Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
2.
Mol Oncol ; 16(19): 3490-3508, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35593080

RESUMO

Despite the initial efficacy of using tyrosine kinase inhibitors of epidermal growth factor receptors (EGFR-TKIs) for treating patients with non-small cell lung cancer (NSCLC), resistance inevitably develops. Recent studies highlight a link between alternative splicing and cancer drug response. Therefore, we aimed to identify deregulated splicing events that play a role in resistance to EGFR-TKI. By using RNA sequencing, reverse-transcription PCR (RT-PCR), and RNA interference, we showed that overexpression of a splice variant of the autophagic gene ATG16-L1 that retains exon 8 and encodes the ß-isoform of autophagy-related protein 16-1 (ATG16-L1 ß) concurs acquired resistance to EGFR-TKI in NSCLC cells. Using matched biopsies, we found increased levels of ATG16-L1 ß at the time of progression in 3 of 11 NSCLC patients treated with EGFR-TKI. Mechanistically, gefitinib-induced autophagy was impaired in resistant cells that accumulated ATG16-L1 ß. Neutralization of ATG16-L1 ß restored autophagy in response to gefitinib, induced apoptosis, and inhibited the growth of in ovo tumor xenografts. Conversely, overexpression of ATG16-L1 ß in parental sensitive cells prevented gefitinib-induced autophagy and increased cell survival. These results support a role of defective autophagy in acquired resistance to EGFR-TKIs and identify splicing regulation of ATG16-L1 as a therapeutic vulnerability that could be explored for improving EGFR-targeted cancer therapy.


Assuntos
Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Antineoplásicos/farmacologia , Autofagia , Proteínas Relacionadas à Autofagia/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Família de Proteínas EGF/farmacologia , Família de Proteínas EGF/uso terapêutico , Receptores ErbB/metabolismo , Gefitinibe/farmacologia , Gefitinibe/uso terapêutico , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico
7.
Front Genet ; 10: 390, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31134126

RESUMO

Lung cancer, including non-small cell lung carcinoma (NSCLC), is the most frequently diagnosed cancer. It is also the leading cause of cancer-related mortality worldwide because of its late diagnosis and its resistance to therapies. Therefore, the identification of biomarkers for early diagnosis, prognosis, and monitoring of therapeutic response is urgently needed. Liquid biopsies, especially blood, are considered as promising tools to detect and quantify circulating cancer biomarkers. Cell-free circulating tumor DNA has been extensively studied. Recently, the possibility to detect and quantify RNAs in tumor biopsies, notably circulating cell-free RNAs, has gained great attention. RNA alternative splicing contributes to the proteome diversity through the biogenesis of several mRNA splice variants from the same pre-mRNA. Circular RNA (circRNA) is a new class of RNAs resulting from pre-mRNA back splicing. Owing to the development of high-throughput transcriptomic analyses, numerous RNA splice variants and, more recently, circRNAs have been identified and found to be differentially expressed in tumor patients compared to healthy controls. The contribution of some of these RNA splice variants and circRNAs to tumor progression, dissemination, or drug response has been clearly demonstrated in preclinical models. In this review, we discuss the potential of circRNAs and mRNA splice variants as candidate biomarkers for the prognosis and the therapeutic response of NSCLC in liquid biopsies.

8.
Sci Rep ; 9(1): 336, 2019 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-30674935

RESUMO

The splice variant sVEGFR1-i13 is a truncated version of the cell membrane-spanning VEGFR1 receptor that is devoid of its transmembrane and tyrosine kinase domains. We recently showed the contribution of sVEGFR1-i13 to the progression and the response of squamous lung carcinoma to anti-angiogenic therapies. In this study, we identify VEGF165, a splice variant of VEGF-A, as a regulator of sVEGFR1-i13 expression in these tumors, and further show that VEGF165 cooperates with the transcription factor SOX2 and the splicing factor SRSF2 to control sVEGFR1-i13 expression. We also demonstrate that anti-angiogenic therapies up-regulate sVEGFR1-i13 protein level in squamous lung carcinoma cells by a mechanism involving the VEGF165/SOX2/SRSF2 network. Collectively, our results identify for the first time a signaling network that controls VEGFR1 pre-mRNA alternative splicing in cancer cells.


Assuntos
Processamento Alternativo , Carcinoma de Células Escamosas/patologia , Neoplasias Pulmonares/patologia , Fatores de Transcrição SOXB1/metabolismo , Fatores de Processamento de Serina-Arginina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Linhagem Celular Tumoral , Redes Reguladoras de Genes , Humanos , Precursores de RNA/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética
9.
Oncogene ; 38(7): 1050-1066, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30194450

RESUMO

Vascular endothelial growth factor-A (VEGF-A) is highly subjected to alternative pre-mRNA splicing that generates several splice variants. The VEGFxxx and VEGFxxxb families encode splice variants of VEGF-A that differ only at the level of six amino acids in their C-terminal part. The expression level of VEGFxxx splice variants and their function as pro-angiogenic factors during tumor neo-angiogenesis have been well-described. The role of VEGFxxxb isoforms is less well known, but they have been shown to inhibit VEGFxxx-mediated angiogenesis, while being partial or weak activators of VEGFR receptors in endothelial cells. On the opposite, their role on tumor cells expressing VEGFRs at their surface remains largely unknown. In this study, we find elevated levels of VEGF165b, the main VEGFxxxb isoform, in 36% of non-small cell lung carcinoma (NSCLC), mainly lung adenocarcinoma (46%), and show that a high VEGF165b/VEGF165 ratio correlates with the presence of lymph node metastases. At the molecular level, we demonstrate that VEGF165b stimulates proliferation and invasiveness of two lung tumor cell lines through a VEGFR/ß1 integrin loop. We further provide evidence that the isoform-specific knockdown of VEGF165b reduces tumor growth, demonstrating a tumor-promoting autocrine role for VEGF165b in lung cancer cells. Importantly, we show that bevacizumab, an anti-angiogenic compound used for the treatment of lung adenocarcinoma patients, increases the expression of VEGF165b and activates the invasive VEGFR/ß1 integrin loop. Overall, these data highlight an unexpected role of the VEGF165b splice variant in the progression of lung tumors and their response to anti-angiogenic therapies.


Assuntos
Processamento Alternativo , Comunicação Autócrina/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Integrina beta1/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/metabolismo , Neovascularização Patológica/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Inibidores da Angiogênese/farmacologia , Bevacizumab/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Feminino , Humanos , Integrina beta1/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Proteínas de Neoplasias/genética , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/genética
10.
Biol Aujourdhui ; 212(1-2): 27-33, 2018.
Artigo em Francês | MEDLINE | ID: mdl-30362453

RESUMO

EGFR (Epidermal Growth Factor Receptor) is one of the most studied molecules in biology. From its early identification and cloning to the discovery of its role in cancer, it has been at the forefront of our understanding of Receptor Tyrosine Kinase (RTK) and cell signals that induce homeostasis, but when overexpressed, facilitate tumorigenesis. While the biological functions of EGFR traditionally involve the activation of a signaling network from the plasma membrane that includes activation of the RAS/MAPK/ERK, PI3K/AKT and STATS pathways, a new mode of EGFR signaling has been progressively decoded in which membrane-associated EGFR is transported after endocytosis from cell surface to the nucleus through endocytosis, retrograde trafficking to the Golgi, the endoplasmic reticulum and the inner nuclear membrane through a series of proteic interactions. In the nucleus, EGFR acts as a transcriptional regulator, a kinase and a physical interactor, transmits signals and is involved in multiple biological functions, including cell proliferation, tumor progression, DNA repair and replication, and resistance to cancer therapies. In this review, we will summarize current knowledge of the EGFR nuclear signaling network, including how it is delivered to the nucleus, the functions it serves in the nucleus and how these functions affect cancer progression, survival and the response to treatment.


Assuntos
Carcinogênese , Núcleo Celular/metabolismo , Neoplasias , Animais , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Receptores ErbB/metabolismo , Receptores ErbB/fisiologia , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Transdução de Sinais/fisiologia , Falha de Tratamento
11.
Exp Cell Res ; 370(2): 264-272, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29959911

RESUMO

Epidermal Growth Factor Receptor (EGFR) signaling regulates multiple cellular processes including proliferation, survival and apoptosis, and is attenuated by lysosomal receptor degradation. EGFR is a potent oncogene and activating mutations of EGFR are critical determinants of oncogenic transformation as well as therapeutic targets in non-small cell lung cancer. We previously demonstrated that wild type and mutant EGFRs repress the expression of the ARF tumor suppressor to promote the survival of lung tumor cells. In this study, using transient transfection systems in CHO EGFR-null cells as well as in various lung tumor cell lines carrying wild type or activated mutant EGFR, we show that ARF downregulates the expression of EGFR protein by reducing its half life. In wild type EGFR cells, ARF promotes canonical lysosomal degradation of the receptor through enhanced phosphorylation of EGFR-Y1045 and Cbl-Y731. In contrast, in mutant EGFR cells, ARF induces EGFR degradation by activating a non-canonical AKT-dependent lysosomal pathway. Taken together, these results uncover a feedback loop by which ARF may control EGFR turnover to restrain oncogenic signaling. They also highlight distinct degradation promoting pathways between wild type and mutant EGFRs in response to ARF.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Fator de Crescimento Epidérmico/genética , Lisossomos/metabolismo , Fases de Leitura/genética , Apoptose/fisiologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo/genética , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Neoplasias Pulmonares/patologia , Lisossomos/genética , Mutação/genética , Fosforilação , Transdução de Sinais/fisiologia
12.
Br J Cancer ; 118(12): 1596-1608, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29795310

RESUMO

BACKGROUND: While lung adenocarcinoma patients can somewhat benefit from anti-angiogenic therapies, patients with squamous cell lung carcinoma (SQLC) cannot. The reasons for this discrepancy remain largely unknown. Soluble VEGF receptor-1, namely sVEGFR1-i13, is a truncated splice variant of the cell membrane-spanning VEGFR1 that has no transmembrane or tyrosine kinase domain. sVEGFR1-i13 is mainly viewed as an anti-angiogenic factor which counteracts VEGF-A/VEGFR signalling in endothelial cells. However, its role in tumour cells is poorly known. METHODS: mRNA and protein status were analysed by Real-Time qPCR, western blotting, ELISA assay, proximity ligation assay or immunohistochemistry in human tumour cell lines, murine tumourgrafts and non small cell lung carcinoma patients samples. RESULTS: We show that anti-angiogenic therapies specifically increase the levels of sVEGFR1-i13 in SQLC cell lines and chemically induced SQLC murine tumourgrafts. At the molecular level, we characterise a sVEGFR1-i13/ß1 integrin/VEGFR autocrine loop which determines whether SQLC cells proliferate or go into apoptosis, in response to anti-angiogenic therapies. Furthermore, we show that high levels of both sVEGFR1-i13 and ß1 integrin mRNAs and proteins are associated with advanced stages in SQLC patients and with a poor clinical outcome in patients with early stage SQLC. CONCLUSIONS: Overall, these results reveal an unexpected pro-tumoural function of sVEGFR1-i13 in SQLC tumour cells, which contributes to their progression and escape from anti-angiogenic therapies. These data might help to understand why some SQLC patients do not respond to anti-angiogenic therapies.


Assuntos
Inibidores da Angiogênese/farmacologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Integrina beta1/metabolismo , Neoplasias Pulmonares/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Comunicação Autócrina/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/irrigação sanguínea , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Progressão da Doença , Humanos , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Camundongos , Isoformas de Proteínas , Receptor Cross-Talk/efeitos dos fármacos , Células Tumorais Cultivadas , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cancer Lett ; 420: 146-155, 2018 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-29421153

RESUMO

Many Receptor Tyrosine Kinases translocate from the cell surface to the nucleus in normal and pathological conditions, including cancer. Here we report the nuclear expression of insulin-like growth factor-1 receptor (IGF1R) in primary human lung tumours. Using lung cancer cell lines and lung tumour xenografts, we demonstrate that the epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) gefitinib induces the nuclear accumulation of IGF1R in mucinous lung adenocarcinoma by a mechanism involving the intracellular re-localization of the growth factor amphiregulin. Amphiregulin allows the binding of IGF1R to importin-ß1 and promotes its nuclear transport. The nuclear accumulation of IGF1R by amphiregulin induces cell cycle arrest through p21WAF1/CIP1 upregulation, and prevents the induction of apoptosis in response to gefitinib. These results identify amphiregulin as the first nuclear localization signal-containing protein that interacts with IGF1R and allows its nuclear translocation. Furthermore they indicate that nuclear expression of IGF1R contributes to EGFR-TKI resistance in lung cancer.


Assuntos
Adenocarcinoma Mucinoso/metabolismo , Anfirregulina/metabolismo , Núcleo Celular/metabolismo , Resistencia a Medicamentos Antineoplásicos , Neoplasias Pulmonares/metabolismo , Receptores de Somatomedina/metabolismo , Células A549 , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Gefitinibe/farmacologia , Humanos , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico , Receptor IGF Tipo 1 , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Int J Mol Sci ; 18(2)2017 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-28208660

RESUMO

Receptor tyrosine kinases (RTKs) belong to a family of transmembrane receptors that display tyrosine kinase activity and trigger the activation of downstream signalling pathways mainly involved in cell proliferation and survival. RTK amplification or somatic mutations leading to their constitutive activation and oncogenic properties have been reported in various tumour types. Numerous RTK-targeted therapies have been developed to counteract this hyperactivation. Alternative splicing of pre-mRNA has recently emerged as an important contributor to cancer development and tumour maintenance. Interestingly, RTKs are alternatively spliced. However, the biological functions of RTK splice variants, as well as the upstream signals that control their expression in tumours, remain to be understood. More importantly, it remains to be determined whether, and how, these splicing events may affect the response of tumour cells to RTK-targeted therapies, and inversely, whether these therapies may impact these splicing events. In this review, we will discuss the role of alternative splicing of RTKs in tumour progression and response to therapies, with a special focus on two major RTKs that control proliferation, survival, and angiogenesis, namely, epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor-1 (VEGFR1).


Assuntos
Processamento Alternativo , Família Multigênica , Neoplasias/genética , Receptores Proteína Tirosina Quinases/genética , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Biomarcadores , Progressão da Doença , Receptores ErbB/sangue , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Terapia de Alvo Molecular , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Prognóstico , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Precursores de RNA/genética , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética
15.
Curr Opin Oncol ; 28(1): 58-64, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26575690

RESUMO

PURPOSE OF REVIEW: PremRNA alternative splicing is more a rule than an exception as it affects more than 90% of multiexons genes and plays a key role in proteome diversity. Here, we discuss some recent studies published in the extensively growing field linking RNA splicing and cancer. RECENT FINDINGS: These last years, the development of high-throughput studies together with appropriate bioinformatic tools have led to the identification of new cancer-specific splicing patterns that allow to distinguish various cancer types, and provide new prognosis biomarkers. In addition, the functional consequences of hot spot mutations affecting various components of the spliceosome machinery in cancers have been described. As an example, missplicing of the enhancer of zeste homolog 2 histone methyltransferase premRNA in response to hot spot mutation of the splicing factor SRSF2 was found to participate to the pathogenesis of myelodysplastic syndrome. Moreover, proofs of principle that targeting the RNA splicing machinery can be used to correct aberrant missplicing, kill oncogene-driven cancer cells, or reverse resistance of tumor cells to targeted therapies have been done. As another example, the core spliceosomal function was recently found to be critical for the survival of Myc-driven breast cancer cells, rendering them hypersensitive to spliceosome inhibitors. SUMMARY: Dysregulation of premRNA alternative splicing appears to be one of the hallmarks of cancer. The characterization of novel splicing signatures in cancer as well as the identification of original signaling networks involving RNA splicing regulators should allow to decipher novel oncogenic mechanisms and to develop new therapeutic strategies.


Assuntos
Mutação , Neoplasias/genética , Neoplasias/patologia , Splicing de RNA , Comunicação Celular/genética , Humanos , Terapia de Alvo Molecular/métodos , Síndromes Mielodisplásicas/genética , Proteínas Nucleares/genética , Ribonucleoproteínas/genética , Fatores de Processamento de Serina-Arginina , Transdução de Sinais/genética , Spliceossomos/genética , Spliceossomos/metabolismo
16.
Cell Cycle ; 12(8): 1267-78, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23518498

RESUMO

The transcription factor E2F1 belongs to the E2F family and plays a crucial role during cell cycle progression and apoptosis. Ser/Arg-Rich (SR) proteins are a family of RNA-binding phosphoproteins that control both constitutive and alternative pre-mRNA splicing events. We previously identified the SR protein SRSF2 as a new transcriptional target of E2F1 and demonstrated that both proteins cooperate to induce apoptosis in non-small cell lung carcinoma. In this study, we postulated that SRSF2 is also involved in the proliferative functions of E2F1. Using IHC, we first demonstrate that SRSF2 and its phosphorylated form (P-SRSF2) are overexpressed in neuroendocrine lung tumors that are highly proliferative tumors expressing high levels of E2F1. Importantly, we show a direct correlation between cyclin E, an E2F1-target gene controlling S phase, and P-SRSF2 proteins levels (p = 0.0083), suggesting a role of SRSF2 in E2F1-mediated cellular proliferation. Accordingly, using neuroendocrine lung carcinoma cell lines, we demonstrate that SRSF2 is a cell cycle-regulated protein involved in entry and progression into S phase. We also provide evidence that SRSF2 interacts with E2F1 and stimulates its transcriptional control of cell cycle target genes such as cyclin E. Finally, we show that inhibition of AKT signaling pathway prevents SRSF2 phosphorylation and activity toward E2F1 transcriptional function. Taken together, these results identify a new role of SRSF2 in the control of cell cycle progression and reinforce the functional link between SRSF2 and E2F1 proteins.


Assuntos
Carcinoma Neuroendócrino/fisiopatologia , Fator de Transcrição E2F1/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias Pulmonares/fisiopatologia , Proteínas Nucleares/metabolismo , Ribonucleoproteínas/metabolismo , Fase S/fisiologia , Western Blotting , Carcinoma Neuroendócrino/metabolismo , Linhagem Celular Tumoral , Ciclina E/metabolismo , Primers do DNA/genética , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Luciferases , Neoplasias Pulmonares/metabolismo , Fosforilação , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Processamento de Serina-Arginina , Transdução de Sinais/fisiologia
17.
PLoS One ; 7(10): e46539, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23071587

RESUMO

Splicing abnormalities frequently occur in cancer. A key role as splice site choice regulator is played by the members of the SR (Ser/Arg-rich) family of proteins. We recently demonstrated that SRSF2 is involved in cisplatin-mediated apoptosis of human lung carcinoma cell lines. In this study, by using immunohistochemistry, we demonstrate that the SR proteins SRSF1 and SRSF2 are overexpressed in 63% and 65% of lung adenocarcinoma (ADC) as well as in 68% and 91% of squamous cell lung carcinoma (SCC), respectively, compared to normal lung epithelial cells. In addition, we show that SRSF2 overexpression correlates with high level of phosphorylated SRSF2 in both ADC (p<0.0001) and SCC (p = 0.02), indicating that SRSF2 mostly accumulates under a phosphorylated form in lung tumors. Consistently, we further show that the SR-phosphorylating kinases SRPK1 and SRPK2 are upregulated in 92% and 94% of ADC as well as in 72% and 68% of SCC, respectively. P-SRSF2 and SRPK2 scores are correlated in ADC (p = 0.01). Using lung adenocarcinoma cell lines, we demonstrate that SRSF1 overexpression leads to a more invasive phenotype, evidenced by activation of PI3K/AKT and p42/44MAPK signaling pathways, increased growth capacity in soft agar, acquisition of mesenchymal markers such as E cadherin loss, vimentin and fibronectin gain, and increased resistance to chemotherapies. Finally, we provide evidence that high levels of SRSF1 and P-SRSF2 proteins are associated with extensive stage (III-IV) in ADC. Taken together, these results indicate that a global deregulation of pre-mRNA splicing regulators occurs during lung tumorigenesis and does not predict same outcome in both Non Small Cell Lung Carcinoma histological sub-types, likely contributing to a more aggressive phenotype in adenocarcinoma.


Assuntos
Adenocarcinoma/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/metabolismo , Adenocarcinoma/patologia , Idoso , Antineoplásicos/farmacologia , Carboplatina/farmacologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Feminino , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Pulmão/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Proteínas Nucleares/genética , Paclitaxel/farmacologia , Fenótipo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas de Ligação a RNA/genética , Ribonucleoproteínas/genética , Fatores de Processamento de Serina-Arginina , Transdução de Sinais
18.
Carcinogenesis ; 33(2): 320-5, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22159227

RESUMO

The Tip60 and E2F1 proteins are key players of the cellular response induced by genotoxic stresses. Here, new insights into the involvement of both proteins during the DNA damage response are provided. We show that Tip60 interacts with E2F1 and promotes its acetylation. We identify the lysine residues 120/125 of the E2F1 protein as the prime target sites of Tip60 and show that acetylation at these sites promotes the accumulation of E2F1. Importantly, we demonstrate that cisplatin induces the accumulation of E2F1 in a Tip60-dependent manner. However, and in contrast to PCAF and p300, Tip60 is not required for the induction of apoptosis in cisplatin-treated cells. Instead, Tip60 and E2F1 are involved in the upregulation of the excision repair cross-complementation group 1 protein expression, an enzyme involved in the repair of cisplatin-induced DNA lesions. These findings identify Tip60 as a direct regulator of E2F1 and support their cooperative role in DNA repair.


Assuntos
Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/metabolismo , Antineoplásicos/farmacologia , Cisplatino/farmacologia , Proteínas de Ligação a DNA/metabolismo , Fator de Transcrição E2F1/metabolismo , Endonucleases/metabolismo , Histona Acetiltransferases/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Acetilação/efeitos dos fármacos , Adenocarcinoma/genética , Adenocarcinoma de Pulmão , Apoptose/efeitos dos fármacos , Sítios de Ligação/efeitos dos fármacos , Linhagem Celular Tumoral , Dano ao DNA , Reparo do DNA , Proteína p300 Associada a E1A/metabolismo , Humanos , Neoplasias Pulmonares/genética , Lisina/metabolismo , Lisina Acetiltransferase 5 , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Fatores de Transcrição de p300-CBP/metabolismo
19.
Cell Cycle ; 10(12): 1968-77, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21555914

RESUMO

Sodium butyrate (NaBu) is a histone deacetylase inhibitor that exhibits numerous antiproliferative activities in various cancer cell lines, notably through the accumulation of the well-known cyclin-dependent kinase inhibitor p21(WAF1) . SRSF2 belongs to the family of SR proteins that are crucial regulators of constitutive and alternative pre-mRNA splicing. Previous studies demonstrated that NaBu alters pre-mRNA splicing patterns through upregulation of SR proteins expression in non-tumor cells. In this study, we show that NaBu also induces the accumulation of SRSF2 in human lung carcinoma cell lines. We recently identified a signaling network involving the acetyltransferase TIP60, the deacetylase HDAC6 and the SRPK kinases that regulates SRSF2 protein turnover through phosphorylation/acetylation modifications in response to cisplatin. Here, we show that the same signaling pathway controls SRSF2 protein expression upon NaBu treatment. Importantly, we further demonstrate that SRSF2 is required for the accumulation of p21(WAF1) at both mRNA and protein levels in response to NaBu. Finally, we provide evidence that a long-term NaBu-treatment triggers senescence in our cellular models, a phenomenon that is prevented by the knockdown of SRSF2. Altogether, these results unravel a new function of SRSF2 in the process of cellular senescence and identify the cyclin-Cdk inhibitor p21(WAF1) as a key target of SRSF2 in this setting.


Assuntos
Ácido Butírico/farmacologia , Senescência Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/genética , Neoplasias Pulmonares/patologia , Proteínas Nucleares/metabolismo , Ribonucleoproteínas/metabolismo , Ativação Transcricional/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidores de Histona Desacetilases , Humanos , RNA Mensageiro , Fatores de Processamento de Serina-Arginina , Transdução de Sinais
20.
EMBO J ; 30(3): 510-23, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21157427

RESUMO

SRSF2 is a serine/arginine-rich protein belonging to the family of SR proteins that are crucial regulators of constitutive and alternative pre-mRNA splicing. Although it is well known that phosphorylation inside RS domain controls activity of SR proteins, other post-translational modifications regulating SRSF2 functions have not been described to date. In this study, we provide the first evidence that the acetyltransferase Tip60 acetylates SRSF2 on its lysine 52 residue inside the RNA recognition motif, and promotes its proteasomal degradation. We also demonstrate that the deacetylase HDAC6 counters this acetylation and acts as a positive regulator of SRSF2 protein level. In addition, we show that Tip60 downregulates SRSF2 phosphorylation by inhibiting the nuclear translocation of both SRPK1 and SRPK2 kinases. Finally, we demonstrate that this acetylation/phosphorylation signalling network controls SRSF2 accumulation as well as caspase-8 pre-mRNA splicing in response to cisplatin and determines whether cells undergo apoptosis or G(2)/M cell cycle arrest. Taken together, these results unravel lysine acetylation as a crucial post-translational modification regulating SRSF2 protein level and activity in response to genotoxic stress.


Assuntos
Diferenciação Celular/fisiologia , Cisplatino/farmacologia , Histona Acetiltransferases/metabolismo , Histona Desacetilases/metabolismo , Proteínas Nucleares/metabolismo , Ribonucleoproteínas/metabolismo , Transdução de Sinais/fisiologia , Acetilação , Processamento Alternativo/efeitos dos fármacos , Western Blotting , Caspase 8/genética , Caspase 8/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Primers do DNA/genética , Desacetilase 6 de Histona , Humanos , Imunoprecipitação , Lisina/metabolismo , Lisina Acetiltransferase 5 , Oligonucleotídeos/genética , Fosforilação , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Processamento de Serina-Arginina , Transdução de Sinais/genética , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA