Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 16.486
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 186(8): 1652-1669, 2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-37059068

RESUMEN

Immune checkpoint therapy (ICT) has dramatically altered clinical outcomes for cancer patients and conferred durable clinical benefits, including cure in a subset of patients. Varying response rates across tumor types and the need for predictive biomarkers to optimize patient selection to maximize efficacy and minimize toxicities prompted efforts to unravel immune and non-immune factors regulating the responses to ICT. This review highlights the biology of anti-tumor immunity underlying response and resistance to ICT, discusses efforts to address the current challenges with ICT, and outlines strategies to guide the development of subsequent clinical trials and combinatorial efforts with ICT.


Asunto(s)
Inmunoterapia , Neoplasias , Humanos , Antígeno B7-H1 , Neoplasias/tratamiento farmacológico , Ensayos Clínicos como Asunto , Inhibidores de Puntos de Control Inmunológico/administración & dosificación
2.
Cell ; 185(2): 345-360.e28, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-35063075

RESUMEN

We present a whole-cell fully dynamical kinetic model (WCM) of JCVI-syn3A, a minimal cell with a reduced genome of 493 genes that has retained few regulatory proteins or small RNAs. Cryo-electron tomograms provide the cell geometry and ribosome distributions. Time-dependent behaviors of concentrations and reaction fluxes from stochastic-deterministic simulations over a cell cycle reveal how the cell balances demands of its metabolism, genetic information processes, and growth, and offer insight into the principles of life for this minimal cell. The energy economy of each process including active transport of amino acids, nucleosides, and ions is analyzed. WCM reveals how emergent imbalances lead to slowdowns in the rates of transcription and translation. Integration of experimental data is critical in building a kinetic model from which emerges a genome-wide distribution of mRNA half-lives, multiple DNA replication events that can be compared to qPCR results, and the experimentally observed doubling behavior.


Asunto(s)
Células/citología , Simulación por Computador , Adenosina Trifosfato/metabolismo , Ciclo Celular/genética , Proliferación Celular/genética , Células/metabolismo , Replicación del ADN/genética , Regulación de la Expresión Génica , Imagenología Tridimensional , Cinética , Lípidos/química , Redes y Vías Metabólicas , Metaboloma , Anotación de Secuencia Molecular , Nucleótidos/metabolismo , Termodinámica , Factores de Tiempo
3.
Cell ; 184(5): 1214-1231.e16, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33636133

RESUMEN

Although enteric helminth infections modulate immunity to mucosal pathogens, their effects on systemic microbes remain less established. Here, we observe increased mortality in mice coinfected with the enteric helminth Heligmosomoides polygyrus bakeri (Hpb) and West Nile virus (WNV). This enhanced susceptibility is associated with altered gut morphology and transit, translocation of commensal bacteria, impaired WNV-specific T cell responses, and increased virus infection in the gastrointestinal tract and central nervous system. These outcomes were due to type 2 immune skewing, because coinfection in Stat6-/- mice rescues mortality, treatment of helminth-free WNV-infected mice with interleukin (IL)-4 mirrors coinfection, and IL-4 receptor signaling in intestinal epithelial cells mediates the susceptibility phenotypes. Moreover, tuft cell-deficient mice show improved outcomes with coinfection, whereas treatment of helminth-free mice with tuft cell-derived cytokine IL-25 or ligand succinate worsens WNV disease. Thus, helminth activation of tuft cell-IL-4-receptor circuits in the gut exacerbates infection and disease of a neurotropic flavivirus.


Asunto(s)
Coinfección , Nematospiroides dubius/fisiología , Transducción de Señal , Infecciones por Strongylida/patología , Virus del Nilo Occidental/fisiología , Animales , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Mucosa Intestinal/parasitología , Mucosa Intestinal/virología , Ratones , Ratones Endogámicos C57BL , Neuronas/parasitología , Neuronas/virología , Receptores de Interleucina-4/metabolismo , Factor de Transcripción STAT6/genética , Índice de Severidad de la Enfermedad , Infecciones por Strongylida/parasitología
4.
Cell ; 183(7): 1826-1847.e31, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33296702

RESUMEN

Inborn errors of human interferon gamma (IFN-γ) immunity underlie mycobacterial disease. We report a patient with mycobacterial disease due to inherited deficiency of the transcription factor T-bet. The patient has extremely low counts of circulating Mycobacterium-reactive natural killer (NK), invariant NKT (iNKT), mucosal-associated invariant T (MAIT), and Vδ2+ γδ T lymphocytes, and of Mycobacterium-non reactive classic TH1 lymphocytes, with the residual populations of these cells also producing abnormally small amounts of IFN-γ. Other lymphocyte subsets develop normally but produce low levels of IFN-γ, with the exception of CD8+ αß T and non-classic CD4+ αß TH1∗ lymphocytes, which produce IFN-γ normally in response to mycobacterial antigens. Human T-bet deficiency thus underlies mycobacterial disease by preventing the development of innate (NK) and innate-like adaptive lymphocytes (iNKT, MAIT, and Vδ2+ γδ T cells) and IFN-γ production by them, with mycobacterium-specific, IFN-γ-producing, purely adaptive CD8+ αß T, and CD4+ αß TH1∗ cells unable to compensate for this deficit.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Interferón gamma/inmunología , Mycobacterium/inmunología , Proteínas de Dominio T Box/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Linaje de la Célula , Preescolar , Cromatina/metabolismo , Islas de CpG/genética , Metilación de ADN/genética , Células Dendríticas/metabolismo , Epigénesis Genética , Femenino , Homocigoto , Humanos , Mutación INDEL/genética , Lactante , Interferón gamma/metabolismo , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Mutación con Pérdida de Función/genética , Masculino , Infecciones por Mycobacterium/genética , Infecciones por Mycobacterium/inmunología , Infecciones por Mycobacterium/microbiología , Linaje , Proteínas de Dominio T Box/química , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética , Linfocitos T Colaboradores-Inductores/inmunología , Transcriptoma/genética
5.
Nat Immunol ; 23(9): 1317-1323, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35999393

RESUMEN

An orchestrated cellular network, including adaptive lymphocytes and group 3 innate lymphoid cells (ILC3s), maintains intestinal barrier integrity and homeostasis. T cells can monitor environmental insults through constitutive circulation, scanning tissues and forming immunological contacts, a process named immunosurveillance. In contrast, the dynamics of intestinal ILC3s are unknown. Using intravital imaging, we observed that villus ILC3s were largely immotile at steady state but acquired migratory 'patrolling' attributes and enhanced cytokine expression in response to inflammation. We showed that T cells, the chemokine CCL25 and bacterial ligands regulated intestinal ILC3 behavior and that loss of patrolling behavior by interleukin-22 (IL-22)-producing ILC3s altered the intestinal barrier through increased epithelial cell death. Collectively, we identified notable differences between the behavior of ILC3s and T cells, with a prominent adaptation of intestinal ILC3s toward mucosal immunosurveillance after inflammation.


Asunto(s)
Inmunidad Innata , Linfocitos , Citocinas/metabolismo , Humanos , Inflamación/metabolismo , Mucosa Intestinal , Ligandos
6.
Nat Immunol ; 22(11): 1367-1374, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34686862

RESUMEN

Group 2 innate lymphoid cells (ILC2s) represent innate homologs of type 2 helper T cells (TH2) that participate in immune defense and tissue homeostasis through production of type 2 cytokines. While T lymphocytes metabolically adapt to microenvironmental changes, knowledge of human ILC2 metabolism is limited, and its key regulators are unknown. Here, we show that circulating 'naive' ILC2s have an unexpected metabolic profile with a higher level of oxidative phosphorylation (OXPHOS) than natural killer (NK) cells. Accordingly, ILC2s are severely reduced in individuals with mitochondrial disease (MD) and impaired OXPHOS. Metabolomic and nutrient receptor analysis revealed ILC2 uptake of amino acids to sustain OXPHOS at steady state. Following activation with interleukin-33 (IL-33), ILC2s became highly proliferative, relying on glycolysis and mammalian target of rapamycin (mTOR) to produce IL-13 while continuing to fuel OXPHOS with amino acids to maintain cellular fitness and proliferation. Our results suggest that proliferation and function are metabolically uncoupled in human ILC2s, offering new strategies to target ILC2s in disease settings.


Asunto(s)
Proliferación Celular , Citocinas/metabolismo , Metabolismo Energético , Inmunidad Innata , Activación de Linfocitos , Enfermedades Mitocondriales/metabolismo , Células Th2/metabolismo , Aminoácidos de Cadena Ramificada/metabolismo , Arginina/metabolismo , Estudios de Casos y Controles , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Metabolismo Energético/efectos de los fármacos , Humanos , Inmunidad Innata/efectos de los fármacos , Interleucina-33/farmacología , Activación de Linfocitos/efectos de los fármacos , Mitocondrias/metabolismo , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/inmunología , Fenotipo , Células Th2/efectos de los fármacos , Células Th2/inmunología
7.
Nat Immunol ; 22(11): 1428-1439, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34471264

RESUMEN

Coordinated local mucosal and systemic immune responses following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection either protect against coronavirus disease 2019 (COVID-19) pathologies or fail, leading to severe clinical outcomes. To understand this process, we performed an integrated analysis of SARS-CoV-2 spike-specific antibodies, cytokines, viral load and bacterial communities in paired nasopharyngeal swabs and plasma samples from a cohort of clinically distinct patients with COVID-19 during acute infection. Plasma viral load was associated with systemic inflammatory cytokines that were elevated in severe COVID-19, and also with spike-specific neutralizing antibodies. By contrast, nasopharyngeal viral load correlated with SARS-CoV-2 humoral responses but inversely with interferon responses, the latter associating with protective microbial communities. Potential pathogenic microorganisms, often implicated in secondary respiratory infections, were associated with mucosal inflammation and elevated in severe COVID-19. Our results demonstrate distinct tissue compartmentalization of SARS-CoV-2 immune responses and highlight a role for the nasopharyngeal microbiome in regulating local and systemic immunity that determines COVID-19 clinical outcomes.


Asunto(s)
COVID-19/inmunología , Microbiota/inmunología , Nasofaringe/inmunología , SARS-CoV-2/fisiología , Enfermedad Aguda , Adolescente , Adulto , Anciano , Anticuerpos Antivirales/sangre , Estudios de Cohortes , Femenino , Humanos , Inmunidad Humoral , Inmunidad Mucosa , Interferones/sangre , Masculino , Persona de Mediana Edad , Nasofaringe/microbiología , Glicoproteína de la Espiga del Coronavirus/inmunología , Carga Viral , Adulto Joven
8.
Cell ; 175(5): 1198-1212.e12, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30293866

RESUMEN

Although chronic gastrointestinal dysmotility syndromes are a common worldwide health problem, underlying causes for these disorders are poorly understood. We show that flavivirus infection of enteric neurons leads to acute neuronal injury and cell death, inflammation, bowel dilation, and slowing of intestinal transit in mice. Flavivirus-primed CD8+ T cells promote these phenotypes, as their absence diminished enteric neuron injury and intestinal transit delays, and their adoptive transfer reestablished dysmotility after flavivirus infection. Remarkably, mice surviving acute flavivirus infection developed chronic gastrointestinal dysmotility that was exacerbated by immunization with an unrelated alphavirus vaccine or exposure to a non-infectious inflammatory stimulus. This model of chronic post-infectious gastrointestinal dysmotility in mice suggests that viral infections with tropism for enteric neurons and the ensuing immune response might contribute to the development of bowel motility disorders in humans. These results suggest an opportunity for unique approaches to diagnosis and therapy of gastrointestinal dysmotility syndromes.


Asunto(s)
Infecciones por Flavivirus/patología , Flavivirus/patogenicidad , Motilidad Gastrointestinal , Intestinos/patología , Animales , Linfocitos T CD8-positivos/inmunología , Flavivirus/genética , Infecciones por Flavivirus/inmunología , Infecciones por Flavivirus/virología , Intestinos/virología , Leucocitos/citología , Leucocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/patología , Neuronas/ultraestructura , ARN Viral/aislamiento & purificación , ARN Viral/metabolismo , Síndrome
9.
Cell ; 174(5): 1054-1066, 2018 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-30142344

RESUMEN

Innate lymphoid cells (ILCs) are lymphocytes that do not express the type of diversified antigen receptors expressed on T cells and B cells. ILCs are largely tissue-resident cells and are deeply integrated into the fabric of tissues. The discovery and investigation of ILCs over the past decade has changed our perception of immune regulation and how the immune system contributes to the maintenance of tissue homeostasis. We now know that cytokine-producing ILCs contribute to multiple immune pathways by, for example, sustaining appropriate immune responses to commensals and pathogens at mucosal barriers, potentiating adaptive immunity, and regulating tissue inflammation. Critically, the biology of ILCs also extends beyond classical immunology to metabolic homeostasis, tissue remodeling, and dialog with the nervous system. The last 10 years have also contributed to our greater understanding of the transcriptional networks that regulate lymphocyte commitment and delineation. This, in conjunction with the recent advances in our understanding of the influence of local tissue microenvironments on the plasticity and function of ILCs, has led to a re-evaluation of their existing categorization. In this review, we distill the advances in ILC biology over the past decade to refine the nomenclature of ILCs and highlight the importance of ILCs in tissue homeostasis, morphogenesis, metabolism, repair, and regeneration.


Asunto(s)
Inmunidad Adaptativa/fisiología , Inmunidad Innata , Linfocitos/citología , Animales , Linfocitos B/inmunología , Citocinas/inmunología , Homeostasis , Humanos , Sistema Hipotálamo-Hipofisario , Inflamación/inmunología , Células Asesinas Naturales/citología , Ratones , Fenotipo , Sistema Hipófiso-Suprarrenal , Regeneración , Linfocitos T/inmunología
10.
Cell ; 170(6): 1120-1133.e17, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28803728

RESUMEN

Immune-checkpoint blockade is able to achieve durable responses in a subset of patients; however, we lack a satisfying comprehension of the underlying mechanisms of anti-CTLA-4- and anti-PD-1-induced tumor rejection. To address these issues, we utilized mass cytometry to comprehensively profile the effects of checkpoint blockade on tumor immune infiltrates in human melanoma and murine tumor models. These analyses reveal a spectrum of tumor-infiltrating T cell populations that are highly similar between tumor models and indicate that checkpoint blockade targets only specific subsets of tumor-infiltrating T cell populations. Anti-PD-1 predominantly induces the expansion of specific tumor-infiltrating exhausted-like CD8 T cell subsets. In contrast, anti-CTLA-4 induces the expansion of an ICOS+ Th1-like CD4 effector population in addition to engaging specific subsets of exhausted-like CD8 T cells. Thus, our findings indicate that anti-CTLA-4 and anti-PD-1 checkpoint-blockade-induced immune responses are driven by distinct cellular mechanisms.


Asunto(s)
Antígeno CTLA-4/antagonistas & inhibidores , Melanoma/inmunología , Melanoma/terapia , Metástasis de la Neoplasia/inmunología , Metástasis de la Neoplasia/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Subgrupos de Linfocitos T/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Regulación de la Expresión Génica , Humanos , Inmunoterapia , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/patología , Análisis de la Célula Individual , Transcripción Genética
11.
Cell ; 168(6): 1086-1100.e10, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28283063

RESUMEN

Innate lymphoid cells (ILCs) represent innate versions of T helper and cytotoxic T cells that differentiate from committed ILC precursors (ILCPs). How ILCPs give rise to mature tissue-resident ILCs remains unclear. Here, we identify circulating and tissue ILCPs in humans that fail to express the transcription factors and cytokine outputs of mature ILCs but have these signature loci in an epigenetically poised configuration. Human ILCPs robustly generate all ILC subsets in vitro and in vivo. While human ILCPs express low levels of retinoic acid receptor (RAR)-related orphan receptor C (RORC) transcripts, these cells are found in RORC-deficient patients and retain potential for EOMES+ natural killer (NK) cells, interferon gamma-positive (IFN-γ+) ILC1s, interleukin (IL)-13+ ILC2s, and for IL-22+, but not for IL-17A+ ILC3s. Our results support a model of tissue ILC differentiation ("ILC-poiesis"), whereby diverse ILC subsets are generated in situ from systemically distributed ILCPs in response to local environmental signals.


Asunto(s)
Linfocitos/citología , Células Madre/citología , Animales , Antígenos CD34/análisis , Diferenciación Celular , Linaje de la Célula , Sangre Fetal/citología , Feto/citología , Humanos , Inmunidad Innata , Interleucina-17 , Hígado/citología , Pulmón/citología , Linfocitos/inmunología , Tejido Linfoide/citología , Ratones , Proteínas Proto-Oncogénicas c-kit/análisis , Transcripción Genética
12.
Immunity ; 55(3): 423-441.e9, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35139355

RESUMEN

Cell death plays an important role during pathogen infections. Here, we report that interferon-γ (IFNγ) sensitizes macrophages to Toll-like receptor (TLR)-induced death that requires macrophage-intrinsic death ligands and caspase-8 enzymatic activity, which trigger the mitochondrial apoptotic effectors, BAX and BAK. The pro-apoptotic caspase-8 substrate BID was dispensable for BAX and BAK activation. Instead, caspase-8 reduced pro-survival BCL-2 transcription and increased inducible nitric oxide synthase (iNOS), thus facilitating BAX and BAK signaling. IFNγ-primed, TLR-induced macrophage killing required iNOS, which licensed apoptotic caspase-8 activity and reduced the BAX and BAK inhibitors, A1 and MCL-1. The deletion of iNOS or caspase-8 limited SARS-CoV-2-induced disease in mice, while caspase-8 caused lethality independent of iNOS in a model of hemophagocytic lymphohistiocytosis. These findings reveal that iNOS selectively licenses programmed cell death, which may explain how nitric oxide impacts disease severity in SARS-CoV-2 infection and other iNOS-associated inflammatory conditions.


Asunto(s)
COVID-19/inmunología , Caspasa 8/metabolismo , Interferón gamma/metabolismo , Linfohistiocitosis Hemofagocítica/inmunología , Macrófagos/inmunología , Mitocondrias/metabolismo , SARS-CoV-2/fisiología , Animales , Caspasa 8/genética , Células Cultivadas , Citotoxicidad Inmunológica , Humanos , Interferón gamma/genética , Activación de Macrófagos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Transducción de Señal , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
13.
Cell ; 167(2): 397-404.e9, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27667683

RESUMEN

Antibody blockade of the inhibitory CTLA-4 pathway has led to clinical benefit in a subset of patients with metastatic melanoma. Anti-CTLA-4 enhances T cell responses, including production of IFN-γ, which is a critical cytokine for host immune responses. However, the role of IFN-γ signaling in tumor cells in the setting of anti-CTLA-4 therapy remains unknown. Here, we demonstrate that patients identified as non-responders to anti-CTLA-4 (ipilimumab) have tumors with genomic defects in IFN-γ pathway genes. Furthermore, mice bearing melanoma tumors with knockdown of IFN-γ receptor 1 (IFNGR1) have impaired tumor rejection upon anti-CTLA-4 therapy. These data highlight that loss of the IFN-γ signaling pathway is associated with primary resistance to anti-CTLA-4 therapy. Our findings demonstrate the importance of tumor genomic data, especially IFN-γ related genes, as prognostic information for patients selected to receive treatment with immune checkpoint therapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígeno CTLA-4/antagonistas & inhibidores , Resistencia a Antineoplásicos/genética , Interferón gamma/genética , Melanoma/tratamiento farmacológico , Receptores de Interferón/genética , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Citocinas/inmunología , Técnicas de Silenciamiento del Gen , Humanos , Ipilimumab , Melanoma/genética , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/genética , Ratones , Ratones Endogámicos C57BL , Neoplasias Cutáneas/genética , Linfocitos T/inmunología , Receptor de Interferón gamma
14.
Cell ; 166(5): 1231-1246.e13, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27545347

RESUMEN

Innate lymphoid cells (ILCs) are critical modulators of mucosal immunity, inflammation, and tissue homeostasis, but their full spectrum of cellular states and regulatory landscapes remains elusive. Here, we combine genome-wide RNA-seq, ChIP-seq, and ATAC-seq to compare the transcriptional and epigenetic identity of small intestinal ILCs, identifying thousands of distinct gene profiles and regulatory elements. Single-cell RNA-seq and flow and mass cytometry analyses reveal compartmentalization of cytokine expression and metabolic activity within the three classical ILC subtypes and highlight transcriptional states beyond the current canonical classification. In addition, using antibiotic intervention and germ-free mice, we characterize the effect of the microbiome on the ILC regulatory landscape and determine the response of ILCs to microbial colonization at the single-cell level. Together, our work characterizes the spectrum of transcriptional identities of small intestinal ILCs and describes how ILCs differentially integrate signals from the microbial microenvironment to generate phenotypic and functional plasticity.


Asunto(s)
Microbioma Gastrointestinal , Inmunidad Innata/genética , Intestinos/inmunología , Intestinos/microbiología , Linfocitos/inmunología , Linfocitos/microbiología , Animales , Secuencia de Bases , Cromatina/metabolismo , Citocinas/inmunología , Epigénesis Genética , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Análisis de la Célula Individual , Transcripción Genética
15.
Nat Immunol ; 19(3): 302-314, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29476184

RESUMEN

The quantification and characterization of circulating immune cells provide key indicators of human health and disease. To identify the relative effects of environmental and genetic factors on variation in the parameters of innate and adaptive immune cells in homeostatic conditions, we combined standardized flow cytometry of blood leukocytes and genome-wide DNA genotyping of 1,000 healthy, unrelated people of Western European ancestry. We found that smoking, together with age, sex and latent infection with cytomegalovirus, were the main non-genetic factors that affected variation in parameters of human immune cells. Genome-wide association studies of 166 immunophenotypes identified 15 loci that showed enrichment for disease-associated variants. Finally, we demonstrated that the parameters of innate cells were more strongly controlled by genetic variation than were those of adaptive cells, which were driven by mainly environmental exposure. Our data establish a resource that will generate new hypotheses in immunology and highlight the role of innate immunity in susceptibility to common autoimmune diseases.


Asunto(s)
Variación Genética/inmunología , Inmunidad Innata/genética , Inmunidad Adaptativa/genética , Adulto , Anciano , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Inmunofenotipificación , Masculino , Persona de Mediana Edad , Adulto Joven
17.
Cell ; 161(2): 205-14, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25860605

RESUMEN

Research in two fronts has enabled the development of therapies that provide significant benefit to cancer patients. One area stems from a detailed knowledge of mutations that activate or inactivate signaling pathways that drive cancer development. This work triggered the development of targeted therapies that lead to clinical responses in the majority of patients bearing the targeted mutation, although responses are often of limited duration. In the second front are the advances in molecular immunology that unveiled the complexity of the mechanisms regulating cellular immune responses. These developments led to the successful targeting of immune checkpoints to unleash anti-tumor T cell responses, resulting in durable long-lasting responses but only in a fraction of patients. In this Review, we discuss the evolution of research in these two areas and propose that intercrossing them and increasing funding to guide research of combination of agents represent a path forward for the development of curative therapies for the majority of cancer patients.


Asunto(s)
Sistemas de Liberación de Medicamentos , Inmunoterapia , Neoplasias/terapia , Animales , Humanos , Mutación , Neoplasias/genética , Neoplasias/inmunología , Medicina de Precisión , Transducción de Señal/efectos de los fármacos , Linfocitos T/inmunología
18.
Nature ; 626(8001): 1125-1132, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38355796

RESUMEN

To conserve energy during starvation and stress, many organisms use hibernation factor proteins to inhibit protein synthesis and protect their ribosomes from damage1,2. In bacteria, two families of hibernation factors have been described, but the low conservation of these proteins and the huge diversity of species, habitats and environmental stressors have confounded their discovery3-6. Here, by combining cryogenic electron microscopy, genetics and biochemistry, we identify Balon, a new hibernation factor in the cold-adapted bacterium Psychrobacter urativorans. We show that Balon is a distant homologue of the archaeo-eukaryotic translation factor aeRF1 and is found in 20% of representative bacteria. During cold shock or stationary phase, Balon occupies the ribosomal A site in both vacant and actively translating ribosomes in complex with EF-Tu, highlighting an unexpected role for EF-Tu in the cellular stress response. Unlike typical A-site substrates, Balon binds to ribosomes in an mRNA-independent manner, initiating a new mode of ribosome hibernation that can commence while ribosomes are still engaged in protein synthesis. Our work suggests that Balon-EF-Tu-regulated ribosome hibernation is a ubiquitous bacterial stress-response mechanism, and we demonstrate that putative Balon homologues in Mycobacteria bind to ribosomes in a similar fashion. This finding calls for a revision of the current model of ribosome hibernation inferred from common model organisms and holds numerous implications for how we understand and study ribosome hibernation.


Asunto(s)
Proteínas Bacterianas , Respuesta al Choque por Frío , Factores de Terminación de Péptidos , Biosíntesis de Proteínas , Psychrobacter , Proteínas Ribosómicas , Ribosomas , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Factor Tu de Elongación Peptídica/química , Factor Tu de Elongación Peptídica/metabolismo , Factor Tu de Elongación Peptídica/ultraestructura , Proteínas Ribosómicas/química , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Proteínas Ribosómicas/ultraestructura , Ribosomas/química , Ribosomas/metabolismo , Ribosomas/ultraestructura , Psychrobacter/química , Psychrobacter/genética , Psychrobacter/metabolismo , Psychrobacter/ultraestructura , Microscopía por Crioelectrón , Factores de Terminación de Péptidos/química , Factores de Terminación de Péptidos/genética , Factores de Terminación de Péptidos/metabolismo , Factores de Terminación de Péptidos/ultraestructura
19.
Nature ; 626(8000): 859-863, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38326609

RESUMEN

Bacteria in the gastrointestinal tract produce amino acid bile acid amidates that can affect host-mediated metabolic processes1-6; however, the bacterial gene(s) responsible for their production remain unknown. Herein, we report that bile salt hydrolase (BSH) possesses dual functions in bile acid metabolism. Specifically, we identified a previously unknown role for BSH as an amine N-acyltransferase that conjugates amines to bile acids, thus forming bacterial bile acid amidates (BBAAs). To characterize this amine N-acyltransferase BSH activity, we used pharmacological inhibition of BSH, heterologous expression of bsh and mutants in Escherichia coli and bsh knockout and complementation in Bacteroides fragilis to demonstrate that BSH generates BBAAs. We further show in a human infant cohort that BBAA production is positively correlated with the colonization of bsh-expressing bacteria. Lastly, we report that in cell culture models, BBAAs activate host ligand-activated transcription factors including the pregnane X receptor and the aryl hydrocarbon receptor. These findings enhance our understanding of how gut bacteria, through the promiscuous actions of BSH, have a significant role in regulating the bile acid metabolic network.


Asunto(s)
Aciltransferasas , Amidohidrolasas , Aminas , Ácidos y Sales Biliares , Biocatálisis , Microbioma Gastrointestinal , Humanos , Aciltransferasas/metabolismo , Amidohidrolasas/metabolismo , Aminas/química , Aminas/metabolismo , Bacteroides fragilis/enzimología , Bacteroides fragilis/genética , Bacteroides fragilis/metabolismo , Ácidos y Sales Biliares/química , Ácidos y Sales Biliares/metabolismo , Estudios de Cohortes , Escherichia coli/enzimología , Escherichia coli/genética , Escherichia coli/metabolismo , Microbioma Gastrointestinal/fisiología , Ligandos , Receptor X de Pregnano/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Factores de Transcripción/metabolismo , Lactante , Técnicas de Cultivo de Célula
20.
Nature ; 627(8003): 347-357, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38374256

RESUMEN

Type 2 diabetes (T2D) is a heterogeneous disease that develops through diverse pathophysiological processes1,2 and molecular mechanisms that are often specific to cell type3,4. Here, to characterize the genetic contribution to these processes across ancestry groups, we aggregate genome-wide association study data from 2,535,601 individuals (39.7% not of European ancestry), including 428,452 cases of T2D. We identify 1,289 independent association signals at genome-wide significance (P < 5 × 10-8) that map to 611 loci, of which 145 loci are, to our knowledge, previously unreported. We define eight non-overlapping clusters of T2D signals that are characterized by distinct profiles of cardiometabolic trait associations. These clusters are differentially enriched for cell-type-specific regions of open chromatin, including pancreatic islets, adipocytes, endothelial cells and enteroendocrine cells. We build cluster-specific partitioned polygenic scores5 in a further 279,552 individuals of diverse ancestry, including 30,288 cases of T2D, and test their association with T2D-related vascular outcomes. Cluster-specific partitioned polygenic scores are associated with coronary artery disease, peripheral artery disease and end-stage diabetic nephropathy across ancestry groups, highlighting the importance of obesity-related processes in the development of vascular outcomes. Our findings show the value of integrating multi-ancestry genome-wide association study data with single-cell epigenomics to disentangle the aetiological heterogeneity that drives the development and progression of T2D. This might offer a route to optimize global access to genetically informed diabetes care.


Asunto(s)
Diabetes Mellitus Tipo 2 , Progresión de la Enfermedad , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Adipocitos/metabolismo , Cromatina/genética , Cromatina/metabolismo , Enfermedad de la Arteria Coronaria/complicaciones , Enfermedad de la Arteria Coronaria/genética , Diabetes Mellitus Tipo 2/clasificación , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/fisiopatología , Nefropatías Diabéticas/complicaciones , Nefropatías Diabéticas/genética , Células Endoteliales/metabolismo , Células Enteroendocrinas , Epigenómica , Predisposición Genética a la Enfermedad/genética , Islotes Pancreáticos/metabolismo , Herencia Multifactorial/genética , Enfermedad Arterial Periférica/complicaciones , Enfermedad Arterial Periférica/genética , Análisis de la Célula Individual
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA