Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Nat Cell Biol ; 21(3): 359-371, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30804505

RESUMO

Loss of apical-basal polarity and activation of epithelial-mesenchymal transition (EMT) both contribute to carcinoma progression and metastasis. Here, we report that apical-basal polarity inhibits EMT to suppress metastatic dissemination. Using mouse and human epithelial three-dimensional organoid cultures, we show that the PAR-atypical protein kinase C (aPKC) polarity complex inhibits EMT and invasion by promoting degradation of the SNAIL family protein SNAI1. Under intact apical-basal polarity, aPKC kinases phosphorylate S249 of SNAI1, which leads to protein degradation. Loss of apical-basal polarity prevents aPKC-mediated SNAI1 phosphorylation and stabilizes the SNAI1 protein to promote EMT and invasion. In human breast tumour xenografts, inhibition of the PAR-complex-mediated SNAI1 degradation mechanism promotes tumour invasion and metastasis. Analyses of human breast tissue samples reveal negative correlations between PAR3 and SNAI1 protein levels. Our results demonstrate that apical-basal polarity functions as a critical checkpoint of EMT to precisely control epithelial-mesenchymal plasticity during tumour metastasis.


Assuntos
Polaridade Celular , Transição Epitelial-Mesenquimal , Complexos Multiproteicos/metabolismo , Neoplasias/metabolismo , Fatores de Transcrição da Família Snail/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Células CACO-2 , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Camundongos Transgênicos , Metástase Neoplásica , Neoplasias/genética , Neoplasias/patologia , Proteína Quinase C/metabolismo , Proteólise , Interferência de RNA , Fatores de Transcrição da Família Snail/genética , Transplante Heterólogo
3.
Nat Cell Biol ; 17(5): 678-88, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25893917

RESUMO

Matrix stiffness potently regulates cellular behaviour in various biological contexts. In breast tumours, the presence of dense clusters of collagen fibrils indicates increased matrix stiffness and correlates with poor survival. It is unclear how mechanical inputs are transduced into transcriptional outputs to drive tumour progression. Here we report that TWIST1 is an essential mechanomediator that promotes epithelial-mesenchymal transition (EMT) in response to increasing matrix stiffness. High matrix stiffness promotes nuclear translocation of TWIST1 by releasing TWIST1 from its cytoplasmic binding partner G3BP2. Loss of G3BP2 leads to constitutive TWIST1 nuclear localization and synergizes with increasing matrix stiffness to induce EMT and promote tumour invasion and metastasis. In human breast tumours, collagen fibre alignment, a marker of increasing matrix stiffness, and reduced expression of G3BP2 together predict poor survival. Our findings reveal a TWIST1-G3BP2 mechanotransduction pathway that responds to biomechanical signals from the tumour microenvironment to drive EMT, invasion and metastasis.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma Intraductal não Infiltrante/metabolismo , Carcinoma Intraductal não Infiltrante/secundário , Proteínas de Transporte/metabolismo , Junções Célula-Matriz/metabolismo , Transição Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Mecanotransdução Celular , Proteínas Nucleares/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Transporte Ativo do Núcleo Celular , Proteínas Adaptadoras de Transdução de Sinal , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/mortalidade , Carcinoma Intraductal não Infiltrante/genética , Carcinoma Intraductal não Infiltrante/mortalidade , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Colágeno/metabolismo , Bases de Dados Genéticas , Intervalo Livre de Doença , Elasticidade , Feminino , Humanos , Estimativa de Kaplan-Meier , Camundongos SCID , Invasividade Neoplásica , Estadiamento de Neoplasias , Proteínas Nucleares/genética , Interferência de RNA , Proteínas de Ligação a RNA , Estudos Retrospectivos , Fatores de Tempo , Transfecção , Microambiente Tumoral , Proteína 1 Relacionada a Twist/genética
4.
Genes Dev ; 27(20): 2192-206, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24142872

RESUMO

Tumor metastasis is a multistep process by which tumor cells disseminate from their primary site and form secondary tumors at a distant site. Metastasis occurs through a series of steps: local invasion, intravasation, transport, extravasation, and colonization. A developmental program termed epithelial-mesenchymal transition (EMT) has been shown to play a critical role in promoting metastasis in epithelium-derived carcinoma. Recent experimental and clinical studies have improved our knowledge of this dynamic program and implicated EMT and its reverse program, mesenchymal-epithelial transition (MET), in the metastatic process. Here, we review the functional requirement of EMT and/or MET during the individual steps of tumor metastasis and discuss the potential of targeting this program when treating metastatic diseases.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Metástase Neoplásica/fisiopatologia , Neoplasias/fisiopatologia , Humanos , Neoplasias/terapia
5.
Cancer Cell ; 22(6): 725-36, 2012 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-23201165

RESUMO

Epithelial-mesenchymal transition (EMT) is implicated in converting stationary epithelial tumor cells into motile mesenchymal cells during metastasis. However, the involvement of EMT in metastasis is still controversial, due to the lack of a mesenchymal phenotype in human carcinoma metastases. Using a spontaneous squamous cell carcinoma mouse model, we show that activation of the EMT-inducing transcription factor Twist1 is sufficient to promote carcinoma cells to undergo EMT and disseminate into blood circulation. Importantly, in distant sites, turning off Twist1 to allow reversion of EMT is essential for disseminated tumor cells to proliferate and form metastases. Our study demonstrates in vivo the requirement of "reversible EMT" in tumor metastasis and may resolve the controversy on the importance of EMT in carcinoma metastasis.


Assuntos
Neoplasias da Mama/patologia , Carcinoma de Células Escamosas/patologia , Transição Epitelial-Mesenquimal/fisiologia , Neoplasias Pulmonares/secundário , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/secundário , Proliferação de Células , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal/genética , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína 1 Relacionada a Twist/genética , Proteína 1 Relacionada a Twist/metabolismo
6.
PLoS One ; 6(7): e22499, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21799874

RESUMO

In the adult organism, cell migration is required for physiological processes such as angiogenesis and immune surveillance, as well as pathological events such as tumor metastasis. The adaptor protein and Src substrate Tks5 is necessary for cancer cell migration through extracellular matrix in vitro and tumorigenicity in vivo. However, a role for Tks5 during embryonic development, where cell migration is essential, has not been examined. We used morpholinos to reduce Tks5 expression in zebrafish embryos, and observed developmental defects, most prominently in neural crest-derived tissues such as craniofacial structures and pigmentation. The Tks5 morphant phenotype was rescued by expression of mammalian Tks5, but not by a variant of Tks5 in which the Src phosphorylation sites have been mutated. We further evaluated the role of Tks5 in neural crest cells and neural crest-derived tissues and found that loss of Tks5 impaired their ventral migration. Inhibition of Src family kinases also led to abnormal ventral patterning of neural crest cells and their derivatives. We confirmed that these effects were likely to be cell autonomous by shRNA-mediated knockdown of Tks5 in a murine neural crest stem cell line. Tks5 was required for neural crest cell migration in vitro, and both Src and Tks5 were required for the formation of actin-rich structures with similarity to podosomes. Additionally, we observed that neural crest cells formed Src-Tks5-dependent cell protrusions in 3-D culture conditions and in vivo. These results reveal an important and novel role for the Src-Tks5 pathway in neural crest cell migration during embryonic development. Furthermore, our data suggests that this pathway regulates neural crest cell migration through the generation of actin-rich pro-migratory structures, implying that similar mechanisms are used to control cell migration during embryogenesis and cancer metastasis.


Assuntos
Movimento Celular , Crista Neural/citologia , Crista Neural/embriologia , Fosfoproteínas/metabolismo , Quinases da Família src/metabolismo , Actinas/metabolismo , Animais , Matriz Extracelular/metabolismo , Humanos , Camundongos , Crista Neural/metabolismo , Proteínas de Ligação a Fosfato , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo
7.
PLoS One ; 4(8): e6539, 2009 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-19657384

RESUMO

BACKGROUND: Epidermal growth factor receptor (EGFR) inhibitors have shown only modest clinical activity when used as single agents to treat cancers. They decrease tumor cell expression of hypoxia-inducible factor 1-alpha (HIF-1alpha) and vascular endothelial growth factor (VEGF). Hypothesizing that this might normalize tumor vasculature, we examined the effects of the EGFR inhibitor erlotinib on tumor vascular function, tumor microenvironment (TME) and chemotherapy and radiotherapy sensitivity. METHODOLOGY/PRINCIPAL FINDINGS: Erlotinib treatment of human tumor cells in vitro and mice bearing xenografts in vivo led to decreased HIF-1alpha and VEGF expression. Treatment altered xenograft vessel morphology assessed by confocal microscopy (following tomato lectin injection) and decreased vessel permeability (measured by Evan's blue extravasation), suggesting vascular normalization. Erlotinib increased tumor blood flow measured by Power Doppler ultrasound and decreased hypoxia measured by EF5 immunohistochemistry and tumor O(2) saturation measured by optical spectroscopy. Predicting that these changes would improve drug delivery and increase response to chemotherapy and radiation, we performed tumor regrowth studies in nude mice with xenografts treated with erlotinib and either radiotherapy or the chemotherapeutic agent cisplatin. Erlotinib therapy followed by cisplatin led to synergistic inhibition of tumor growth compared with either treatment by itself (p<0.001). Treatment with erlotinib before cisplatin led to greater tumor growth inhibition than did treatment with cisplatin before erlotinib (p = 0.006). Erlotinib followed by radiation inhibited tumor regrowth to a greater degree than did radiation alone, although the interaction between erlotinib and radiation was not synergistic. CONCLUSIONS/SIGNIFICANCE: EGFR inhibitors have shown clinical benefit when used in combination with conventional cytotoxic therapy. Our studies show that targeting tumor cells with EGFR inhibitors may modulate the TME via vascular normalization to increase response to chemotherapy and radiotherapy. These studies suggest ways to assess the response of tumors to EGFR inhibition using non-invasive imaging of the TME.


Assuntos
Receptores ErbB/antagonistas & inibidores , Neoplasias Experimentais/irrigação sanguínea , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Cloridrato de Erlotinib , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Camundongos Nus , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/radioterapia , Oxigênio/metabolismo , Quinazolinas/farmacologia , Quinazolinas/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Mol Cancer Res ; 7(3): 300-10, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19276184

RESUMO

Signaling through the Tie2 receptor on endothelial cells has been shown to play an important role in normal and pathologic vascular development. We generated K1735 murine melanoma tumor cells that inducibly express soluble Tie2 receptor (Tie2Ex) to study the effects of inhibiting Tie2 signaling on tumor vasculature. Tie2Ex induction rapidly decreased AKT activation but not extracellular signal-regulated kinase (ERK) activation in tumor endothelial cells as detected by immunostaining. This was accompanied by an increase in endothelial cell TUNEL staining but no change in Ki-67 expression. Together with a decrease in the percentage of perfused vessels, this suggested that tumor vessel regression and impaired vascular function rather than angiogenesis inhibition was responsible for the delay in tumor growth following Tie2Ex treatment. However, Tie2Ex failed to inhibit the growth of larger, more established K1735 tumors. These tumors were additionally treated with sorafenib, a multikinase inhibitor that inhibits tumor endothelial cell ERK activation but not AKT activation. Combining Tie2Ex and sorafenib decreased both endothelial cell AKT and ERK activation, decreased endothelial cell survival and proliferation, and significantly inhibited growth of the more established tumors. These studies indicate that activity of specific signaling pathways and prosurvival effects are brought about by Tie2 activation in tumor endothelial cells, and knowledge of the effects of Tie2 inhibition can lead to development of more effective therapeutic regimens for inhibiting tumor neovascularization.


Assuntos
Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/terapia , Receptor TIE-2/metabolismo , Angiopoietina-1/farmacologia , Animais , Apoptose/fisiologia , Benzenossulfonatos/farmacologia , Doxiciclina/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Técnicas In Vitro , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C3H , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neovascularização Patológica/terapia , Niacinamida/análogos & derivados , Compostos de Fenilureia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/farmacologia , Receptor TIE-2/antagonistas & inibidores , Receptor TIE-2/genética , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sorafenibe , Transfecção
9.
Acad Radiol ; 15(9): 1133-41, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18692754

RESUMO

RATIONALE AND OBJECTIVES: The goal was to determine whether the tumor vascular disrupting actions of low-intensity ultrasound were frequency dependent. MATERIALS AND METHODS: The effect of the frequency (1 MHz at 2.2 W/cm2 or 3 MHz at 2.4 W/cm2) of low-intensity ultrasound as a neovascular disrupting modality was investigated in 15 murine melanomas (K1735(22)) insonated for 3 minutes after the intravenous injection of a microbubble contrast agent (Definity). In contrast-enhanced power Doppler observations of each tumor (before and after treatment), measurements were made of the size of the area of the tumor that was perfused with blood containing the ultrasound contrast agent (percentage area of flow [PAF]), and the volume of contrast agent flowing through the unit volume of the tumor (color-weighted fractional area [CWFA]). During insonation of the tumor, the temperature was measured with a fine wire thermocouple in an additional eight mice. RESULTS: The antivascular action of low-intensity ultrasound was significantly enhanced (PAF by 64%; CWFA by 106%) when the tumor was treated with 3-MHz ultrasound rather than 1 MHz (analysis of variance: PAF, P=.02; CWFA, P=.04). The average rate of tumor temperature increase was 2.6+/-1.3 degrees C/min for 1 MHz and 5.0+/-1.7 degrees C/min for 3 MHz; these increases were significantly different (P=.04). CONCLUSIONS: Insonation of the tumor at a higher frequency amplified the heating of the neoplasm and led to greater disruption of the tumor vasculature; 3-MHz ultrasound was more efficacious than 1 MHz for antivascular cancer therapy.


Assuntos
Melanoma Experimental/terapia , Neovascularização Patológica/terapia , Terapia por Ultrassom/métodos , Animais , Temperatura Corporal , Meios de Contraste , Feminino , Fluorocarbonos , Camundongos , Camundongos Endogâmicos C3H
10.
Ultrasound Med Biol ; 33(12): 1901-10, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17720299

RESUMO

This study investigated whether a microbubble-containing ultrasound contrast agent had a role in the antivascular action of physiotherapy ultrasound on tumor neovasculature. Ultrasound images (B-mode and contrast-enhanced power Doppler [0.02 mL Definity]) were made of 22 murine melanomas (K1735(22)). The tumor was insonated (I(SATA) = 1.7 W cm(-2), 1 MHz, continuous output) for 3 min and the power Doppler observations of the pre- and postinsonation tumor vascularities were analyzed. Significant reductions (p = 0.005 for analyses of color-weighted fractional area) in vascularity occurred when a contrast-enhanced power Doppler study occurred before insonation. Vascularity was unchanged in tumors without a pretherapy Doppler study. Histologic studies revealed tissue structural changes that correlated with the ultrasound findings. The underlying etiology of the interaction between the physiotherapy ultrasound beam, the microbubble-containing contrast agent and the tumor neovasculature is unknown. It was concluded that contrast agents play an important role in the antivascular effects induced by physiotherapy ultrasound.


Assuntos
Fluorocarbonos/uso terapêutico , Melanoma Experimental/terapia , Neovascularização Patológica/terapia , Terapia por Ultrassom/métodos , Animais , Meios de Contraste/uso terapêutico , Feminino , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/diagnóstico por imagem , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C3H , Microbolhas , Transplante de Neoplasias , Neovascularização Patológica/diagnóstico por imagem , Neovascularização Patológica/patologia , Ultrassonografia Doppler/métodos
11.
Cancer Biol Ther ; 4(12): 1395-1400, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16676437

RESUMO

The vascular effects of ionizing radiation were examined in K1735 murine melanoma tumors. Single-fraction and fractionated radiation virtually arrested growth of these tumors for about a week, after which they resumed more rapid growth. Tumor microvessel density (MVD) and blood perfusion was unchanged seven days after radiation but decreased at later time points after irradiation, when they had grown 10-fold or more. Together with the finding of severe tumor hypoxia and VEGF induction in the latter tumors, the evidence pointed to vascular insufficiency and inhibited neovascularization in tumors that had grown substantially after radiation. Endothelial cell (EC) death detected by TUNEL staining only transiently increased the day following radiation, whereas EC proliferation detected by Ki-67 staining was increased in irradiated tumors that had grown substantially. The fact that increased EC proliferative activity produced fewer vessels suggests that angiogenesis is defective or ineffective after radiation. These results complement recent genetic evidence that EC damage from radiation plays a major role in tissue damage and antitumor efficacy to highlight the importance of EC and vasculature in radiation response. Our studies further show that radiation impact on tumor vasculature extends beyond near-term induction of EC death to more prolonged effects on their ability to support angiogenesis.


Assuntos
Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/radioterapia , Neovascularização Patológica/radioterapia , Radiação Ionizante , Animais , Hipóxia Celular/efeitos da radiação , Citoproteção/efeitos da radiação , Fracionamento da Dose de Radiação , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Microcirculação/efeitos da radiação , Transplante de Neoplasias , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Immunol Res ; 27(2-3): 219-34, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12857970

RESUMO

Anti-double-stranded DNA (dsDNA) B cells persist even in nonautoimmune- prone animals. In this review, we summarize data regarding the activation potential of these cells. Provision of cognate CD4 T cell help to anti-dsDNA B cells in nonautoimmune mice not only drives their maturation and entry into the B cell follicle, but also leads to secretion of anti-dsDNA autoantibodies. Intriguingly, if T regulatory cells are provided along with T helper cells, the antibody response of anti-dsDNA B cells is diminished. We have also found that T-independent stimulation with CpG oligodeoxynucleotides leads to the proliferation and enhanced recovery of antidsDNA B cells in vitro. These data suggest that control of anti-dsDNA antibody production may rely on elements from both the innate and adaptive arms of the immune system.


Assuntos
Anticorpos Antinucleares/imunologia , Autoimunidade , Linfócitos B/imunologia , Ativação Linfocitária/imunologia , Animais , DNA/imunologia , Cooperação Linfocítica/imunologia , Camundongos , Linfócitos T/imunologia
13.
J Endotoxin Res ; 9(6): 395-400, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14733728

RESUMO

Lipid A is the pro-inflammatory component of bacterial lipopolysaccharide, the major surface component of Gram-negative bacteria. Gram-negative bacteria alter the structure of lipid A in response to specific environmental conditions including those found upon colonization of a host. The opportunistic pathogen Pseudomonas aeruginosa synthesizes a unique hexa-acylated lipid A containing palmitate and aminoarabinose during adaptation to the cystic fibrosis airway. Different lipid A species are observed in P. aeruginosa isolated from non-cystic fibrosis associated infections. Here we report that P. aeruginosa isolates from the airway of a cystic fibrosis patient with severe pulmonary disease synthesized a novel hepta-acylated lipid A. Cystic fibrosis-specific P. aeruginosa lipid A modifications result in resistance to host antimicrobial peptides and increased recognition by human Toll-like receptor 4 (TLR4). Using P. aeruginosa lipid A with different levels of acylation, we identified a 222 amino acid region in the extracellular portion of human TLR4 that is required for the differential recognition of cystic fibrosis-specific lipid A. P. aeruginosa adaptation to the human airway may, therefore, play a fundamental role in the progressive lung damage associated with cystic fibrosis.


Assuntos
Variação Genética , Lipídeo A/imunologia , Lipopolissacarídeos/farmacologia , Glicoproteínas de Membrana/imunologia , Pseudomonas aeruginosa/química , Receptores de Superfície Celular/imunologia , Sequência de Aminoácidos , Linhagem Celular , Fibrose Cística/complicações , Fibrose Cística/microbiologia , Humanos , Rim/citologia , Lipídeo A/química , Lipídeo A/genética , Lipopolissacarídeos/química , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/isolamento & purificação , Espectrometria de Massas , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Peso Molecular , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/crescimento & desenvolvimento , Pseudomonas aeruginosa/isolamento & purificação , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Receptor 4 Toll-Like , Receptores Toll-Like , Transfecção
15.
Nat Immunol ; 3(4): 354-9, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11912497

RESUMO

Lipopolysaccharide (LPS) is the principal proinflammatory component of the Gram-negative bacterial envelope and is recognized by the Toll-like receptor 4 (TLR4)-MD-2 receptor complex. Bacteria can alter the acylation state of their LPS in response to environmental changes. One opportunistic bacterium, Pseudomonas aeruginosa, synthesizes more highly acylated (hexa-acylated) LPS structures during adaptation to the cystic fibrosis airway. Here we show that human, but not murine, TLR4-MD-2 recognizes this adaptation and transmits robust proinflammatory signals in response to hexa-acylated but not penta-acylated LPS from P. aeruginosa. Whereas responses to lipidIVA and taxol are dependent on murine MD-2, discrimination of P. aeruginosa LPS structures is mediated by an 82-amino-acid region of human TLR4 that is hypervariable across species. Thus, in contrast to mice, humans use TLR4 to recognize a molecular signature of bacterial-host adaptation to modulate the innate immune response.


Assuntos
Antígenos de Superfície/imunologia , Proteínas de Drosophila , Lipídeo A/imunologia , Glicoproteínas de Membrana/imunologia , Receptores de Superfície Celular/imunologia , Animais , Antígenos de Superfície/genética , Sítios de Ligação , Sequência de Carboidratos , Diferenciação Celular , Linhagem Celular , Humanos , Lactente , Lipídeo A/química , Antígeno 96 de Linfócito , Glicoproteínas de Membrana/genética , Camundongos , Dados de Sequência Molecular , Pseudomonas aeruginosa/imunologia , Receptores de Superfície Celular/genética , Receptor 4 Toll-Like , Receptores Toll-Like
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...