Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Immunol ; 25(2): 226-239, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38191855

RESUMO

Sepsis is a systemic response to infection with life-threatening consequences. Our understanding of the molecular and cellular impact of sepsis across organs remains rudimentary. Here, we characterize the pathogenesis of sepsis by measuring dynamic changes in gene expression across organs. To pinpoint molecules controlling organ states in sepsis, we compare the effects of sepsis on organ gene expression to those of 6 singles and 15 pairs of recombinant cytokines. Strikingly, we find that the pairwise effects of tumor necrosis factor plus interleukin (IL)-18, interferon-gamma or IL-1ß suffice to mirror the impact of sepsis across tissues. Mechanistically, we map the cellular effects of sepsis and cytokines by computing changes in the abundance of 195 cell types across 9 organs, which we validate by whole-mouse spatial profiling. Our work decodes the cytokine cacophony in sepsis into a pairwise cytokine message capturing the gene, cell and tissue responses of the host to the disease.


Assuntos
Citocinas , Sepse , Camundongos , Animais , Interleucina-6/genética , Fator de Necrose Tumoral alfa/metabolismo , Interferon gama , Sepse/genética
2.
bioRxiv ; 2023 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-36778287

RESUMO

Sepsis is a systemic response to infection with life-threatening consequences. Our understanding of the impact of sepsis across organs of the body is rudimentary. Here, using mouse models of sepsis, we generate a dynamic, organism-wide map of the pathogenesis of the disease, revealing the spatiotemporal patterns of the effects of sepsis across tissues. These data revealed two interorgan mechanisms key in sepsis. First, we discover a simplifying principle in the systemic behavior of the cytokine network during sepsis, whereby a hierarchical cytokine circuit arising from the pairwise effects of TNF plus IL-18, IFN-γ, or IL-1ß explains half of all the cellular effects of sepsis on 195 cell types across 9 organs. Second, we find that the secreted phospholipase PLA2G5 mediates hemolysis in blood, contributing to organ failure during sepsis. These results provide fundamental insights to help build a unifying mechanistic framework for the pathophysiological effects of sepsis on the body.

3.
Nat Commun ; 12(1): 6715, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34795271

RESUMO

Progress in biological imaging is intrinsically linked to advances in labeling methods. The explosion in the development of high-resolution and super-resolution imaging calls for new approaches to label targets with small probes. These should allow to faithfully report the localization of the target within the imaging resolution - typically nowadays a few nanometers - and allow access to any epitope of the target, in the native cellular and tissue environment. We report here the development of a complete labeling and imaging pipeline using genetic code expansion and non-canonical amino acids in neurons that allows to fluorescently label masked epitopes in target transmembrane proteins in live neurons, both in dissociated culture and organotypic brain slices. This allows us to image the differential localization of two AMPA receptor (AMPAR) auxiliary subunits of the transmembrane AMPAR regulatory protein family in complex with their partner with a variety of methods including widefield, confocal, and dSTORM super-resolution microscopy.


Assuntos
Aminoácidos/metabolismo , Epitopos/metabolismo , Proteínas de Membrana/metabolismo , Neurônios/metabolismo , Coloração e Rotulagem/métodos , Animais , Células COS , Chlorocebus aethiops , Feminino , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Microscopia Confocal/métodos , Imagem Óptica/métodos , Ratos Sprague-Dawley , Receptores de AMPA/metabolismo
4.
Cell Syst ; 11(5): 495-508.e10, 2020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33113356

RESUMO

The immune system makes decisions in response to combinations of multiple microbial inputs. We do not understand the combinatorial logic governing how higher-order combinations of microbial signals shape immune responses. Here, using coculture experiments and statistical analyses, we discover a general property for the combinatorial sensing of microbial signals, whereby the effects of triplet combinations of microbial signals on immune responses can be predicted by combining the effects of single and pairs. Mechanistically, we find that singles and pairs dictate the information signaled by triplets in mouse and human DCs at the levels of transcription, chromatin, and protein secretion. We exploit this simplifying property to develop cell-based immunotherapies prepared with adjuvant combinations that trigger protective responses in mouse models of cancer. We conclude that the processing of multiple input signals by innate immune cells is governed by pairwise effects, which will inform the rationale combination of adjuvants to manipulate immunity.


Assuntos
Imunidade Inata/fisiologia , Imunidade/fisiologia , Receptores de Reconhecimento de Padrão/fisiologia , Adjuvantes Imunológicos/farmacologia , Animais , Feminino , Imunidade/imunologia , Imunidade Inata/imunologia , Imunoterapia/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Reconhecimento de Padrão/imunologia
5.
Nat Protoc ; 15(4): 1459-1483, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32076350

RESUMO

The immune system operates at the scale of the whole organism in mammals. We currently lack experimental approaches to systematically track and study organism-wide molecular processes in mice. Here we describe an integrated toolkit for measuring gene expression in whole tissues, 3-prime mRNA extension sequencing, that is applicable to most mouse organs and any mouse model of interest. Further, the methods of RNA-seq described in this protocol are broadly applicable to other sample types beyond whole organs, such as tissue samples or isolated cell populations. We report procedures to collect, store and lyse a dozen organ types using conditions compatible with the extraction of high-quality RNA. In addition, we detail protocols to perform high-throughput and low-cost RNA extraction and sequencing, as well as downstream data analysis. The protocol takes 5 d to process 384 mouse organs from collecting tissues to obtaining raw sequencing data, with additional time required for data analysis and mining. The protocol is accessible to individuals with basic skills in (i) mouse perfusion and dissection for sample collection and (ii) computation using Unix and R for data analysis. Overall, the methods presented here fill a gap in our toolbox for studying organism-wide processes in immunology and physiology.


Assuntos
Perfilação da Expressão Gênica/métodos , RNA-Seq/métodos , Análise Serial de Tecidos/métodos , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos/genética , RNA Mensageiro/análise , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
6.
Curr Opin Syst Biol ; 18: 19-26, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32490290

RESUMO

The immune system is a dynamic mesh of molecules, cells and tissues spanning the entire organism. Despite a wealth of knowledge about the components of the immune system, little is known about the general rules governing the organismal circuitry of immunity. Deciphering the immune system at the scale of the whole organism is crucial to understanding fundamental problems in immunobiology and physiology, and to manipulate immunity for maintaining health and preventing disease. Here I discuss the emerging principles of inter-organ communications during immune responses by focusing on three common themes that are the regulation of the (i) composition, (ii) condition and (iii) coordination of communicating organs by molecular and cellular factors. Based on these common principles, I emphasize fundamental gaps in our knowledge of organismal immune processes and the outlook to tackle immunity at the scale of the whole organism.

7.
Cell ; 171(2): 398-413.e21, 2017 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-28942919

RESUMO

A fundamental challenge in immunology is to decipher the principles governing immune responses at the whole-organism scale. Here, using a comparative infection model, we observe immune signal propagation within and between organs to obtain a dynamic map of immune processes at the organism level. We uncover two inter-organ mechanisms of protective immunity mediated by soluble and cellular factors. First, analyzing ligand-receptor connectivity across tissues reveals that type I IFNs trigger a whole-body antiviral state, protecting the host within hours after skin vaccination. Second, combining parabiosis, single-cell analyses, and gene knockouts, we uncover a multi-organ web of tissue-resident memory T cells that functionally adapt to their environment to stop viral spread across the organism. These results have implications for manipulating tissue-resident memory T cells through vaccination and open up new lines of inquiry for the analysis of immune responses at the organism level.


Assuntos
Memória Imunológica , Interferon Tipo I/imunologia , Vaccinia virus/fisiologia , Vacínia/imunologia , Vacínia/prevenção & controle , Vacinas Virais/imunologia , Administração Cutânea , Animais , Feminino , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Organismos Livres de Patógenos Específicos , Linfócitos T/imunologia , Vacinas Virais/administração & dosagem
8.
Cell Rep ; 19(13): 2853-2866, 2017 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-28658630

RESUMO

Building an integrated view of cellular responses to environmental cues remains a fundamental challenge due to the complexity of intracellular networks in mammalian cells. Here, we introduce an integrative biochemical and genetic framework to dissect signal transduction events using multiple data types and, in particular, to unify signaling and transcriptional networks. Using the Toll-like receptor (TLR) system as a model cellular response, we generate multifaceted datasets on physical, enzymatic, and functional interactions and integrate these data to reveal biochemical paths that connect TLR4 signaling to transcription. We define the roles of proximal TLR4 kinases, identify and functionally test two dozen candidate regulators, and demonstrate a role for Ap1ar (encoding the Gadkin protein) and its binding partner, Picalm, potentially linking vesicle transport with pro-inflammatory responses. Our study thus demonstrates how deciphering dynamic cellular responses by integrating datasets on various regulatory layers defines key components and higher-order logic underlying signaling-to-transcription pathways.


Assuntos
Células Dendríticas/metabolismo , Receptores Toll-Like/metabolismo , Humanos , Fosforilação , Transdução de Sinais
9.
Nat Immunol ; 17(12): 1436-1446, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27695002

RESUMO

Follicular regulatory T cells (TFR cells) inhibit follicular helper T cell (TFH cell)-mediated antibody production. The mechanisms by which TFR cells exert their key immunoregulatory functions are largely unknown. Here we found that TFR cells induced a distinct suppressive state in TFH cells and B cells, in which effector transcriptional signatures were maintained but key effector molecules and metabolic pathways were suppressed. The suppression of B cell antibody production and metabolism by TFR cells was durable and persisted even in the absence of TFR cells. This durable suppression was due in part to epigenetic changes. The cytokine IL-21 was able to overcome TFR cell-mediated suppression and inhibited TFR cells and stimulated B cells. By determining mechanisms of TFR cell-mediated suppression, we have identified methods for modulating the function of TFR cells and antibody production.


Assuntos
Subpopulações de Linfócitos B/imunologia , Centro Germinativo/imunologia , Tolerância Imunológica , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia , Animais , Formação de Anticorpos , Células Cultivadas , Epigênese Genética , Fatores de Transcrição Forkhead/metabolismo , Subunidade alfa de Receptor de Interleucina-21/genética , Interleucinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
10.
Cell ; 164(3): 564-78, 2016 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-26824662

RESUMO

Type 1 interferon (IFN) is a key mediator of organismal responses to pathogens, eliciting prototypical "interferon signature genes" that encode antiviral and inflammatory mediators. For a global view of IFN signatures and regulatory pathways, we performed gene expression and chromatin analyses of the IFN-induced response across a range of immunocyte lineages. These distinguished ISGs by cell-type specificity, kinetics, and sensitivity to tonic IFN and revealed underlying changes in chromatin configuration. We combined 1,398 human and mouse datasets to computationally infer ISG modules and their regulators, validated by genetic analysis in both species. Some ISGs are controlled by Stat1/2 and Irf9 and the ISRE DNA motif, but others appeared dependent on non-canonical factors. This regulatory framework helped to interpret JAK1 blockade pharmacology, different clusters being affected under tonic or IFN-stimulated conditions, and the IFN signatures previously associated with human diseases, revealing unrecognized subtleties in disease footprints, as affected by human ancestry.


Assuntos
Redes Reguladoras de Genes , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Animais , Linfócitos T CD4-Positivos/metabolismo , Conjuntos de Dados como Assunto , Humanos , Janus Quinases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Interferon alfa e beta/metabolismo
11.
Science ; 347(6226): 1259038, 2015 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-25745177

RESUMO

Protein expression is regulated by the production and degradation of messenger RNAs (mRNAs) and proteins, but their specific relationships remain unknown. We combine measurements of protein production and degradation and mRNA dynamics so as to build a quantitative genomic model of the differential regulation of gene expression in lipopolysaccharide-stimulated mouse dendritic cells. Changes in mRNA abundance play a dominant role in determining most dynamic fold changes in protein levels. Conversely, the preexisting proteome of proteins performing basic cellular functions is remodeled primarily through changes in protein production or degradation, accounting for more than half of the absolute change in protein molecules in the cell. Thus, the proteome is regulated by transcriptional induction for newly activated cellular functions and by protein life-cycle changes for remodeling of preexisting functions.


Assuntos
Células da Medula Óssea/imunologia , Células Dendríticas/imunologia , Interações Hospedeiro-Patógeno/imunologia , Simulação de Dinâmica Molecular , Biossíntese de Proteínas , Proteólise , Aminoácidos/química , Aminoácidos/metabolismo , Animais , Técnicas de Cultura de Células , Marcação por Isótopo/métodos , Lipopolissacarídeos/imunologia , Camundongos , Proteínas Mitocondriais/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Análise de Sequência de RNA
12.
Cell Rep ; 5(2): 323-30, 2013 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-24120862

RESUMO

Bisphosphonates are a class of drugs that are widely used to inhibit loss of bone mass in patients. We show here that the administration of clinically relevant doses of bisphosphonates in mice increases antibody responses to live and inactive viruses, proteins, haptens, and existing commercial vaccine formulations. Bisphosphonates exert this adjuvant-like activity in the absence of CD4(+) and γδ T cells, neutrophils, or dendritic cells, and their effect does not rely on local macrophage depletion, Toll-like receptor signaling, or the inflammasome. Rather, bisphosphonates target directly B cells and enhance B cell expansion and antibody production upon antigen encounter. These data establish bisphosphonates as an additional class of adjuvants that boost humoral immune responses.


Assuntos
Linfócitos B/metabolismo , Difosfonatos/farmacologia , Imunidade Humoral/efeitos dos fármacos , Adjuvantes Imunológicos , Animais , Formação de Anticorpos/efeitos dos fármacos , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Humanos , Imunoglobulina G/metabolismo , Inflamassomos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transdução de Sinais , Receptores Toll-Like/metabolismo , Vesiculovirus/imunologia
13.
Nat Biotechnol ; 31(4): 342-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23503680

RESUMO

Individual genetic variation affects gene responsiveness to stimuli, often by influencing complex molecular circuits. Here we combine genomic and intermediate-scale transcriptional profiling with computational methods to identify variants that affect the responsiveness of genes to stimuli (responsiveness quantitative trait loci or reQTLs) and to position these variants in molecular circuit diagrams. We apply this approach to study variation in transcriptional responsiveness to pathogen components in dendritic cells from recombinant inbred mouse strains. We identify reQTLs that correlate with particular stimuli and position them in known pathways. For example, in response to a virus-like stimulus, a trans-acting variant responds as an activator of the antiviral response; using RNA interference, we identify Rgs16 as the likely causal gene. Our approach charts an experimental and analytic path to decipher the mechanisms underlying genetic variation in circuits that control responses to stimuli.


Assuntos
Células Dendríticas/virologia , Redes Reguladoras de Genes , Variação Genética , Locos de Características Quantitativas/genética , Transcrição Gênica , Animais , Cromossomos de Mamíferos/genética , Células Dendríticas/metabolismo , Feminino , Regulação da Expressão Gênica , Pleiotropia Genética , Camundongos , Camundongos Endogâmicos , Proteínas RGS/genética , Receptores Toll-Like/metabolismo
14.
J Infect Dis ; 207(7): 1075-83, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23303808

RESUMO

Brucellae replicate in a vacuole derived from the endoplasmic reticulum (ER) in epithelial cells, macrophages, and dendritic cells. In animals, trophoblasts are also key cellular targets where brucellae efficiently replicate in association with the ER. Therefore, we investigated the ability of Brucella spp. to infect human trophoblasts using both immortalized and primary trophoblasts. Brucella extensively proliferated within different subpopulations of trophoblasts, suggesting that they constitute an important niche in cases where the fetal-maternal barrier is breached. In extravillous trophoblasts (EVTs), B. abortus and B. suis replicated within single-membrane acidic lysosomal membrane-associated protein 1-positive inclusions, whereas B. melitensis replicated in the ER-derived compartment. Furthermore, B. melitensis but not B. abortus nor B. suis interfered with the invasive capacity of EVT-like cells in vitro. Because EVTs are essential for implantation during early stages of pregnancy, the nature of the replication niche may have a central role during Brucella-associated abortion in infected women.


Assuntos
Brucella abortus/crescimento & desenvolvimento , Brucella suis/crescimento & desenvolvimento , Trofoblastos/microbiologia , Autofagia , Carga Bacteriana , Brucella abortus/metabolismo , Brucella abortus/patogenicidade , Brucella melitensis/crescimento & desenvolvimento , Brucella melitensis/metabolismo , Brucella melitensis/patogenicidade , Brucella suis/metabolismo , Brucella suis/patogenicidade , Brucelose/microbiologia , Brucelose/patologia , Calnexina/metabolismo , Células Cultivadas , Feminino , Humanos , Proteínas de Membrana Lisossomal/metabolismo , Viabilidade Microbiana , Microscopia de Fluorescência , Placenta/metabolismo , Placenta/microbiologia , Placenta/patologia , Gravidez , Tetraspanina 30/metabolismo , Trofoblastos/metabolismo , Trofoblastos/patologia
15.
Nano Lett ; 12(12): 6498-504, 2012 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-23190424

RESUMO

A circuit level understanding of immune cells and hematological cancers has been severely impeded by a lack of techniques that enable intracellular perturbation without significantly altering cell viability and function. Here, we demonstrate that vertical silicon nanowires (NWs) enable gene-specific manipulation of diverse murine and human immune cells with negligible toxicity. To illustrate the power of the technique, we then apply NW-mediated gene silencing to investigate the role of the Wnt signaling pathway in chronic lymphocytic leukemia (CLL). Remarkably, CLL-B cells from different patients exhibit tremendous heterogeneity in their response to the knockdown of a single gene, LEF1. This functional heterogeneity defines three distinct patient groups not discernible by conventional CLL cytogenetic markers and provides a prognostic indicator for patients' time to first therapy. Analyses of gene expression signatures associated with these functional patient subgroups reveal unique insights into the underlying molecular basis for disease heterogeneity. Overall, our findings suggest a functional classification that can potentially guide the selection of patient-specific therapies in CLL and highlight the opportunities for nanotechnology to drive biological inquiry.


Assuntos
Leucemia Linfocítica Crônica de Células B/genética , Nanofios/química , RNA Interferente Pequeno/administração & dosagem , Silício/química , Animais , Linfócitos B/metabolismo , Células Cultivadas , Humanos , Fator 1 de Ligação ao Facilitador Linfoide/genética , Camundongos , Nanofios/toxicidade , Interferência de RNA , RNA Interferente Pequeno/genética , Silício/toxicidade
16.
Mol Cell ; 47(5): 810-22, 2012 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-22940246

RESUMO

Understanding the principles governing mammalian gene regulation has been hampered by the difficulty in measuring in vivo binding dynamics of large numbers of transcription factors (TF) to DNA. Here, we develop a high-throughput Chromatin ImmunoPrecipitation (HT-ChIP) method to systematically map protein-DNA interactions. HT-ChIP was applied to define the dynamics of DNA binding by 25 TFs and 4 chromatin marks at 4 time-points following pathogen stimulus of dendritic cells. Analyzing over 180,000 TF-DNA interactions we find that TFs vary substantially in their temporal binding landscapes. This data suggests a model for transcription regulation whereby TF networks are hierarchically organized into cell differentiation factors, factors that bind targets prior to stimulus to prime them for induction, and factors that regulate specific gene programs. Overlaying HT-ChIP data on gene-expression dynamics shows that many TF-DNA interactions are established prior to the stimuli, predominantly at immediate-early genes, and identified specific TF ensembles that coordinately regulate gene-induction.


Assuntos
Imunoprecipitação da Cromatina/métodos , Células Dendríticas/metabolismo , Regulação da Expressão Gênica , Ensaios de Triagem em Larga Escala , Animais , DNA/genética , DNA/metabolismo , Camundongos , Fatores de Transcrição/metabolismo
17.
Immunity ; 36(3): 415-26, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22386268

RESUMO

Neutralizing antibodies have been thought to be required for protection against acutely cytopathic viruses, such as the neurotropic vesicular stomatitis virus (VSV). Utilizing mice that possess B cells but lack antibodies, we show here that survival upon subcutaneous (s.c.) VSV challenge was independent of neutralizing antibody production or cell-mediated adaptive immunity. However, B cells were absolutely required to provide lymphotoxin (LT) α1ß2, which maintained a protective subcapsular sinus (SCS) macrophage phenotype within virus draining lymph nodes (LNs). Macrophages within the SCS of B cell-deficient LNs, or of mice that lack LTα1ß2 selectively in B cells, displayed an aberrant phenotype, failed to replicate VSV, and therefore did not produce type I interferons, which were required to prevent fatal VSV invasion of intranodal nerves. Thus, although B cells are essential for survival during VSV infection, their contribution involves the provision of innate differentiation and maintenance signals to macrophages, rather than adaptive immune mechanisms.


Assuntos
Linfócitos B/imunologia , Macrófagos/imunologia , Estomatite Vesicular/imunologia , Imunidade Adaptativa , Animais , Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/metabolismo , Imunidade Inata , Interferon Tipo I/biossíntese , Linfonodos/imunologia , Heterotrímero de Linfotoxina alfa1 e beta2/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/imunologia , Vesiculovirus/imunologia , Vesiculovirus/patogenicidade
18.
Cell ; 147(4): 853-67, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-22078882

RESUMO

Deciphering the signaling networks that underlie normal and disease processes remains a major challenge. Here, we report the discovery of signaling components involved in the Toll-like receptor (TLR) response of immune dendritic cells (DCs), including a previously unkown pathway shared across mammalian antiviral responses. By combining transcriptional profiling, genetic and small-molecule perturbations, and phosphoproteomics, we uncover 35 signaling regulators, including 16 known regulators, involved in TLR signaling. In particular, we find that Polo-like kinases (Plk) 2 and 4 are essential components of antiviral pathways in vitro and in vivo and activate a signaling branch involving a dozen proteins, among which is Tnfaip2, a gene associated with autoimmune diseases but whose role was unknown. Our study illustrates the power of combining systematic measurements and perturbations to elucidate complex signaling circuits and discover potential therapeutic targets.


Assuntos
Células Dendríticas/imunologia , Transdução de Sinais , Receptores Toll-Like/metabolismo , Vírus , Animais , Células Dendríticas/metabolismo , Feminino , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferons/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo
19.
Sci Signal ; 4(167): ra21, 2011 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-21467299

RESUMO

Natural killer (NK) cell tolerance to self is partly ensured by major histocompatibility complex (MHC) class I-specific inhibitory receptors on NK cells, which dampen their reactivity when engaged. However, NK cells that do not detect self MHC class I are not autoreactive. We used dynamic fluorescence correlation spectroscopy to show that MHC class I-independent NK cell tolerance in mice was associated with the presence of hyporesponsive NK cells in which both activating and inhibitory receptors were confined in an actin meshwork at the plasma membrane. In contrast, the recognition of self MHC class I by inhibitory receptors "educated" NK cells to become fully reactive, and activating NK cell receptors became dynamically compartmentalized in membrane nanodomains. We propose that the confinement of activating receptors at the plasma membrane is pivotal to ensuring the self-tolerance of NK cells.


Assuntos
Membrana Celular/metabolismo , Células Matadoras Naturais/imunologia , Tolerância a Antígenos Próprios/imunologia , Transdução de Sinais/imunologia , Actinas/metabolismo , Animais , Cálcio/metabolismo , Células Cultivadas , Primers do DNA/genética , Citometria de Fluxo , Interferon gama/metabolismo , Células Matadoras Naturais/metabolismo , Camundongos , Espectrometria de Fluorescência
20.
Science ; 326(5950): 257-63, 2009 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-19729616

RESUMO

Models of mammalian regulatory networks controlling gene expression have been inferred from genomic data but have largely not been validated. We present an unbiased strategy to systematically perturb candidate regulators and monitor cellular transcriptional responses. We applied this approach to derive regulatory networks that control the transcriptional response of mouse primary dendritic cells to pathogens. Our approach revealed the regulatory functions of 125 transcription factors, chromatin modifiers, and RNA binding proteins, which enabled the construction of a network model consisting of 24 core regulators and 76 fine-tuners that help to explain how pathogen-sensing pathways achieve specificity. This study establishes a broadly applicable, comprehensive, and unbiased approach to reveal the wiring and functions of a regulatory network controlling a major transcriptional response in primary mammalian cells.


Assuntos
Bactérias/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Inflamação/metabolismo , Vírus/imunologia , Animais , Montagem e Desmontagem da Cromatina , DNA de Cadeia Simples/imunologia , Retroalimentação Fisiológica , Perfilação da Expressão Gênica , Inflamação/imunologia , Lipopeptídeos/imunologia , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Poli I-C/imunologia , Proteínas de Ligação a RNA/metabolismo , Receptores Toll-Like/agonistas , Fatores de Transcrição/metabolismo , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...