Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
BMC Biol ; 19(1): 173, 2021 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-34433435

RESUMO

BACKGROUND: Angiogenesis is the process by which new blood vessels arise from pre-existing ones. Fibroblast growth factor-2 (FGF-2), a leading member of the FGF family of heparin-binding growth factors, contributes to normal as well as pathological angiogenesis. Pre-mRNA alternative splicing plays a key role in the regulation of cellular and tissular homeostasis and is highly controlled by splicing factors, including SRSFs. SRSFs belong to the SR protein family and are regulated by serine/threonine kinases such as SRPK1. Up to now, the role of SR proteins and their regulators in the biology of endothelial cells remains elusive, in particular upstream signals that control their expression. RESULTS: By combining 2D endothelial cells cultures, 3D collagen sprouting assay, a model of angiogenesis in cellulose sponges in mice and a model of angiogenesis in zebrafish, we collectively show that FGF-2 promotes proliferation, survival, and sprouting of endothelial cells by activating a SRSF1/SRSF3/SRPK1-dependent axis. In vitro, we further demonstrate that this FGF-2-dependent signaling pathway controls VEGFR1 pre-mRNA splicing and leads to the generation of soluble VEGFR1 splice variants, in particular a sVEGFR1-ex12 which retains an alternative last exon, that contribute to FGF-2-mediated angiogenic functions. Finally, we show that sVEGFR1-ex12 mRNA level correlates with that of FGF-2/FGFR1 in squamous lung carcinoma patients and that sVEGFR1-ex12 is a poor prognosis marker in these patients. CONCLUSIONS: We demonstrate that FGF-2 promotes angiogenesis by activating a SRSF1/SRSF3/SRPK1 network that regulates VEGFR1 alternative splicing in endothelial cells, a process that could also contribute to lung tumor progression.


Assuntos
Fator 2 de Crescimento de Fibroblastos , Neoplasias Pulmonares , Animais , Células Endoteliais , Fator 2 de Crescimento de Fibroblastos/genética , Humanos , Camundongos , Neovascularização Patológica/genética , Proteínas Serina-Treonina Quinases , Precursores de RNA , Fatores de Processamento de Serina-Arginina/genética , Peixe-Zebra/genética
2.
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
3.
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
5.
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
6.
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
7.
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
8.
Mol Cancer ; 13: 9, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24428911

RESUMO

Alternative pre-mRNA splicing (AS) widely expands proteome diversity through the combinatorial assembly of exons. The analysis of AS on a large scale, by using splice-sensitive microarrays, is a highly efficient method to detect the majority of known and predicted alternative transcripts for a given gene. The response to targeted anticancer therapies cannot easily be anticipated without prior knowledge of the expression, by the tumor, of target proteins or genes. To analyze, in depth, transcript structure and levels for genes involved in these responses, including AKT1-3, HER1-4, HIF1A, PIK3CA, PIK3R1-2, VEGFA-D and PIR, we engineered a dedicated gene chip with coverage of an average 185 probes per gene and, especially, exon-exon junction probes. As a proof of concept, we demonstrated the ability of such a chip to detect the effects of over-expressed SRSF2 RNA binding protein on the structure and abundance of mRNA products in H358 lung cancer cells conditionally over-expressing SRSF2. Major splicing changes were observed, including in HER1/EGFR pre-mRNA, which were also seen in human lung cancer samples over-expressing the SRSF2 protein. In addition, we showed that variations in HER1/EGFR pre-mRNA splicing triggered by SRSF2 overexpression in H358 cells resulted in a drop in HER1/EGFR protein level, which correlated with increased sensitivity to gefitinib, an EGFR tyrosine kinase inhibitor. We propose, therefore, that this novel tool could be especially relevant for clinical applications, with the aim to predict the response before treatment.


Assuntos
Terapia de Alvo Molecular , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Precursores de RNA/genética , Adenocarcinoma/genética , Processamento Alternativo , Western Blotting , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/genética , Proteínas Nucleares/genética , Reação em Cadeia da Polimerase em Tempo Real , Ribonucleoproteínas/genética , Fatores de Processamento de Serina-Arginina
9.
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
10.
J Biol Chem ; 286(30): 26406-17, 2011 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-21653699

RESUMO

The inhibitor of apoptosis protein cIAP1 (cellular inhibitor of apoptosis protein-1) is a potent regulator of the tumor necrosis factor (TNF) receptor family and NF-κB signaling pathways in the cytoplasm. However, in some primary cells and tumor cell lines, cIAP1 is expressed in the nucleus, and its nuclear function remains poorly understood. Here, we show that the N-terminal part of cIAP1 directly interacts with the DNA binding domain of the E2F1 transcription factor. cIAP1 dramatically increases the transcriptional activity of E2F1 on synthetic and CCNE promoters. This function is not conserved for cIAP2 and XIAP, which are cytoplasmic proteins. Chromatin immunoprecipitation experiments demonstrate that cIAP1 is recruited on E2F binding sites of the CCNE and CCNA promoters in a cell cycle- and differentiation-dependent manner. cIAP1 silencing inhibits E2F1 DNA binding and E2F1-mediated transcriptional activation of the CCNE gene. In cells that express a nuclear cIAP1 such as HeLa, THP1 cells and primary human mammary epithelial cells, down-regulation of cIAP1 inhibits cyclin E and A expression and cell proliferation. We conclude that one of the functions of cIAP1 when localized in the nucleus is to regulate E2F1 transcriptional activity.


Assuntos
Núcleo Celular/metabolismo , Ciclina A/biossíntese , Ciclina E/biossíntese , Fator de Transcrição E2F1/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Elementos de Resposta/fisiologia , Transcrição Gênica/fisiologia , Animais , Núcleo Celular/genética , Proliferação de Células , Ciclina A/genética , Ciclina E/genética , Fator de Transcrição E2F1/genética , Inativação Gênica , Células HeLa , Humanos , Proteínas Inibidoras de Apoptose/genética , Camundongos , Estrutura Terciária de Proteína
11.
Biochim Biophys Acta Mol Cell Res ; 1869(7): 119253, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35259425

RESUMO

Basic FGF (bFGF) was discovered as a typical inducer of angiogenesis and has already been studied for 3 decades. Recent evidence indicates that bFGF plays different roles and controls signaling pathways that participate in the hallmarks of cancer, underscoring bFGF an appealing target for anti-cancer therapy. However, the early clinical trials designed to block bFGF signaling showed safety without satisfiable benefits for cancer patients. In this review, we firstly discuss bFGF's canonical signaling pathways and later review newly identified bFGF's functions that contribute to the cancer hallmarks besides its typical role in angiogenesis. After, we summarize the role of bFGF as a therapeutic target in response to different cancer therapies including radiotherapy, chemotherapy, targeted therapy, immunotherapy, and highlight the difficulties we must solve regarding the design of drugs targeting specifically bFGF. We also emphasize the need, especially for natural bFGF traps, to deepen their molecular mechanisms of action considering the specific context of cancer with different FGFR status, as well as the urgence of stratifying patients for both anti-bFGF first line and second line anti-cancer therapy. Finally, a perspective on potential feed-forward oncogenic signaling pathways mediated by bFGF is made. We discuss the importance of developing additional robust biomarkers to select patients who will benefit from bFGF-targeted therapy, as well as the rationale of developing combinatory therapies targeting either bFGF and/or its intracellular (co)effectors. This would ultimately provide novel therapeutic strategies to fight cancer.


Assuntos
Neoplasias , Terapia Combinada , Humanos , Imunoterapia , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Transdução de Sinais
12.
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
13.
Biochem Pharmacol ; 189: 114039, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-32417188

RESUMO

Pre-mRNA splicing is the removal of introns and ligation of exons to form mature mRNAs, and it provides a critical mechanism by which eukaryotic cells can regulate their gene expression. Strikingly, more than 90% of protein-encoding transcripts are alternatively spliced, through exon inclusion/skipping, differential use of 5' or 3' alternative splice sites, intron retention or selection of an alternative promoter, thereby drastically increasing protein diversity. Splicing is altered in various pathological conditions, including cancers. In the last decade, high-throughput transcriptomic analyses have identified thousands of splice variants in cancers, which can distinguish between tumoral and normal tissues as well as identify tumor types, subtypes and clinical stages. These abnormal or aberrantly expressed splice variants, found in all cancer hallmarks, can result from mutations in splice sites, deregulated expression or even somatic mutations of components of the spliceosome machinery. Therefore, and based on these recent observations, a new anti-cancer strategy of targeting the spliceosome machinery with small molecules has emerged; however, the potential for these therapies is still a matter of great debate. Notably, more preclinical studies are needed to clarify which splicing patterns are mainly affected by these compounds, which cancer patients could be the most eligible for these treatments and whether using these spliceosome inhibitors alone or in combination with chemotherapies or targeted therapies would provide better therapeutic benefits. In this commentary, I will discuss all of these aspects.


Assuntos
Sistemas de Liberação de Medicamentos , Terapia Genética , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Spliceossomos/metabolismo , Processamento Alternativo/efeitos dos fármacos , Processamento Alternativo/fisiologia , Antineoplásicos/administração & dosagem , Antineoplásicos/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Terapia Genética/métodos , Humanos , Macrolídeos/administração & dosagem , Macrolídeos/metabolismo , Neoplasias/genética , Piperazinas/administração & dosagem , Piperazinas/metabolismo , Piridinas/administração & dosagem , Piridinas/metabolismo , Spliceossomos/efeitos dos fármacos , Spliceossomos/genética
14.
Nanomaterials (Basel) ; 11(1)2021 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-33445812

RESUMO

The synthesis of silica nanoparticles (SiNPs) decorated on their surface with a range of various elements (e.g., ligands, drugs, fluorophores, vectors, etc.) in a controlled ratio remains a big challenge. We have previously developed an efficient strategy to obtain in one-step, well-defined multifunctional fluorescent SiNPs displaying fluorophores and two peptides ligands as targeting elements, allowing selective detection of cancer cells. In this paper, we demonstrate that additional level of controlled multifunctionality can be achieved, getting even closer to the original concept of "magic bullet", using solely sol-gel chemistry to achieve conjugation of PEG chains for stealth, along with three different ligands. In addition, we have answered the recurrent question of the surface ungrafting by investigating the stability of different siloxane linkages with the ERETIC Method (Electronic Reference to Access In Vivo Concentrations) by 19F NMR quantification. We also compared the efficiency of the hybrid silylated fluorophore covalent linkage in the core of the SiNP to conventional methods. Finally, the tumor-cell-targeting efficiency of these multi-ligand NPs on human endothelial cells (HUVEC or HDMEC) and mixed spheroids of human melanoma cells and HUVEC displaying different types of receptors were evaluated in vitro.

15.
Int J Cancer ; 127(10): 2239-47, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20549699

RESUMO

The INK4b-ARF-INK4a locus encodes two members of the INK4 family of cyclin-dependent kinase inhibitors, p15(INK4b) and p16(INK4a), and a completely unrelated protein called ARF. ARF is a nucleolar protein with unusual structure that exhibits tumor suppressive functions. There is growing evidence that ARF signaling is complex, and involves p53-dependent or -independent pathways aiming mainly at restraining abnormal cell growth and at maintaining genomic stability. As such, ARF is a critical component of tumor surveillance, and its expression is decreased in human tumors. In this review, we present the current knowledge on ARF regulation and major functions. The ARF status in human tumors is also briefly summarized.


Assuntos
Neoplasias/metabolismo , Neoplasias/patologia , Proteína Supressora de Tumor p14ARF/fisiologia , Animais , Humanos , Neoplasias/genética , Proteína Supressora de Tumor p14ARF/genética , Proteína Supressora de Tumor p14ARF/metabolismo
16.
Matrix Biol ; 94: 18-30, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32682018

RESUMO

Type V collagen (ColV) is a component of the endothelial basement membrane zone. During angiogenesis, extracellular matrix remodelling results in the release of active protein fragments that display pro- or anti-angiogenic properties. The latter often exert their activity through their heparin-binding site. We previously characterized a ColVα1-derived fragment called HEPV that contains a high affinity-binding site for heparin and heparan sulphate chains. Here we show that HEPV binds to FGF2 through its heparin-binding site. Using in vitro and in vivo angiogenesis assays, we show that HEPV but not the HEPV mutant at the heparin-binding site, inhibits FGF2-dependant angiogenesis. On the opposite, HEPV does not bind to VEGFA and has no effect on VEGFA-mediated angiogenesis. In 3D collagen gels, the addition of HEPV abrogates endothelial cell invasion and sprouting induced by FGF2. Interestingly, in vivo experiments reveal that HEPV anti-angiogenic activity is associated with the appearance of endothelial to mesenchymal transition (EndMT) markers. Together, these findings indicate that the ColVα1-derived fragment HEPV functions as an anti-angiogenic factor that represses FGF2-mediated angiogenesis through the regulation of endothelial cell plasticity. Previous observations showing that ColV overexpression negatively regulates pathological angiogenesis were left unexplained. Our data provide insights into the possible molecular mechanisms.


Assuntos
Colágeno Tipo V/genética , Fator 2 de Crescimento de Fibroblastos/genética , Morfogênese/genética , Neovascularização Patológica/genética , Fator A de Crescimento do Endotélio Vascular/genética , Sequência de Aminoácidos/genética , Inibidores da Angiogênese/farmacologia , Animais , Sítios de Ligação/genética , Linhagem Celular Tumoral , Plasticidade Celular/genética , Células Endoteliais/efeitos dos fármacos , Heparina/genética , Heparitina Sulfato/genética , Xenoenxertos , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Morfogênese/efeitos dos fármacos , Neovascularização Patológica/patologia , Ligação Proteica/genética
17.
Mol Cell Biol ; 26(11): 4339-50, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16705183

RESUMO

p14ARF is a tumor suppressor that controls a well-described p53/Mdm2-dependent checkpoint in response to oncogenic signals. Here, new insights into the tumor-suppressive function of p14ARF are provided. We previously showed that p14ARF can induce a p53-independent G2 cell cycle arrest. In this study, we demonstrate that the activation of ATM/ATR/CHK signaling pathways contributes to this G2 checkpoint and highlight the interrelated roles of p14ARF and the Tip60 protein in the initiation of this DNA damage-signaling cascade. We show that Tip60 is a new direct p14ARF binding partner and that its expression is upregulated and required for ATM/CHK2 activation in response to p14ARF. Strikingly, both p14ARF and Tip60 products accumulate following a cell treatment with alkylating agents and are absolutely required for ATM/CHK2 activation in this setting. Moreover, and consistent with p14ARF being a determinant of CHK2 phosphorylation in lung carcinogenesis, a strong correlation between p14ARF and phospho-CHK2 (Thr68) protein expression is observed in human lung tumors (P < 0.00006). Overall, these data point to a novel regulatory pathway that mediates the p53-independent negative-cell-growth control of p14ARF. Inactivation of this pathway is likely to contribute to lung carcinogenesis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Histona Acetiltransferases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Células COS , Cafeína/farmacologia , Proliferação de Células/efeitos dos fármacos , Quinase do Ponto de Checagem 2 , Chlorocebus aethiops , Ativação Enzimática/efeitos dos fármacos , Fase G2 , Humanos , Neoplasias Pulmonares/patologia , Lisina Acetiltransferase 5 , Mutagênicos/farmacologia , Proteínas de Neoplasias/metabolismo , Fosforilação , Ligação Proteica , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/genética
18.
Clin Cancer Res ; 14(22): 7237-45, 2008 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-18974389

RESUMO

PURPOSE: Epigenetic modifications of histone have crucial roles in the control of gene activity, nuclear architecture, and genomic stability. In this respect, they may contribute to the development and progression of cancer. We investigated whether epigenetic changes of histone H4 are involved in lung carcinogenesis. EXPERIMENTAL DESIGN: Epigenetic modifications of histone H4 were studied by immunohistochemistry in normal lung and 157 lung carcinoma using antibodies specifically recognizing the acetylated (Ac) lysines 5 (K5), K8, K12, K16, and trimethylated (me3) K20 residues of histone H4. Western blotting was used to validate the immunohistochemistry results. H4K20me3 was also studied in 17 preneoplastic lesions. Expression of the Suv4-20h1/2 trimethyltransferases was analyzed by quantitative reverse transcription-PCR in a subset of tumor samples. RESULTS: As compared with normal lung, cancer cells displayed an aberrant pattern of histone H4 modifications with hyperacetylation of H4K5/H4K8, hypoacetylation of H4K12/H4K16, and loss of H4K20 trimethylation. Alteration of H4K20 trimethylation was frequent in squamous cell carcinoma (67%) and was observed in early precursors lesions in which the level of H4K20me3 staining strongly decreased with disease progression. In adenocarcinoma, the down-regulation of H4K20me3 was less frequent (28%) but allowed the identification of a subgroup of stage I adenocarcinoma patients with reduced survival (P = 0.007). Loss of H4K20 trimethylation was associated with decreased expression of Suv4-20h2, a specific H4K20 trimethyltransferase involved in telomere length maintenance. CONCLUSIONS: Our findings indicate an important role of histone H4 modifications in bronchial carcinogenesis and highlight H4K20me3 as a candidate biomarker for early detection of and therapeutic approaches to lung cancer.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Histonas/metabolismo , Neoplasias Pulmonares/metabolismo , Lesões Pré-Cancerosas/metabolismo , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/genética , Epigênese Genética , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/genética , Metilação , Lesões Pré-Cancerosas/genética , Prognóstico , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
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
20.
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.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA