Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Breast Cancer Res ; 25(1): 144, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37968653

RESUMO

BACKGROUND: Breast cancer stem cells (BCSCs) are resistant to standard therapies, facilitate tumor dissemination, and contribute to relapse and progression. Super-enhancers are regulators of stemness, and BET proteins, which are critical for super-enhancer function, are a potential therapeutic target. Here, we investigated the effects of BET proteins on the regulation of breast cancer stemness using the pan-BET degrader ZBC260. METHODS: We evaluated the effect of ZBC260 on CSCs in TNBC cell lines. We assessed the effect of ZBC260 on cellular viability and tumor growth and measured its effects on cancer stemness. We used RNA sequencing and stemness index to determine the global transcriptomic changes in CSCs and bulk cells and further validated our findings by qPCR, western blot, and ELISA. RESULTS: ZBC260 potently inhibited TNBC growth both in vitro and in vivo. ZBC260 reduced stemness as measured by cell surface marker expression, ALDH activity, tumorsphere number, and stemness index while increasing differentiated cells. GSEA analysis indicated preferential downregulation of stemness-associated and inflammatory genes by ZBC260 in ALDH+ CSCs. CONCLUSIONS: The BET degrader ZBC260 is an efficient degrader of BET proteins that suppresses tumor progression and decreases CSCs through the downregulation of inflammatory genes and pathways. Our findings support the further development of BET degraders alone and in combination with other therapeutics as CSC targeting agents.


Assuntos
Neoplasias da Mama , Neoplasias de Mama Triplo Negativas , Humanos , Feminino , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Recidiva Local de Neoplasia/patologia , Proteínas/metabolismo , Proteínas/farmacologia , Proteínas/uso terapêutico , Transformação Celular Neoplásica/metabolismo , Diferenciação Celular/genética , Células-Tronco Neoplásicas/patologia
3.
EBioMedicine ; 41: 320-332, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30745168

RESUMO

BACKGROUND: Hyper-activation of TGF-ß signaling is critically involved in progression of hepatocellular carcinoma (HCC). However, the events that contribute to the dysregulation of TGF-ß pathway in HCC, especially at the post-translational level, are not well understood. METHODS: Associations of deubiquitinase POH1 with TGF-ß signaling activity and the outcomes of HCC patients were examined by data mining of online HCC datasets, immunohistochemistry analyses using human HCC specimens, spearman correlation and survival analyses. The effects of POH1 on the ubiquitination and stability of the TGF-ß receptors (TGFBR1 and TGFBR2) and the activation of downstream effectors were tested by western blotting. Primary mouse liver tissues from polyinosinic:polycytidylic acid (poly I:C)- treated Mx-Cre+, poh1f/f mice and control mice were used to detect the TGF-ß receptors. The metastatic-related capabilities of HCC cells were studied in vitro and in mice. FINDINGS: Here we show that POH1 is a critical regulator of TGF-ß signaling and promotes tumor metastasis. Integrative analyses of HCC subgroups classified with unsupervised transcriptome clustering of the TGF-ß response, metastatic potential and outcomes, reveal that POH1 expression positively correlates with activities of TGF-ß signaling in tumors and with malignant disease progression. Functionally, POH1 intensifies TGF-ß signaling delivery and, as a consequence, promotes HCC cell metastatic properties both in vitro and in vivo. The expression of the TGF-ß receptors was severely downregulated in POH1-deficient mouse hepatocytes. Mechanistically, POH1 deubiquitinates the TGF-ß receptors and CAV1, therefore negatively regulates lysosome pathway-mediated turnover of TGF-ß receptors. CONCLUSION: Our study highlights the pathological significance of aberrantly expressed POH1 in TGF-ß signaling hyperactivation and aggressive progression in HCC.


Assuntos
Carcinoma Hepatocelular/patologia , Caveolina 1/metabolismo , Neoplasias Hepáticas/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Humanos , Estimativa de Kaplan-Meier , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Prognóstico , Complexo de Endopeptidases do Proteassoma/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Transativadores/antagonistas & inibidores , Transativadores/genética , Ubiquitinação
4.
Cell Death Differ ; 26(4): 751-762, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30038387

RESUMO

Foxp3-expressing regulatory T (Treg) cells are essential for averting autoimmune diseases and maintaining immune homeostasis. However, the molecular mechanisms underlying the development and maintenance of Treg cells are still unclear. Here, we found that T cell-specific deletion of the gene encoding the deubiquitinase POH1 compromised the development of mature T cells, especially CD4+Foxp3+ Treg cells. Moreover, POH1 deficiency significantly attenuated the transition of CD25+ Treg cell precursors into Foxp3+ Treg cells accompanied by downregulation of interleukin 2 (IL-2)-STAT5 signaling. Deletion of POH1 in generated CD4+Foxp3+ Treg cells led to an early onset of fetal autoimmune disorders and a decrease in the pool size of peripheral Treg cells in mice, which were mostly due to decreased expansion of these cells. Thus, these results revealed that POH1 has a pivotal role in the development and maintenance of CD4+Foxp3+ Treg cells and contributes to immune tolerance.


Assuntos
Autoimunidade/genética , Diferenciação Celular/genética , Fatores de Transcrição Forkhead/metabolismo , Tolerância Imunológica/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Linfócitos T Reguladores/metabolismo , Transativadores/metabolismo , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/imunologia , Proliferação de Células/genética , Fatores de Transcrição Forkhead/imunologia , Interleucina-2/metabolismo , Subunidade alfa de Receptor de Interleucina-2/imunologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexo de Endopeptidases do Proteassoma/genética , RNA-Seq , Fator de Transcrição STAT5/metabolismo , Baço/imunologia , Baço/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Timo/imunologia , Timo/metabolismo , Transativadores/genética
5.
Oncogene ; 38(13): 2405-2419, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30531833

RESUMO

Metastatic progression is the main cause of mortality in breast cancer, necessitating the determination of the molecular events driving this process for the development of new therapeutic approaches. Here, we demonstrate that hyperactivation of the deubiquitinase USP1 contributes to breast cancer metastasis. Upregulated USP1 expression in primary breast cancer specimens correlates with metastatic progression and poor prognosis in breast cancer patients. USP1 enhances the expression of a number of pro-metastatic genes in breast cancer cells, promotes cell migration and invasion in vitro, and facilitates lung metastasis of breast cancer cells. Moreover, USP1-mediated deubiquitination and stabilization of KPNA2 are revealed as the downstream events crucial for USP1-pro-metastatic function. Most importantly, pharmacological intervention of USP1 function by pimozide or ML323 significantly represses breast cancer metastasis in mice, suggesting a rationale for using USP1 inhibitors for treatment of patients with breast cancer. Taken together, our results establish USP1 as a promoter of breast cancer metastasis and provide evidence for the potential practice of USP1 targeting in the treatment of breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Metástase Neoplásica/patologia , Proteases Específicas de Ubiquitina/antagonistas & inibidores , alfa Carioferinas/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Progressão da Doença , Feminino , Células HEK293 , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica/patologia , Prognóstico , Regiões Promotoras Genéticas/fisiologia
6.
Nat Commun ; 9(1): 4225, 2018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30315153

RESUMO

Inflammasome activation is essential for host defence against invading pathogens, but is also involved in various forms of inflammatory diseases. The processes that control inflammasome activity are thus important for averting excessive immune responses and tissue damage. Here we show that the deubiquitinase POH1 negatively regulates the immune response triggered by inflammasome activation. POH1 deficiency in macrophages enhances mature IL-1ß production without significant alterations in inflammasome priming and ASC-caspase-1 activation. In WT macrophages, POH1 interacts with and deubiquitinates pro-IL-1ß by decreasing the K63-linked polyubiquitin chains, as well as decreases the efficacy of pro-IL-1ß cleavage. Furthermore, myeloid cell-specific deletion of POH1 aggravates lipopolysaccharide-induced systemic inflammation and alum-induced peritonitis inflammatory responses in vivo. Our study thereby reveals that POH1-mediated deubiquitination of pro-IL-1ß is an important regulatory event that restrains inflammatory responses for the maintenance of immune homeostasis.


Assuntos
Inflamassomos/metabolismo , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Transativadores/metabolismo , Animais , Caspase 1/genética , Caspase 1/metabolismo , Sistemas Computacionais , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Células HEK293 , Humanos , Imunoprecipitação , Inflamassomos/efeitos dos fármacos , Inflamação/induzido quimicamente , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos/genética , Poliubiquitina/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Transativadores/genética
7.
Sci Rep ; 6: 33039, 2016 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-27633997

RESUMO

Hepatitis B virus (HBV) infection is a major factor that contributes to the development of hepatocellular carcinoma (HCC). HBV X protein (HBx) has been shown to accelerate HCC progression by promoting tumour growth and metastasis. In the clinic, carboxyl-terminal truncated HBx (Ct-HBx) proteins are frequently present in HCC tumour tissues, but not in non-tumorous tissues. In this study, we analysed deubiquitinase expression profiles in cells with or without ectopic expression of the Ct-HBx proteins and observed that the expression of ubiquitin specific peptidase 16 (USP16) was substantially inhibited by Ct-HBx proteins. Liver tumour cells with forced down-regulation of USP16 exhibited increased capabilities for colony formation and tumour growth in vivo. In addition, USP16 inhibition promoted stem-like properties in tumour cells, as evidenced by their spheroid formation and chemo-responsiveness. Furthermore, ectopic expression of USP16 in tumour cells significantly abrogated the tumour promoting activities of the Ct-HBx proteins (HBxΔ35), leading to decreased tumour cell viability and tumour growth. In human HCCs, USP16 was frequently downregulated, and the decreased expression of USP16 was correlated with high tumour stages and poor differentiation status. Taken together, our study suggests that USP16 downregulation is a critical event in Ct-HBx-mediated promotion of HCC tumorigenicity and malignancy.


Assuntos
Carcinoma Hepatocelular/genética , Regulação para Baixo/genética , Vírus da Hepatite B/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Transativadores/genética , Ubiquitina Tiolesterase/genética , Animais , Carcinoma Hepatocelular/patologia , Diferenciação Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Sobrevivência Celular/genética , Transformação Celular Neoplásica/genética , Regulação Neoplásica da Expressão Gênica/genética , Células HEK293 , Células Hep G2 , Hepatite B/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Virais Reguladoras e Acessórias
8.
Nat Commun ; 6: 8704, 2015 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-26510456

RESUMO

Hyperactivation of the transcriptional factor E2F1 occurs frequently in human cancers and contributes to malignant progression. E2F1 activity is regulated by proteolysis mediated by the ubiquitin-proteasome system. However, the deubiquitylase that controls E2F1 ubiquitylation and stability remains undefined. Here we demonstrate that the deubiquitylase POH1 stabilizes E2F1 protein through binding to and deubiquitylating E2F1. Conditional knockout of Poh1 alleles results in reduced E2F1 expression in primary mouse liver cells. The POH1-mediated regulation of E2F1 expression strengthens E2F1-downstream prosurvival signals, including upregulation of Survivin and FOXM1 protein levels, and efficiently facilitates tumour growth of liver cancer cells in nude mice. Importantly, human hepatocellular carcinomas (HCCs) recapitulate POH1 regulation of E2F1 expression, as nuclear abundance of POH1 is increased in HCCs and correlates with E2F1 overexpression and tumour growth. Thus, our study suggests that the hyperactivated POH1-E2F1 regulation may contribute to the development of liver cancer.


Assuntos
Proliferação de Células , Fator de Transcrição E2F1/metabolismo , Neoplasias Hepáticas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Transativadores/metabolismo , Ubiquitina/metabolismo , Animais , Carcinogênese , Fator de Transcrição E2F1/química , Fator de Transcrição E2F1/genética , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Complexo de Endopeptidases do Proteassoma/genética , Ligação Proteica , Estabilidade Proteica , Transativadores/genética , Ubiquitinação
9.
Oncotarget ; 6(32): 33636-47, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26378039

RESUMO

Cellular senescence evasion caused by the inactivation of tumor suppressive programs is implicated in tumor initiation and therapeutic resistance. Our previous study has shown that the downregulation of growth arrest and DNA damage 45G (GADD45G) contributes to senescence bypass in hepatocellular carcinoma (HCC). Here, we report that the Smad-interacting protein-1 (SIP1) is transcriptionally activated and functions critically in the GADD45G-induced tumor cell senescence. Knockdown of SIP1 significantly abrogates the suppressive effects of GADD45G on the growth of xenografted liver tumor in vivo. The essential role of SIP1 in GADD45G activities is further validated in the model of the proteasome inhibitor MG132-induced cell senescence. We further show that JNK but not p38 MAPK activation is involved in the GADD45G-mediated SIP1 upregulation, and that JNK inhibition counteracts the GADD45G-induced cellular senescence. More importantly, we show that GADD45G and SIP1 expression are coincidently downregulated in primary human HCC tissues. Together, our results establish that the dowregulation of GADD45G-SIP1 axis may contribute to cellular senescence evasion and HCC development.


Assuntos
Carcinoma Hepatocelular/metabolismo , Proteínas de Homeodomínio/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Repressoras/metabolismo , Animais , Antineoplásicos/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Senescência Celular/efeitos dos fármacos , Senescência Celular/fisiologia , Inibidores de Cisteína Proteinase/farmacologia , Células HEK293 , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Leupeptinas/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , MAP Quinase Quinase 4/metabolismo , Camundongos , Camundongos Nus , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Homeobox 2 de Ligação a E-box com Dedos de Zinco
10.
Cancer Lett ; 355(1): 25-33, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25242357

RESUMO

Krüppel-like factor 9 (KLF9) is known to be a tumor suppressor gene in colorectal tumors and glioblastoma; however, the functional status and significance of KLF9 in hepatocellular carcinoma (HCC) is unclear. We report here that KLF9 is downregulated in HCC tissues. Restoration of KLF9 significantly inhibited growth and caused apoptosis in SK-Hep1 and HepG2 cells. We found that KLF9 positively regulated p53 levels by directly binding to GC boxes within the proximal region of the p53 promoter. Moreover, in the presence of cycloheximide, KLF9 significantly increased p53 stability in HCC cells. Remarkably, ectopic expression of KLF9 was sufficient to delay the onset of tumors and to promote regression of the established tumors in vivo, suggesting that KLF9 plays a critical role in HCC development and that pharmacological or genetic activation of KLF9 may have potential in the treatment of HCC.


Assuntos
Carcinoma Hepatocelular/metabolismo , Proliferação de Células , Fatores de Transcrição Kruppel-Like/metabolismo , Neoplasias Hepáticas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Sítios de Ligação , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Humanos , Fatores de Transcrição Kruppel-Like/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Regiões Promotoras Genéticas , Estabilidade Proteica , Transdução de Sinais , Fatores de Tempo , Transcrição Gênica , Transfecção , Proteína Supressora de Tumor p53/genética
11.
Int J Mol Med ; 34(1): 262-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24820432

RESUMO

Strategies for peroxisome proliferator-activated receptor (PPAR) activation or survivin inhibition have potential for cancer therapy. However, whether the combination of these two approaches can be developed as a rational regimen with enhanced efficiency in the inhibition of tumor cells remains to be determined. In this study, the combinatory effect of PPAR-γ agonist and survivin inhibition on bladder cancer cells was investigated. T24 and 5637 cells were treated with 15d-PGJ(2) to determine whether 15d-PGJ(2) had an inhibitory effect. Cell viability and proliferation were analyzed and efficiency of survivin siRNAs was assessed using western blot analysis. The results showed that, in the human bladder cancer cell lines T24 and 5637, the natural PPAR-γ ligand 15d-PGJ(2) significantly decreased cell proliferation and loci formation. The increase in the proportion of apoptotic cells was observed in the cells 48 h after 15d-PGJ(2) treatment. Furthermore, 15d-PGJ(2) substantially inhibited the levels of stemness-related genes in these cells. The ability of sphere formation was markedly suppressed in the cells treated with 15d-PGJ(2). More importantly, the downregulation of survivin with siRNAs significantly enhanced the 15d-PGJ(2)-mediated induction of cell apoptosis and inhibition of sphere formation. Accordingly, we also found that survivin inhibition significantly enhanced 15d-PGJ(2)-induced production of reactive oxygen species (ROS) in bladder cancer cells. Taken together, these findings suggest that the combination of 15d-PGJ(2) and survivin inhibition play a potentially role in the therapeutical manipulation of bladder cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Células-Tronco Neoplásicas/efeitos dos fármacos , PPAR gama/agonistas , Prostaglandina D2/análogos & derivados , RNA Interferente Pequeno/genética , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Proteínas Inibidoras de Apoptose/genética , Proteínas Inibidoras de Apoptose/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , PPAR gama/genética , PPAR gama/metabolismo , Fenótipo , Prostaglandina D2/farmacologia , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Survivina , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/metabolismo , Bexiga Urinária/patologia
12.
Hepatology ; 59(1): 178-89, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23897841

RESUMO

UNLABELLED: Growth arrest and DNA damage 45G (GADD45G), a stress sensor with multiple implications in various biological processes, is down-regulated in a broad spectrum of cancers. However, little is known about the biological effects of GADD45G on hepatocellular carcinoma (HCC) cells and the related mechanisms. In the present study, we found that GADD45G was commonly down-regulated in oncogene-transformed mouse liver cells and in human and mouse HCC. Ectopic expression of GADD45G robustly elicited senescence in HCC cells and suppressed tumor growth in vivo. Furthermore, GADD45G-induced senescence occurred in HCC cells independently of p53, p16(INK4a) (p16), and retinoblastoma (Rb). Instead, the prompt inhibition of Janus kinase 2 (Jak2), tyrosine kinase 2 (Tyk2), and signal transducer and activator of transcription 3 (Stat3) activation was observed in cells undergoing senescence. Impairment of Jak-Stat3 activation caused by GADD45G expression was associated with activation of SH2 domain-containing protein tyrosine phosphatase-2 (Shp2). Expression of constitutively activated Stat3 or human telomerase reverse transcriptase (hTERT), as well as knockdown of Shp2f, efficiently counteracted GADD45G-induced senescence. More important, in clinical HCC specimens, we found that GADD45G expression was inversely correlated with phosphorylated Stat3 expression in tumor cells and disease progression. CONCLUSION: GADD45G functions as a negative regulator of the Jak-Stat3 pathway and inhibits HCC by inducing cellular senescence. The decrease or absence of GADD45G expression may be a key event for tumor cells or premalignant liver cells to bypass cellular senescence.


Assuntos
Carcinoma Hepatocelular/metabolismo , Proteínas de Transporte/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Janus Quinases/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Linhagem Celular Tumoral , Senescência Celular , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Regulação para Baixo , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Fosforilação , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo
13.
Eur J Immunol ; 44(2): 521-32, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24165986

RESUMO

The generation of CD4⁺Foxp3⁺ Treg cells in the thymus is crucial for immune homeostasis and self-tolerance. Recent studies have shown Treg-cell plasticity when Th-related transcriptional factors and cytokines are present. However, the mechanisms that maintain the stability of Treg cells are poorly understood. Here, using mice with a T-cell-specific deletion of the transforming growth factor-ß receptor 2 (Tgfbr2⁻/⁻ mice), we identify the restriction of AKT activation as a key event for the control of Treg-cell stability in Th1 inflammation. AKT regulation was evident in thymic CD4⁺Foxp3⁺ Treg cells before they egressed to peripheral tissues. CD4⁺Foxp3⁺ thymocytes from mice with the Tgfbr2 deletion expressed high levels of CXCR3 and T-bet, and produced IFN-γ and TNF-α. Thymic Tgfbr2⁻/⁻ Treg cells also showed an increase in the activation of AKT pathway. Enhanced AKT activity induced the expression of IFN-γ both in natural and inducible Treg cells. Inhibition of AKT activity markedly attenuated the expression of IFN-γ and TNF-α in thymic Tgfbr2⁻/⁻ Treg cells in vivo. In addition, mixed bone marrow transplantation showed that TGF-ß signaling maintained Treg-cell stability in an intrinsic manner. Our results demonstrate that AKT hyperactivation contributes to the conversion of Treg cells to a Th1 phenotype.


Assuntos
Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Linfócitos T Reguladores/metabolismo , Células Th1/metabolismo , Animais , Transplante de Medula Óssea , Linfócitos T CD4-Positivos/metabolismo , Regulação para Baixo/genética , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Expressão Gênica/genética , Inflamação/genética , Inflamação/metabolismo , Interferon gama/genética , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Fosforilação/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Transdução de Sinais/genética , Timo/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
14.
Carcinogenesis ; 34(9): 2099-108, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23695722

RESUMO

Myeloid cells have a critical role in maintaining intestinal homeostasis and regulating the development of inflammatory bowel disease and colitis-associated cancer (CAC). However, the signaling pathways that control the function of colonic myeloid cells in these pathological processes are still poorly defined. In this study, we demonstrate that transforming growth factor-ß (TGF-ß) signaling in colonic myeloid cells is significantly involved in the development of CAC. Myeloid TGF-ß receptor II (Tgfbr2)-deficient mice showed reduced susceptibility to chemically induced colitis-associated tumorigenesis, as evidenced by decreases in number and size of tumors. Myeloid Tgfbr2 deficiency markedly decreased the production of interleukin-6 and tumor necrosis factor-α, two proinflammatory cytokines that are essential for colonic tumorigenesis; in addition, a marked increase in the proportions of Foxp3+CD4+ regulatory T cells was observed in the colonic lamina propria in the initial stage of CAC. Loss of myeloid Tgfbr2 was associated with a decrease in the presence of F4/80 positive macrophages and a downregulation of phosphorylated STAT3, proliferative cell nuclear antigen and cyclin D1 expression in colonic adenoma tissues. TGF-ß enhanced macrophage recruitment, at least in part, through modulating the expression of the chemokine (C-C motif) receptor 2 (CCR2) ligands in tumor environment and the CCR2 signaling in macrophages. Collectively, these results suggest that myeloid TGF-ß signaling modulates intestinal inflammation and significantly promotes tumorigenesis in the development of colitis-associated colon cancer.


Assuntos
Carcinogênese , Colite/patologia , Células Mieloides/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/genética , Fator de Necrose Tumoral alfa/metabolismo , Animais , Colite/induzido quimicamente , Colite/genética , Colo/metabolismo , Colo/patologia , Humanos , Inflamação/metabolismo , Inflamação/patologia , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Camundongos , Células Mieloides/patologia , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais
15.
Cancer Lett ; 336(2): 290-8, 2013 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-23523871

RESUMO

Transforming growth factor activated kinase 1 (TAK1) provides prosurvival signals in various types of cells, and emerging evidence indicates that targeting TAK1 is a promising means to eliminate certain types of cancer cells. Here, we show that TAK1 is required for efficient tumorigenicity of AKT-transformed cells. TAK1 inhibition accelerates cell apoptosis of AKT-transformed cells in anchorage-independent cell growth accompanying by the downregulation of Mcl-1 and Bcl-2 expression. On the contrary, the tumorigenicity of c-Myc-transformed cells is not significantly affected by TAK1 inhibition. Moreover, AKT-transformed cells with c-Myc overexpression tolerate TAK1 inhibition in anchorage-independent growth and tumorigenicity in vivo. Together, our results provide evidence that TAK1-dependency in the tumorigenicity of AKT-transformed cells can be alleviated by c-Myc overexpression. These findings suggest that dual-targeting TAK1 and c-Myc might be a rational therapeutic strategy for treatment of certain types of cancer.


Assuntos
MAP Quinase Quinase Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-myc/biossíntese , Animais , Apoptose/fisiologia , Processos de Crescimento Celular/fisiologia , Sobrevivência Celular/fisiologia , Transformação Celular Neoplásica/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Técnicas de Silenciamento de Genes , Humanos , MAP Quinase Quinase Quinases/deficiência , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Células NIH 3T3 , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-myc/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
16.
PLoS One ; 6(12): e28405, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22145042

RESUMO

The mammalian target of the rapamycin (mTOR) pathway, which drives cell proliferation, is frequently hyperactivated in a variety of malignancies. Therefore, the inhibition of the mTOR pathway has been considered as an appropriate approach for cancer therapy. In this study, we examined the roles of mTOR in the maintenance and differentiation of cancer stem-like cells (CSCs), the conversion of conventional cancer cells to CSCs and continuous tumor growth in vivo. In H-Ras-transformed mouse liver tumor cells, we found that pharmacological inhibition of mTOR with rapamycin greatly increased not only the CD133+ populations both in vitro and in vivo but also the expression of stem cell-like genes. Enhancing mTOR activity by over-expressing Rheb significantly decreased CD133 expression, whereas knockdown of the mTOR yielded an opposite effect. In addition, mTOR inhibition severely blocked the differentiation of CD133+ to CD133- liver tumor cells. Strikingly, single-cell culture experiments revealed that CD133- liver tumor cells were capable of converting to CD133+ cells and the inhibition of mTOR signaling substantially promoted this conversion. In serial implantation of tumor xenografts in nude BALB/c mice, the residual tumor cells that were exposed to rapamycin in vivo displayed higher CD133 expression and had increased secondary tumorigenicity compared with the control group. Moreover, rapamycin treatment also enhanced the level of stem cell-associated genes and CD133 expression in certain human liver tumor cell lines, such as Huh7, PLC/PRC/7 and Hep3B. The mTOR pathway is significantly involved in the generation and the differentiation of tumorigenic liver CSCs. These results may be valuable for the design of more rational strategies to control clinical malignant HCC using mTOR inhibitors.


Assuntos
Antígenos CD/metabolismo , Carcinoma Hepatocelular/patologia , Proliferação de Células , Glicoproteínas/metabolismo , Neoplasias Hepáticas/patologia , Células-Tronco Neoplásicas/patologia , Peptídeos/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Antígeno AC133 , Animais , Antibióticos Antineoplásicos/farmacologia , Antígenos CD/genética , Apoptose/efeitos dos fármacos , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Glicoproteínas/genética , Humanos , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Peptídeos/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Proteína Supressora de Tumor p53/fisiologia
17.
Anal Bioanal Chem ; 382(1): 28-36, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15900448

RESUMO

This paper describes the preparation for the first time of lipobead-based micrometric fluorescence biosensors and the optimization of their analytical properties. The study focused on the well-established urea biosensors as a model system. Fluorescence-sensing lipobeads were prepared by coating carboxyl-functionalized silica microspheres with phospholipids. The enzyme urease and the pH indicator fluorescein-5-thiosemicarbazide were then attached covalently to the phospholipid membrane of the lipobeads. Urease converts urea to ammonia, which results in a pH increase in the analyte solution and to a urea concentration-dependent increase in the fluorescence intensity of the sensing lipobeads. Previous fluorescence-sensing lipobeads were synthesized by coating polystyrene particles with a phospholipid membrane. The membrane was physically attached to the particles and the fluorophores were entrapped in the membrane. In this study, we prepared improved fluorescence-sensing lipobeads by utilizing covalent chemistry to bind the phospholipid membrane to the silica particles and the fluorophores to the membrane. This led to improvement in the stability of the newly developed urea-sensing lipobeads compared to previously developed miniaturized fluorescence biosensors.


Assuntos
Técnicas Biossensoriais , Fluorescência , Concentração de Íons de Hidrogênio , Microscopia Eletrônica de Varredura , Microscopia de Fluorescência , Espectroscopia de Infravermelho com Transformada de Fourier
18.
J Am Chem Soc ; 127(15): 5276-7, 2005 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-15826138

RESUMO

In this communication, we report a successful synthesis of quasi-monodisperse In2O3 nanocrystals with high crystallinity in a high-temperature organic solution. The average size of nanocrystals can be tuned using a dynamic injection technique. TEM and XRD investigations indicate that each nanocrystal is a single crystal. The optical determination implies that the photoluminescence behavior of these In2O3 nanocrystals is different from that of the bulk, probably due to the combination of weak quantum-confinement-effects and the nature of high crystallinity in nanocrystals.

19.
Anal Chem ; 76(3): 569-75, 2004 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-14750848

RESUMO

This paper describes the preparation and optimization of the analytical properties of fluorescence-based submicrometric chloride ion sensing lipobeads. Fluorescence sensing lipobeads are polystyrene nanoparticles that are coated with a phospholipid membrane that contains a fluorescent indicator for a targeted analyte. In this study, the halide-specific fluorescence dye, lucigenin, was immobilized into the phospholipid membrane of the lipobeads to enable chloride ion detection. The fluorescence intensity of lucigenin decreases with increasing chloride ion concentration due to dynamic quenching. Lipobeads that contained only lucigenin were ineffective as chloride ion sensors due to poor partition of the water-soluble lucigenin molecules into the phospholipid membrane and high leakage rate of immobilized lucigenin molecules to the aqueous solution. To stabilize the chloride ion sensing lipobeads we coimmobilized hexadecanesulfonate molecules into the phospholipid membrane. The formation of ion pairs between hexadecanesulfonate and lucigenin decreased the hydrophilicity of the dye, increased its partition rate into the membrane, increased the brightness of the particles, and significantly decreased the leakage rate of the hydrophobic ion pair from the membrane to the solution. To further improve their chloride ion sensitivity, we also immobilized the chloride ionophore [9] mercuracarborand-3 into the lipobead membrane. The study resulted in a unique submicrometric chloride ion sensor, which is suitable for chloride ion measurements in biological fluids.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...