Inmunológica/genética , Histona Demetilasas/genética , Memoria Inmunológica/genética , Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Receptor 2 Celular del Virus de la Hepatitis A/genética , Receptor 2 Celular del Virus de la Hepatitis A/inmunología , Histona Demetilasas/deficiencia , Histona Demetilasas/inmunología , Histonas/genética , Histonas/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Transducción de Señal , Linfocitos T Citotóxicos/virología , Carga Viral/genética , Carga Viral/inmunología , Proteína del Gen 3 de Activación de Linfocitos
14.
Mol Cell ; 81(11): 2477-2493.e10, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-33891860

RESUMEN

CD8 T cells play an essential role in defense against viral and bacterial infections and in tumor immunity. Deciphering T cell loss of functionality is complicated by the conspicuous heterogeneity of CD8 T cell states described across experimental and clinical settings. By carrying out a unified analysis of over 300 assay for transposase-accessible chromatin sequencing (ATAC-seq) and RNA sequencing (RNA-seq) experiments from 12 studies of CD8 T cells in cancer and infection, we defined a shared differentiation trajectory toward dysfunction and its underlying transcriptional drivers and revealed a universal early bifurcation of functional and dysfunctional T cell states across models. Experimental dissection of acute and chronic viral infection using single-cell ATAC (scATAC)-seq and allele-specific single-cell RNA (scRNA)-seq identified state-specific drivers and captured the emergence of similar TCF1+ progenitor-like populations at an early branch point, at which functional and dysfunctional T cells diverge. Our atlas of CD8 T cell states will facilitate mechanistic studies of T cell immunity and translational efforts.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epigénesis Genética/inmunología , Inmunidad Celular , Coriomeningitis Linfocítica/genética , Neoplasias/genética , Factores de Transcripción/genética , Enfermedad Aguda , Atlas como Asunto , Linfocitos T CD8-positivos/clasificación , Linfocitos T CD8-positivos/patología , Cromatina/química , Cromatina/inmunología , Enfermedad Crónica , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Activación de Linfocitos , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Neoplasias/inmunología , Neoplasias/patología , Análisis de Componente Principal , Análisis de la Célula Individual , Factores de Transcripción/clasificación , Factores de Transcripción/inmunología , Transcripción Genética , Transposasas/genética , Transposasas/metabolismo
15.
J Exp Med ; 218(3)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33201171

RESUMEN

Tracking how individual naive T cells from a natural TCR repertoire clonally expand, differentiate, and make lineage choices in response to an infection has not previously been possible. Here, using single-cell sequencing technology to identify clones by their unique TCR sequences, we were able to trace the clonal expansion, differentiation trajectory, and lineage commitment of individual virus-specific CD4 T cells during an acute lymphocytic choriomeningitis virus (LCMV) infection. Notably, we found previously unappreciated clonal diversity and cellular heterogeneity among virus-specific helper T cells. Interestingly, although most naive CD4 T cells gave rise to multiple lineages at the clonal level, ∼28% of naive cells exhibited a preferred lineage choice toward either Th1 or TFH cells. Mechanistically, we found that TCR structure, in particular the CDR3 motif of the TCR α chain, skewed lineage decisions toward the TFH cell fate.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Linaje de la Célula , Virus de la Coriomeningitis Linfocítica/inmunología , Secuencias de Aminoácidos , Animales , Células Clonales , Subgrupos Linfocitarios/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/patología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/fisiología , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Especificidad de la Especie
16.
Proc Natl Acad Sci U S A ; 117(52): 33446-33454, 2020 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-33318189

RESUMEN

Reduced nutrient intake is a widely conserved manifestation of sickness behavior with poorly characterized effects on adaptive immune responses. During infectious challenges, naive T cells encountering their cognate antigen become activated and differentiate into highly proliferative effector T cells. Despite their evident metabolic shift upon activation, it remains unclear how effector T cells respond to changes in nutrient availability in vivo. Here, we show that spontaneous or imposed feeding reduction during infection decreases the numbers of splenic lymphocytes. Effector T cells showed cell-intrinsic responses dependent on the nuclear receptor Farnesoid X Receptor (FXR). Deletion of FXR in T cells prevented starvation-induced loss of lymphocytes and increased effector T cell fitness in nutrient-limiting conditions, but imparted greater weight loss to the host. FXR deficiency increased the contribution of glutamine and fatty acids toward respiration and enhanced cell survival under low-glucose conditions. Provision of glucose during anorexia of infection rescued effector T cells, suggesting that this sugar is a limiting nutrient for activated lymphocytes and that alternative fuel usage may affect cell survival in starved animals. Altogether, we identified a mechanism by which the host scales immune responses according to food intake, featuring FXR as a T cell-intrinsic sensor.


Asunto(s)
Conducta Alimentaria , Coriomeningitis Linfocítica/inmunología , Receptores Citoplasmáticos y Nucleares/metabolismo , Linfocitos T/inmunología , Animales , Anorexia/virología , Ayuno , Coriomeningitis Linfocítica/patología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/fisiología , Ratones Endogámicos C57BL , Nutrientes/metabolismo , Bazo/patología , Transcripción Genética
17.
Front Immunol ; 11: 600012, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33343572

RESUMEN

IL4I1 is an immunoregulatory enzyme that inhibits CD8 T-cell proliferation in vitro and in the tumoral context. Here, we dissected the effect of IL4I1 on CD8 T-cell priming by studying the differentiation of a transgenic CD8 T-cell clone and the endogenous repertoire in a mouse model of acute lymphocytic choriomeningitis virus (LCMV) infection. Unexpectedly, we show that IL4I1 accelerates the expansion of functional effector CD8 T cells during the first several days after infection and increases the average affinity of the elicited repertoire, supporting more efficient LCMV clearance in WT mice than IL4I1-deficient mice. Conversely, IL4I1 restrains the differentiation of CD8 T-cells into long-lived memory precursors and favors the memory response to the most immunodominant peptides. IL4I1 expression does not affect the phenotype or antigen-presenting functions of dendritic cells (DCs), but directly reduces the stability of T-DC immune synapses in vitro, thus dampening T-cell activation. Overall, our results support a model in which IL4I1 increases the threshold of T-cell activation, indirectly promoting the priming of high-affinity clones while limiting memory T-cell differentiation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Memoria Inmunológica , L-Aminoácido Oxidasa/inmunología , Activación de Linfocitos , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Enfermedad Aguda , Animales , Linfocitos T CD8-positivos/patología , Células Dendríticas/inmunología , Células Dendríticas/patología , Sinapsis Inmunológicas/genética , Sinapsis Inmunológicas/inmunología , Sinapsis Inmunológicas/patología , L-Aminoácido Oxidasa/genética , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/genética , Ratones , Ratones Noqueados
18.
PLoS Pathog ; 16(10): e1008461, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33002089

RESUMEN

The induction of an interferon-mediated response is the first line of defense against pathogens such as viruses. Yet, the dynamics and extent of interferon alpha (IFNα)-induced antiviral genes vary remarkably and comprise three expression clusters: early, intermediate and late. By mathematical modeling based on time-resolved quantitative data, we identified mRNA stability as well as a negative regulatory loop as key mechanisms endogenously controlling the expression dynamics of IFNα-induced antiviral genes in hepatocytes. Guided by the mathematical model, we uncovered that this regulatory loop is mediated by the transcription factor IRF2 and showed that knock-down of IRF2 results in enhanced expression of early, intermediate and late IFNα-induced antiviral genes. Co-stimulation experiments with different pro-inflammatory cytokines revealed that this amplified expression dynamics of the early, intermediate and late IFNα-induced antiviral genes can also be achieved by co-application of IFNα and interleukin1 beta (IL1ß). Consistently, we found that IL1ß enhances IFNα-mediated repression of viral replication. Conversely, we observed that in IL1ß receptor knock-out mice replication of viruses sensitive to IFNα is increased. Thus, IL1ß is capable to potentiate IFNα-induced antiviral responses and could be exploited to improve antiviral therapies.


Asunto(s)
Regulación Viral de la Expresión Génica/efectos de los fármacos , Factor 2 Regulador del Interferón/metabolismo , Interferón-alfa/farmacología , Coriomeningitis Linfocítica/tratamiento farmacológico , Virus de la Coriomeningitis Linfocítica/efectos de los fármacos , Receptores Tipo I de Interleucina-1/fisiología , Replicación Viral/efectos de los fármacos , Animales , Antivirales/farmacología , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/inmunología , Hepatocitos/virología , Humanos , Factor 2 Regulador del Interferón/genética , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/patología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/aislamiento & purificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estabilidad del ARN
19.
Proc Natl Acad Sci U S A ; 117(40): 24998-25007, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32958643

RESUMEN

Infections elicit immune adaptations to enable pathogen resistance and/or tolerance and are associated with compositional shifts of the intestinal microbiome. However, a comprehensive understanding of how infections with pathogens that exhibit distinct capability to spread and/or persist differentially change the microbiome, the underlying mechanisms, and the relative contribution of individual commensal species to immune cell adaptations is still lacking. Here, we discovered that mouse infection with a fast-spreading and persistent (but not a slow-spreading acute) isolate of lymphocytic choriomeningitis virus induced large-scale microbiome shifts characterized by increased Verrucomicrobia and reduced Firmicute/Bacteroidetes ratio. Remarkably, the most profound microbiome changes occurred transiently after infection with the fast-spreading persistent isolate, were uncoupled from sustained viral loads, and were instead largely caused by CD8 T cell responses and/or CD8 T cell-induced anorexia. Among the taxa enriched by infection with the fast-spreading virus, Akkermansia muciniphila, broadly regarded as a beneficial commensal, bloomed upon starvation and in a CD8 T cell-dependent manner. Strikingly, oral administration of A. muciniphila suppressed selected effector features of CD8 T cells in the context of both infections. Our findings define unique microbiome differences after chronic versus acute viral infections and identify CD8 T cell responses and downstream anorexia as driver mechanisms of microbial dysbiosis after infection with a fast-spreading virus. Our data also highlight potential context-dependent effects of probiotics and suggest a model in which changes in host behavior and downstream microbiome dysbiosis may constitute a previously unrecognized negative feedback loop that contributes to CD8 T cell adaptations after infections with fast-spreading and/or persistent pathogens.


Asunto(s)
Anorexia/inmunología , Antígenos CD8/inmunología , Memoria Inmunológica/inmunología , Coriomeningitis Linfocítica/inmunología , Virosis/inmunología , Akkermansia , Animales , Anorexia/microbiología , Anorexia/virología , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/microbiología , Disbiosis/inmunología , Disbiosis/microbiología , Disbiosis/virología , Firmicutes/inmunología , Firmicutes/metabolismo , Microbioma Gastrointestinal/inmunología , Humanos , Coriomeningitis Linfocítica/microbiología , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones , Linfocitos T/inmunología , Linfocitos T/microbiología , Verrucomicrobia/inmunología , Verrucomicrobia/patogenicidad , Virosis/microbiología , Virosis/patología
20.
J Clin Invest ; 130(12): 6523-6538, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32897877

RESUMEN

Chronic viral infections are often established by the exploitation of immune-regulatory mechanisms that result in nonfunctional T cell responses. Viruses that establish persistent infections remain a serious threat to human health. Sphingosine kinase 2 (SphK2) generates sphingosine 1-phosphate, which is a molecule known to regulate multiple cellular processes. However, little is known about SphK2's role during the host immune responses to viral infection. Here, we demonstrate that SphK2 functions during lymphocytic choriomeningitis virus Cl 13 (LCMV Cl 13) infection to limit T cell immune pathology, which subsequently aids in the establishment of virus-induced immunosuppression and the resultant viral persistence. The infection of Sphk2-deficient (Sphk2-/-) mice with LCMV Cl 13 led to the development of nephropathy and mortality via T cell-mediated immunopathology. Following LCMV infection, Sphk2-/- CD4+ T cells displayed increased activity and proliferation, and these cells promoted overactive LCMV Cl 13-specific CD8+ T cell responses. Notably, oral instillation of an SphK2-selective inhibitor promoted protective T cell responses and accelerated the termination of LCMV Cl 13 persistence in mice. Thus, SphK2 is indicated as an immunotherapeutic target for the control of persistent viral infections.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Enfermedades Renales/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Fosfotransferasas (Aceptor de Grupo Alcohol)/inmunología , Animales , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Enfermedades Renales/genética , Enfermedades Renales/patología , Enfermedades Renales/virología , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/patología , Ratones , Ratones Noqueados , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA