Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.799
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 23(2): 186-193, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35105982

RESUMEN

The adaptive immune response is a major determinant of the clinical outcome after SARS-CoV-2 infection and underpins vaccine efficacy. T cell responses develop early and correlate with protection but are relatively impaired in severe disease and are associated with intense activation and lymphopenia. A subset of T cells primed against seasonal coronaviruses cross reacts with SARS-CoV-2 and may contribute to clinical protection, particularly in early life. T cell memory encompasses broad recognition of viral proteins, estimated at around 30 epitopes within each individual, and seems to be well sustained so far. This breadth of recognition can limit the impact of individual viral mutations and is likely to underpin protection against severe disease from viral variants, including Omicron. Current COVID-19 vaccines elicit robust T cell responses that likely contribute to remarkable protection against hospitalization or death, and novel or heterologous regimens offer the potential to further enhance cellular responses. T cell immunity plays a central role in the control of SARS-CoV-2 and its importance may have been relatively underestimated thus far.


Asunto(s)
COVID-19/inmunología , Inmunidad Celular , Activación de Linfocitos , SARS-CoV-2/inmunología , Linfocitos T/inmunología , Animales , Antígenos Virales/inmunología , COVID-19/metabolismo , COVID-19/virología , Reacciones Cruzadas , Interacciones Huésped-Patógeno , Humanos , Memoria Inmunológica , Fenotipo , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , Linfocitos T/metabolismo , Linfocitos T/virología
2.
Nat Immunol ; 23(2): 210-216, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35027728

RESUMEN

A proportion of patients surviving acute coronavirus disease 2019 (COVID-19) infection develop post-acute COVID syndrome (long COVID (LC)) lasting longer than 12 weeks. Here, we studied individuals with LC compared to age- and gender-matched recovered individuals without LC, unexposed donors and individuals infected with other coronaviruses. Patients with LC had highly activated innate immune cells, lacked naive T and B cells and showed elevated expression of type I IFN (IFN-ß) and type III IFN (IFN-λ1) that remained persistently high at 8 months after infection. Using a log-linear classification model, we defined an optimal set of analytes that had the strongest association with LC among the 28 analytes measured. Combinations of the inflammatory mediators IFN-ß, PTX3, IFN-γ, IFN-λ2/3 and IL-6 associated with LC with 78.5-81.6% accuracy. This work defines immunological parameters associated with LC and suggests future opportunities for prevention and treatment.


Asunto(s)
Linfocitos B/inmunología , COVID-19/complicaciones , Inmunidad Innata , SARS-CoV-2/inmunología , Linfocitos T/inmunología , Adulto , Anciano , Linfocitos B/metabolismo , Linfocitos B/virología , Biomarcadores/sangre , COVID-19/sangre , COVID-19/inmunología , COVID-19/virología , Estudios de Casos y Controles , Citocinas/sangre , Femenino , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/sangre , Masculino , Persona de Mediana Edad , Pronóstico , SARS-CoV-2/patogenicidad , Índice de Severidad de la Enfermedad , Linfocitos T/metabolismo , Linfocitos T/virología , Factores de Tiempo , Síndrome Post Agudo de COVID-19
3.
Nat Immunol ; 22(1): 86-98, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33235385

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for an unprecedented global pandemic of COVID-19. Animal models are urgently needed to study the pathogenesis of COVID-19 and to screen vaccines and treatments. We show that African green monkeys (AGMs) support robust SARS-CoV-2 replication and develop pronounced respiratory disease, which may more accurately reflect human COVID-19 cases than other nonhuman primate species. SARS-CoV-2 was detected in mucosal samples, including rectal swabs, as late as 15 days after exposure. Marked inflammation and coagulopathy in blood and tissues were prominent features. Transcriptome analysis demonstrated stimulation of interferon and interleukin-6 pathways in bronchoalveolar lavage samples and repression of natural killer cell- and T cell-associated transcripts in peripheral blood. Despite a slight waning in antibody titers after primary challenge, enhanced antibody and cellular responses contributed to rapid clearance after re-challenge with an identical strain. These data support the utility of AGM for studying COVID-19 pathogenesis and testing medical countermeasures.


Asunto(s)
COVID-19/inmunología , Modelos Animales de Enfermedad , Reinfección/inmunología , SARS-CoV-2/inmunología , Linfocitos T/inmunología , Animales , Anticuerpos Antivirales/inmunología , COVID-19/epidemiología , COVID-19/virología , Chlorocebus aethiops , Epidemias/prevención & control , Expresión Génica/genética , Expresión Génica/inmunología , Perfilación de la Expresión Génica , Humanos , Interferones/genética , Interferones/inmunología , Interferones/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Reinfección/virología , SARS-CoV-2/fisiología , Linfocitos T/metabolismo , Linfocitos T/virología
4.
Annu Rev Cell Dev Biol ; 35: 381-406, 2019 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-31283378

RESUMEN

Innate immunity and adaptive immunity consist of highly specialized immune lineages that depend on transcription factors for both function and development. In this review, we dissect the similarities between two innate lineages, innate lymphoid cells (ILCs) and dendritic cells (DCs), and an adaptive immune lineage, T cells. ILCs, DCs, and T cells make up four functional immune modules and interact in concert to produce a specified immune response. These three immune lineages also share transcriptional networks governing the development of each lineage, and we discuss the similarities between ILCs and DCs in this review.


Asunto(s)
Inmunidad Adaptativa , Células Dendríticas/inmunología , Redes Reguladoras de Genes , Inmunidad Innata/genética , Linfocitos/inmunología , Animales , Diferenciación Celular/inmunología , Citocinas/metabolismo , Regulación de la Expresión Génica/inmunología , Humanos , Linfocitos T/inmunología , Linfocitos T/microbiología , Linfocitos T/parasitología , Linfocitos T/virología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Cell ; 170(4): 637-648.e10, 2017 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-28757252

RESUMEN

Non-neutralizing antibodies (nnAbs) to HIV-1 show little measurable activity in prevention or therapy in animal models yet were the only correlate of protection in the RV144 vaccine trial. To investigate the role of nnAbs on HIV-1 infection in vivo, we devised a replication-competent HIV-1 reporter virus that expresses a heterologous HA-tag on the surface of infected cells and virions. Anti-HA antibodies bind to, but do not neutralize, the reporter virus in vitro. However, anti-HA protects against infection in humanized mice and strongly selects for nnAb-resistant viruses in an entirely Fc-dependent manner. Similar results were also obtained with tier 2 HIV-1 viruses using a human anti-gp41 nnAb, 246D. While nnAbs are demonstrably less effective than broadly neutralizing antibodies (bNAbs) against HIV-1 in vitro and in vivo, the data show that nnAbs can protect against and alter the course of HIV-1 infection in vivo. PAPERCLIP.


Asunto(s)
Anticuerpos Anti-VIH/inmunología , Infecciones por VIH/inmunología , VIH-1/fisiología , Vacunas contra el SIDA/inmunología , Animales , Antígenos CD4/química , Antígenos CD4/metabolismo , Modelos Animales de Enfermedad , Proteína gp120 de Envoltorio del VIH/química , Proteína gp120 de Envoltorio del VIH/metabolismo , VIH-1/genética , Humanos , Ratones , Mutación , Receptores Fc/inmunología , Linfocitos T/virología
6.
Cell ; 164(4): 695-709, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26830877

RESUMEN

Whereas human dendritic cells (DCs) are largely resistant to productive infection with HIV-1, they have a unique ability to take up the virus and transmit it efficiently to T lymphocytes through a process of trans-infection or trans-enhancement. To elucidate the molecular and cell biological mechanism for trans-enhancement, we performed an shRNA screen of several hundred genes involved in organelle and membrane trafficking in immature human monocyte-derived dendritic cells (MDDCs). We identified TSPAN7 and DNM2, which control actin nucleation and stabilization, as having important and distinct roles in limiting HIV-1 endocytosis and in maintaining virus particles on dendrites, which is required for efficient transfer to T lymphocytes. Further characterization of this process may provide insights not only into the role of DCs in transmission and dissemination of HIV-1 but also more broadly into mechanisms controlling capture and internalization of pathogens.


Asunto(s)
Actinas/metabolismo , Células Dendríticas/inmunología , Infecciones por VIH/inmunología , VIH-1/fisiología , Linfocitos T/inmunología , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Actomiosina/metabolismo , Citoesqueleto/efectos de los fármacos , Células Dendríticas/virología , Dinamina II , Dinaminas/metabolismo , Endocitosis , Técnicas de Silenciamiento del Gen , Infecciones por VIH/virología , Humanos , Sinapsis Inmunológicas , Monocitos/inmunología , Proteínas del Tejido Nervioso/metabolismo , Linfocitos T/virología , Tetraspaninas/metabolismo
7.
Nature ; 631(8019): 189-198, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38898278

RESUMEN

The COVID-19 pandemic is an ongoing global health threat, yet our understanding of the dynamics of early cellular responses to this disease remains limited1. Here in our SARS-CoV-2 human challenge study, we used single-cell multi-omics profiling of nasopharyngeal swabs and blood to temporally resolve abortive, transient and sustained infections in seronegative individuals challenged with pre-Alpha SARS-CoV-2. Our analyses revealed rapid changes in cell-type proportions and dozens of highly dynamic cellular response states in epithelial and immune cells associated with specific time points and infection status. We observed that the interferon response in blood preceded the nasopharyngeal response. Moreover, nasopharyngeal immune infiltration occurred early in samples from individuals with only transient infection and later in samples from individuals with sustained infection. High expression of HLA-DQA2 before inoculation was associated with preventing sustained infection. Ciliated cells showed multiple immune responses and were most permissive for viral replication, whereas nasopharyngeal T cells and macrophages were infected non-productively. We resolved 54 T cell states, including acutely activated T cells that clonally expanded while carrying convergent SARS-CoV-2 motifs. Our new computational pipeline Cell2TCR identifies activated antigen-responding T cells based on a gene expression signature and clusters these into clonotype groups and motifs. Overall, our detailed time series data can serve as a Rosetta stone for epithelial and immune cell responses and reveals early dynamic responses associated with protection against infection.


Asunto(s)
COVID-19 , Multiómica , SARS-CoV-2 , Análisis de la Célula Individual , Femenino , Humanos , Masculino , COVID-19/genética , COVID-19/inmunología , COVID-19/patología , COVID-19/virología , Células Epiteliales/inmunología , Perfilación de la Expresión Génica , Interferones/inmunología , Macrófagos/inmunología , Macrófagos/virología , Nasofaringe/virología , Nasofaringe/inmunología , SARS-CoV-2/crecimiento & desarrollo , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , SARS-CoV-2/fisiología , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología , Factores de Tiempo , Replicación Viral
8.
Immunity ; 50(2): 403-417.e4, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30709740

RESUMEN

The tolerogenic microenvironment of the liver is associated with impaired hepatic T cell function. Here, we examined the contribution of liver-resident natural killer (LrNK) cells, a prominent hepatic NK cell compartment, to T cell antiviral responses in the liver. The number of virus-specific T cells increased in LrNK-cell-deficient mice during both acute and chronic lymphocytic choriomeningitis virus infection. Upon infection with adenovirus, hepatic T cells from these mice produced more cytokines, which was accompanied by reduced viral loads. Transfer of LrNK cells into LrNK-cell-deficient or wild-type mice inhibited hepatic T cell function, resulting in impaired viral clearance, whereas transfer of conventional NK cells promoted T cell antiviral responses. LrNK-cell-mediated inhibition of T cell function was dependent on the PD-1-PD-L1 axis. Our findings reveal a role for LrNK cells in the regulation of T cell immunity and provide insight into the mechanisms of immune tolerance in the liver.


Asunto(s)
Antígeno B7-H1/inmunología , Células Asesinas Naturales/inmunología , Hígado/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T/inmunología , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepatocitos/virología , Células Asesinas Naturales/metabolismo , Hígado/metabolismo , Hígado/virología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/metabolismo , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Transducción de Señal/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología , Transcriptoma/genética , Transcriptoma/inmunología
9.
Mol Cell ; 73(6): 1162-1173.e5, 2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30712990

RESUMEN

The MHC class I antigen presentation system enables T cell immunosurveillance of cancers and viruses. A substantial fraction of the immunopeptidome derives from rapidly degraded nascent polypeptides (DRiPs). By knocking down each of the 80 ribosomal proteins, we identified proteins that modulate peptide generation without altering source protein expression. We show that 60S ribosomal proteins L6 (RPL6) and RPL28, which are adjacent on the ribosome, play opposite roles in generating an influenza A virus-encoded peptide. Depleting RPL6 decreases ubiquitin-dependent peptide presentation, whereas depleting RPL28 increases ubiquitin-dependent and -independent peptide presentation. 40S ribosomal protein S28 (RPS28) knockdown increases total peptide supply in uninfected cells by increasing DRiP synthesis from non-canonical translation of "untranslated" regions and non-AUG start codons and sensitizes tumor cells for T cell targeting. Our findings raise the possibility of modulating immunosurveillance by pharmaceutical targeting ribosomes.


Asunto(s)
Presentación de Antígeno , Antígenos de Histocompatibilidad Clase I/biosíntesis , Proteínas Ribosómicas/metabolismo , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Subunidades Ribosómicas Pequeñas de Eucariotas/metabolismo , Linfocitos T/metabolismo , Animales , Línea Celular Tumoral , Técnicas de Cocultivo , Células HEK293 , Antígenos de Histocompatibilidad Clase I/inmunología , Interacciones Huésped-Patógeno , Humanos , Vigilancia Inmunológica , Virus de la Influenza A/inmunología , Virus de la Influenza A/patogenicidad , Melanoma/inmunología , Melanoma/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Ribosómicas/genética , Subunidades Ribosómicas Grandes de Eucariotas/genética , Subunidades Ribosómicas Pequeñas de Eucariotas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Linfocitos T/inmunología , Linfocitos T/virología
10.
PLoS Pathog ; 20(9): e1012555, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39283919

RESUMEN

Manipulation of immune cell functions, independently of direct infection of these cells, emerges as a key process in viral pathophysiology. Chronic infection by Human T-cell Leukemia Virus type 1 (HTLV-1) is associated with immune dysfunctions, including misdirected responses of dendritic cells (DCs). Here, we interrogate the ability of transformed HTLV-1-infected T cells to manipulate human DC functions. We show that exposure to transformed HTLV-1-infected T cells induces a biased and peculiar transcriptional signature in monocyte-derived DCs, associated with an inefficient maturation and a poor responsiveness to subsequent stimulation by a TLR4 agonist. This poor responsiveness is also associated with a unique transcriptional landscape characterized by a set of genes whose expression is either conferred, impaired or abolished by HTLV-1 pre-exposure. Induction of this functional impairment requires several hours of coculture with transformed HTLV-1-infected cells, and associated mechanisms driven by viral capture, cell-cell contacts, and soluble mediators. Altogether, this cross-talk between infected T cells and DCs illustrate how HTLV-1 might co-opt communications between cells to induce a unique local tolerogenic immune microenvironment suitable for its own persistence.


Asunto(s)
Células Dendríticas , Infecciones por HTLV-I , Virus Linfotrópico T Tipo 1 Humano , Humanos , Células Dendríticas/inmunología , Células Dendríticas/virología , Células Dendríticas/metabolismo , Virus Linfotrópico T Tipo 1 Humano/inmunología , Infecciones por HTLV-I/inmunología , Infecciones por HTLV-I/virología , Infecciones por HTLV-I/genética , Linfocitos T/inmunología , Linfocitos T/virología , Reprogramación Celular
11.
Mol Cell ; 67(6): 1001-1012.e6, 2017 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-28844864

RESUMEN

BET proteins commonly activate cellular gene expression, yet inhibiting their recruitment paradoxically reactivates latent HIV-1 transcription. Here we identify the short isoform of BET family member BRD4 (BRD4S) as a corepressor of HIV-1 transcription. We found that BRD4S was enriched in chromatin fractions of latently infected T cells, and it was more rapidly displaced from chromatin upon BET inhibition than the long isoform. BET inhibition induced marked nucleosome remodeling at the latent HIV-1 promoter, which was dependent on the activity of BRG1-associated factors (BAF), an SWI/SNF chromatin-remodeling complex with known repressive functions in HIV-1 transcription. BRD4S directly bound BRG1, a catalytic subunit of BAF, via its bromodomain and extraterminal (ET) domain, and this isoform was necessary for BRG1 recruitment to latent HIV-1 chromatin. Using chromatin immunoprecipitation sequencing (ChIP-seq) combined with assay for transposase-accessible chromatin coupled to high-throughput sequencing (ATAC-seq) data, we found that the latent HIV-1 promoter phenotypically resembles endogenous long terminal repeat (LTR) sequences, pointing to a select role of BRD4S-BRG1 complexes in genomic silencing of invasive retroelements.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , ADN Viral/metabolismo , VIH-1/metabolismo , Proteínas Nucleares/metabolismo , Linfocitos T/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Latencia del Virus , Azepinas/farmacología , Proteínas de Ciclo Celular , Cromatina/genética , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Inmunoprecipitación de Cromatina , Proteínas Cromosómicas no Histona/efectos de los fármacos , Proteínas Cromosómicas no Histona/genética , ADN Helicasas/genética , ADN Helicasas/metabolismo , ADN Viral/genética , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Regulación Viral de la Expresión Génica , Células HEK293 , VIH-1/efectos de los fármacos , VIH-1/genética , VIH-1/inmunología , Secuenciación de Nucleótidos de Alto Rendimiento , Interacciones Huésped-Patógeno , Humanos , Células Jurkat , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Unión Proteica , Isoformas de Proteínas , Interferencia de ARN , Retroelementos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/virología , Factores de Tiempo , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Transfección , Triazoles/farmacología , Latencia del Virus/efectos de los fármacos
12.
J Biol Chem ; 299(6): 104743, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37100283

RESUMEN

Fc receptors are involved in a variety of physiologically and disease-relevant responses. Among them, FcγRIIA (CD32a) is known for its activating functions in pathogen recognition and platelet biology, and, as potential marker of T lymphocytes latently infected with HIV-1. The latter has not been without controversy due to technical challenges complicated by T-B cell conjugates and trogocytosis as well as a lack of antibodies distinguishing between the closely related isoforms of FcγRII. To generate high-affinity binders specific for FcγRIIA, libraries of designed ankyrin repeat proteins (DARPins) were screened for binding to its extracellular domains by ribosomal display. Counterselection against FcγRIIB eliminated binders cross-reacting with both isoforms. The identified DARPins bound FcγRIIA with no detectable binding for FcγRIIB. Their affinities for FcγRIIA were in the low nanomolar range and could be enhanced by cleavage of the His-tag and dimerization. Interestingly, complex formation between DARPin and FcγRIIA followed a two-state reaction model, and discrimination from FcγRIIB was based on a single amino acid residue. In flow cytometry, DARPin F11 detected FcγRIIA+ cells even when they made up less than 1% of the cell population. Image stream analysis of primary human blood cells confirmed that F11 caused dim but reliable cell surface staining of a small subpopulation of T lymphocytes. When incubated with platelets, F11 inhibited their aggregation equally efficient as antibodies unable to discriminate between both FcγRII isoforms. The selected DARPins are unique novel tools for platelet aggregation studies as well as the role of FcγRIIA for the latent HIV-1 reservoir.


Asunto(s)
Proteínas de Repetición de Anquirina Diseñadas , Agregación Plaquetaria , Receptores de IgG , Humanos , Anticuerpos/metabolismo , Plaquetas/metabolismo , Proteínas de Repetición de Anquirina Diseñadas/metabolismo , VIH-1 , Isoformas de Proteínas/metabolismo , Receptores de IgG/metabolismo , Latencia del Virus , Linfocitos T/virología
13.
J Virol ; 97(12): e0117923, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-37991367

RESUMEN

IMPORTANCE: The traditional view of retrovirus assembly posits that packaging of gRNA by HIV-1 Gag occurs in the cytoplasm or at the plasma membrane. However, our previous studies showing that HIV-1 Gag enters the nucleus and binds to USvRNA at transcription sites suggest that gRNA selection may occur in the nucleus. In the present study, we observed that HIV-1 Gag trafficked to the nucleus and co-localized with USvRNA within 8 hours of expression. In infected T cells (J-Lat 10.6) reactivated from latency and in a HeLa cell line stably expressing an inducible Rev-dependent HIV-1 construct, we found that Gag preferentially localized with euchromatin histone marks associated with enhancer and promoter regions near the nuclear periphery, which is the favored site HIV-1 integration. These observations support the innovative hypothesis that HIV-1 Gag associates with euchromatin-associated histones to localize to active transcription sites, promoting capture of newly synthesized gRNA for packaging.


Asunto(s)
Núcleo Celular , Eucromatina , VIH-1 , Código de Histonas , Histonas , Empaquetamiento del Genoma Viral , Productos del Gen gag del Virus de la Inmunodeficiencia Humana , Humanos , Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo , Elementos de Facilitación Genéticos/genética , Eucromatina/genética , Eucromatina/metabolismo , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo , Células HeLa , Histonas/metabolismo , VIH-1/genética , VIH-1/crecimiento & desarrollo , VIH-1/metabolismo , Regiones Promotoras Genéticas/genética , Linfocitos T/virología , Transcripción Genética , Activación Viral
14.
Cytometry A ; 105(7): 488-492, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38747672

RESUMEN

We introduce a 35-marker imaging mass cytometry (IMC) panel for a detailed examination of immune cell populations and HIV RNA in formalin fixed paraffin embedded (FFPE) human intestinal tissue. The panel has broad cell type coverage and particularly excels in delineating subsets of mononuclear phagocytes and T cells. Markers for key tissue structures are included, enabling identification of epithelium, blood vessels, lymphatics, and musculature. The described method for HIV RNA detection can be generalized to other low abundance RNA targets, whether endogenous or pathogen derived. As such, the panel presented here is useful for high parameter spatial mapping of intestinal immune cells and their interactions with pathogens such as HIV.


Asunto(s)
Infecciones por VIH , Citometría de Imagen , Adhesión en Parafina , Humanos , Adhesión en Parafina/métodos , Citometría de Imagen/métodos , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Infecciones por VIH/diagnóstico , Infecciones por VIH/patología , Biomarcadores , Formaldehído/química , ARN Viral/genética , ARN Viral/análisis , Citometría de Flujo/métodos , Intestinos/virología , Intestinos/inmunología , Fijación del Tejido/métodos , VIH-1/inmunología , Linfocitos T/inmunología , Linfocitos T/virología
15.
Cell Commun Signal ; 22(1): 349, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965547

RESUMEN

T lymphocytes play a primary role in the adaptive antiviral immunity. Both lymphocytosis and lymphopenia were found to be associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While lymphocytosis indicates an active anti-viral response, lymphopenia is a sign of poor prognosis. T-cells, in essence, rarely express ACE2 receptors, making the cause of cell depletion enigmatic. Moreover, emerging strains posed an immunological challenge, potentially alarming for the next pandemic. Herein, we review how possible indirect and direct key mechanisms could contribute to SARS-CoV-2-associated-lymphopenia. The fundamental mechanism is the inflammatory cytokine storm elicited by viral infection, which alters the host cell metabolism into a more acidic state. This "hyperlactic acidemia" together with the cytokine storm suppresses T-cell proliferation and triggers intrinsic/extrinsic apoptosis. SARS-CoV-2 infection also results in a shift from steady-state hematopoiesis to stress hematopoiesis. Even with low ACE2 expression, the presence of cholesterol-rich lipid rafts on activated T-cells may enhance viral entry and syncytia formation. Finally, direct viral infection of lymphocytes may indicate the participation of other receptors or auxiliary proteins on T-cells, that can work alone or in concert with other mechanisms. Therefore, we address the role of CD147-a novel route-for SARS-CoV-2 and its new variants. CD147 is not only expressed on T-cells, but it also interacts with other co-partners to orchestrate various biological processes. Given these features, CD147 is an appealing candidate for viral pathogenicity. Understanding the molecular and cellular mechanisms behind SARS-CoV-2-associated-lymphopenia will aid in the discovery of potential therapeutic targets to improve the resilience of our immune system against this rapidly evolving virus.


Asunto(s)
Basigina , COVID-19 , Linfopenia , SARS-CoV-2 , Humanos , Linfopenia/inmunología , Linfopenia/virología , COVID-19/inmunología , COVID-19/virología , COVID-19/patología , SARS-CoV-2/metabolismo , Basigina/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología , Síndrome de Liberación de Citoquinas/inmunología , Animales
16.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33431697

RESUMEN

GPR15 is a G protein-coupled receptor (GPCR) proposed to play a role in mucosal immunity that also serves as a major entry cofactor for HIV-2 and simian immunodeficiency virus (SIV). To discover novel endogenous GPR15 ligands, we screened a hemofiltrate (HF)-derived peptide library for inhibitors of GPR15-mediated SIV infection. Our approach identified a C-terminal fragment of cystatin C (CysC95-146) that specifically inhibits GPR15-dependent HIV-1, HIV-2, and SIV infection. In contrast, GPR15L, the chemokine ligand of GPR15, failed to inhibit virus infection. We found that cystatin C fragments preventing GPR15-mediated viral entry do not interfere with GPR15L signaling and are generated by proteases activated at sites of inflammation. The antiretroviral activity of CysC95-146 was confirmed in primary CD4+ T cells and is conserved in simian hosts of SIV infection. Thus, we identified a potent endogenous inhibitor of GPR15-mediated HIV and SIV infection that does not interfere with the physiological function of this GPCR.


Asunto(s)
Cistatina C/genética , Infecciones por VIH/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Péptidos/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Animales , Infecciones por VIH/patología , Infecciones por VIH/virología , VIH-1/genética , VIH-1/patogenicidad , Humanos , Receptores Virales/genética , Transducción de Señal/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Linfocitos T/metabolismo , Linfocitos T/virología , Internalización del Virus
17.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34001664

RESUMEN

Comprehensive and accurate comparisons of transcriptomic distributions of cells from samples taken from two different biological states, such as healthy versus diseased individuals, are an emerging challenge in single-cell RNA sequencing (scRNA-seq) analysis. Current methods for detecting differentially abundant (DA) subpopulations between samples rely heavily on initial clustering of all cells in both samples. Often, this clustering step is inadequate since the DA subpopulations may not align with a clear cluster structure, and important differences between the two biological states can be missed. Here, we introduce DA-seq, a targeted approach for identifying DA subpopulations not restricted to clusters. DA-seq is a multiscale method that quantifies a local DA measure for each cell, which is computed from its k nearest neighboring cells across a range of k values. Based on this measure, DA-seq delineates contiguous significant DA subpopulations in the transcriptomic space. We apply DA-seq to several scRNA-seq datasets and highlight its improved ability to detect differences between distinct phenotypes in severe versus mildly ill COVID-19 patients, melanomas subjected to immune checkpoint therapy comparing responders to nonresponders, embryonic development at two time points, and young versus aging brain tissue. DA-seq enabled us to detect differences between these phenotypes. Importantly, we find that DA-seq not only recovers the DA cell types as discovered in the original studies but also reveals additional DA subpopulations that were not described before. Analysis of these subpopulations yields biological insights that would otherwise be undetected using conventional computational approaches.


Asunto(s)
Envejecimiento/genética , COVID-19/genética , Linaje de la Célula/genética , Melanoma/genética , ARN Citoplasmático Pequeño/genética , Neoplasias Cutáneas/genética , Envejecimiento/metabolismo , Linfocitos B/inmunología , Linfocitos B/virología , Encéfalo/citología , Encéfalo/metabolismo , COVID-19/inmunología , COVID-19/patología , COVID-19/virología , Linaje de la Célula/inmunología , Citocinas/genética , Citocinas/inmunología , Conjuntos de Datos como Asunto , Células Dendríticas/inmunología , Células Dendríticas/virología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Melanoma/inmunología , Melanoma/patología , Monocitos/inmunología , Monocitos/virología , Fenotipo , ARN Citoplasmático Pequeño/inmunología , SARS-CoV-2/patogenicidad , Índice de Severidad de la Enfermedad , Análisis de la Célula Individual/métodos , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Linfocitos T/inmunología , Linfocitos T/virología , Transcriptoma
18.
J Virol ; 96(6): e0206521, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35107369

RESUMEN

Recent evidence indicates that viral components of the microbiota can contribute to intestinal homeostasis and protection from local inflammatory or infectious insults. However, host-derived mechanisms that regulate the virome remain largely unknown. In this study, we used colonization with the model commensal murine norovirus (MNV; strain CR6) to interrogate host-directed mechanisms of viral regulation, and we show that STAT1 is a central coordinator of both viral replication and antiviral T cell responses. In addition to restricting CR6 replication to the intestinal tract, we show that STAT1 regulates antiviral CD4+ and CD8+ T cell responses and prevents systemic viral-induced tissue damage and disease. Despite altered T cell responses that resemble those that mediate lethal immunopathology in systemic viral infections in STAT1-deficient mice, depletion of adaptive immune cells and their associated effector functions had no effect on CR6-induced disease. However, therapeutic administration of an antiviral compound limited viral replication, preventing virus-induced tissue damage and death without impacting the generation of inflammatory antiviral T cell responses. Collectively, our data show that STAT1 restricts MNV CR6 replication within the intestinal mucosa and that uncontrolled viral replication mediates disease rather than the concomitant development of dysregulated antiviral T cell responses in STAT1-deficient mice. IMPORTANCE The intestinal microbiota is a collection of bacteria, archaea, fungi, and viruses that colonize the mammalian gut. Coevolution of the host and microbiota has required development of immunological tolerance to prevent ongoing inflammatory responses against intestinal microbes. Breakdown of tolerance to bacterial components of the microbiota can contribute to immune activation and inflammatory disease. However, the mechanisms that are necessary to maintain tolerance to viral components of the microbiome, and the consequences of loss of tolerance, are less well understood. Here, we show that STAT1 is integral for preventing escape of a commensal-like virus, murine norovirus CR6 (MNV CR6), from the gut and that in the absence of STAT1, mice succumb to infection-induced disease. In contrast to the case with other systemic viral infections, mortality of STAT1-deficient mice is not driven by immune-mediated pathology. Our data demonstrate the importance of host-mediated geographical restriction of commensal-like viruses.


Asunto(s)
Infecciones por Caliciviridae , Norovirus , Factor de Transcripción STAT1 , Linfocitos T , Replicación Viral , Animales , Infecciones por Caliciviridae/mortalidad , Infecciones por Caliciviridae/fisiopatología , Mucosa Intestinal/virología , Ratones , Norovirus/fisiología , Factor de Transcripción STAT1/deficiencia , Factor de Transcripción STAT1/genética , Linfocitos T/inmunología , Linfocitos T/virología
19.
J Virol ; 96(5): e0142721, 2022 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-34936483

RESUMEN

Latency is a hallmark of herpesviruses, allowing them to persist in their host without virion production. Acute exposure to hypoxia (below 3% O2) was identified as a trigger of latent-to-lytic switch (reactivation) for human oncogenic gammaherpesviruses (Kaposi's sarcoma-associated virus [KSHV] and Epstein-Barr virus [EBV]). Therefore, we hypothesized that hypoxia could also induce reactivation of Marek's disease virus (MDV), which shares biological properties with EBV and KSHV (notably oncogenic properties), in lymphocytes. Acute exposure to hypoxia (1% O2) of two MDV-latently infected cell lines derived from MD tumors (3867K and MSB-1) induced MDV reactivation. A bioinformatic analysis of the RB-1B MDV genome revealed 214 putative hypoxia response element consensus sequences on 119 open reading frames. Reverse transcriptase quantitative PCR (RT-qPCR) analysis showed five MDV genes strongly upregulated early after hypoxia. In 3867K cells under normoxia, pharmacological agents mimicking hypoxia (MLN4924 and CoCl2) increased MDV reactivation, but to a lower level than real hypoxia. Overexpression of wild-type or stabilized human hypoxia inducible factor 1α (HIF-1α) in MSB-1 cells in normoxia also promoted MDV reactivation. Under such conditions, the lytic cycle was detected in cells with a sustainable HIF-1α expression but also in HIF-1α-negative cells, indicating that MDV reactivation is mediated by HIF-1 in a direct and/or indirect manner. Lastly, we demonstrated by a reporter assay that HIF-1α overexpression induced the transactivation of two viral promoters, shown to be upregulated in hypoxia. These results suggest that hypoxia may play a crucial role in the late lytic replication phase observed in vivo in MDV-infected chickens exhibiting tumors, since a hypoxic microenvironment is a hallmark of most solid tumors. IMPORTANCE Latent-to-lytic switch of herpesviruses (also known as reactivation) is responsible for pathology recurrences and/or viral shedding. Studying physiological triggers of reactivation is therefore important for health to limit lesions and viral transmission. Marek's disease virus (MDV) is a potent oncogenic alphaherpesvirus establishing latency in T lymphocytes and causing lethal T lymphomas in chickens. In vivo, a second lytic phase is observed during the tumoral stage. Hypoxia being a hallmark of tumors, we wondered whether hypoxia induces MDV reactivation in latently infected T lymphocytes, like previously shown for EBV and KSHV in B lymphocytes. In this study, we demonstrated that acute hypoxia (1% O2) triggers MDV reactivation in two MDV transformed T-cell lines. We provide some molecular basis of this reactivation by showing that hypoxia inducible factor 1 (HIF-1) overexpression induces MDV reactivation to an extent similar to that of hypoxia after 24 h. Hypoxia is therefore a reactivation stimulus shared by mammalian and avian oncogenic herpesviruses of different genera.


Asunto(s)
Herpesvirus Gallináceo 2 , Factor 1 Inducible por Hipoxia , Hipoxia , Enfermedad de Marek , Linfocitos T , Activación Viral , Animales , Línea Celular Tumoral , Pollos , Herpesvirus Gallináceo 2/genética , Hipoxia/virología , Factor 1 Inducible por Hipoxia/metabolismo , Linfoma , Enfermedad de Marek/virología , Linfocitos T/virología
20.
J Virol ; 96(12): e0039422, 2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-35612313

RESUMEN

The main target cells for Epstein-Barr virus (EBV) infection and persistence are B lymphocytes, although T and NK cells can also become infected. In this paper, we characterize the EBV present in 21 pediatric and adult patients who were treated in France for a range of diseases that involve infection of T or NK cells. Of these 21 cases, 5 pediatric patients (21%) and 11 adult patients (52%) were of Caucasian origin. In about 30% of the cases, some of the EBV genomes contain a large deletion. The deletions are different in every patient but tend to cluster near the BART region of the viral genome. Detailed investigation of a family in which several members have persistent T or NK cell infection by EBV indicates that the virus genome deletions arise or are selected independently in each individual patient. Genome sequence polymorphisms in the EBV in these T or NK cell diseases reflect the geographic origin of the patient and not a distinct type of EBV (the 21 cases studied included examples of both type 1 and type 2 EBV infection). Using virus produced from type 1 or type 2 EBV genomes cloned in bacterial artificial chromosome (BAC) vectors, we demonstrate infection of T cells in cord blood from healthy donors. Our results are consistent with transient infection of some T cells being part of normal asymptomatic infection by EBV in young children. IMPORTANCE EBV contributes to several types of human cancer. Some cancers and nonmalignant lymphoproliferative diseases involving T or NK cells contain EBV. These diseases are relatively frequent in Japan and China and have been shown sometimes to have deletions in the EBV genome in the disease cells. We identify further examples of deletions within the EBV genome associated with T or NK cell diseases, and we provide evidence that the virus genomes with these deletions are most likely selected in the individual cases, rather than being transmitted between people during infection. We demonstrate EBV infection of cord blood T cells by highly characterized, cloned EBV genomes and suggest that transient infection of T cells may be part of normal asymptomatic infection by EBV in young children.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Eliminación de Gen , Genoma Viral , Herpesvirus Humano 4 , Trastornos Linfoproliferativos , Adulto , Infecciones Asintomáticas , Niño , Herpesvirus Humano 4/genética , Humanos , Células Asesinas Naturales/virología , Trastornos Linfoproliferativos/virología , Linfocitos T/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA