Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cells ; 13(7)2024 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-38607004

RESUMO

Leucine-rich repeat kinase 2 (LRRK2) is a serine-threonine protein kinase belonging to the ROCO protein family. Within the kinase domain of LRRK2, a point mutation known as LRRK2 G2019S has emerged as the most prevalent variant associated with Parkinson's disease. Recent clinical studies have indicated that G2019S carriers have an elevated risk of cancers, including colon cancer. Despite this observation, the underlying mechanisms linking LRRK2 G2019S to colon cancer remain elusive. In this study, employing a colitis-associated cancer (CAC) model and LRRK2 G2019S knock-in (KI) mouse model, we demonstrate that LRRK2 G2019S promotes the pathogenesis of colon cancer, characterized by increased tumor number and size in KI mice. Furthermore, LRRK2 G2019S enhances intestinal epithelial cell proliferation and inflammation within the tumor microenvironment. Mechanistically, KI mice exhibit heightened susceptibility to DSS-induced colitis, with inhibition of LRRK2 kinase activity ameliorating colitis severity and CAC progression. Our investigation also reveals that LRRK2 G2019S promotes inflammasome activation and exacerbates gut epithelium necrosis in the colitis model. Notably, GSDMD inhibitors attenuate colitis in LRRK2 G2019S KI mice. Taken together, our findings offer experimental evidence indicating that the gain-of-kinase activity in LRRK2 promotes colorectal tumorigenesis, suggesting LRRK2 as a potential therapeutic target in colon cancer patients exhibiting hyper LRRK2 kinase activity.


Assuntos
Colite , Neoplasias do Colo , Gasderminas , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Animais , Camundongos , Colite/induzido quimicamente , Colite/complicações , Colite/genética , Neoplasias do Colo/genética , Inflamação/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Microambiente Tumoral
2.
bioRxiv ; 2023 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-37425755

RESUMO

LRRK2 G2019S is the most prevalent variant associated with Parkinson's disease (PD), found in 1-3% of sporadic and 4-8% of familial PD cases. Intriguingly, emerging clinical studies have suggested that LRRK2 G2019S carriers have an increased risk of cancers including colorectal cancer. However, the underlying mechanisms of the positive correlation between LRRK2-G2019S and colorectal cancer remain unknown. Using a mouse model of colitis-associated cancer (CAC) and LRRK2 G2019S knockin (KI) mice, here we report that LRRK2 G2019S promotes the pathogenesis of colon cancer as evidenced by increased tumor number and tumor size in LRRK2 G2019S KI mice. LRRK2 G2019S promoted intestinal epithelial cell proliferation and inflammation within the tumor microenvironment. Mechanistically, we found that LRRK2 G2019S KI mice are more susceptible to dextran sulfate sodium (DSS)-induced colitis. Suppressing the kinase activity of LRRK2 ameliorated the severity of colitis in both LRRK2 G2019S KI and WT mice. At the molecular level, our investigation unveiled that LRRK2 G2019S promotes the production of reactive oxygen species, triggers inflammasome activation, and induces cell necrosis in the gut epithelium in a mouse model of colitis. Collectively, our data provide direct evidence that gain-of-kinase activity in LRRK2 promotes colorectal tumorigenesis, implicating LRRK2 as a potential target in colon cancer patients with hyper LRRK2 kinase activity.

3.
Nat Commun ; 13(1): 2748, 2022 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-35585086

RESUMO

Toll-like receptors/Interleukin-1 receptor signaling plays an important role in high-fat diet-induced adipose tissue dysfunction contributing to obesity-associated metabolic syndromes. Here, we show an unconventional IL-1R-IRAKM-Slc25a1 signaling axis in adipocytes that reprograms lipogenesis to promote diet-induced obesity. Adipocyte-specific deficiency of IRAKM reduces high-fat diet-induced body weight gain, increases whole body energy expenditure and improves insulin resistance, associated with decreased lipid accumulation and adipocyte cell sizes. IL-1ß stimulation induces the translocation of IRAKM Myddosome to mitochondria to promote de novo lipogenesis in adipocytes. Mechanistically, IRAKM interacts with and phosphorylates mitochondrial citrate carrier Slc25a1 to promote IL-1ß-induced mitochondrial citrate transport to cytosol and de novo lipogenesis. Moreover, IRAKM-Slc25a1 axis mediates IL-1ß induced Pgc1a acetylation to regulate thermogenic gene expression in adipocytes. IRAKM kinase-inactivation also attenuates high-fat diet-induced obesity. Taken together, our study suggests that the IL-1R-IRAKM-Slc25a1 signaling axis tightly links inflammation and adipocyte metabolism, indicating a potential therapeutic target for obesity.


Assuntos
Resistência à Insulina , Lipogênese , Adipócitos/metabolismo , Animais , Dieta Hiperlipídica , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/metabolismo , Obesidade/etiologia , Obesidade/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Receptores de Interleucina-1/metabolismo , Termogênese
4.
J Clin Invest ; 130(8): 4218-4234, 2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32597834

RESUMO

Gasdermin D (GSDMD) induces pyroptosis via the pore-forming activity of its N-terminal domain, cleaved by activated caspases associated with the release of IL-1ß. Here, we report a nonpyroptotic role of full-length GSDMD in guiding the release of IL-1ß-containing small extracellular vesicles (sEVs) from intestinal epithelial cells (IECs). In response to caspase-8 inflammasome activation, GSDMD, chaperoned by Cdc37/Hsp90, recruits the E3 ligase, NEDD4, to catalyze polyubiquitination of pro-IL-1ß, serving as a signal for cargo loading into secretory vesicles. GSDMD and IL-1ß colocalize with the exosome markers CD63 and ALIX intracellularly, and GSDMD and NEDD4 are required for release of CD63+ sEVs containing IL-1ß, GSDMD, NEDD4, and caspase-8. Importantly, increased expression of epithelial-derived GSDMD is observed both in patients with inflammatory bowel disease (IBD) and those with experimental colitis. While GSDMD-dependent release of IL-1ß-containing sEVs is detected in cultured colonic explants from colitic mice, GSDMD deficiency substantially attenuates disease severity, implicating GSDMD-mediated release of IL-1ß sEVs in the pathogenesis of intestinal inflammation, such as that observed in IBD.


Assuntos
Colite/metabolismo , Células Epiteliais/metabolismo , Doenças Inflamatórias Intestinais/metabolismo , Interleucina-1beta/metabolismo , Mucosa Intestinal/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Animais , Linhagem Celular , Colite/genética , Colite/patologia , Células Epiteliais/patologia , Exossomos/genética , Exossomos/metabolismo , Exossomos/patologia , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patologia , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Interleucina-1beta/genética , Mucosa Intestinal/patologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Knockout , Ubiquitina-Proteína Ligases Nedd4/genética , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Proteínas de Ligação a Fosfato/genética , Tetraspanina 30/genética , Tetraspanina 30/metabolismo
5.
J Immunol ; 200(5): 1889-1900, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29352002

RESUMO

Dysregulation of the immune barrier function of the intestinal epithelium can often result in dysbiosis. In this study we report a novel role of intestinal epithelial cell (IEC)-derived liver kinase B1 (LKB1) in suppressing colitogenic microbiota. IEC-specific deletion of LKB1 (LKB1ΔIEC) resulted in an increased susceptibility to dextran sodium sulfate (DSS)-induced colitis and a definitive shift in the composition of the microbial population in the mouse intestine. Importantly, transfer of the microbiota from LKB1ΔIEC mice was sufficient to confer increased susceptibility to DSS-induced colitis in wild-type recipient mice. Collectively, the data indicate that LKB1 deficiency in intestinal epithelial cells nurtures the outgrowth of colitogenic bacteria in the commensal community. In addition, LKB1 deficiency in the intestinal epithelium reduced the production of IL-18 and antimicrobial peptides in the colon. Administration of exogenous IL-18 restored the expression of antimicrobial peptides, corrected the outgrowth of several bacterial genera, and rescued the LKB1ΔIEC mice from increased sensitivity to DSS challenge. Taken together, our study reveals an important function of LKB1 in IECs for suppressing colitogenic microbiota by IL-18 expression.


Assuntos
Células Epiteliais/imunologia , Mucosa Intestinal/imunologia , Intestinos/imunologia , Microbiota/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Quinases Ativadas por AMP , Animais , Colite/induzido quimicamente , Colite/imunologia , Colo/efeitos dos fármacos , Colo/imunologia , Sulfato de Dextrana/farmacologia , Disbiose/imunologia , Interleucina-18/imunologia , Intestinos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL
6.
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
7.
J Exp Med ; 214(10): 3051-3066, 2017 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-28821568

RESUMO

Although genetic polymorphisms in the LRRK2 gene are associated with a variety of diseases, the physiological function of LRRK2 remains poorly understood. In this study, we report a crucial role for LRRK2 in the activation of the NLRC4 inflammasome during host defense against Salmonella enteric serovar Typhimurium infection. LRRK2 deficiency reduced caspase-1 activation and IL-1ß secretion in response to NLRC4 inflammasome activators in macrophages. Lrrk2-/- mice exhibited impaired clearance of pathogens after acute S. Typhimurium infection. Mechanistically, LRRK2 formed a complex with NLRC4 in the macrophages, and the formation of the LRRK2-NLRC4 complex led to the phosphorylation of NLRC4 at Ser533. Importantly, the kinase activity of LRRK2 is required for optimal NLRC4 inflammasome activation. Collectively, our study reveals an important role for LRRK2 in the host defense by promoting NLRC4 inflammasome activation.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Proteínas de Ligação ao Cálcio/fisiologia , Inflamassomos/imunologia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/fisiologia , Salmonelose Animal/imunologia , Salmonella typhimurium/imunologia , Animais , Caspase 1/fisiologia , Inflamassomos/fisiologia , Interleucina-1beta/fisiologia , Macrófagos/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação
8.
Nat Commun ; 8: 15508, 2017 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-28561022

RESUMO

NOTCH1 signalling contributes to defective remyelination by impairing differentiation of oligodendrocyte progenitor cells (OPCs). Here we report that IL-17 stimulation induces NOTCH1 activation in OPCs, contributing to Th17-mediated demyelinating disease. Mechanistically, IL-17R interacts with NOTCH1 via the extracellular domain, which facilitates the cleavage of NOTHC1 intracellular domain (NICD1). IL-17-induced NOTCH1 activation results in the interaction of IL-17R adaptor Act1 with NICD1, followed by the translocation of the Act1-NICD1 complex into the nucleus. Act1-NICD1 are recruited to the promoters of several NOTCH1 target genes (including STEAP4, a metalloreductase important for inflammation and cell proliferation) that are specifically induced in the spinal cord by Th17 cells. A decoy peptide disrupting the IL-17RA-NOTCH1 interaction inhibits IL-17-induced NOTCH1 activation and attenuates Th17-mediated experimental autoimmune encephalitis (EAE). Taken together, these findings demonstrate critical crosstalk between the IL-17 and NOTCH1 pathway, regulating Th17-induced inflammatory and proliferative genes to promote demyelinating disease.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Interleucina-17/metabolismo , Esclerose Múltipla/imunologia , Células Precursoras de Oligodendrócitos/fisiologia , Receptor Notch1/imunologia , Células Th17/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Astrócitos , Diferenciação Celular/imunologia , Proliferação de Células/fisiologia , Técnicas de Cocultura , Feminino , Células HEK293 , Células HeLa , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/imunologia , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Cultura Primária de Células , Ligação Proteica/imunologia , Domínios Proteicos/fisiologia , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptores de Interleucina-17/metabolismo , Remielinização/fisiologia , Transdução de Sinais/imunologia , Células Th1/imunologia , Células Th17/metabolismo , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo
9.
Nat Immunol ; 17(5): 583-92, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26998763

RESUMO

Interleukin 1ß (IL-1ß) is critical for the in vivo survival, expansion and effector function of IL-17-producing helper T (T(H)17) cells during autoimmune responses, including experimental autoimmune encephalomyelitis (EAE). However, the spatiotemporal role and cellular source of IL-1ß during EAE pathogenesis are poorly defined. In the present study, we uncovered a T cell-intrinsic inflammasome that drives IL-1ß production during T(H)17-mediated EAE pathogenesis. Activation of T cell antigen receptors induced expression of pro-IL-1ß, whereas ATP stimulation triggered T cell production of IL-1ß via ASC-NLRP3-dependent caspase-8 activation. IL-1R was detected on T(H)17 cells but not on type 1 helper T (T(H)1) cells, and ATP-treated T(H)17 cells showed enhanced survival compared with ATP-treated T(H)1 cells, suggesting autocrine action of T(H)17-derived IL-1ß. Together these data reveal a critical role for IL-1ß produced by a T(H)17 cell-intrinsic ASC-NLRP3-caspase-8 inflammasome during inflammation of the central nervous system.


Assuntos
Proteínas Reguladoras de Apoptose/imunologia , Encefalomielite Autoimune Experimental/imunologia , Linfócitos T/imunologia , Células Th17/imunologia , Trifosfato de Adenosina/farmacologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Adaptadoras de Sinalização CARD , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Caspase 8/genética , Caspase 8/imunologia , Caspase 8/metabolismo , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Células Cultivadas , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/metabolismo , Citometria de Fluxo , Expressão Gênica/imunologia , Immunoblotting , Inflamassomos/genética , Inflamassomos/imunologia , Inflamassomos/metabolismo , Interleucina-17/genética , Interleucina-17/imunologia , Interleucina-17/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Células Th17/efeitos dos fármacos , Células Th17/metabolismo
10.
Am J Stem Cells ; 1(2): 146-153, 2012 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-22754757

RESUMO

Interleukin (IL)-6 family cytokine signaling through gp130 and signal transducer and activator of transcription (STAT) activation is believed important for early hematopoiesis. To determine whether gp130/STAT1/3 physical interaction is required, we compared hematopoietic repopulating activities of embryonic day (E)14.5 fetal liver cells from gp130(FXXQ/FXXQ) knock-in mice, which have four mutated STAT1/3 binding sites. In hematopoietic cells, failure to tyrosine phosphorylate STAT3 by gp130 did not cause any significant effects on myeloid progenitor colony forming units (CFU) in vitro and or on competitive multilineage hematopoietic reconstitution. Serial transplantation of fetal liver (FL) cells was unaffected throughout primary, secondary, and tertiary transplants indicating normal self-renewal capacity. Even gp130(FXXQ/FXXQ) on the background of STAT5 deficiency, with known hematopoietic stem cell (HSC) repopulating dysfunction, did not further impair HSCs beyond that of STAT5 alone. Overall, the defective gp130-mediated STAT1/3 signaling is surprisingly dispensable for HSC function. However, since these mice lack both STAT1/3 binding sites there are several possible explanations for this result and these are discussed.

11.
J Immunol ; 189(1): 33-7, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22649194

RESUMO

The effector T cell subset, Th17, plays a significant role in the pathogenesis of multiple sclerosis and of other autoimmune diseases. The signature cytokine, IL-17, engages the IL-17R and recruits the E3-ligase NF-κB activator 1 (Act1) upon stimulation. In this study, we examined the role of TNFR-associated factor (TRAF)4 in IL-17 signaling and Th17-mediated autoimmune encephalomyelitis. Primary cells from TRAF4-deficient mice displayed markedly enhanced IL-17-activated signaling pathways and induction of chemokine mRNA. Adoptive transfer of MOG35-55 specific wild-type Th17 cells into TRAF4-deficient recipient mice induced an earlier onset of disease. Mechanistically, we found that TRAF4 and TRAF6 used the same TRAF binding sites on Act1, allowing the competition of TRAF4 with TRAF6 for the interaction with Act1. Taken together, the results of this study reveal the necessity of a unique role of TRAF4 in restricting the effects of IL-17 signaling and Th17-mediated disease.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Interleucina-17/antagonistas & inibidores , Interleucina-17/fisiologia , Transdução de Sinais/imunologia , Fator 4 Associado a Receptor de TNF/fisiologia , Animais , Encefalomielite Autoimune Experimental/genética , Células HEK293 , Células HeLa , Humanos , Interleucina-17/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/biossíntese , Transdução de Sinais/genética , Fator 4 Associado a Receptor de TNF/deficiência , Fator 4 Associado a Receptor de TNF/genética , Células Th17/imunologia , Células Th17/patologia
12.
Immunity ; 36(5): 821-33, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22608496

RESUMO

Interleukin-25 (IL-25 or IL-17E), a member of the structurally related IL-17 family, functions as an important mediator of T helper 2 cell-type (type 2) responses. We examined the cell type-specific role of IL-25-induced Act1-mediated signaling in protective immunity against helminth infection. Targeted Act1 deficiency in epithelial cells resulted in a marked delay in worm expulsion and abolished the expansion of the Lin(-)c-Kit(+) innate cell population in the mesenteric lymph node, lung, and liver. Th2 cell-inducing cytokine (IL-25 and IL-33) expression were reduced in the intestinal epithelial cells from the infected and IL-25-injected epithelial-specific Act1-deficient mice. Adoptive transfer of Lin(-)c-Kit(+) cells or combined injection of IL-25 and IL-33 restored the type 2 responses in these mice. Taken together, these results suggest that epithelial-specific Act1 mediates the expansion of the Lin(-)c-Kit(+) innate cell population through the positive-feedback loop of IL-25, initiating the type 2 immunity against helminth infection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Células Epiteliais/imunologia , Helmintíase/imunologia , Helmintos/imunologia , Interleucinas/imunologia , Células Th2/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem da Célula , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Helmintíase/metabolismo , Helmintos/metabolismo , Imunidade Inata/imunologia , Interleucinas/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Fígado/citologia , Fígado/imunologia , Fígado/metabolismo , Pulmão/citologia , Pulmão/imunologia , Pulmão/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Th2/metabolismo
13.
Sci Signal ; 4(197): ra72, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22045852

RESUMO

Interleukin-17 (IL-17) and IL-25 signaling induce the expression of genes encoding inflammatory factors and are implicated in the pathology of various inflammatory diseases. Nuclear factor κB (NF-κB) activator 1 (Act1) is an adaptor protein and E3 ubiquitin ligase that is critical for signaling by either IL-17 or IL-25, and it is recruited to their receptors (IL-17R and IL-25R) through heterotypic interactions between the SEFIR [SEF (similar expression to fibroblast growth factor genes) and IL-17R] domain of Act1 and that of the receptor. SEFIR domains have structural similarity with the Toll-IL-1 receptor (TIR) domains of Toll-like receptors and IL-1R. Whereas the BB' loop of TIR is required for TIR-TIR interactions, we found that deletion of the BB' loop from Act1 or IL-17RA (a common subunit of both IL-17R and IL-25R) did not affect Act1-IL-17RA interactions; rather, deletion of the CC' loop from Act1 or IL-17RA abolished the interaction between both proteins. Surface plasmon resonance measurements showed that a peptide corresponding to the CC' loop of Act1 bound directly to IL-17RA. A cell-permeable decoy peptide based on the CC' loop sequence inhibited IL-17- or IL-25-mediated signaling in vitro, as well as IL-17- and IL-25-induced pulmonary inflammation in mice. Together, these findings provide the molecular basis for the specificity of SEFIR-SEFIR versus TIR-TIR domain interactions and consequent signaling. Moreover, we suggest that the CC' loop motif of SEFIR domains is a promising target for therapeutic strategies against inflammatory diseases associated with IL-17 or IL-25 signaling.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Peptídeos/farmacologia , Pneumonia/prevenção & controle , Receptores de Interleucina-17/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Western Blotting , Células Cultivadas , Feminino , Células HEK293 , Células HeLa , Humanos , Interleucina-17/toxicidade , Interleucinas/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Peptídeos/química , Peptídeos/genética , Pneumonia/induzido quimicamente , Pneumonia/metabolismo , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Receptores de Interleucina-17/química , Receptores de Interleucina-17/genética , Transdução de Sinais/efeitos dos fármacos , Ressonância de Plasmônio de Superfície
14.
J Immunol ; 186(5): 3006-14, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21270393

RESUMO

Bone marrow-derived plasmacytoid dendritic cells (pDCs) from IL-1R-associated kinase (IRAK)2-deficient mice produced more IFNs than did wild-type pDCs upon stimulation with the TLR9 ligand CpG. Furthermore, in CpG-stimulated IRAK2-deficient pDCs there was increased nuclear translocation of IFN regulatory factor 7, the key transcription factor for IFN gene transcription in these cells. In IRAK2-deficient macrophages, enhanced NF-κB activation and increased expression of CpG-induced genes were detected within 2 h after treatment. However, at later times, NF-κB activation was decreased and, in contrast to the results with IFN, there was less secretion of other proinflammatory cytokines (such as TNF-α) and chemokines in CpG-stimulated IRAK2-deficient pDCs and macrophages. Therefore, although IRAK2 is a negative regulator of TLR9-mediated IFN production through its modulation of the transcriptional activity of IFN regulatory factor 7, it is also a positive regulator of TLR9-mediated proinflammatory cytokine and chemokine production at some level subsequent to transcription.


Assuntos
Citocinas/biossíntese , Mediadores da Inflamação/metabolismo , Interferon Tipo I/biossíntese , Interferon beta/biossíntese , Quinases Associadas a Receptores de Interleucina-1/fisiologia , Receptor Toll-Like 9/fisiologia , Animais , Células Cultivadas , Ilhas de CpG/imunologia , Citocinas/antagonistas & inibidores , Citocinas/fisiologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Regulação para Baixo/imunologia , Infecções por Herpesviridae/enzimologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/patologia , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/fisiologia , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/fisiologia , Interferon Tipo I/antagonistas & inibidores , Interferon-alfa , Interferon beta/antagonistas & inibidores , Quinases Associadas a Receptores de Interleucina-1/deficiência , Ligantes , Camundongos , Camundongos Knockout , Proteínas Recombinantes , Rhadinovirus/imunologia , Receptor Toll-Like 9/metabolismo , Infecções Tumorais por Vírus/enzimologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/patologia
15.
Immunity ; 32(3): 414-25, 2010 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-20303295

RESUMO

Interleukin-17 (IL-17) secreted by T helper 17 (Th17) cells is essential in the development of experimental autoimmune encephalomyelitis (EAE). However, it remains unclear how IL-17-mediated signaling in different cellular compartments participates in the central nervous system (CNS) inflammatory process. We examined CNS inflammation in mice with specific deletion of Act1, a critical component required for IL-17 signaling, in endothelial cells, macrophages and microglia, and neuroectoderm (neurons, astrocytes, and oligodendrocytes). In Act1-deficient mice, Th17 cells showed normal infiltration into the CNS but failed to recruit lymphocytes, neutrophils, and macrophages. Act1 deficiency in endothelial cells or in macrophages and microglia did not substantially impact the development of EAE. However, targeted Act1 deficiency in neuroectoderm-derived CNS-resident cells resulted in markedly reduced severity in EAE. Specifically, Act1-deficient astrocytes showed impaired IL-17-mediated inflammatory gene induction. Thus, astroctyes are critical in IL-17-Act1-mediated leukocyte recruitment during autoimmune-induced inflammation of the CNS.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Astrócitos/imunologia , Encefalomielite Autoimune Experimental/imunologia , Interleucina-17/imunologia , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Astrócitos/metabolismo , Sobrevivência Celular , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Regulação da Expressão Gênica , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/imunologia
16.
Am J Hematol ; 84(5): 308-9, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19396856

RESUMO

Nearly half of the patients with newly diagnosed acute myeloid leukemia have normal cytogenetics (NC-AML) and are classified as intermediate risk, but their 5-year overall survival (OS) ranges from 24 to 42%. Therefore, molecular biomarkers to identify poor-risk patients are needed. Elevated AF1q expression in the absence of specific poor cytogenetics is associated with poor outcomes in pediatric patients with AML and adult patients with myelodysplastic syndrome. We examined AF1q expression in 290 patients with NC-AML. We found that patients with low AF1q (n = 73) expression (AF1q(low)) have better OS (P = 0.026), disease-free survival (P = 0.1), and complete remission rate (P = 0.06) when compared with patients with high AF1q expression (AF1q(high) n = 217). The patients with AF1q(high) had significantly greater incidence of concurrent tyrosine kinase3 internal tandem duplication. A subgroup of the patients with AF1q(high) who received allogeneic stem cell transplantation (SCT) had a significant better relapse-free survival when compared with patients who received chemotherapy/autologous SCT (P = 0.04). This study suggests that high AF1q expression is a poor prognostic marker for adult patients with NC-AML.


Assuntos
Biomarcadores Tumorais , Proteínas Sanguíneas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/terapia , Proteínas de Neoplasias/metabolismo , Adolescente , Adulto , Proteínas Sanguíneas/análise , Terapia Combinada , Análise Citogenética , Intervalo Livre de Doença , Feminino , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/análise , Prognóstico , Proteínas Proto-Oncogênicas , Transplante de Células-Tronco , Transplante Autólogo , Transplante Homólogo , Adulto Jovem , Tirosina Quinase 3 Semelhante a fms/genética
17.
Cancer Res ; 68(11): 4369-76, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18519698

RESUMO

As the pathophysiology of acute myelogenous leukemia (AML) involves a block of myeloid maturation, a desirable therapeutic strategy is to induce leukemic cell maturation to increase the efficacy and to avoid the side effects of traditional chemotherapeutics. Through a compound library screen, 6-benzylthioinosine (6BT) was identified as a promising differentiation-inducing agent. 6BT induces monocytic differentiation of myeloid leukemia cell lines such as HL-60 and OCI-AML3, as well as primary patient samples as evidenced by morphology, immunophenotyping, and nitroblue tetrazolium reduction. Not only can 6BT induce differentiation but a subset of AML cell lines such as MV4-11 and HNT34 instead undergo 6BT-mediated cell death. Despite inducing cell death in some leukemic cells, 6BT exhibits extremely low toxicity on several nonmalignant cells such as fibroblasts, normal bone marrow, and endothelial cells. This toxicity profile may relate to the function of 6BT as an inhibitor of the nucleoside transporter, ent1, which is thought to prevent it from entering many cell types. In contrast, 6BT likely enters at least some leukemic cell lines as shown by its requirement for phosphorylation for its differentiation activity. 6BT is also able to synergize with currently used myeloid differentiation agents such as ATRA and decitabine. Early studies indicate that the mechanism of action of this compound may involve ATP depletion that leads to growth inhibition and subsequent differentiation. Besides in vitro activity, 6BT also shows the ability to impair HL-60 and MV4-11 tumor growth in nude mice. 6BT is a promising new monocytic differentiation agent with apparent leukemic cell-specific activity.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Leucemia Mieloide Aguda/patologia , Tioinosina/análogos & derivados , Animais , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Humanos , Masculino , Camundongos , Camundongos Nus , Fosforilação , Reação em Cadeia da Polimerase , Tioinosina/metabolismo , Tioinosina/farmacologia , Transplante Heterólogo
18.
Exp Hematol ; 35(11): 1684-94, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17976521

RESUMO

OBJECTIVE: Signal transducer and activator of transcription 5 (STAT5) is a critical regulator of hematopoietic development and its impaired activation is associated with hematopoietic and immune cell defects. However, much of this information has been learned from knockout mice that still retain the potential for expression of STAT5 proteins that are N-terminally truncated due to alternative internal translation initiation codons. The goal of these studies was to use transplantation-based assays to analyze the degree of STAT5 deltaN activity in hematopoietic stem cells (HSC) and throughout lymphomyeloid development. METHODS: We have directly compared E14.5 fetal liver cells from mice with potential to express STAT5ab deltaN (STAT5ab(deltaN/deltaN)) with mice completely lacking STAT5a and STAT5b (STAT5abnull/null). We have also utilized retroviral complementation of STAT5abnull/null fetal liver HSC to enforce expression of full-length STAT5a or STAT5a lacking the first 136 amino acids (STAT5a deltaN). RESULTS: We report that STAT5 is required for HSC, lymphocyte, and erythrocyte development. We demonstrate that restored expression of STAT5a in STAT5abnull/null HSC provides a strong selective advantage, correcting T- and B-lymphocyte and erythrocyte development. Interestingly, Gr-1(+) blood cells were inversely correlated with B lymphocytes and both were normalized by STAT5a expression. In contrast, transduction of STAT5a deltaN only provided partial B-lymphocyte development. CONCLUSIONS: These studies define the role of STAT5 in maintaining normal lymphoid vs myeloid balance during hematopoiesis and highlight a major role for the N-domain in HSC function. The platform of retroviral complementation described here will be particularly useful for future studies to subdefine the N-domain regions that are critical for hematopoiesis.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas/citologia , Fator de Transcrição STAT5/fisiologia , Animais , Linhagem da Célula , Células Cultivadas , Linfócitos/citologia , Camundongos , Camundongos Knockout , Células Mieloides/citologia , Estrutura Terciária de Proteína , Fator de Transcrição STAT5/química , Fator de Transcrição STAT5/genética , Deleção de Sequência
19.
Blood ; 110(1): 116-24, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17374739

RESUMO

Gab2 is an important adapter molecule for cytokine signaling. Despite its major role in signaling by receptors associated with hematopoiesis, the role of Gab2 in hematopoiesis has not been addressed. We report that despite normal numbers of peripheral blood cells, bone marrow cells, and c-Kit(+)Lin(-)Sca-1(+) (KLS) cells, Gab2-deficient hematopoietic cells are deficient in cytokine responsiveness. Significant reductions in the number of colony-forming units in culture (CFU-C) in the presence of limiting cytokine concentrations were observed, and these defects could be completely corrected by retroviral complementation. In earlier hematopoiesis, Gab2-deficient KLS cells isolated in vitro responded poorly to hematopoietic growth factors, resulting in an up to 11-fold reduction in response to a cocktail of stem cell factor, flt3 ligand, and thrombopoietin. Gab2-deficient c-Kit(+)Lin(-) cells also demonstrate impaired activation of extracellular signal-regulated kinase (ERK) and S6 in response to IL-3, which supports defects in activating the phosphatidylinositol-3 kinase (PI-3K) and mitogen-associated protein kinase (MAPK) signaling cascades. Associated with the early defects in cytokine response, competitive transplantation of Gab2(-/-) bone marrow cells resulted in defective long-term multilineage repopulation. Therefore, we demonstrate that Gab2 adapter function is intrinsically required for hematopoietic cell response to early-acting cytokines, resulting in defective hematopoiesis in Gab2-deficient mice.


Assuntos
Hematopoese , Fosfoproteínas/deficiência , Fosfoproteínas/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Transplante de Medula Óssea , Células Cultivadas , Citocinas/farmacologia , Células-Tronco Hematopoéticas/patologia , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Mutantes , Fosfoproteínas/genética , Células-Tronco/citologia
20.
Zhonghua Xue Ye Xue Za Zhi ; 24(2): 74-7, 2003 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-12697100

RESUMO

OBJECTIVE: To explore the ex vivo expansion characteristics of selected CD(34)(+) cells and mononuclear cells (MNC). METHODS: CD(34)(+) cells were isolated from umbilical cord blood MNC by MiniMACS system, expanded under the same conditions as that for MNC. The effects of re-isolation and the MNC supernatant (MNC-SN) on the selected CD(34)(+) cells were investigated. And the CD(34)(-) cells of MNC were cultured ex vivo. RESULTS: In the culture of selected CD(34)(+) cells, both the colony density and the proportion of the CD(34)(+) cells declined continuously with the culturing, although they presented a high proliferation potential. However, in the culture of the MNC, from day 0 to day 7, the colony density and the proportion of CD(34)(+) cells were increased from 412 +/- 167/10(5) cells and (1.12 +/- 0.42)% to 1 162 +/- 566/10(5) cells and (4.17 +/- 1.44)%, respectively. It was found that both the total cells and the CD(34)(+) cells restored expansion potential by re-isolating. CD(34)(-) cells of MNC had the ability to form colony and could transform to CD(34)(+) cells. MNC-SN can promote colony forming ability and lead to CD(34)(+) cells differentiation at the same time. CONCLUSIONS: In ex vivo culture, selected CD(34)(+) cells presented a high proliferation and differentiation potentials, and the CD(34)(-) cells produced during the cultivation had inhibition effect on CD(34)(+) cells expansion. CD(34)(-) cell population from cord blood MNC contained hematopoietic stem/progenitor cells and the cytokines secreted by CD(34)(-) cells could induce CD(34)(+) cells to more mature colony-forming cells.


Assuntos
Antígenos CD34/análise , Sangue Fetal/citologia , Leucócitos Mononucleares/citologia , Contagem de Células , Diferenciação Celular/imunologia , Divisão Celular/imunologia , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Sangue Fetal/imunologia , Células-Tronco Hematopoéticas/citologia , Humanos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA