Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.594
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 42(1): 235-258, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38271641

RESUMEN

The choice of developing thymocytes to become CD8+ cytotoxic or CD4+ helper T cells has been intensely studied, but many of the underlying mechanisms remain to be elucidated. Recent multiomics approaches have provided much higher resolution analysis of gene expression in developing thymocytes than was previously achievable, thereby offering a fresh perspective on this question. Focusing on our recent studies using CITE-seq (cellular indexing of transcriptomes and epitopes) analyses of mouse thymocytes, we present a detailed timeline of RNA and protein expression changes during CD8 versus CD4 T cell differentiation. We also revisit our current understanding of the links between T cell receptor signaling and expression of the lineage-defining transcription factors ThPOK and RUNX3. Finally, we propose a sequential selection model to explain the tight linkage between MHC-I versus MHC-II recognition and T cell lineage choice. This model incorporates key aspects of previously proposed kinetic signaling, instructive, and stochastic/selection models.


Asunto(s)
Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Diferenciación Celular , Linaje de la Célula , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Humanos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Ratones , Factores de Transcripción/metabolismo , Transcriptoma , Multiómica
2.
Annu Rev Immunol ; 42(1): 317-345, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38941605

RESUMEN

Regionalized immune surveillance relies on the concerted efforts of diverse memory T cell populations. Of these, tissue-resident memory T (TRM) cells are strategically positioned in barrier tissues, where they enable efficient frontline defense against infections and cancer. However, the long-term persistence of these cells has been implicated in a variety of immune-mediated pathologies. Consequently, modulating TRM cell populations represents an attractive strategy for novel vaccination and therapeutic interventions against tissue-based diseases. Here, we provide an updated overview of TRM cell heterogeneity and function across tissues and disease states. We discuss mechanisms of TRM cell-mediated immune protection and their potential contributions to autoimmune disorders. Finally, we examine how TRM cell responses might be durably boosted or dampened for therapeutic gain.


Asunto(s)
Memoria Inmunológica , Células T de Memoria , Humanos , Animales , Células T de Memoria/inmunología , Células T de Memoria/metabolismo , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Especificidad de Órganos/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Vigilancia Inmunológica
3.
Annu Rev Immunol ; 41: 483-512, 2023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-36750317

RESUMEN

Transforming growth factor ß (TGF-ß) is a key cytokine regulating the development, activation, proliferation, differentiation, and death of T cells. In CD4+ T cells, TGF-ß maintains the quiescence and controls the activation of naive T cells. While inhibiting the differentiation and function of Th1 and Th2 cells, TGF-ß promotes the differentiation of Th17 and Th9 cells. TGF-ß is required for the induction of Foxp3 in naive T cells and the development of regulatory T cells. TGF-ß is crucial in the differentiation of tissue-resident memory CD8+ T cells and their retention in the tissue, whereas it suppresses effector T cell function. In addition, TGF-ß also regulates the generation or function of natural killer T cells, γδ T cells, innate lymphoid cells, and gut intraepithelial lymphocytes. Here I highlight the major findings and recent advances in our understanding of TGF-ß regulation of T cells and provide a personal perspective of the field.


Asunto(s)
Linfocitos T CD8-positivos , Factor de Crecimiento Transformador beta1 , Animales , Humanos , Diferenciación Celular , Inmunidad Innata , Linfocitos/metabolismo , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
4.
Annu Rev Immunol ; 38: 229-247, 2020 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-31928469

RESUMEN

Neonatal CD4+ and CD8+ T cells have historically been characterized as immature or defective. However, recent studies prompt a reinterpretation of the functions of neonatal T cells. Rather than a population of cells always falling short of expectations set by their adult counterparts, neonatal T cells are gaining recognition as a distinct population of lymphocytes well suited for the rapidly changing environment in early life. In this review, I will highlight new evidence indicating that neonatal T cells are not inert or less potent versions of adult T cells but instead are a broadly reactive layer of T cells poised to quickly develop into regulatory or effector cells, depending on the needs of the host. In this way, neonatal T cells are well adapted to provide fast-acting immune protection against foreign pathogens, while also sustaining tolerance to self-antigens.


Asunto(s)
Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Inmunidad Adaptativa , Animales , Biomarcadores , Diferenciación Celular/inmunología , Interacciones Huésped-Patógeno , Humanos , Memoria Inmunológica , Activación de Linfocitos/inmunología , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/inmunología , Células Progenitoras Linfoides/metabolismo , Fenotipo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/citología
5.
Annu Rev Immunol ; 35: 149-176, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28125356

RESUMEN

To monitor the health of cells, the immune system tasks antigen-presenting cells with gathering antigens from other cells and bringing them to CD8 T cells in the form of peptides bound to MHC-I molecules. Most cells would be unable to perform this function because they use their MHC-I molecules to exclusively present peptides derived from the cell's own proteins. However, the immune system evolved mechanisms for dendritic cells and some other phagocytes to sample and present antigens from the extracellular milieu on MHC-I through a process called cross-presentation. How this important task is accomplished, its role in health and disease, and its potential for exploitation are the subject of this review.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Reactividad Cruzada , Células Dendríticas/inmunología , Animales , Antígenos/inmunología , Antígenos/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Vigilancia Inmunológica , Activación de Linfocitos , Fagocitosis
6.
Cell ; 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38897196

RESUMEN

Reversing CD8+ T cell dysfunction is crucial in treating chronic hepatitis B virus (HBV) infection, yet specific molecular targets remain unclear. Our study analyzed co-signaling receptors during hepatocellular priming and traced the trajectory and fate of dysfunctional HBV-specific CD8+ T cells. Early on, these cells upregulate PD-1, CTLA-4, LAG-3, OX40, 4-1BB, and ICOS. While blocking co-inhibitory receptors had minimal effect, activating 4-1BB and OX40 converted them into antiviral effectors. Prolonged stimulation led to a self-renewing, long-lived, heterogeneous population with a unique transcriptional profile. This includes dysfunctional progenitor/stem-like (TSL) cells and two distinct dysfunctional tissue-resident memory (TRM) populations. While 4-1BB expression is ubiquitously maintained, OX40 expression is limited to TSL. In chronic settings, only 4-1BB stimulation conferred antiviral activity. In HBeAg+ chronic patients, 4-1BB activation showed the highest potential to rejuvenate dysfunctional CD8+ T cells. Targeting all dysfunctional T cells, rather than only stem-like precursors, holds promise for treating chronic HBV infection.

7.
Cell ; 187(7): 1651-1665.e21, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38490195

RESUMEN

The immune checkpoint blockade (ICB) response in human cancers is closely linked to the gut microbiota. Here, we report that the abundance of commensal Lactobacillus johnsonii is positively correlated with the responsiveness of ICB. Supplementation with Lactobacillus johnsonii or tryptophan-derived metabolite indole-3-propionic acid (IPA) enhances the efficacy of CD8+ T cell-mediated αPD-1 immunotherapy. Mechanistically, Lactobacillus johnsonii collaborates with Clostridium sporogenes to produce IPA. IPA modulates the stemness program of CD8+ T cells and facilitates the generation of progenitor exhausted CD8+ T cells (Tpex) by increasing H3K27 acetylation at the super-enhancer region of Tcf7. IPA improves ICB responsiveness at the pan-cancer level, including melanoma, breast cancer, and colorectal cancer. Collectively, our findings identify a microbial metabolite-immune regulatory pathway and suggest a potential microbial-based adjuvant approach to improve the responsiveness of immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos , Inmunoterapia , Lactobacillus , Neoplasias , Humanos , Lactobacillus/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Indoles/metabolismo , Inhibidores de Puntos de Control Inmunológico/uso terapéutico
8.
Cell ; 186(6): 1127-1143.e18, 2023 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-36931243

RESUMEN

CD8+ T cell responses are critical for anti-tumor immunity. While extensively profiled in the tumor microenvironment, recent studies in mice identified responses in lymph nodes (LNs) as essential; however, the role of LNs in human cancer patients remains unknown. We examined CD8+ T cells in human head and neck squamous cell carcinomas, regional LNs, and blood using mass cytometry, single-cell genomics, and multiplexed ion beam imaging. We identified progenitor exhausted CD8+ T cells (Tpex) that were abundant in uninvolved LN and clonally related to terminally exhausted cells in the tumor. After anti-PD-L1 immunotherapy, Tpex in uninvolved LNs reduced in frequency but localized near dendritic cells and proliferating intermediate-exhausted CD8+ T cells (Tex-int), consistent with activation and differentiation. LN responses coincided with increased circulating Tex-int. In metastatic LNs, these response hallmarks were impaired, with immunosuppressive cellular niches. Our results identify important roles for LNs in anti-tumor immune responses in humans.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Humanos , Animales , Ratones , Ganglios Linfáticos , Neoplasias/terapia , Neoplasias/patología , Inmunoterapia/métodos , Microambiente Tumoral
9.
Annu Rev Immunol ; 33: 1-28, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25581308

RESUMEN

Herman Eisen and Sondra Schlesinger spent several days together in September 2007 in Woods Hole, Massachusetts, talking about immunology, focusing on his remembrances of the field over the more than 60 years of his involvement. This article is an abridged version of those discussions (the full version is available on the Annual Reviews website). It is both an oral history and a written memory of some important but selected areas of immunology.


Asunto(s)
Alergia e Inmunología , Alergia e Inmunología/historia , Alergia e Inmunología/organización & administración , Alergia e Inmunología/tendencias , Animales , Anticuerpos , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Sistema Inmunológico , Linfocitos T
10.
Cell ; 185(22): 4049-4066.e25, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36208623

RESUMEN

Blocking PD-1/PD-L1 signaling transforms cancer therapy and is assumed to unleash exhausted tumor-reactive CD8+ T cells in the tumor microenvironment (TME). However, recent studies have also indicated that the systemic tumor-reactive CD8+ T cells may respond to PD-1/PD-L1 immunotherapy. These discrepancies highlight the importance of further defining tumor-specific CD8+ T cell responders to PD-1/PD-L1 blockade. Here, using multiple preclinical tumor models, we revealed that a subset of tumor-specific CD8+ cells in the tumor draining lymph nodes (TdLNs) was not functionally exhausted but exhibited canonical memory characteristics. TdLN-derived tumor-specific memory (TTSM) cells established memory-associated epigenetic program early during tumorigenesis. More importantly, TdLN-TTSM cells exhibited superior anti-tumor therapeutic efficacy after adoptive transfer and were characterized as bona fide responders to PD-1/PD-L1 blockade. These findings highlight that TdLN-TTSM cells could be harnessed to potentiate anti-tumor immunotherapy.


Asunto(s)
Antígeno B7-H1 , Neoplasias , Humanos , Receptor de Muerte Celular Programada 1 , Linfocitos T CD8-positivos , Inhibidores de Puntos de Control Inmunológico , Microambiente Tumoral , Neoplasias/terapia , Neoplasias/patología , Ganglios Linfáticos/patología
11.
Cell ; 185(16): 2936-2951.e19, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35931021

RESUMEN

We studied the prevalent cytotoxic CD8 T cell response mounted against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Spike glycoprotein269-277 epitope (sequence YLQPRTFLL) via the most frequent human leukocyte antigen (HLA) class I worldwide, HLA A∗02. The Spike P272L mutation that has arisen in at least 112 different SARS-CoV-2 lineages to date, including in lineages classified as "variants of concern," was not recognized by the large CD8 T cell response seen across cohorts of HLA A∗02+ convalescent patients and individuals vaccinated against SARS-CoV-2, despite these responses comprising of over 175 different individual T cell receptors. Viral escape at prevalent T cell epitopes restricted by high frequency HLAs may be particularly problematic when vaccine immunity is focused on a single protein such as SARS-CoV-2 Spike, providing a strong argument for inclusion of multiple viral proteins in next generation vaccines and highlighting the need for monitoring T cell escape in new SARS-CoV-2 variants.


Asunto(s)
COVID-19 , SARS-CoV-2 , Linfocitos T CD8-positivos , Epítopos de Linfocito T , Antígenos HLA-A , Antígenos de Histocompatibilidad Clase I , Humanos
12.
Cell ; 185(4): 585-602.e29, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-35051368

RESUMEN

The relevance of extracellular magnesium in cellular immunity remains largely unknown. Here, we show that the co-stimulatory cell-surface molecule LFA-1 requires magnesium to adopt its active conformation on CD8+ T cells, thereby augmenting calcium flux, signal transduction, metabolic reprogramming, immune synapse formation, and, as a consequence, specific cytotoxicity. Accordingly, magnesium-sufficiency sensed via LFA-1 translated to the superior performance of pathogen- and tumor-specific T cells, enhanced effectiveness of bi-specific T cell engaging antibodies, and improved CAR T cell function. Clinically, low serum magnesium levels were associated with more rapid disease progression and shorter overall survival in CAR T cell and immune checkpoint antibody-treated patients. LFA-1 thus directly incorporates information on the composition of the microenvironment as a determinant of outside-in signaling activity. These findings conceptually link co-stimulation and nutrient sensing and point to the magnesium-LFA-1 axis as a therapeutically amenable biologic system.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Magnesio/metabolismo , Animales , Infecciones Bacterianas/inmunología , Restricción Calórica , Línea Celular Tumoral , Citotoxicidad Inmunológica , Células HEK293 , Humanos , Memoria Inmunológica , Sinapsis Inmunológicas/metabolismo , Inmunoterapia , Activación de Linfocitos/inmunología , Sistema de Señalización de MAP Quinasas , Magnesio/administración & dosificación , Masculino , Ratones Endogámicos C57BL , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Fenotipo , Fosforilación , Proteínas Proto-Oncogénicas c-jun/metabolismo
13.
Cell ; 185(7): 1189-1207.e25, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35325594

RESUMEN

Macrophage infiltration is a hallmark of solid cancers, and overall macrophage infiltration correlates with lower patient survival and resistance to therapy. Tumor-associated macrophages, however, are phenotypically and functionally heterogeneous. Specific subsets of tumor-associated macrophage might be endowed with distinct roles on cancer progression and antitumor immunity. Here, we identify a discrete population of FOLR2+ tissue-resident macrophages in healthy mammary gland and breast cancer primary tumors. FOLR2+ macrophages localize in perivascular areas in the tumor stroma, where they interact with CD8+ T cells. FOLR2+ macrophages efficiently prime effector CD8+ T cells ex vivo. The density of FOLR2+ macrophages in tumors positively correlates with better patient survival. This study highlights specific roles for tumor-associated macrophage subsets and paves the way for subset-targeted therapeutic interventions in macrophages-based cancer therapies.


Asunto(s)
Neoplasias de la Mama , Macrófagos , Mama/inmunología , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/inmunología , Linfocitos T CD8-positivos , Femenino , Receptor 2 de Folato , Humanos , Linfocitos Infiltrantes de Tumor , Pronóstico
14.
Cell ; 185(1): 131-144.e18, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34919814

RESUMEN

Two HIV fusion-inhibitory lipopeptides (LP-97 and LP-98) were designed with highly potent, long-acting antiviral activity. Monotherapy using a low dose of LP-98 sharply reduced viral loads and maintained long-term viral suppression in 21 SHIVSF162P3-infected rhesus macaques. We found that five treated monkeys achieved potential posttreatment control (PTC) efficacy and had lower viral DNA in deep lymph nodes, whereas monkeys with a stable viral rebound had higher viral DNA in superficial lymph nodes. The tissues of PTC monkeys exhibited significantly decreased quantitative viral outgrowth and fewer PD-1+ central memory CD4+ T cells, and CD8+ T cells contributed to virologic control efficacy. Moreover, LP-98 administrated as a pre-exposure prophylaxis (PrEP) provided complete protection against SHIVSF162P3 and SIVmac239 infections in 51 monkeys via intrarectal, intravaginal, or intravenous challenge. In conclusion, our lipopeptides exhibit high potential as an efficient HIV treatment or prevention strategy.


Asunto(s)
Inhibidores de Fusión de VIH/administración & dosificación , Lipopéptidos/administración & dosificación , Profilaxis Pre-Exposición/métodos , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Femenino , Células HEK293 , Humanos , Macaca mulatta , Masculino , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Respuesta Virológica Sostenida , Células U937 , Carga Viral/efectos de los fármacos
15.
Cell ; 184(5): 1262-1280.e22, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33636129

RESUMEN

Improving effector activity of antigen-specific T cells is a major goal in cancer immunotherapy. Despite the identification of several effector T cell (TEFF)-driving transcription factors (TFs), the transcriptional coordination of TEFF biology remains poorly understood. We developed an in vivo T cell CRISPR screening platform and identified a key mechanism restraining TEFF biology through the ETS family TF, Fli1. Genetic deletion of Fli1 enhanced TEFF responses without compromising memory or exhaustion precursors. Fli1 restrained TEFF lineage differentiation by binding to cis-regulatory elements of effector-associated genes. Loss of Fli1 increased chromatin accessibility at ETS:RUNX motifs, allowing more efficient Runx3-driven TEFF biology. CD8+ T cells lacking Fli1 provided substantially better protection against multiple infections and tumors. These data indicate that Fli1 safeguards the developing CD8+ T cell transcriptional landscape from excessive ETS:RUNX-driven TEFF cell differentiation. Moreover, genetic deletion of Fli1 improves TEFF differentiation and protective immunity in infections and cancer.


Asunto(s)
Linfocitos T CD8-positivos/citología , Proteína Proto-Oncogénica c-fli-1/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Sistemas CRISPR-Cas , Diferenciación Celular , Enfermedad Crónica , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Epigénesis Genética , Redes Reguladoras de Genes , Infecciones/inmunología , Ratones , Neoplasias/inmunología
16.
Cell ; 184(25): 6101-6118.e13, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34852236

RESUMEN

CD4 T follicular helper (TFH) cells support B cells, which are critical for germinal center (GC) formation, but the importance of TFH-B cell interactions in cancer is unclear. We found enrichment of TFH cell transcriptional signature correlates with GC B cell signature and with prolonged survival in individuals with lung adenocarcinoma (LUAD). We further developed a murine LUAD model in which tumor cells express B cell- and T cell-recognized neoantigens. Interactions between tumor-specific TFH and GC B cells, as well as interleukin (IL)-21 primarily produced by TFH cells, are necessary for tumor control and effector CD8 T cell function. Development of TFH cells requires B cells and B cell-recognized neoantigens. Thus, tumor neoantigens can regulate the fate of tumor-specific CD4 T cells by facilitating their interactions with tumor-specific B cells, which in turn promote anti-tumor immunity by enhancing CD8 T cell effector functions.


Asunto(s)
Adenocarcinoma/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Interleucinas/inmunología , Neoplasias Pulmonares/inmunología , Animales , Linfocitos B/citología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
17.
Cell ; 184(19): 4996-5014.e26, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34534464

RESUMEN

CD8 T cell responses against different tumor neoantigens occur simultaneously, yet little is known about the interplay between responses and its impact on T cell function and tumor control. In mouse lung adenocarcinoma, we found that immunodominance is established in tumors, wherein CD8 T cell expansion is predominantly driven by the antigen that most stably binds MHC. T cells responding to subdominant antigens were enriched for a TCF1+ progenitor phenotype correlated with response to immune checkpoint blockade (ICB) therapy. However, the subdominant T cell response did not preferentially benefit from ICB due to a dysfunctional subset of TCF1+ cells marked by CCR6 and Tc17 differentiation. Analysis of human samples and sequencing datasets revealed that CCR6+ TCF1+ cells exist across human cancers and are not correlated with ICB response. Vaccination eliminated CCR6+ TCF1+ cells and dramatically improved the subdominant response, highlighting a strategy to optimally engage concurrent neoantigen responses against tumors.


Asunto(s)
Adenocarcinoma del Pulmón/inmunología , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Neoplasias Pulmonares/inmunología , Células Madre/inmunología , Secuencia de Aminoácidos , Animales , Antígeno CTLA-4/metabolismo , Epítopos , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Pulmonares/patología , Ratones , Péptidos/química , Fenotipo , Receptor de Muerte Celular Programada 1/metabolismo , RNA-Seq , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores CCR6/metabolismo , Análisis de la Célula Individual , Vacunación
18.
Cell ; 184(11): 2988-3005.e16, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-34019793

RESUMEN

Clear cell renal carcinoma (ccRCC) is a heterogeneous disease with a variable post-surgical course. To assemble a comprehensive ccRCC tumor microenvironment (TME) atlas, we performed single-cell RNA sequencing (scRNA-seq) of hematopoietic and non-hematopoietic subpopulations from tumor and tumor-adjacent tissue of treatment-naive ccRCC resections. We leveraged the VIPER algorithm to quantitate single-cell protein activity and validated this approach by comparison to flow cytometry. The analysis identified key TME subpopulations, as well as their master regulators and candidate cell-cell interactions, revealing clinically relevant populations, undetectable by gene-expression analysis. Specifically, we uncovered a tumor-specific macrophage subpopulation characterized by upregulation of TREM2/APOE/C1Q, validated by spatially resolved, quantitative multispectral immunofluorescence. In a large clinical validation cohort, these markers were significantly enriched in tumors from patients who recurred following surgery. The study thus identifies TREM2/APOE/C1Q-positive macrophage infiltration as a potential prognostic biomarker for ccRCC recurrence, as well as a candidate therapeutic target.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Recurrencia Local de Neoplasia/genética , Macrófagos Asociados a Tumores/metabolismo , Adulto , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Biomarcadores de Tumor/genética , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Estudios de Cohortes , Femenino , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Riñón/metabolismo , Neoplasias Renales/patología , Linfocitos Infiltrantes de Tumor/patología , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Persona de Mediana Edad , Recurrencia Local de Neoplasia/metabolismo , Pronóstico , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Microambiente Tumoral , Macrófagos Asociados a Tumores/fisiología
19.
Cell ; 184(17): 4401-4413.e10, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34265281

RESUMEN

The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants that escape convalescent and vaccine-induced antibody responses has renewed focus on the development of broadly protective T-cell-based vaccines. Here, we apply structure-based network analysis and assessments of HLA class I peptide stability to define mutationally constrained CD8+ T cell epitopes across the SARS-CoV-2 proteome. Highly networked residues are conserved temporally among circulating variants and sarbecoviruses and disproportionately impair spike pseudotyped lentivirus infectivity when mutated. Evaluation of HLA class I stabilizing activity for 18 globally prevalent alleles identifies CD8+ T cell epitopes within highly networked regions with limited mutational frequencies in circulating SARS-CoV-2 variants and deep-sequenced primary isolates. Moreover, these epitopes elicit demonstrable CD8+ T cell reactivity in convalescent individuals but reduced recognition in recipients of mRNA-based vaccines. These data thereby elucidate key mutationally constrained regions and immunogenic epitopes in the SARS-CoV-2 proteome for a global T-cell-based vaccine against emerging variants and SARS-like coronaviruses.


Asunto(s)
Vacunas contra la COVID-19/inmunología , Epítopos de Linfocito T , Linfocitos T CD8-positivos/inmunología , COVID-19/inmunología , COVID-19/prevención & control , Vacunas contra la COVID-19/química , Antígenos HLA/inmunología , Humanos , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo
20.
Cell ; 184(7): 1804-1820.e16, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33691139

RESUMEN

SARS-CoV-2 has caused the global COVID-19 pandemic. Although passively delivered neutralizing antibodies against SARS-CoV-2 show promise in clinical trials, their mechanism of action in vivo is incompletely understood. Here, we define correlates of protection of neutralizing human monoclonal antibodies (mAbs) in SARS-CoV-2-infected animals. Whereas Fc effector functions are dispensable when representative neutralizing mAbs are administered as prophylaxis, they are required for optimal protection as therapy. When given after infection, intact mAbs reduce SARS-CoV-2 burden and lung disease in mice and hamsters better than loss-of-function Fc variant mAbs. Fc engagement of neutralizing antibodies mitigates inflammation and improves respiratory mechanics, and transcriptional profiling suggests these phenotypes are associated with diminished innate immune signaling and preserved tissue repair. Immune cell depletions establish that neutralizing mAbs require monocytes and CD8+ T cells for optimal clinical and virological benefit. Thus, potently neutralizing mAbs utilize Fc effector functions during therapy to mitigate lung infection and disease.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Linfocitos T CD8-positivos , COVID-19 , Fragmentos Fc de Inmunoglobulinas/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/uso terapéutico , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Células CHO , COVID-19/inmunología , COVID-19/terapia , Chlorocebus aethiops , Cricetulus , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , SARS-CoV-2/inmunología , Células Vero , Carga Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA