Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
2.
Int J Mol Sci ; 24(12)2023 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-37373496

RESUMO

The standard-of-care treatment of T-cell acute lymphoblastic leukaemia (T-ALL) with chemotherapy usually achieves reasonable rates of initial complete response. However, patients who relapse or do not respond to conventional therapy show dismal outcomes, with cure rates below 10% and limited therapeutic options. To ameliorate the clinical management of these patients, it is urgent to identify biomarkers able to predict their outcomes. In this work, we investigate whether NRF2 activation constitutes a biomarker with prognostic value in T-ALL. Using transcriptomic, genomic, and clinical data, we found that T-ALL patients with high NFE2L2 levels had shorter overall survival. Our results demonstrate that the PI3K-AKT-mTOR pathway is involved in the oncogenic signalling induced by NRF2 in T-ALL. Furthermore, T-ALL patients with high NFE2L2 levels displayed genetic programs of drug resistance that may be provided by NRF2-induced biosynthesis of glutathione. Altogether, our results indicate that high levels of NFE2L2 may be a predictive biomarker of poor treatment response in T-ALL patients, which would explain the poor prognosis associated with these patients. This enhanced understanding of NRF2 biology in T-ALL may allow a more refined stratification of patients and the proposal of targeted therapies, with the ultimate goal of improving the outcome of relapsed/refractory T-ALL patients.


Assuntos
Leucemia-Linfoma Linfoblástico de Células T Precursoras , Humanos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Fator 2 Relacionado a NF-E2/genética , Prognóstico , Fosfatidilinositol 3-Quinases , Recidiva Local de Neoplasia , Linfócitos T
3.
Nat Commun ; 14(1): 3130, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37253733

RESUMO

Clinical management of breast cancer (BC) metastasis remains an unmet need as it accounts for 90% of BC-associated mortality. Although the luminal subtype, which represents >70% of BC cases, is generally associated with a favorable outcome, it is susceptible to metastatic relapse as late as 15 years after treatment discontinuation. Seeking therapeutic approaches as well as screening tools to properly identify those patients with a higher risk of recurrence is therefore essential. Here, we report that the lipid-degrading enzyme fatty acid amide hydrolase (FAAH) is a predictor of long-term survival in patients with luminal BC, and that it blocks tumor progression and lung metastasis in cell and mouse models of BC. Together, our findings highlight the potential of FAAH as a biomarker with prognostic value in luminal BC and as a therapeutic target in metastatic disease.


Assuntos
Amidoidrolases , Biomarcadores Tumorais , Neoplasias Pulmonares , Animais , Camundongos , Amidoidrolases/genética , Neoplasias Pulmonares/patologia , Recidiva Local de Neoplasia/patologia
4.
Br J Haematol ; 201(4): 718-724, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36786170

RESUMO

Despite the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway being frequently altered in T-ALL/LBL, no specific therapy has been approved for T-ALL/LBL patients with constitutive signalling by JAK/STAT, so there is an urgent need to identify pathway members that may be potential therapeutic targets. In the present study, we searched for JAK/STAT pathway members potentially modulated through aberrant methylation and identified SOCS3 hypermethylation as a recurrent event in T-ALL/LBL. Additionally, we explored the implications of SOCS3 deregulation in T-ALL/LBL and demonstrated that SOCS3 counteracts the constitutive activation of the JAK/STAT pathway through different molecular mechanisms. Therefore, SOCS3 emerges as a potential therapeutic target in T-ALL/LBL.


Assuntos
Leucemia-Linfoma de Células T do Adulto , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Humanos , Janus Quinases/metabolismo , Transdução de Sinais , Proteína 3 Supressora da Sinalização de Citocinas/genética , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Fatores de Transcrição STAT/metabolismo , Fator de Transcrição STAT3/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Linfócitos T/metabolismo
5.
Int J Mol Sci ; 23(23)2022 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-36499482

RESUMO

A reduction in FADD levels has been reported in precursor T-cell neoplasms and other tumor types. Such reduction would impact on the ability of tumor cells to undergo apoptosis and has been associated with poor clinical outcomes. However, FADD is also known to participate in non-apoptotic functions, but these mechanisms are not well-understood. Linking FADD expression to the severity of precursor T-cell neoplasms could indicate its use as a prognostic marker and may open new avenues for targeted therapeutic strategies. Using transcriptomic and clinical data from patients with precursor T-cell neoplasms, complemented by in vitro analysis of cellular functions and by high-throughput interactomics, our results allow us to propose a dual role for FADD in precursor T-cell neoplasms, whereby resisting cell death and chemotherapy would be a canonical consequence of FADD deficiency in these tumors, whereas deregulation of the cellular metabolism would be a relevant non-canonical function in patients expressing FADD. These results reveal that evaluation of FADD expression in precursor T-cell neoplasms may aid in the understanding of the biological processes that are affected in the tumor cells. The altered biological processes can be of different natures depending on the availability of FADD influencing its ability to exert its canonical or non-canonical functions. Accordingly, specific therapeutic interventions would be needed in each case.


Assuntos
Apoptose , Neoplasias , Humanos , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/metabolismo , Apoptose/genética , Perfilação da Expressão Gênica , Morte Celular , Linfócitos T/metabolismo
6.
Carcinogenesis ; 41(8): 1113-1122, 2020 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-31734690

RESUMO

Precursor T-cell lymphoblastic neoplasms are aggressive malignancies in need for more effective and specific therapeutic treatments. A significant fraction of these neoplasms harbor deletions on the locus 9p21, targeting the tumor suppressor CDKN2A but also deleting the aconitase 1 (ACO1) gene, a neighboring housekeeping gene involved in cytoplasm and mitochondrial metabolism. Here we show that reducing the aconitase activity with fluorocitrate decreases the viability of T-cell lymphoblastic neoplasia cells in correlation to the differential aconitase expression. The consequences of the treatment were evidenced in vitro using T-cell lymphoblastic neoplasia cell lines exhibiting 9p21 deletions and variable levels of ACO1 expression or activity. Similar results were observed in melanoma cell lines, suggesting a true potential for fluorocitrate in different cancer types. Notably, ectopic expression of ACO1 alleviated the susceptibility of cell lines to fluorocitrate and, conversely, knockdown experiments increased susceptibility of resistant cell lines. These findings were confirmed in vivo on athymic nude mice by using tumor xenografts derived from two T-cell lines with different levels of ACO1. Taken together, our results indicate that the non-targeted ACO1 deficiency induced by common deletions exerts a collateral cellular lethality that can be used as a novel therapeutic strategy in the treatment of several types of cancer.


Assuntos
Cromossomos Humanos Par 9/genética , Citratos/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Inibidores Enzimáticos/farmacologia , Deleção de Genes , Proteína 1 Reguladora do Ferro/deficiência , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citratos/uso terapêutico , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidores Enzimáticos/uso terapêutico , Feminino , Xenoenxertos , Humanos , Proteína 1 Reguladora do Ferro/antagonistas & inibidores , Proteína 1 Reguladora do Ferro/genética , Melanoma/genética , Camundongos , Camundongos Nus , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Neoplasias Cutâneas/genética
7.
Cancers (Basel) ; 11(10)2019 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-31569512

RESUMO

FADD was initially described as an adaptor molecule for death receptor-mediated apoptosis, but subsequently it has been implicated in nonapoptotic cellular processes such as proliferation and cell cycle control. During the last decade, FADD has been shown to play a pivotal role in most of the signalosome complexes, such as the necroptosome and the inflammasome. Interestingly, various mechanisms involved in regulating FADD functions have been identified, essentially posttranslational modifications and secretion. All these aspects have been thoroughly addressed in previous reviews. However, FADD implication in cancer is complex, due to pleiotropic effects. It has been reported either as anti- or protumorigenic, depending on the cell type. Regulation of FADD expression in cancer is a complex issue since both overexpression and downregulation have been reported, but the mechanisms underlying such alterations have not been fully unveiled. Posttranslational modifications also constitute a relevant mechanism controlling FADD levels and functions in tumor cells. In this review, we aim to provide detailed, updated information on alterations leading to changes in FADD expression and function in cancer. The participation of FADD in various biological processes is recapitulated, with a mention of interesting novel functions recently proposed for FADD, such as regulation of gene expression and control of metabolic pathways. Finally, we gather all the available evidence regarding the clinical implications of FADD alterations in cancer, especially as it has been proposed as a potential biomarker with prognostic value.

8.
Sci Rep ; 9(1): 5179, 2019 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-30914738

RESUMO

Fusions transcripts have been proven to be strong drivers for neoplasia-associated mutations, although their incidence in T-cell lymphoblastic lymphoma needs to be determined yet. Using RNA-Seq we have selected 55 fusion transcripts identified by at least two of three detection methods in the same tumour. We confirmed the existence of 24 predicted novel fusions that had not been described in cancer or normal tissues yet, indicating the accuracy of the prediction. Of note, one of them involves the proto oncogene TAL1. Other confirmed fusions could explain the overexpression of driver genes such as COMMD3-BMI1, LMO1 or JAK3. Five fusions found exclusively in tumour samples could be considered pathogenic (NFYG-TAL1, RIC3-TCRBC2, SLC35A3-HIAT1, PICALM MLLT10 and MLLT10-PICALM). However, other fusions detected simultaneously in normal and tumour samples (JAK3-INSL3, KANSL1-ARL17A/B and TFG-ADGRG7) could be germ-line fusions genes involved in tumour-maintaining tasks. Notably, some fusions were confirmed in more tumour samples than predicted, indicating that the detection methods underestimated the real number of existing fusions. Our results highlight the potential of RNA-Seq to identify new cryptic fusions, which could be drivers or tumour-maintaining passenger genes. Such novel findings shed light on the searching for new T-LBL biomarkers in these haematological disorders.


Assuntos
Proteínas de Fusão Oncogênica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , RNA-Seq , Algoritmos , Sequência de Bases , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas de Fusão Oncogênica/metabolismo , Proto-Oncogene Mas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
9.
Oncogene ; 38(23): 4620-4636, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30742097

RESUMO

FBXW7 is a driver gene in T-cell lymphoblastic neoplasia acting through proteasome degradation of key proto-oncogenes. FBXW7 encodes three isoforms, α, ß and γ, which differ only in the N-terminus. In this work, massive sequencing revealed significant downregulation of FBXW7 in a panel of primary T-cell lymphoblastic lymphomas characterised by the absence of mutations in its sequence. We observed that decreased expression mainly affected the FBXW7ß isoform and to a lesser extent FBXW7α and may be attributed to the combined effect of epigenetic changes, alteration of upstream factors and upregulation of miRNAs. Transient transfections with miRNA mimics in selected cell lines resulted in a significant decrease of total FBXW7 expression and its different isoforms separately, with the consequent increment of critical substrates and the stimulation of cell proliferation. Transient inhibition of endogenous miRNAs in a T-cell lymphoblastic-derived cell line (SUP-T1) was capable of reversing these proliferative effects. Finally, we show how FBXW7 isoforms display different roles within the cell. Simultaneous downregulation of the α and γ isoforms modulates the amount of CCNE1, whilst the ß-isoform alone was found to have a prominent role in modulating the amount of c-MYC. Our data also revealed that downregulation of all isoforms is a sine qua non condition to induce a proliferative pattern in our cell model system. Taking these data into account, potential new treatments to reverse downregulation of all or a specific FBXW7 isoform may be an effective strategy to counteract the proliferative capacity of these tumour cells.


Assuntos
Proteína 7 com Repetições F-Box-WD/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Linhagem Celular Tumoral , Regulação para Baixo/genética , Epigênese Genética , Perfilação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Humanos , Isoenzimas/genética , Células Jurkat , MicroRNAs/genética , Análise em Microsséries , Leucemia-Linfoma Linfoblástico de Células T Precursoras/enzimologia
10.
Carcinogenesis ; 40(10): 1260-1268, 2019 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-30805584

RESUMO

T-cell lymphoblastic lymphoma is a haematological disease with an urgent need for reliable prognostic biomarkers that allow therapeutic stratification and dose adjustment. The scarcity of human samples is responsible for the delayed progress in the study and the clinical management of this disease, especially compared with T-cell acute lymphoblastic leukaemia, its leukemic counterpart. In the present work, we have determined by immunohistochemistry that S194-P-FADD protein is significantly reduced in a cohort of 22 samples from human T-cell lymphoblastic lymphoma. Notably, the extent of such reduction varies significantly among samples and has revealed determinant for the outcome of the tumour. We demonstrate that Fas-associated protein with death domain (FADD) phosphorylation status affects protein stability, subcellular localization and non-apoptotic functions, specifically cell proliferation. Phosphorylated FADD would be more stable and preferentially localized to the cell nucleus; there, it would favour cell proliferation. We show that patients with higher levels of S194-P-FADD exhibit more proliferative tumours and that they present worse clinical characteristics and a significant enrichment to an oncogenic signature. This supports that FADD phosphorylation may serve as a predictor for T-cell lymphoblastic lymphoma aggressiveness and clinical status. In summary, we propose FADD phosphorylation as a new biomarker with prognostic value in T-cell lymphoblastic lymphoma.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Regulação Neoplásica da Expressão Gênica , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Apoptose , Estudos de Casos e Controles , Proliferação de Células , Estudos de Coortes , Proteína de Domínio de Morte Associada a Fas/química , Seguimentos , Humanos , Fosforilação , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Prognóstico , Estabilidade Proteica , Taxa de Sobrevida , Células Tumorais Cultivadas
11.
BMC Cancer ; 18(1): 430, 2018 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-29661169

RESUMO

BACKGROUND: Precursor T-cell lymphoblastic lymphomas (T-LBL) are rare aggressive hematological malignancies that mainly develop in children. As in other cancers, the loss of cell cycle control plays a prominent role in the pathogenesis in these malignancies that is primarily attributed to loss of CDKN2A (encoding protein p16INK4A). However, the impact of the deregulation of other genes such as CDKN1C, E2F1, and TP53 remains to be clarified. Interestingly, experiments in mouse models have proven that conditional T-cell specific deletion of Cdkn1c gene may induce a differentiation block at the DN3 to DN4 transition, and that the loss of this gene in the absence of Tp53 led to aggressive thymic lymphomas. RESULTS: In this manuscript, we demonstrated that the simultaneous deregulation of CDKN1C, E2F1, and TP53 genes by epigenetic mechanisms and/or the deregulation of specific microRNAs, together with additional impairing of TP53 function by the expression of dominant-negative isoforms are common features in primary human T-LBLs. CONCLUSIONS: Previous experimental work in mice revealed that T-cell specific deletion of Cdkn1c accelerates lymphomagenesis in the absence of Tp53. If, as expected, the consequences of the deregulation of the CDKN1C-E2F1-TP53 axis were the same as those experimentally demonstrated in mouse models, the disruption of this axis might be useful to predict tumor aggressiveness, and to provide the basis towards the development of potential therapeutic strategiesin human T-LBL.


Assuntos
Inibidor de Quinase Dependente de Ciclina p57/genética , Fator de Transcrição E2F1/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteína Supressora de Tumor p53/genética , Adolescente , Adulto , Animais , Criança , Epigênese Genética/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Análise de Sequência de RNA , Transdução de Sinais/genética , Adulto Jovem
12.
Oncotarget ; 7(38): 61485-61499, 2016 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-27556297

RESUMO

In the present work, we show that T-cell lymphoblastic lymphoma cells exhibit a reduction of FADD availability in the cytoplasm, which may contribute to impaired apoptosis. In addition, we observe a reduction of FADD phosphorylation that inversely correlates with the proliferation capacity and tumor aggressiveness. The resultant balance between FADD-dependent apoptotic and non-apoptotic abilities may define the outcome of the tumor. Thus, we propose that FADD expression and phosphorylation can be reliable biomarkers with prognostic value for T-LBL stratification.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Regulação Neoplásica da Expressão Gênica , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Animais , Apoptose , Biomarcadores Tumorais/genética , Caseína Quinase Ialfa/metabolismo , Proliferação de Células , Citoplasma/metabolismo , DNA Complementar/genética , DNA Complementar/isolamento & purificação , Regulação para Baixo , Fosfatases de Especificidade Dupla/metabolismo , Proteína de Domínio de Morte Associada a Fas/genética , Feminino , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células Jurkat , Estimativa de Kaplan-Meier , Leucemia Experimental/genética , Leucemia Experimental/mortalidade , Leucemia Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Fosforilação , Leucemia-Linfoma Linfoblástico de Células T Precursoras/mortalidade , Prognóstico , Proteínas Serina-Treonina Quinases/metabolismo , Medição de Risco/métodos , Análise de Sequência de DNA , Serina/metabolismo , Timócitos/metabolismo , Timócitos/patologia , Regulação para Cima
13.
J Natl Cancer Inst ; 107(6): djv077, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25855725

RESUMO

BACKGROUND: Pharmacological activation of cannabinoid receptors elicits antitumoral responses in different cancer models. However, the biological role of these receptors in tumor physio-pathology is still unknown. METHODS: We analyzed CB2 cannabinoid receptor protein expression in two series of 166 and 483 breast tumor samples operated in the University Hospitals of Kiel, Tübingen, and Freiburg between 1997 and 2010 and CB2 mRNA expression in previously published DNA microarray datasets. The role of CB2 in oncogenesis was studied by generating a mouse line that expresses the human V-Erb-B2 Avian Erythroblastic Leukemia Viral Oncogene Homolog 2 (HER2) rat ortholog (neu) and lacks CB2 and by a variety of biochemical and cell biology approaches in human breast cancer cells in culture and in vivo, upon modulation of CB2 expression by si/shRNAs and overexpression plasmids. CB2-HER2 molecular interaction was studied by colocalization, coimmunoprecipitation, and proximity ligation assays. Statistical tests were two-sided. RESULTS: We show an association between elevated CB2 expression in HER2+ breast tumors and poor patient prognosis (decreased overall survival, hazard ratio [HR] = 0.29, 95% confidence interval [CI] = 0.09 to 0.71, P = .009) and higher probability to suffer local recurrence (HR = 0.09, 95% CI = 0.049 to 0.54, P = .003) and to develop distant metastases (HR = 0.33, 95% CI = 0.13 to 0.75, P = .009). We also demonstrate that genetic inactivation of CB2 impairs tumor generation and progression in MMTV-neu mice. Moreover, we show that HER2 upregulates CB2 expression by activating the transcription factor ELK1 via the ERK cascade and that an increased CB2 expression activates the HER2 pro-oncogenic signaling at the level of the tyrosine kinase c-SRC. Finally, we show HER2 and CB2 form heteromers in cancer cells. CONCLUSIONS: Our findings reveal an unprecedented role of CB2 as a pivotal regulator of HER2 pro-oncogenic signaling in breast cancer, and they suggest that CB2 may be a biomarker with prognostic value in these tumors.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Receptor ErbB-2/metabolismo , Transdução de Sinais , Animais , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Alemanha , Humanos , Imuno-Histoquímica , Imunoprecipitação , Estimativa de Kaplan-Meier , Camundongos , Prognóstico , RNA Mensageiro/metabolismo , Receptor CB2 de Canabinoide/genética , Receptor ErbB-2/genética , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Análise Serial de Tecidos , Transcrição Gênica
14.
Carcinogenesis ; 34(4): 902-8, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23288923

RESUMO

Inappropriate activation of the GLI/hedgehog (GLI/Hh) signalling occurs in several human cancers, including haematological neoplasms. However, little is known about its relevance in precursor T-cell lymphoblastic lymphomas (T-LBL) development. Moreover, the mechanisms whereby GLI/Hh signalling is activated in haematological malignancies are not fully clear. Here, we show that the gene Smoothened (SMO), the only non-redundant gene of this pathway, is up-regulated in mouse and human T-LBL. Interestingly, down-regulation of micro-RNAs mmu-miR-30a and mmu-miR-141 as well as hsa-miR-193b clearly contributes to enhance the expression of this gene in mouse and human lymphomas and, subsequently, to activate the GLI/Hh signalling. Activation of the GLI/Hh signalling in T-LBL promotes cell survival and proliferation, since inhibition of the pathway using short-hairpin-RNA-mediated SMO knockdown, or cyclopamine as a specific antagonist, significantly reduces these cellular processes. These findings suggest that sustained SMO up-regulation may contribute to T-LBL development and advocate the use of specific SMO inhibitors or microRNAs-based therapies as an attractive possibility to treat an important subset of T-LBL.


Assuntos
Linfoma de Células T/genética , MicroRNAs/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Receptores Acoplados a Proteínas G/genética , Células 3T3 , Animais , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Regulação para Baixo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Células Jurkat , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Interferência de RNA , RNA Interferente Pequeno , Receptores Acoplados a Proteínas G/biossíntese , Transdução de Sinais/genética , Receptor Smoothened , Regulação para Cima , Alcaloides de Veratrum/farmacologia , Proteína GLI1 em Dedos de Zinco
15.
Expert Opin Ther Targets ; 16(1): 85-101, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22239437

RESUMO

INTRODUCTION: The Fas/FasL system plays a significant role in tumorigenesis. Research has shown that its impairment in cancer cells may lead to apoptosis resistance and contribute to tumor progression. Thus, the development of effective therapies targeting the Fas/FasL system may play an important role in the fight against cancer. AREAS COVERED: In this review the recent literature on targeting the Fas/FasL system for therapeutic exploitation at different levels is reviewed. Promising pre-clinical approaches and various exceptions are highlighted. The potential of combined therapies is also explored, whereby tumor sensitivity to Fas-mediated apoptosis is restored, before an effective targeted therapy is employed. EXPERT OPINION: The success of the Fas/FasL system targeting for therapeutics will require a better understanding of the alterations conferring resistance, in order to use the most appropriate sensitizing chemotherapeutic or radiotherapeutic agents in combination with effective targeted therapies.


Assuntos
Proteína Ligante Fas/metabolismo , Neoplasias/terapia , Receptor fas/metabolismo , Animais , Antineoplásicos/uso terapêutico , Apoptose , Terapia Genética , Humanos , Neoplasias/metabolismo , Transdução de Sinais/efeitos dos fármacos
16.
Carcinogenesis ; 33(2): 452-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22114070

RESUMO

Cryptic deletions at chromosome 6q are common cytogenetic abnormalities in T-cell lymphoblastic leukemia/lymphoma (T-LBL), but the target genes have not been formally identified. Our results build on detection of specific chromosomal losses in a mouse model of γ-radiation-induced T-LBLs and provide interesting clues for new putative susceptibility genes in a region orthologous to human 6q15-6q16.3. Among these, Epha7 emerges as a bona fide candidate tumor suppressor gene because it is inactivated in practically all the T-LBLs analyzed (100% in mouse and 95.23% in human). We provide evidence showing that Epha7 downregulation may occur, at least in part, by loss of heterozygosity (19.35% in mouse and 12.5% in human) or promoter hypermethylation (51.61% in mouse and 43.75% in human) or a combination of both mechanisms (12.90% in mouse and 6.25% in human). These results indicate that EPHA7 might be considered a new tumor suppressor gene for 6q deletions in T-LBLs. Notably, this gene is located in 6q16.1 proximal to GRIK2 and CASP8AP2, other candidate genes identified in this region. Thus, del6q seems to be a complex region where inactivation of multiple genes may cooperatively contribute to the onset of T-cell lymphomas.


Assuntos
Leucemia de Células T/genética , Leucemia-Linfoma de Células T do Adulto/genética , Linfoma de Células T/genética , Receptor EphA7/genética , Deleção de Sequência , Animais , Proteínas de Ligação ao Cálcio/genética , Linhagem Celular Tumoral , Deleção Cromossômica , Cromossomos Humanos Par 4 , Cromossomos Humanos Par 6/genética , Metilação de DNA , Regulação para Baixo , Feminino , Genes Supressores de Tumor , Humanos , Células Jurkat , Perda de Heterozigosidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Receptores de Ácido Caínico/genética , Receptor de GluK2 Cainato
17.
Carcinogenesis ; 31(12): 2165-71, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20889682

RESUMO

In previous reports, we described germ line functional polymorphisms that differentiate Fas and FasL genes in two mouse strains (SEG/Pas and C57BL/6J) exhibiting extreme differences in susceptibility to γ radiation-induced T-cell lymphomas. Here, we provide new data reinforcing the importance of the extrinsic pathway of apoptosis mediated by Fas in T-cell lymphoma development and about the functional significance of polymorphisms located at intracellular and extracellular domains of Fas and FasL. Using DNA recombinant technology, we generate chimerical Fas and FasL proteins by combination of protein regions derived from the two strains and demonstrate that any Fas-FasL interaction involving chimerical proteins drive cell apoptosis to a significant lower extent than the wild-type SEG/Pas and C57BL/6J Fas-FasL systems. In addition, we report new polymorphisms in the coding sequence of Fadd and demonstrate that the interaction between Fas and Fadd is significantly stronger if Fas and Fadd are of SEG/Pas origin compared with the C57BL/6J system. Altogether, these results suggest a model in which functional polymorphisms at the three genes collaborate on the global ability of the Fas/FasL system to induce apoptosis. A complete analysis of these three genes in the pathway appears to be a sine qua non condition to accurately predict the effectiveness of the Fas system and to estimate susceptibility to T-cell lymphoma.


Assuntos
Apoptose , Proteína Ligante Fas/genética , Proteína de Domínio de Morte Associada a Fas/genética , Predisposição Genética para Doença , Polimorfismo Genético , Leucemia-Linfoma Linfoblástico de Células T Precursoras/etiologia , Transdução de Sinais/fisiologia , Receptor fas/genética , Animais , Proteína Ligante Fas/fisiologia , Proteína de Domínio de Morte Associada a Fas/química , Proteína de Domínio de Morte Associada a Fas/fisiologia , Células HEK293 , Humanos , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Receptor fas/fisiologia
18.
Cancer Res ; 69(6): 2577-87, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19244132

RESUMO

Cancer susceptibility is essentially attributable to multiple low-penetrance genes. Using interspecific consomic and congenic mice between the tumor-resistant SEG/Pas and the tumor-sensitive C57BL/6J strains, a region on chromosome 19 involved in the genetic resistance to gamma-irradiation-induced T-cell lymphomas (Tlyr1) has been identified. Through the development of nonoverlapping subcongenic strains, it has been further shown that Anxa1 may be a candidate resistance gene on the basis of its differential expression in thymus stroma cells after gamma-radiation exposure. In addition, thymus stroma cells of thymic lymphomas exhibited a significant reduction in the expression levels of Anxa1. Interestingly, the activity of Anxa1 relies on prostaglandin E(2) (PGE(2)) induction that brings about apoptosis in thymocytes. In fact, in vitro transfection experiments revealed that PGE(2) production was enhanced when HEK 293 cells were transfected with full-length cDNAs of Anxa1, with PGE(2) production in the cells transfected with the allele of the resistant strain (Anxa1(Tyr)) being higher than that in cells transfected with the allele of the susceptible strain (Anxa1(Phe)). Furthermore, the presence of this compound in the medium induced apoptosis of immature CD4(+)CD8(+)CD3(low) cells in a dose-dependent manner. These results improve our knowledge of the molecular mechanisms triggering T-cell lymphoblastic lymphoma development while highlighting the relevance of the stroma in controlling genetic susceptibility and the use of PGE(2) as a new therapeutic approach in T-cell hematologic malignancies.


Assuntos
Anexina A1/genética , Dinoprostona/metabolismo , Linfoma de Células T/genética , Neoplasias do Timo/genética , Animais , Mapeamento Cromossômico , Dinoprostona/biossíntese , Dinoprostona/farmacologia , Raios gama , Predisposição Genética para Doença , Perda de Heterozigosidade , Linfoma de Células T/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Induzidas por Radiação/genética , Neoplasias Induzidas por Radiação/patologia , Células Estromais/metabolismo , Células Estromais/patologia , Timo/efeitos dos fármacos , Timo/patologia , Timo/efeitos da radiação , Neoplasias do Timo/patologia , Transfecção
19.
Clin Transl Oncol ; 10(12): 794-803, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19068450

RESUMO

Genetically modified animals represent a resource of immense potential for cancer research. Classically, genetic modifications in mice were obtained through selected breeding experiments or treatments with powerful carcinogens capable of inducing random mutagenesis. A new era began in the early 1980s when genetic modifications by inserting foreign DNA genes into the cells of an animal allowed for the development of transgenic mice. Since that moment, genetic modifications have been able to be made in a predetermined way. Gene targeting emerged later as a method of in vivo mutagenesis whereby the sequence of a predetermined gene is selectively modified within an intact cell. In this review we focus on how genetically modified mice can be created to study tumour development, and how these models have contributed to an understanding of the genetic alterations involved in human cancer. We also discuss the strengths and weaknesses of the different mouse models for identifying cancer genes, and understanding the consequences of their alterations in order to obtain the maximum benefit for cancer patients.


Assuntos
Modelos Animais de Doenças , Camundongos Transgênicos , Neoplasias/genética , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/isolamento & purificação , Biomarcadores Tumorais/metabolismo , Técnicas de Silenciamento de Genes/métodos , Técnicas de Silenciamento de Genes/veterinária , Marcação de Genes/métodos , Predisposição Genética para Doença , Camundongos , Camundongos Transgênicos/fisiologia , Modelos Biológicos , Neoplasias/patologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
20.
Cancer Res ; 67(21): 10268-77, 2007 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17974968

RESUMO

Endoglin is a membrane glycoprotein that acts as a coreceptor for transforming growth factor-beta. We and others have previously suggested a function of endoglin as a tumor suppressor in epithelial cancer. Here, we study the expression of endoglin during chemical mouse skin carcinogenesis. We find that shedding of membrane endoglin, allowing the secretion of a soluble endoglin form, is a late event associated with progression from squamous to spindle cell carcinomas. Knockdown of endoglin in transformed keratinocytes activates the Smad2/3 signaling pathway resulting in cell growth arrest, delayed tumor latencies, and a squamous to spindle phenotypic conversion. Forced expression of the long endoglin isoform in spindle carcinoma cells blocks transforming growth factor-beta1 stimulation of Smad2/3 signaling and prevents tumor formation. In contrast, expression of the short endoglin isoform has no effect on spindle cell growth in vitro or in vivo. Our results show that endoglin behaves as a suppressor of malignancy during the late stages of carcinogenesis. Therefore, disruption of membrane endoglin emerges as a crucial event for progression to spindle cell carcinomas.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neoplasias Cutâneas/prevenção & controle , Proteínas Supressoras de Tumor/fisiologia , Animais , Caderinas/genética , Movimento Celular , Endoglina , Peptídeos e Proteínas de Sinalização Intracelular/genética , Queratinócitos/patologia , Camundongos , Camundongos SCID , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/análise , RNA Interferente Pequeno/genética , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/genética , Transdução de Sinais , Neoplasias Cutâneas/patologia , Proteína Smad2/fisiologia , Proteína Smad3/fisiologia , Fator de Crescimento Transformador beta1/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...