Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Nat Immunol ; 19(4): 354-365, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29563620

RESUMO

Mechanisms that degrade inflammatory mRNAs are well known; however, stabilizing mechanisms are poorly understood. Here, we show that Act1, an interleukin-17 (IL-17)-receptor-complex adaptor, binds and stabilizes mRNAs encoding key inflammatory proteins. The Act1 SEFIR domain binds a stem-loop structure, the SEFIR-binding element (SBE), in the 3' untranslated region (UTR) of Cxcl1 mRNA, encoding an inflammatory chemokine. mRNA-bound Act1 directs formation of three compartmentally distinct RNA-protein complexes (RNPs) that regulate three disparate events in inflammatory-mRNA metabolism: preventing mRNA decay in the nucleus, inhibiting mRNA decapping in P bodies and promoting translation. SBE RNA aptamers decreased IL-17-mediated mRNA stabilization in vitro, IL-17-induced skin inflammation and airway inflammation in a mouse asthma model, thus providing a therapeutic strategy for autoimmune diseases. These results reveal a network in which Act1 assembles RNPs on the 3' UTRs of select mRNAs and consequently controls receptor-mediated mRNA stabilization and translation during inflammation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Inflamação/imunologia , Interleucina-17/metabolismo , Estabilidade de RNA/fisiologia , Transdução de Sinais/imunologia , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Regulação da Expressão Gênica/imunologia , Inflamação/metabolismo , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo , Receptores de Interleucina-17/metabolismo
2.
Am J Transplant ; 22(3): 955-965, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34679256

RESUMO

The importance of PD-1/PD-L1 interaction to alloimmune response is unknown in intestinal transplantation. We tested whether PD-L1 regulates allograft tissue injury in murine intestinal transplantation. PD-L1 expression was observed on the endothelium and immune cells in the intestinal allograft. Monoclonal antibody treatment against PD-L1 led to accelerated allograft tissue damage, characterized by severe cellular infiltrations, massive destruction of villi, and increased crypt apoptosis in the graft. Interestingly, PD-L1-/- allografts were more severely rejected than wild-type allografts, but the presence or absence of PD-L1 in recipients did not affect the degree of allograft injury. PD-L1-/- allografts showed increased infiltrating Ly6G+ and CD11b+ cells in lamina propria on day 4, whereas the degree of CD4+ or CD8+ T cell infiltration was comparable to wild-type allografts. Gene expression analysis revealed that PD-L1-/- allografts had increased mRNA expressions of Cxcr2, S100a8/9, Nox1, IL1rL1, IL1r2, and Nos2 in the lamina propria cells on day 4. Taken together, study results suggest that PD-L1 expression in the intestinal allograft, but not in the recipient, plays a critical role in mitigating allograft tissue damage in the early phase after transplantation. The PD-1/PD-L1 interaction may contribute to immune regulation of the intestinal allograft via the innate immune system.


Assuntos
Antígeno B7-H1 , Receptor de Morte Celular Programada 1 , Aloenxertos/metabolismo , Animais , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Rejeição de Enxerto , Proteína 1 Semelhante a Receptor de Interleucina-1 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Morte Celular Programada 1/genética
3.
J Immunol ; 201(1): 157-166, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29760195

RESUMO

Human Ag R (HuR) is an RNA binding protein in the ELAVL protein family. To study the neuron-specific function of HuR, we generated inducible, neuron-specific HuR-deficient mice of both sexes. After tamoxifen-induced deletion of HuR, these mice developed a phenotype consisting of poor balance, decreased movement, and decreased strength. They performed significantly worse on the rotarod test compared with littermate control mice, indicating coordination deficiency. Using the grip-strength test, it was also determined that the forelimbs of neuron-specific HuR-deficient mice were much weaker than littermate control mice. Immunostaining of the brain and cervical spinal cord showed that HuR-deficient neurons had increased levels of cleaved caspase-3, a hallmark of cell apoptosis. Caspase-3 cleavage was especially strong in pyramidal neurons and α motor neurons of HuR-deficient mice. Genome-wide microarray and real-time PCR analysis further indicated that HuR deficiency in neurons resulted in altered expression of genes in the brain involved in cell growth, including trichoplein keratin filament-binding protein, Cdkn2c, G-protein signaling modulator 2, immediate early response 2, superoxide dismutase 1, and Bcl2. The additional enriched Gene Ontology terms in the brain tissues of neuron-specific HuR-deficient mice were largely related to inflammation, including IFN-induced genes and complement components. Importantly, some of these HuR-regulated genes were also significantly altered in the brain and spinal cord of patients with amyotrophic lateral sclerosis. Additionally, neuronal HuR deficiency resulted in the redistribution of TDP43 to cytosolic granules, which has been linked to motor neuron disease. Taken together, we propose that this neuron-specific HuR-deficient mouse strain can potentially be used as a motor neuron disease model.


Assuntos
Caspase 3/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteína Semelhante a ELAV 1/genética , Doença dos Neurônios Motores/genética , Doença dos Neurônios Motores/patologia , Neurônios Motores/patologia , Esclerose Lateral Amiotrófica/genética , Animais , Ataxia/genética , Células Cultivadas , Modelos Animais de Doenças , Feminino , Força da Mão/fisiologia , Humanos , Masculino , Camundongos , Camundongos Knockout
4.
J Immunol ; 199(11): 3849-3857, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29070673

RESUMO

This study identifies a novel mechanism linking IL-17A with colon tissue repair and tumor development. Abrogation of IL-17A signaling in mice attenuated tissue repair of dextran sulfate sodium (DSS)-induced damage in colon epithelium and markedly reduced tumor development in an azoxymethane/DSS model of colitis-associated cancer. A novel IL-17A target gene, PLET1 (a progenitor cell marker involved in wound healing), was highly induced in DSS-treated colon tissues and tumors in an IL-17RC-dependent manner. PLET1 expression was induced in LGR5+ colon epithelial cells after DSS treatment. LGR5+PLET1+ marks a highly proliferative cell population with enhanced expression of IL-17A target genes. PLET1 deficiency impaired tissue repair of DSS-induced damage in colon epithelium and reduced tumor formation in an azoxymethane/DSS model of colitis-associated cancer. Our results suggest that IL-17A-induced PLET1 expression contributes to tissue repair and colon tumorigenesis.


Assuntos
Colite/imunologia , Colo/metabolismo , Neoplasias do Colo/imunologia , Células Epiteliais/imunologia , Interleucina-17/metabolismo , Proteínas da Gravidez/metabolismo , Animais , Azoximetano , Carcinogênese , Células Cultivadas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Colite/induzido quimicamente , Colo/patologia , Neoplasias do Colo/induzido quimicamente , Sulfato de Dextrana , Regulação Neoplásica da Expressão Gênica , Interleucina-17/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas da Gravidez/genética , Receptores de Interleucina/genética , Cicatrização
5.
Proc Natl Acad Sci U S A ; 113(52): 14988-14993, 2016 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-27956611

RESUMO

The CRISPR/Cas9 nuclease is commonly used to make gene knockouts. The blunt DNA ends generated by cleavage can be efficiently ligated by the classical nonhomologous end-joining repair pathway (c-NHEJ), regenerating the target site. This repair creates a cycle of cleavage, ligation, and target site regeneration that persists until sufficient modification of the DNA break by alternative NHEJ prevents further Cas9 cutting, generating a heterogeneous population of insertions and deletions typical of gene knockouts. Here, we develop a strategy to escape this cycle and bias events toward defined length deletions by creating an RNA-guided dual active site nuclease that generates two noncompatible DNA breaks at a target site, effectively deleting the majority of the target site such that it cannot be regenerated. The TevCas9 nuclease, a fusion of the I-TevI nuclease domain to Cas9, functions robustly in HEK293 cells and generates 33- to 36-bp deletions at frequencies up to 40%. Deep sequencing revealed minimal processing of TevCas9 products, consistent with protection of the DNA ends from exonucleolytic degradation and repair by the c-NHEJ pathway. Directed evolution experiments identified I-TevI variants with broadened targeting range, making TevCas9 an easy-to-use reagent. Our results highlight how the sequence-tolerant cleavage properties of the I-TevI homing endonuclease can be harnessed to enhance Cas9 applications, circumventing the cleavage and ligation cycle and biasing genome-editing events toward defined length deletions.


Assuntos
Sistemas CRISPR-Cas , Endodesoxirribonucleases/genética , Deleção de Genes , Edição de Genes , Quebras de DNA de Cadeia Dupla , Reparo de Erro de Pareamento de DNA , Endopeptidase K/química , Escherichia coli , Genoma , Células HEK293 , Humanos , RNA Guia de Cinetoplastídeos/genética , Análise de Sequência de DNA , Deleção de Sequência
6.
J Immunol ; 196(1): 428-36, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26597012

RESUMO

Macrophage phagocytosis of particles and pathogens is an essential aspect of innate host defense. Phagocytic function requires cytoskeletal rearrangements that depend on the interaction between macrophage surface receptors, particulates/pathogens, and the extracellular matrix. In the present study we determine the role of a mechanosensitive ion channel, transient receptor potential vanilloid 4 (TRPV4), in integrating the LPS and matrix stiffness signals to control macrophage phenotypic change for host defense and resolution from lung injury. We demonstrate that active TRPV4 mediates LPS-stimulated murine macrophage phagocytosis of nonopsonized particles (Escherichia coli) in vitro and opsonized particles (IgG-coated latex beads) in vitro and in vivo in intact mice. Intriguingly, matrix stiffness in the range seen in inflamed or fibrotic lung is required to sensitize the TRPV4 channel to mediate the LPS-induced increment in macrophage phagocytosis. Furthermore, TRPV4 is required for the LPS induction of anti-inflammatory/proresolution cytokines. These findings suggest that signaling through TRPV4, triggered by changes in extracellular matrix stiffness, cooperates with LPS-induced signals to mediate macrophage phagocytic function and lung injury resolution. These mechanisms are likely to be important in regulating macrophage function in the context of pulmonary infection and fibrosis.


Assuntos
Lipopolissacarídeos/imunologia , Lesão Pulmonar/imunologia , Macrófagos/imunologia , Fagocitose/imunologia , Canais de Cátion TRPV/imunologia , Animais , Células Cultivadas , Citocinas/biossíntese , Citocinas/imunologia , Escherichia coli/imunologia , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Matriz Extracelular/metabolismo , Imunoglobulina G/imunologia , Lesão Pulmonar/patologia , Fenômenos Mecânicos , Camundongos , Camundongos Endogâmicos C57BL , Microesferas , Fibrose Pulmonar/imunologia , Transdução de Sinais/imunologia
7.
J Immunol ; 193(2): 879-88, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24920846

RESUMO

The impact of environmental stressors on the magnitude of specific chemokine gene expression was examined in mouse bone marrow-derived macrophages stimulated through various TLRs. Levels of TLR-stimulated CXCL1 and CXCL2 but not CXCL10 or CCL5 mRNAs were selectively enhanced (>10-fold) in stressed macrophages. The amplification was also manifested for other proinflammatory cytokines, including TNF-α, IL-1α, and IL-6. Responses through TLR3 and TLR4 exhibited the greatest sensitivity, reflecting a requirement for Toll/IL-IR domain-containing adaptor-inducing IFN-ß (TRIF), the adaptor protein selectively associated with these TLRs. IFN regulatory factor 3, a transcription factor that is downstream of TLR4/TRIF signaling, was not required for sensitivity to stress-induced chemokine amplification. c/EBP homologous protein and X box binding protein 1 have been reported to enhance inflammatory cytokine responses but are not required for amplification of TLR3/4-induced CXCL1 expression. Rather, receptor-interacting protein kinase 1, a kinase also linked with TLR3/4/TRIF signaling, is required and involves a stress-dependent increase in its abundance and ubiquitination. Whereas NF-κB activation is necessary for TLR-induced chemokine gene transcription, this factor does not appear to be the primary mechanistic target of environmental stress. The application of stress also enhanced chemokine expression in macrophages infiltrating the peritoneal cavity but was not observed in the resident peritoneal cells or in the liver. These findings identify novel mechanisms for modulating the magnitude and duration of selective TLR-induced chemokine and cytokine gene expression and further establish the importance of cell stress pathways in coordinating the outcomes of cellular and tissue injury.


Assuntos
Quimiocina CXCL1/genética , Quimiocina CXCL2/genética , Macrófagos/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Receptor 3 Toll-Like/genética , Receptor 4 Toll-Like/genética , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Western Blotting , Linhagem Celular , Células Cultivadas , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dimetil Sulfóxido/farmacologia , Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , Interferência de RNA , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Fatores de Transcrição de Fator Regulador X , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética , Receptor 3 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Tunicamicina/farmacologia
8.
J Immunol ; 192(11): 5098-108, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24790153

RESUMO

Hepatic stellate cells (HSC) are a major source of the immunoregulatory metabolite all-trans retinoic acid (ATRA), which may contribute to the generation of tolerogenic dendritic cells (DCs) in the liver. The present study seeks to clarify the mechanism(s) through which ATRA promotes the development of tolerogenic DCs. Although bone marrow-derived ATRA-treated DCs (RA-DCs) and conventional DCs had comparable surface phenotype, RA-DCs had diminished stimulatory capacity and could directly inhibit the expansion of DC/OVA-stimulated OT-II T cells. Arginase-1 (Arg-1) was found promote suppression because 1) ATRA was a potent inducer of Arg-1 protein and activity, 2) the Arg-1 inhibitor N(w)-hydroxy nor-l-arginine partially reversed suppression, and 3) the suppressive function of RA-DCs was partially compromised using OT-II T cells from GCN2(-/-) mice, which are insensitive to Arg-1. Inducible NO synthase (iNOS), however, was found to be a more significant contributor to RA-DC function because 1) ATRA potentiated the expression of IFN-γ-induced iNOS, 2) suppressive function in RA-DCs was blocked by the iNOS inhibitor N(G)-monomethyl-l-arginine, monoacetate salt, and 3) RA-DCs derived from iNOS(-/-) mice exhibited near complete loss of tolerogenic function, despite sustained Arg-1 activity. The expression of iNOS and the suppressive function of RA-DCs were dependent on both IFN-γ and ATRA. Furthermore, the in vivo behavior of RA-DCs proved to be consistent with their in vitro behavior. Thus, we conclude that ATRA enhances both Arg-1 and iNOS expression in IFN-γ-treated DCs, resulting in a tolerogenic phenotype. These findings elucidate mechanisms through which ATRA may contribute to liver immune tolerance.


Assuntos
Antineoplásicos/farmacologia , Arginase/imunologia , Células da Medula Óssea/imunologia , Células Dendríticas/imunologia , Tolerância Imunológica/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/imunologia , Tretinoína/farmacologia , Animais , Arginase/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Regulação Enzimológica da Expressão Gênica/imunologia , Tolerância Imunológica/genética , Tolerância Imunológica/imunologia , Interferon gama/genética , Interferon gama/imunologia , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/genética , Linfócitos T/imunologia
10.
J Immunol ; 191(2): 640-9, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23772036

RESUMO

IL-17, a major inflammatory cytokine plays a critical role in the pathogenesis of many autoimmune inflammatory diseases. In this study, we report a new function of RNA-binding protein HuR in IL-17-induced Act1-mediated chemokine mRNA stabilization. HuR deficiency markedly reduced IL-17-induced chemokine expression due to increased mRNA decay. Act1-mediated HuR polyubiquitination was required for the binding of HuR to CXCL1 mRNA, leading to mRNA stabilization. Although IL-17 induced the coshift of Act1 and HuR to the polysomal fractions in a sucrose gradient, HuR deficiency reduced the ratio of translation-active/translation-inactive IL-17-induced chemokine mRNAs. Furthermore, HuR deletion in distal lung epithelium attenuated IL-17-induced neutrophilia. In summary, HuR functions to couple receptor-proximal signaling to posttranscriptional machinery, contributing to IL-17-induced inflammation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Quimiocina CXCL1/genética , Quimiocina CXCL5/genética , Proteínas ELAV/metabolismo , Interleucina-17/metabolismo , Estabilidade de RNA , Animais , Linhagem Celular , Proteínas ELAV/genética , Células HeLa , Humanos , Inflamação/imunologia , Pulmão/metabolismo , Camundongos , Camundongos Knockout , Ligação Proteica , RNA Mensageiro/metabolismo , Mucosa Respiratória/metabolismo , Transdução de Sinais , Ubiquitinação
11.
ACS Synth Biol ; 12(12): 3578-3590, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38049144

RESUMO

Metagenomic sequences represent an untapped source of genetic novelty, particularly for conjugative systems that could be used for plasmid-based delivery of Cas9-derived antimicrobial agents. However, unlocking the functional potential of conjugative systems purely from metagenomic sequences requires the identification of suitable candidate systems as starting scaffolds for de novo DNA synthesis. Here, we developed a bioinformatics approach that searches through the metagenomic "trash bin" for genes associated with conjugative systems present on contigs that are typically excluded from common metagenomic analysis pipelines. Using a human metagenomic gut data set representing 2805 taxonomically distinct units, we identified 1598 contigs containing conjugation genes with a differential distribution in human cohorts. We synthesized de novo an entire Citrobacter spp. conjugative system of 54 kb containing at least 47 genes and assembled it into a plasmid, pCitro. We found that pCitro conjugates from Escherichia coli to Citrobacter rodentium with a 30-fold higher frequency than to E. coli, and is compatible with Citrobacter resident plasmids. Mutations in the traV and traY conjugation components of pCitro inhibited conjugation. We showed that pCitro can be repurposed as an antimicrobial delivery agent by programming it with the TevCas9 nuclease and Citrobacter-specific sgRNAs to kill C. rodentium. Our study reveals a trove of uncharacterized conjugative systems in metagenomic data and describes an experimental framework to animate these large genetic systems as novel target-adapted delivery vectors for Cas9-based editing of bacterial genomes.


Assuntos
Anti-Infecciosos , Escherichia coli , Humanos , Escherichia coli/genética , Sistemas CRISPR-Cas/genética , RNA Guia de Sistemas CRISPR-Cas , Conjugação Genética/genética , Plasmídeos/genética
12.
J Immunol ; 182(3): 1660-6, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19155515

RESUMO

IL-17 alone is a relatively weak inducer of gene expression, but cooperates with other cytokines, including TNF-alpha, to generate a strong response in part via prolongation of mRNA t(1/2). Because TNFR-associated factor 6 (TRAF6) has been reported to be essential for signaling by IL-17, we examined its involvement in IL-17-mediated mRNA stabilization. Although overexpression of TRAF6 in HeLa cells activates NF-kappaB, it does not stabilize transfected KC mRNA. Furthermore, a dominant-negative TRAF6 abrogates NF-kappaB activation, but does not block IL-17-induced chemokine mRNA stabilization. IL-17 can stabilize KC and MIP-2 mRNAs comparably in TNF-alpha-treated mouse embryo fibroblasts from TRAF6(+/+) and TRAF6(-/-) mice. TRAF6 is known to couple upstream signals with activation of p38 MAPK and mitogen activated protein kinase activated protein kinase 2, both of which have been shown to be important for Toll/IL-1R-mediated mRNA stabilization in various cell types. Inhibition of p38 MAPK, however, does not block IL-17-induced KC mRNA stabilization, and IL-17 can stabilize KC mRNA equally in mouse embryo fibroblasts from both wild-type and mitogen activated protein kinase activated protein kinase 2/3 doubly-deficient mice. Finally, IL-17 can amplify the levels of multiple TNF-alpha-stimulated mRNAs in wild-type and TRAF6-deficient cells, but not in cells from Act1(-/-) mice. Collectively, these findings demonstrate the existence of a TRAF6/p38 MAPK-independent pathway that couples the IL-17R with enhanced mRNA stability. Because the most potent effects of IL-17 on gene expression are obtained in cooperation with other cytokines such as TNF-alpha, these findings suggest that this pathway is a major contributing mechanism for response to IL-17.


Assuntos
Interleucina-17/fisiologia , Estabilidade de RNA/imunologia , RNA Mensageiro/metabolismo , Transdução de Sinais/imunologia , Fator 6 Associado a Receptor de TNF/fisiologia , Animais , Células Cultivadas , Quimiocinas/biossíntese , Quimiocinas/genética , Regulação da Expressão Gênica/imunologia , Células HeLa , Humanos , Mediadores da Inflamação/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/fisiologia , RNA Mensageiro/biossíntese , Transdução de Sinais/genética , Fator 6 Associado a Receptor de TNF/biossíntese , Fator 6 Associado a Receptor de TNF/deficiência , Fator 6 Associado a Receptor de TNF/genética , Fator de Necrose Tumoral alfa/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
13.
Nat Cancer ; 2(11): 1136-1151, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-35122055

RESUMO

Glioblastoma (GBM) contains abundant tumor-associated macrophages (TAMs). The majority of TAMs are tumor-promoting macrophages (pTAMs), while tumor-suppressive macrophages (sTAMs) are the minority. Thus, reprogramming pTAMs into sTAMs represents an attractive therapeutic strategy. By screening a collection of small-molecule compounds, we find that inhibiting ß-site amyloid precursor protein-cleaving enzyme 1 (BACE1) with MK-8931 potently reprograms pTAMs into sTAMs and promotes macrophage phagocytosis of glioma cells; moreover, low-dose radiation markedly enhances TAM infiltration and synergizes with MK-8931 treatment to suppress malignant growth. BACE1 is preferentially expressed by pTAMs in human GBMs and is required to maintain pTAM polarization through trans-interleukin 6 (IL-6)-soluble IL-6 receptor (sIL-6R)-signal transducer and activator of transcription 3 (STAT3) signaling. Because MK-8931 and other BACE1 inhibitors have been developed for Alzheimer's disease and have been shown to be safe for humans in clinical trials, these inhibitors could potentially be streamlined for cancer therapy. Collectively, this study offers a promising therapeutic approach to enhance macrophage-based therapy for malignant tumors.


Assuntos
Glioblastoma , Secretases da Proteína Precursora do Amiloide , Ácido Aspártico Endopeptidases , Glioblastoma/tratamento farmacológico , Humanos , Macrófagos/patologia , Fagocitose
14.
Nat Commun ; 10(1): 4544, 2019 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-31586051

RESUMO

The selective regulation of bacteria in complex microbial populations is key to controlling pathogenic bacteria. CRISPR nucleases can be programmed to kill bacteria, but require an efficient and broad-host range delivery system to be effective. Here, using an Escherichia coli and Salmonella enterica co-culture system, we show that plasmids based on the IncP RK2 conjugative system can be used as delivery vectors for a TevSpCas9 dual nuclease. Notably, a cis-acting plasmid that encodes the conjugation and CRISPR machinery conjugates from E. coli to S. enterica with high frequency compared to a trans system that separates conjugation and CRISPR machinery. In culture conditions that enhance cell-to-cell contact, conjugation rates approach 100% with the cis-acting plasmid. Targeting of single or multiplexed sgRNAs to non-essential genes results in high S. enterica killing efficiencies. Our data highlight the potential of cis-acting conjugative plasmids as a delivery system for CRISPR nucleases or other microbial-altering agents for targeted bacterial killing.


Assuntos
Anti-Infecciosos/administração & dosagem , Proteína 9 Associada à CRISPR/administração & dosagem , Conjugação Genética , Sistemas de Liberação de Medicamentos/métodos , Técnicas de Transferência de Genes , Biofilmes/efeitos dos fármacos , Proteína 9 Associada à CRISPR/genética , Técnicas de Cocultura , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Testes de Sensibilidade Microbiana , Plasmídeos/genética , RNA Guia de Cinetoplastídeos/genética , Saccharomyces cerevisiae , Salmonella enterica/efeitos dos fármacos , Salmonella enterica/genética
15.
J Exp Med ; 216(1): 195-214, 2019 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-30578323

RESUMO

Lrig1 marks a distinct population of stem cells restricted to the upper pilosebaceous unit in normal epidermis. Here we report that IL-17A-mediated activation of EGFR plays a critical role in the expansion and migration of Lrig1+ stem cells and their progenies in response to wounding, thereby promoting wound healing and skin tumorigenesis. Lrig1-specific deletion of the IL-17R adaptor Act1 or EGFR in mice impairs wound healing and reduces tumor formation. Mechanistically, IL-17R recruits EGFR for IL-17A-mediated signaling in Lrig1+ stem cells. While TRAF4, enriched in Lrig1+ stem cells, tethers IL-17RA and EGFR, Act1 recruits c-Src for IL-17A-induced EGFR transactivation and downstream activation of ERK5, which promotes the expansion and migration of Lrig1+ stem cells. This study demonstrates that IL-17A activates the IL-17R-EGFR axis in Lrig1+ stem cells linking wound healing to tumorigenesis.


Assuntos
Carcinogênese/metabolismo , Epiderme/metabolismo , Receptores ErbB/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores de Interleucina-17/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Cicatrização , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteína Tirosina Quinase CSK , Carcinogênese/genética , Carcinogênese/patologia , Epiderme/patologia , Receptores ErbB/genética , Células HeLa , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Receptores de Interleucina-17/genética , Células-Tronco/patologia , Quinases da Família src/genética , Quinases da Família src/metabolismo
16.
J Leukoc Biol ; 82(2): 213-9, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17409125

RESUMO

The magnitude and character of the inflammatory process are determined in part via the trafficking of leukocytes into sites of injury and infection, and this process depends on proper control of the expression of genes encoding chemoattractant peptides and their receptors. Although these controls operate at multiple mechanistic levels, recent evidence indicates that post-transcriptional events governing the half-life of select mRNAs are important determinants. Adenine-uridine rich elements (AREs) located within 3' untranslated regions (UTRs) confer constitutive mRNA instability and in some cases, stabilization following stimulation by ligands of the Toll-IL-1 receptor (TIR) family. Although the importance of AREs in determining activity and mRNA half-life is well-recognized, the mechanistic scope and diversity remain poorly understood. Using the mouse KC or CXCL1 gene as a model, we have demonstrated that the abundance of mRNA and protein produced during an inflammatory response depends on multiple mechanistically distinct AREs present in the 3' UTR of the mRNA. The mRNA encoding the receptor for N-terminal formyl-methionine-containing peptides is also unstable and subject to stabilization in response to TIR ligands. These two models can, however, be readily distinguished from one another on the basis of specific stimulus sensitivity and the signaling pathways, through which such stimuli couple to the control of mRNA decay. These models demonstrate the substantial diversity operative in the post-transcriptional regulation of inflammatory gene expression.


Assuntos
Quimiocinas/genética , Quimiocinas/metabolismo , Regulação da Expressão Gênica , Processamento Pós-Transcricional do RNA , Receptores de Formil Peptídeo/genética , Animais , Previsões , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Modelos Biológicos , RNA Mensageiro/química , RNA Mensageiro/metabolismo
17.
Elife ; 62017 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-28990926

RESUMO

Expression of inflammatory genes is determined in part by post-transcriptional regulation of mRNA metabolism but how stimulus- and transcript-dependent nuclear export influence is poorly understood. Here, we report a novel pathway in which LPS/TLR4 engagement promotes nuclear localization of IRAK2 to facilitate nuclear export of a specific subset of inflammation-related mRNAs for translation in murine macrophages. IRAK2 kinase activity is required for LPS-induced RanBP2-mediated IRAK2 sumoylation and subsequent nuclear translocation. Array analysis showed that an SRSF1-binding motif is enriched in mRNAs dependent on IRAK2 for nuclear export. Nuclear IRAK2 phosphorylates SRSF1 to reduce its binding to target mRNAs, which promotes the RNA binding of the nuclear export adaptor ALYREF and nuclear export receptor Nxf1 loading for the export of the mRNAs. In summary, LPS activates a nuclear function of IRAK2 that facilitates the assembly of nuclear export machinery to export selected inflammatory mRNAs to the cytoplasm for translation.


Assuntos
Transporte Ativo do Núcleo Celular , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Macrófagos/imunologia , RNA Mensageiro/metabolismo , Animais , Lipopolissacarídeos/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Fosforilação , Proteínas de Ligação a RNA/metabolismo , Fatores de Processamento de Serina-Arginina/metabolismo , Sumoilação
18.
Cancer Res ; 62(9): 2606-10, 2002 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11980656

RESUMO

The cytokine interleukin (IL)-10 has potent antitumor activity in many model systems when expressed locally at very high levels from the time of tumor transplantation. We now demonstrate that systemic administration of recombinant human IL-10 to animals bearing established highly malignant mammary tumors also leads to significant growth inhibition. We had shown previously that expression of the CXC chemokines Mig (monokine induced by IFN-gamma) and IP-10 (inducible protein 10) is observed in IL-10 transduced but not neo-vector control tumors. We now demonstrate that treatment of IL-10-tumor-bearing mice with antibodies to either chemokine partially reverses the therapeutic effect of IL-10. Tumor growth in animals treated with both antibodies is comparable with that of vector control tumors. Direct transduction of Mig cDNA into the parental tumor cell line before transplantation also results in smaller tumors. This tumor growth inhibition is associated with increased numbers of CD4+ cells, consistent with a T-cell chemoattractant activity for Mig. No change in vascularization, as indicated by CD31+ cells, was observed in either Mig or IL-10-transfected tumors. Thus, an antiangiogenic activity for either cytokine could not be confirmed. Mig and IP-10 are critical to the therapeutic response resulting from high levels of IL-10, and, furthermore, Mig as a single agent also has tumor-inhibitory activity in a model of breast cancer.


Assuntos
Adenocarcinoma/imunologia , Quimiocinas CXC/imunologia , Peptídeos e Proteínas de Sinalização Intercelular , Interleucina-10/farmacologia , Neoplasias Mamárias Experimentais/imunologia , Adenocarcinoma/genética , Adenocarcinoma/terapia , Sequência de Aminoácidos , Animais , Quimiocina CXCL10 , Quimiocina CXCL9 , Quimiocinas CXC/genética , Imunoterapia , Interleucina-10/genética , Interleucina-10/imunologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Dados de Sequência Molecular , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Transdução Genética , Transfecção
19.
J Interferon Cytokine Res ; 25(1): 11-9, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15684618

RESUMO

Regulation of leukocyte recruitment is an important determinant of the host response to microbial infection. Because tissue infiltration by inflammatory cells represents a potential source of unnecessary tissue damage, the process may be controlled by modulating the expression of chemoattractants and the receptors through which they promote directed leukocyte migration. In the present report, we show that expression of the receptor for chemotactic formylated peptides (FPR1)is negatively regulated in both macrophages and neutrophils by interleukin-4 (IL-4). The reduction of FPR1 mRNA occurs rapidly in response to both IL-4 and IL-13 but endures for <4 h after the removal of IL-4. As with many other responses to IL-4 and IL-13, suppression of FPR1 expression is dependent on the activation of Stat6. The inhibitory effect exhibits relative stimulus specificity in that other Stat-activating cytokines, such as interferon-gamma (IFN-gamma), IFN-beta, and IL-10, have no effect. Using nuclear run-on analysis, the rate of FPR1 gene transcription is high but is not suppressed by IL-4. Moreover, IL-4 does not appear to alter the rate of FPR mRNA decay. Nevertheless, FPR mRNA exhibits a short half-life (< or =2 h), and this appears to be a critical feature of the ability of IL-4 to reduce expression. Taken together, the data suggest that IL-4 and IL-13 suppress the expression of FPR1 mRNA via a mechanism that operates to eliminate primary transcripts prior to maturation and depends on the constitutive instability of preexisiting mRNA.


Assuntos
Regulação da Expressão Gênica/fisiologia , Interleucina-4/metabolismo , Macrófagos Peritoneais/metabolismo , Receptores de Formil Peptídeo/genética , Animais , Camundongos , RNA Mensageiro/metabolismo , Receptores de Formil Peptídeo/biossíntese , Fator de Transcrição STAT6 , Transativadores/metabolismo
20.
J Leukoc Biol ; 75(4): 641-8, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14704366

RESUMO

KC and macrophage-inflammatory protein-2 (MIP-2) are CXC chemokines that exhibit distinct temporal patterns of expression in the skin following surgical injury. In situ hybridization analysis demonstrates that these two chemokines are expressed by distinct cell types at different times following injury. Dermal fibroblasts and endothelial cells are primarily responsible for KC expression in the skin 6 h following surgery. In contrast, MIP-2 production appears to be restricted to infiltrating inflammatory leukocytes including neutrophils and monocytes, which appear later in the response. This cell type-specific pattern of chemokine expression is recapitulated in vitro using isolated primary- and long-term-cultured cell types. Primary dermal fibroblasts stimulated with interleukin-1alpha express predominantly KC and very little MIP-2, and peritoneal exudate neutrophils produce as much or more MIP-2 as KC following stimulation in vitro. Although a collection of exogenous stimuli can induce expression of KC and MIP-2, the quantitative ratio for expression reflects the cell type and not the stimulus. The selective expression of KC over MIP-2 in endothelial cells results from markedly greater KC gene transcription and not from alterations in the rate of mRNA decay. These results demonstrate that distinct CXC chemokines show restricted expression in myeloid versus nonmyeloid cell types and that patterns of chemokine expression at sites of inflammation in vivo reflect the temporally ordered contribution of these distinct cell types.


Assuntos
Quimiocinas/genética , Quimiotaxia de Leucócito/imunologia , Citocinas/genética , Inflamação/imunologia , Neutrófilos/metabolismo , Ferimentos e Lesões/imunologia , Animais , Células Cultivadas , Quimiocina CXCL1 , Quimiocina CXCL2 , Quimiocinas/biossíntese , Quimiocinas CXC , Citocinas/biossíntese , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação da Expressão Gênica/genética , Inflamação/fisiopatologia , Mediadores da Inflamação/metabolismo , Interleucina-1/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Neutrófilos/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Ferimentos e Lesões/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA