Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.033
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Mol Ther ; 31(2): 331-343, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36575793

RESUMO

Leukemia inhibitory factor (LIF) is a pleiotropic cytokine of the interleukin-6 (IL-6) superfamily. LIF was initially discovered as a factor to induce the differentiation of myeloid leukemia cells and thus inhibit their proliferation. Subsequent studies have highlighted the multi-functions of LIF under a wide variety of physiological and pathological conditions in a highly cell-, tissue-, and context-dependent manner. Emerging evidence has demonstrated that LIF plays an essential role in the stem cell niche, where it maintains the homeostasis and regeneration of multiple somatic tissues, including intestine, neuron, and muscle. Further, LIF exerts a crucial regulatory role in immunity and functions as a protective factor against many immunopathological diseases, such as infection, inflammatory bowel disease (IBD), and graft-verse-host disease (GVHD). It is worth noting that while LIF displays a tumor-suppressive function in leukemia, recent studies have highlighted the oncogenic role of LIF in many types of solid tumors, further demonstrating the complexities and context-dependent effects of LIF. In this review, we summarize the recent insights into the roles and mechanisms of LIF in stem cell homeostasis and regeneration, immunity, and cancer, and discuss the potential therapeutic options for human diseases by modulating LIF levels and functions.


Assuntos
Inibidores do Crescimento , Interleucina-6 , Humanos , Fator Inibidor de Leucemia/genética , Fator Inibidor de Leucemia/metabolismo , Inibidores do Crescimento/farmacologia , Inibidores do Crescimento/fisiologia , Diferenciação Celular , Subunidade alfa de Receptor de Fator Inibidor de Leucemia , Linfocinas/farmacologia , Linfocinas/fisiologia
2.
Gut ; 64(7): 1028-39, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25107557

RESUMO

OBJECTIVE: In this study, we investigated the role of Trefoil factor 1 (TFF1) in regulating cell proliferation and tumour development through ß-catenin signalling using in vivo and in vitro models of gastric tumorigenesis. DESIGN: Tff1-knockout (Tff1-KO) mice, immunohistochemistry, luciferase reporter, qRT-PCR, immunoblot, and phosphatase assays were used to examine the role of TFF1 on ß-catenin signalling pathway. RESULTS: Nuclear localisation of ß-catenin with transcriptional upregulation of its target genes, c-Myc and Ccnd1, was detected in hyperplastic tissue at an early age of 4-6 weeks and maintained during all stages of gastric tumorigenesis in the Tff1-KO mice. The reconstitution of TFF1 or TFF1 conditioned media significantly inhibited the ß-catenin/T-cell factor (TCF) transcription activity in MKN28 gastric cancer cells. In agreement with these results, we detected a reduction in the levels of nuclear ß-catenin with downregulation of c-MYC and CCND1 mRNA. Analysis of signalling molecules upstream of ß-catenin revealed a decrease in phosphorylated glycogen synthase kinase 3ß (p-GSK3ß) (Ser9) and p-AKT (Ser473) protein levels following the reconstitution of TFF1 expression; this was consistent with the increase of p-ß-catenin (Ser33/37/Thr41) and decrease of p-ß-catenin (Ser552). This TFF1-induced reduction in phosphorylation of GSK3ß, and AKT was dependent on protein phosphatase 2A (PP2A) activity. The treatment with okadaic acid or knockdown of PP2A abrogated these effects. Consistent with the mouse data, we observed loss of TFF1 and an increase in nuclear localisation of ß-catenin in stages of human gastric tumorigenesis. CONCLUSIONS: Our data indicate that loss of TFF1 promotes ß-catenin activation and gastric tumorigenesis through regulation of PP2A, a major regulator of AKT-GSK3ß signalling.


Assuntos
Inibidores do Crescimento/fisiologia , Peptídeos/fisiologia , Proteína Fosfatase 2/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Neoplasias Gástricas/patologia , Neoplasias Gástricas/fisiopatologia , beta Catenina/fisiologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo/fisiologia , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Ativação Transcricional/fisiologia , Fator Trefoil-1
3.
Curr Alzheimer Res ; 10(7): 706-13, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23905992

RESUMO

Sequential cleavages of APP by ß-secretase and γ-secretase release ß-amyloid (Aß) and one secreted form of APP (sAPP-ß) in Alzheimer' s disease (AD). Alternatively, in non-pathological situations, APP is predominantly cleaved by α-secretase within the amyloid sequence, to release the other soluble form of APP, sAPP-α. However, the functions of the two types of sAPP are still unclear. We performed this study to compare the function of sAPP-α and sAPP-ß in differentiation of the glioma cell line U251. We found that sAPP-α suppressed astrocytic differentiation and promoted neuronal differentiation in U251 cells. Additionally, sAPP-α enhanced U251 terminal differentiation into a cholinergic-like neuronal phenotype. In contrast, sAPP-ß suppressed neuronal differentiation and promoted the astrocytic differentiation of U251 cells. These findings could not only enrich the knowledge of the potential physiological function of sAPP-α and sAPP-ß, but also indicate that they may be connected to the pathological mechanism of AD. Furthermore, these findings suggest that new strategies, such as increasing the level of sAPP-α and/or decreasing the level of sAPP-ß in brain, or transplanting stem cells with increased sAPP-α and/or decreased sAPP-ß, may have potential value for AD treatment.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Astrócitos/fisiologia , Diferenciação Celular/fisiologia , Neurônios/fisiologia , Precursor de Proteína beta-Amiloide/metabolismo , Astrócitos/metabolismo , Astrócitos/patologia , Linhagem Celular Tumoral , Glioma/química , Glioma/patologia , Inibidores do Crescimento/fisiologia , Humanos , Neurônios/metabolismo , Neurônios/patologia
4.
J Neurosci ; 33(28): 11479-93, 2013 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-23843519

RESUMO

Toll-like receptors (TLRs) recognize both pathogen- and danger-associated molecular patterns and induce innate immune responses. Some TLRs are expressed in neurons and regulate neurodevelopment and neurodegeneration. However, the downstream signaling pathways and effectors for TLRs in neurons are still controversial. In this report, we provide evidence that TLR7 negatively regulates dendrite growth through the canonical myeloid differentiation primary response gene 88 (Myd88)-c-Fos-interleukin (IL)-6 pathway. Although both TLR7 and TLR8 recognize single-stranded RNA (ssRNA), the results of quantitative reverse transcription-PCR suggested that TLR7 is the major TLR recognizing ssRNA in brains. In both in vitro cultures and in utero electroporation experiments, manipulation of TLR7 expression levels was sufficient to alter neuronal morphology, indicating the presence of intrinsic TLR7 ligands. Besides, the RNase A treatment that removed ssRNA in cultures promoted dendrite growth. We also found that the addition of ssRNA and synthetic TLR7 agonists CL075 and loxoribine, but not R837 (imiquimod), to cultured neurons specifically restricted dendrite growth via TLR7. These results all suggest that TLR7 negatively regulates neuronal differentiation. In cultured neurons, TLR7 activation induced IL-6 and TNF-α expression through Myd88. Using Myd88-, IL-6-, and TNF-α-deficient neurons, we then demonstrated the essential roles of Myd88 and IL-6, but not TNF-α, in the TLR7 pathway to restrict dendrite growth. In addition to neuronal morphology, TLR7 knockout also affects mouse behaviors, because young mutant mice ∼2 weeks of age exhibited noticeably lower exploratory activity in an open field. In conclusion, our study suggests that TLR7 negatively regulates dendrite growth and influences cognition in mice.


Assuntos
Dendritos/fisiologia , Regulação para Baixo/fisiologia , Inibidores do Crescimento/fisiologia , Interleucina-6/fisiologia , Glicoproteínas de Membrana/fisiologia , Fator 88 de Diferenciação Mieloide/fisiologia , Proteínas Proto-Oncogênicas c-fos/fisiologia , Transdução de Sinais/fisiologia , Receptor 7 Toll-Like/fisiologia , Animais , Células Cultivadas , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez
5.
Anticancer Drugs ; 24(7): 667-76, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23695011

RESUMO

Malignant melanoma is the most aggressive form of skin cancer. Although various antimelanoma approaches have been used in the clinics to treat the disease over the last three decades, none of the drugs significantly prolonged the survival of metastatic melanoma patients; hence, effective drugs against metastatic melanoma are highly desired. In this study, we explored an antimetastatic melanoma agent derived from traditional Chinese medicinal herbs and found that jatrorrhizine hydrochloride (JH), an active component of the traditional Chinese medicinal herb Coptis chinensis, inhibited the proliferation and neovascularization of C8161 human metastatic melanoma cells. JH suppressed C8161 cell proliferation in a dose-dependent manner, with a half-maximal inhibitory concentration of 47.4±1.6 µmol/l; however, it did not induce significant cellular apoptosis at doses up to 320 µmol/l. Mechanistic studies showed that JH-induced C8161 cell cycle arrest at the G0/G1 transition, which was accompanied by overexpression of the cell cycle-suppressive genes p21 and p27 at higher doses. Moreover, JH reduced C8161 cell-mediated neovascularization in vitro and in vivo and impeded the expression of the gene for VE-cadherin, a key protein in tumor vasculogenic mimicry and angiogenesis. Taken together, the effective inhibitory effects of JH on metastatic melanoma cell proliferation and neovascularization with low toxicity suggest that JH is a potential new antimelanoma drug candidate.


Assuntos
Berberina/análogos & derivados , Inibidores do Crescimento/fisiologia , Melanoma/tratamento farmacológico , Melanoma/prevenção & controle , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Animais , Berberina/farmacologia , Berberina/uso terapêutico , Linhagem Celular Tumoral , Inibidores do Crescimento/farmacologia , Humanos , Melanoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neovascularização Patológica/patologia
6.
PLoS One ; 8(1): e52143, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23320069

RESUMO

BACKGROUND: Recent studies show the importance of interactions between CD47 expressed on acute myeloid leukemia (AML) cells and the inhibitory immunoreceptor, signal regulatory protein-alpha (SIRPα) on macrophages. Although AML cells express SIRPα, its function has not been investigated in these cells. In this study we aimed to determine the role of the SIRPα in acute myeloid leukemia. DESIGN AND METHODS: We analyzed the expression of SIRPα, both on mRNA and protein level in AML patients and we further investigated whether the expression of SIRPα on two low SIRPα expressing AML cell lines could be upregulated upon differentiation of the cells. We determined the effect of chimeric SIRPα expression on tumor cell growth and programmed cell death by its triggering with an agonistic antibody in these cells. Moreover, we examined the efficacy of agonistic antibody in combination with established antileukemic drugs. RESULTS: By microarray analysis of an extensive cohort of primary AML samples, we demonstrated that SIRPα is differentially expressed in AML subgroups and its expression level is dependent on differentiation stage, with high levels in FAB M4/M5 AML and low levels in FAB M0-M3. Interestingly, AML patients with high SIRPα expression had a poor prognosis. Our results also showed that SIRPα is upregulated upon differentiation of NB4 and Kasumi cells. In addition, triggering of SIRPα with an agonistic antibody in the cells stably expressing chimeric SIRPα, led to inhibition of growth and induction of programmed cell death. Finally, the SIRPα-derived signaling synergized with the activity of established antileukemic drugs. CONCLUSIONS: Our data indicate that triggering of SIRPα has antileukemic effect and may function as a potential therapeutic target in AML.


Assuntos
Antígenos de Diferenciação/metabolismo , Apoptose , Inibidores do Crescimento/fisiologia , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Receptores Imunológicos/metabolismo , Adulto , Anticorpos Monoclonais/administração & dosagem , Antígenos de Diferenciação/genética , Antineoplásicos/administração & dosagem , Apoptose/genética , Diferenciação Celular/genética , Linhagem Celular Tumoral , Criança , Regulação Neoplásica da Expressão Gênica , Humanos , Leucemia Mieloide Aguda/terapia , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patologia , Leucemia Promielocítica Aguda/terapia , Terapia de Alvo Molecular , Prognóstico , Receptores Imunológicos/genética , Transdução de Sinais/genética
7.
Toxicol Appl Pharmacol ; 267(3): 228-37, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23352504

RESUMO

Accumulated evidence has revealed a tight link between arsenic trioxide (ATO)-induced apoptosis and mitotic arrest in cancer cells. AKT, a serine/threonine kinase frequently over-activated in diverse tumors, plays critical roles in stimulating cell cycle progression, abrogating cell cycle checkpoints, suppressing apoptosis, and regulating mitotic spindle assembly. Inhibition of AKT may therefore enhance ATO cytotoxicity and thus its clinical utility. We show that AKT was activated by ATO in HeLa-S3 cells. Inhibition of AKT by inhibitors of the phosphatidyl inositol 3-kinase/AKT pathway significantly enhanced cell sensitivity to ATO by elevating mitotic cell apoptosis. Ectopic expression of the constitutively active AKT1 had no effect on ATO-induced spindle abnormalities but reduced kinetochore localization of BUBR1 and MAD2 and accelerated mitosis exit, prevented mitotic cell apoptosis, and enhanced the formation of micro- or multi-nuclei in ATO-treated cells. These results indicate that AKT1 activation may prevent apoptosis of ATO-arrested mitotic cells by attenuating the function of the spindle checkpoint and therefore allowing the formation of micro- or multi-nuclei in surviving daughter cells. In addition, AKT1 activation upregulated the expression of aurora kinase B (AURKB) and survivin, and depletion of AURKB or survivin reversed the resistance of AKT1-activated cells to ATO-induced apoptosis. Thus, AKT1 activation suppresses ATO-induced mitotic cell apoptosis, despite the presence of numerous spindle abnormalities, probably by upregulating AURKB and survivin and attenuating spindle checkpoint function. Inhibition of AKT therefore effectively sensitizes cancer cells to ATO by enhancing mitotic cell apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Mitose/efeitos dos fármacos , Óxidos/toxicidade , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/fisiologia , Regulação para Cima/efeitos dos fármacos , Apoptose/fisiologia , Trióxido de Arsênio , Arsenicais , Inibidores do Crescimento/antagonistas & inibidores , Inibidores do Crescimento/fisiologia , Inibidores do Crescimento/toxicidade , Células HeLa , Humanos , Mitose/fisiologia , Regulação para Cima/fisiologia
8.
J Immunol ; 190(4): 1447-56, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23296704

RESUMO

Nucleic acid-reactive B cells frequently arise in the bone marrow but are tolerized by mechanisms including receptor editing, functional anergy, and/or deletion. TLR9, a sensor of endosomal dsDNA, both promotes and regulates systemic autoimmunity in vivo, but the precise nature of its apparently contradictory roles in autoimmunity remained unclear. In this study, using the 3H9 anti-DNA BCR transgene in the autoimmune-prone MRL.Fas(lpr) mouse model of systemic lupus erythematosus, we identify the stages at which TLR9 contributes to establishing and breaking B cell tolerance. Although TLR9 is dispensable for L chain editing during B cell development in the bone marrow, TLR9 limits anti-DNA B cell life span in the periphery and is thus tolerogenic. In the absence of TLR9, anti-DNA B cells have much longer life spans and accumulate in the follicle, neither activated nor deleted. These cells retain some characteristics of anergic cells, in that they have elevated basal BCR signaling but impaired induced responses and downregulate their cell-surface BCR expression. In contrast, whereas TLR9-intact anergic B cells accumulate near the T/B border, TLR9-deficient anti-DNA B cells are somewhat more dispersed throughout the follicle. Nonetheless, in older autoimmune-prone animals, TLR9 expression specifically within the B cell compartment is required for spontaneous peripheral activation of anti-DNA B cells and their differentiation into Ab-forming cells via an extrafollicular pathway. Thus, TLR9 has paradoxical roles in regulating anti-DNA B cells: it helps purge the peripheral repertoire of autoreactive cells, yet is also required for their activation.


Assuntos
Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Anergia Clonal/imunologia , DNA/imunologia , Inibidores do Crescimento/fisiologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Receptor Toll-Like 9/fisiologia , Animais , Subpopulações de Linfócitos B/citologia , Anergia Clonal/genética , Inibidores do Crescimento/genética , Humanos , Camundongos , Camundongos Endogâmicos MRL lpr , Camundongos Knockout , Camundongos Transgênicos , Quimera por Radiação/imunologia , Receptor Toll-Like 9/genética
9.
Development ; 140(1): 66-75, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23222438

RESUMO

There is a gradient of ß-catenin expression along the colonic crypt axis with the highest levels at the crypt bottom. In addition, colorectal cancers show a heterogeneous subcellular pattern of ß-catenin accumulation. However, it remains unclear whether different levels of Wnt signalling exert distinct roles in the colonic epithelium. Here, we investigated the dose-dependent effect of canonical Wnt activation on colonic epithelial differentiation by controlling the expression levels of stabilised ß-catenin using a doxycycline-inducible transgenic system in mice. We show that elevated levels of Wnt signalling induce the amplification of Lgr5+ cells, which is accompanied by crypt fission and a reduction in cell proliferation among progenitor cells. By contrast, lower levels of ß-catenin induction enhance cell proliferation rates of epithelial progenitors without affecting crypt fission rates. Notably, slow-cycling cells produced by ß-catenin activation exhibit activation of Notch signalling. Consistent with the interpretation that the combination of Notch and Wnt signalling maintains crypt cells in a low proliferative state, the treatment of ß-catenin-expressing mice with a Notch inhibitor turned such slow-cycling cells into actively proliferating cells. Our results indicate that the activation of the canonical Wnt signalling pathway is sufficient for de novo crypt formation, and suggest that different levels of canonical Wnt activations, in cooperation with Notch signalling, establish a hierarchy of slower-cycling stem cells and faster-cycling progenitor cells characteristic for the colonic epithelium.


Assuntos
Ciclo Celular/fisiologia , Colo/citologia , Mucosa Intestinal/citologia , Transdução de Sinais/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Linhagem Celular , Proliferação de Células , Colo/patologia , Colo/fisiologia , Técnicas de Introdução de Genes , Inibidores do Crescimento/fisiologia , Mucosa Intestinal/patologia , Mucosa Intestinal/fisiologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Animais , Células-Tronco/citologia , Células-Tronco/metabolismo , beta Catenina/biossíntese , beta Catenina/fisiologia
10.
J Immunol ; 190(1): 159-67, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23225884

RESUMO

MEK1 phosphorylates ERK1/2 and regulates T cell generation, differentiation, and function. MEK1 has recently been shown to translocate to the nucleus. Its nuclear function is largely unknown. By studying human CD4 T cells, we demonstrate that a low level of MEK1 is present in the nucleus of CD4 T cells under basal conditions. T cell activation further increases the nuclear translocation of MEK1. MEK1 interacts with the nuclear receptor corepressor silencing mediator of retinoid and thyroid hormone receptor (SMRT). MEK1 reduces the nuclear level of SMRT in an activation-dependent manner. MEK1 is recruited to the promoter of c-Fos upon TCR stimulation. Conversely, SMRT is bound to the c-Fos promoter under basal conditions and is removed upon TCR stimulation. We examined the role of SMRT in regulation of T cell function. Small interfering RNA-mediated knockdown of SMRT results in a biphasic effect on cytokine production. The production of the cytokines IL-2, IL-4, IL-10, and IFN-γ increases in the early phase (8 h) and then decreases in the late phase (48 h). The late-phase decrease is associated with inhibition of T cell proliferation. The late-phase inhibition of T cell activation is, in part, mediated by IL-10 that is produced in the early phase and, in part, by ß-catenin signaling. Thus, we have identified a novel nuclear function of MEK1. MEK1 triggers a complex pattern of early T cell activation, followed by a late inhibition through its interaction with SMRT. This biphasic dual effect most likely reflects a homeostatic regulation of T cell function by MEK1.


Assuntos
Transporte Ativo do Núcleo Celular/imunologia , Linfócitos T CD4-Positivos/imunologia , MAP Quinase Quinase 1/fisiologia , Correpressor 1 de Receptor Nuclear/fisiologia , Correpressor 2 de Receptor Nuclear/antagonistas & inibidores , Correpressor 2 de Receptor Nuclear/fisiologia , Transporte Ativo do Núcleo Celular/genética , Linfócitos T CD4-Positivos/enzimologia , Linfócitos T CD4-Positivos/metabolismo , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Inativação Gênica/imunologia , Inibidores do Crescimento/genética , Inibidores do Crescimento/metabolismo , Inibidores do Crescimento/fisiologia , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 1/metabolismo , Correpressor 2 de Receptor Nuclear/genética , Regiões Promotoras Genéticas/imunologia , Ligação Proteica/genética , Ligação Proteica/imunologia , Proteínas Proto-Oncogênicas c-fos/metabolismo
11.
J Immunol ; 190(1): 138-46, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23225885

RESUMO

Adenosine is a key endogenous signaling molecule that regulates immune responses. A(2B) adenosine receptor (AR) is a relatively low-affinity receptor for adenosine, and the activation of A(2B)AR is believed to require pathological level of adenosine that is associated with ischemia, inflammation, trauma, or other types of stress. The role of A(2B)AR in the pathogenesis of multiple sclerosis (MS) is still unclear. In this study, we discovered that A(2B)AR was upregulated both in the peripheral blood leukocytes of MS patients and the peripheral lymphoid tissues of experimental autoimmune encephalomyelitis (EAE) mice. A(2B)AR-specific antagonists, CVT-6883 and MRS-1754, alleviated the clinical symptoms of EAE and protected the CNS from immune damage. A(2B)AR-knockout mice also developed less severe EAE. Further study indicated that blocking or deleting A(2B)AR inhibited Th17 cell differentiation by blocking IL-6 production from APCs such as dendritic cells. In dendritic cells, A(2B)AR was also upregulated during the development of EAE. CVT-6883 and genetic deletion of A(2B)AR significantly reduced adenosine-mediated IL-6 production. The phospholipase Cß-protein kinase C and p38 MAPK pathways were found to be involved in the A(2B)AR-mediated IL-6 production. Our findings not only revealed the pathological role of A(2B)AR in EAE, but also suggested that this receptor might be a new therapeutic target for the development of anti-MS drugs.


Assuntos
Diferenciação Celular/imunologia , Encefalomielite Autoimune Experimental/imunologia , Interleucina-6/antagonistas & inibidores , Receptor A2B de Adenosina/metabolismo , Células Th17/imunologia , Adulto , Animais , Células Cultivadas , Técnicas de Cocultura , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/prevenção & controle , Feminino , Inibidores do Crescimento/antagonistas & inibidores , Inibidores do Crescimento/fisiologia , Humanos , Interleucina-6/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Receptor A2B de Adenosina/deficiência , Células Th17/metabolismo , Células Th17/patologia , Regulação para Cima/imunologia
12.
J Immunol ; 190(1): 21-6, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23225888

RESUMO

Signaling lymphocytic activation molecule family receptors and the specific adapter signaling lymphocytic activation molecule-associated protein modulate the development of innate-like lymphocytes. In this study, we show that the thymus of Ly9-deficient mice contains an expanded population of CD8 single-positive cells with the characteristic phenotype of innate memory-like CD8(+) T cells. Moreover, the proportion of these innate CD8(+) T cells increased dramatically postinfection with mouse CMV. Gene expression profiling of Ly9-deficient mice thymi showed a significant upregulation of IL-4 and promyelocytic leukemia zinc finger. Analyses of Ly9(-/-)IL4ra(-/-) double-deficient mice revealed that IL-4 was needed to generate the thymic innate CD8(+) T cell subset. Furthermore, increased numbers of invariant NKT cells were detected in Ly9-deficient thymi. In wild-type mice, IL-4 levels induced by α-galactosylceramide injection could be inhibited by a mAb against Ly9. Thus, Ly9 plays a unique role as an inhibitory cell surface receptor regulating the size of the thymic innate CD8(+) T cell pool and the development of invariant NKT cells.


Assuntos
Antígenos CD/fisiologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Regulação para Baixo/imunologia , Memória Imunológica , Células T Matadoras Naturais/imunologia , Receptores de Superfície Celular/fisiologia , Timo/imunologia , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/genética , Regulação para Baixo/genética , Feminino , Inibidores do Crescimento/genética , Inibidores do Crescimento/fisiologia , Imunidade Inata/genética , Memória Imunológica/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células T Matadoras Naturais/citologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Família de Moléculas de Sinalização da Ativação Linfocitária , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Timo/citologia
13.
Invest Ophthalmol Vis Sci ; 53(13): 8433-46, 2012 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-23139280

RESUMO

PURPOSE: The secreted Ly6/uPAR-related protein-1 (Slurp1), associated with the hyperkeratotic disorder mal de Meleda, is abundantly expressed in corneas. Here, we examine its corneal expression and functions. METHODS: Gene expression was quantified by quantitative PCR (qPCR), immunoblots, and immunofluorescent staining. Effect of Kruppel-like factor 4 (Klf4) on Slurp1 promoter was evaluated by chromatin immunoprecipitation (ChIP) and transient transfections. Adenoviral vectors were used to express Slurp1 in corneas. Leukocytic infiltration in bacterial lipopolysaccharide (LPS)-, herpes simplex virus type 1 (HSV-1)-, or adenovirus (serotype 5)-treated mouse corneas was characterized by flow cytometry. RESULTS: Corneal expression of Slurp1 increased sharply upon mouse eyelid opening, concurrent with the elevated expression of Klf4. Slurp1 was significantly decreased in Klf4 conditional null (Klf4CN) corneas that displayed elevated expression of cytokines and cytokine receptors, as well as neutrophil influx consistent with a proinflammatory environment. In additional models of corneal inflammation, Slurp1 expression was abrogated within 24 hours of LPS injection or HSV-1 or adenoviral infection, accompanied by a predominantly neutrophilic infiltrate. Neutrophilic infiltration was enhanced in HSV-1-infected Klf4CN corneas lacking Slurp1. SLURP1 promoter activity was stimulated by KLF4, suppressed by IL-4, IL-13, and TNFα, and unperturbed by IFN-γ. Slurp1 downregulation and neutrophil influx were comparable in HSV-1-infected wild-type (WT) and Ifng-/- mouse corneas. Mouse corneas infected with Slurp1-expressing adenoviral vectors displayed reduced signs of inflammation and restricted neutrophilic infiltration compared with those infected with control vectors. CONCLUSIONS: Klf4 regulates the expression of Slurp1, a key immunomodulatory peptide that is abundantly expressed in healthy corneas and is downregulated in proinflammatory conditions.


Assuntos
Antígenos Ly/genética , Córnea/metabolismo , Regulação da Expressão Gênica/fisiologia , Inibidores do Crescimento/fisiologia , Fatores de Transcrição Kruppel-Like/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/genética , Adenoviridae/genética , Animais , Antígenos Ly/metabolismo , Sequência de Bases , Imunoprecipitação da Cromatina , Citocinas/farmacologia , Feminino , Citometria de Fluxo , Técnica Indireta de Fluorescência para Anticorpo , Vetores Genéticos , Herpesvirus Humano 1/fisiologia , Doenças do Sistema Imunitário , Immunoblotting , Fatores Imunológicos/genética , Fatores Imunológicos/metabolismo , Ceratite Herpética/genética , Ceratite Herpética/metabolismo , Fator 4 Semelhante a Kruppel , Transtornos Leucocíticos , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Transfecção , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
14.
J Immunol ; 189(9): 4379-86, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23002443

RESUMO

B lymphopoiesis declines with age, and in rabbits this occurs by 8 wk of age. We found that CFU fibroblasts (CFU-Fs) in the bone marrow (BM) decrease 10-fold by a few weeks of age and that the CFU-Fs preferentially differentiate into adipocytes instead of osteoblasts. BM becomes filled with fat spaces during this time, making rabbit a unique model to study the effects of accelerated fat accumulation on B lymphopoiesis. We show that adipocytes of both rabbit and human secrete a soluble factor(s) that inhibits B lymphopoiesis, and we tested if this inhibition was due to effects on the BM stroma or hematopoietic progenitors. Pretreatment of BM mononuclear cells with adipocyte conditioned medium dramatically inhibited their differentiation into proB cells in cocultures with OP9 stromal cells. In contrast, pretreatment of OP9 stromal cells with adipocyte conditioned medium had no effect on B lymphopoiesis. Using human hematopoietic stem cells, we show that inhibition by the adipocyte-derived factor occurred at the common lymphoid progenitor to preproB cell stage. We propose that the age-related decline in B lymphopoiesis is due to a decrease in CFU-Fs, an increase in adipocytes, and an adipocyte-derived factor that blocks B lymphopoiesis at the common lymphoid progenitor to preproB cell stage.


Assuntos
Adipócitos/fisiologia , Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/imunologia , Proteínas de Ligação a Ácido Graxo/fisiologia , Inibidores do Crescimento/fisiologia , Linfopoese/imunologia , Animais , Subpopulações de Linfócitos B/metabolismo , Linhagem Celular , Fibroblastos/citologia , Fibroblastos/imunologia , Fibroblastos/metabolismo , Humanos , Osteoblastos/citologia , Osteoblastos/imunologia , Osteoblastos/metabolismo , Coelhos , Células-Tronco/citologia , Células-Tronco/imunologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia
15.
J Immunol ; 189(3): 1118-22, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22753931

RESUMO

The CTLA-4 pathway is a key regulator of T cell activation and a critical failsafe against autoimmunity. Although early models postulated that CTLA-4 transduced a negative signal, in vivo evidence suggests that CTLA-4 functions in a cell-extrinsic manner. That multiple cell-intrinsic mechanisms have been attributed to CTLA-4, yet its function in vivo appears to be cell-extrinsic, has been an ongoing paradox in the field. Although CTLA-4 expressed on conventional T cells (Tconv) can mediate inhibitory function, it is unclear why this fails to manifest as an intrinsic effect. In this study, we show that Tconv-expressed CTLA-4 can function in a cell-extrinsic manner in vivo. CTLA-4(+/+) T cells, from DO11/rag(-/-) mice that lack regulatory T cells, were able to regulate the response of CTLA-4(-/-) T cells in cotransfer experiments. This observation provides a potential resolution to the above paradox and suggests CTLA-4 function on both Tconv and regulatory T cells can be achieved through cell-extrinsic mechanisms.


Assuntos
Antígeno CTLA-4/fisiologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Transferência Adotiva , Animais , Transplante de Medula Óssea/imunologia , Antígeno CTLA-4/deficiência , Antígeno CTLA-4/genética , Inibidores do Crescimento/deficiência , Inibidores do Crescimento/genética , Inibidores do Crescimento/fisiologia , Tolerância Imunológica/genética , Imunidade Celular/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Quimera por Radiação/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
16.
Mol Endocrinol ; 26(8): 1252-67, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22745190

RESUMO

Androgen receptor (AR) signaling exerts an antiestrogenic, growth-inhibitory influence in normal breast tissue, and this role may be sustained in estrogen receptor α (ERα)-positive luminal breast cancers. Conversely, AR signaling may promote growth of a subset of ERα-negative, AR-positive breast cancers with a molecular apocrine phenotype. Understanding the molecular mechanisms whereby androgens can elicit distinct gene expression programs and opposing proliferative responses in these two breast cancer phenotypes is critical to the development of new therapeutic strategies to target the AR in breast cancer.


Assuntos
Androgênios/fisiologia , Neoplasias da Mama/metabolismo , Inibidores do Crescimento/fisiologia , Glândulas Mamárias Humanas/metabolismo , Receptores Androgênicos/fisiologia , Animais , Neoplasias da Mama/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Genes Supressores de Tumor , Humanos , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Oncogenes , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Transdução de Sinais
17.
Arch Pharm Res ; 35(5): 867-76, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22644854

RESUMO

Snake venom toxin from Vipera lebetina turanica induces apoptosis in many cancer cell lines, but there is no study about the apoptotic effect of snake venom toxin on human ovarian cancer cells. In this study, we investigated the apoptotic effect of snake venom toxin in human ovarian cancer PA-1 and SK-OV3 cells. Snake venom toxin dose dependently (0∼10 µg/mL) inhibited ovarian cancer cell growth with IC(50) values 4.5 µg/mL in PA-1 cells, and 6.5 µg/mL in SK-OV3 cells. Our results also showed that apoptotic cell death increased by snake venom toxin in a dose dependent manner (0∼10 µg/mL). Consistent with increased cell death, snake venom toxin increased the expression of pro-apoptotic protein Bax and caspase-3, but down-regulated anti-apoptotic protein Bcl-2. Untreated ovarian cancer cells showed a high DNA binding activity of nuclear factor B (NF-κB), but it was inhibited by snake venom toxin accompanied by inhibition of p50 and p65 translocation into the nucleus as well as phosphorylation of inhibitory κB. Snake venom toxin also inhibited DNA binding activity of the signal transducer and activator of transcription 3 (STAT3). Moreover, the combination treatment of NF-κB (salicylic acid, 1 or 5 µM) and STAT3 (stattic, 1 µM) with snake venom toxin (1 µg/mL) further enhanced cell growth inhibitory effects of snake venom toxin. These results showed that snake venom toxin from Vipera lebetina turanica caused apoptotic cell death of ovarian cancer cells through the inhibition of NF-κB and STAT3 signal, and suggested that snake venom toxin may be applicable as an anticancer agent for ovarian cancer.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores do Crescimento/fisiologia , NF-kappa B/antagonistas & inibidores , Neurotoxinas/farmacologia , Neoplasias Ovarianas/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Venenos de Víboras/farmacologia , Animais , Apoptose/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Feminino , Inibidores do Crescimento/isolamento & purificação , Inibidores do Crescimento/uso terapêutico , Humanos , NF-kappa B/metabolismo , Neurotoxinas/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Fator de Transcrição STAT3/metabolismo , Venenos de Víboras/isolamento & purificação , Venenos de Víboras/uso terapêutico
18.
Cytokine Growth Factor Rev ; 23(3): 119-25, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22579070

RESUMO

Prostate cancer (PCa), the most common non-skin cancer in men, is a worldwide health concern. Treatment options for aggressive PCa are limited to androgen deprivation therapies (ADT), which are ineffective, with robust diagnostic options also being limited. The prostate specific antigen (PSA) test, for instance, is subject to high levels of false positive results and cannot distinguish between cancer confined to the prostate and aggressive metastatic cancer. As such, additional therapeutic and diagnostic options are urgently required. In recent years, a clear association between activins and prostate cancer has become evident. Activins are members of the TGF-ß superfamily and are responsible for a plethora of physiological processes, including cell proliferation, apoptosis, immune surveillance, embryonic development, and follicle stimulating hormone (FSH) regulation. Activin A normally inhibits cancer development and progression, however, cancer cell growth in high-grade PCa is not inhibited by this protein. The mechanism for this apparent acquired capability to resist activin A-mediated growth inhibition is currently not well understood. Thus, the aim of this review is to analyse the role of activin A in PCa progression and to present mechanisms by which transformed cells may escape its effects. The overarching hypothesis is that insensitivity to the growth inhibitory effects of activin A is an acquired capability in PCa progression. Therefore, local and genetic elements that may be responsible for this change in cellular sensitivity to activin A during cancer progression will be highlighted with a view to identifying potential diagnostic or therapeutic targets.


Assuntos
Ativinas/fisiologia , Neoplasias da Próstata/fisiopatologia , Ativinas/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Progressão da Doença , Inibidores do Crescimento/fisiologia , Humanos , Masculino , MicroRNAs/metabolismo , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/terapia
19.
Pharm Biol ; 50(7): 839-49, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22486657

RESUMO

CONTEXT: Estrogen is well-known as an important factor in the physiological functions and pathological processes of breast. Estrogen receptor ß (ERß) is expressed in the majority of breast cancers at lower levels compared with the normal breast tissue. OBJECTIVE: The effect of ERß on the characteristics of breast tumor cells and its prognostication for the use of ERß-selective therapy were investigated here for the first time. MATERIALS AND METHODS: ERß was overexpressed in ERα positive MCF-7 breast cancer cells by gene transfection. The proliferation, motility, and xenografts growth of MCF-7 cells were investigated by MTT assays, wound-healing assay and animal study. RESULTS: Results demonstrated that ERß-GFP localized in both the cytoplasm and the nucleus in the presence of 17ß-estradiol (E2), with stronger fluorescence-signal intensity in the nucleus, 2.8-times higher than that in the cytoplasm. The ERß overexpressed MCF-7 cells resulted in a 38.7% decreased growth rate and motility in vitro. Furthermore, ERß overexpression enhanced the antiproliferative effects of phytoestrogen, antiestrogen, and histone deacetylase inhibitor. Exogenous ERß expression reduced tumor volume by 99% at 27 days postadministration, indicated that overexpression of ERß led to retardation of tumor formation and growth in immunodeficient mice. DISCUSSION AND CONCLUSION: This study provided a relatively new evidence to support that ERß is an important modulator of proliferation and motility of breast cancer cells, and implied for the first time a possibility for the use of novel ERß-selective therapies in breast cancer treatment.


Assuntos
Neoplasias da Mama/metabolismo , Proliferação de Células , Receptor beta de Estrogênio/fisiologia , Inibidores do Crescimento/fisiologia , Animais , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Receptor beta de Estrogênio/administração & dosagem , Receptor beta de Estrogênio/uso terapêutico , Medicina Baseada em Evidências/métodos , Feminino , Inibidores do Crescimento/administração & dosagem , Inibidores do Crescimento/uso terapêutico , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica/patologia , Invasividade Neoplásica/prevenção & controle , Prognóstico , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
20.
J Immunol ; 188(11): 5337-47, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22539794

RESUMO

Protein kinase C (PKC)-θ has been shown to be a critical TCR signaling molecule that promotes the activation and differentiation of naive T cells into inflammatory effector T cells. In this study, we demonstrate that PKC-θ-mediated signals inhibit inducible regulatory T cell (iTreg) differentiation via an AKT-Foxo1/3A pathway. TGF-ß-induced iTreg differentiation was enhanced in PKC-θ(-/-) T cells or wild-type cells treated with a specific PKC-θ inhibitor, but was inhibited by the PKC-θ activator PMA, or by CD28 crosslinking, which enhances PKC-θ activation. PKC-θ(-/-) T cells had reduced activity of the AKT kinase, and the expression of a constitutively active form of AKT in PKC-θ(-/-) T cells restored the ability to inhibit iTreg differentiation. Furthermore, knockdown or overexpression of the AKT downstream targets Foxo1 and Foxo3a was found to inhibit or promote iTreg differentiation in PKC-θ(-/-) T cells accordingly, indicating that the AKT-Foxo1/3A pathway is responsible for the inhibition of iTreg differentiation of iTregs downstream of PKC-θ. We conclude that PKC-θ is able to control T cell-mediated immune responses by shifting the balance between the differentiation of effector T cells and inhibitory Tregs.


Assuntos
Fatores de Transcrição Forkhead/fisiologia , Inibidores do Crescimento/fisiologia , Isoenzimas/fisiologia , Ativação Linfocitária/imunologia , Proteína Quinase C/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Animais , Comunicação Celular/genética , Comunicação Celular/imunologia , Células Cultivadas , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Inibidores do Crescimento/deficiência , Isoenzimas/deficiência , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteína Quinase C/deficiência , Proteína Quinase C-theta , Transdução de Sinais/genética , Linfócitos T Reguladores/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA