Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
IUBMB Life ; 76(1): 4-25, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37623925

RESUMO

Systemic modalities are crucial in the management of disseminated malignancies and liquid tumours. However, patient responses and tolerability to treatment are generally poor and those that enter remission often return with refractory disease. Combination therapies provide a methodology to overcome chemoresistance mechanisms and address dose-limiting toxicities. A deeper understanding of tumorigenic processes at the molecular level has brought a targeted therapy approach to the forefront of cancer research, and novel cancer biomarkers are being identified at a rapid rate, with some showing potential therapeutic benefits. The Karyopherin superfamily of proteins is soluble receptors that mediate nucleocytoplasmic shuttling of proteins and RNAs, and recently, nuclear transport receptors have been recognized as novel anticancer targets. Inhibitors against nuclear export have been approved for clinical use against certain cancer types, whereas inhibitors against nuclear import are in preclinical stages of investigation. Mechanistically, targeting nucleocytoplasmic shuttling has shown to abrogate oncogenic signalling and restore tumour suppressor functions through nuclear sequestration of relevant proteins and mRNAs. Hence, nuclear transport inhibitors display broad spectrum anticancer activity and harbour potential to engage in synergistic interactions with a wide array of cytotoxic agents and other targeted agents. This review is focussed on the most researched nuclear transport receptors in the context of cancer, XPO1 and KPNB1, and highlights how inhibitors targeting these receptors can enhance the therapeutic efficacy of standard of care therapies and novel targeted agents in a combination therapy approach. Furthermore, an updated review on the therapeutic targeting of lesser characterized karyopherin proteins is provided and resistance to clinically approved nuclear export inhibitors is discussed.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Transporte Ativo do Núcleo Celular/fisiologia , Proteína Exportina 1 , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Carioferinas/genética , Carioferinas/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Quimioterapia Combinada
2.
Sci Rep ; 12(1): 20171, 2022 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-36418423

RESUMO

Karyopherin beta 1 (Kpnß1) is the principal nuclear importer of cargo proteins and plays a role in many cellular processes. Its expression is upregulated in cancer and essential for cancer cell viability, thus the identification of its binding partners might help in the discovery of anti-cancer therapeutic targets and cancer biomarkers. Herein, we applied immunoprecipitation coupled to mass spectrometry (IP-MS) to identify Kpnß1 binding partners in normal and cancer cells. IP-MS identified 100 potential Kpnß1 binding partners in non-cancer hTERT-RPE1, 179 in HeLa cervical cancer, 147 in WHCO5 oesophageal cancer and 176 in KYSE30 oesophageal cancer cells, including expected and novel interaction partners. 38 binding proteins were identified in all cell lines, with the majority involved in RNA metabolism. 18 binding proteins were unique to the cancer cells, with many involved in protein translation. Western blot analysis validated the interaction of known and novel binding partners with Kpnß1 and revealed enriched interactions between Kpnß1 and select proteins in cancer cells, including proteins involved in cancer development, such as Kpnα2, Ran, CRM1, CCAR1 and FUBP1. Together, this study shows that Kpnß1 interacts with numerous proteins, and its enhanced interaction with certain proteins in cancer cells likely contributes to the cancer state.


Assuntos
Neoplasias Esofágicas , Neoplasias do Colo do Útero , Feminino , Humanos , beta Carioferinas , Espectrometria de Massas , Imunoprecipitação , Proteínas de Ciclo Celular , Proteínas Reguladoras de Apoptose , Proteínas de Ligação a DNA , Proteínas de Ligação a RNA
3.
Int J Cancer ; 150(2): 347-361, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34591985

RESUMO

Previous studies have identified increased expression of members of the nuclear transport protein family in cancer cells. Recently, certain nuclear transport proteins have been reported to be secreted by cells and found in the serum. The aims of our study were to investigate the levels of multiple nuclear transport proteins secreted from cancer cells, and to determine their potential as diagnostic markers for cervical and oesophageal cancer. Mass spectrometry identified 10 nuclear transport proteins in the secretome and exosomes of cultured cancer cells, and Western blot analysis confirmed increased secreted levels in cancer cells compared to normal. To investigate their presence in patient serum, enzyme-linked immunosorbent assays were performed and revealed significantly increased levels of KPNß1, CRM1, CAS, IPO5 and TNPO1 in cervical and oesophageal cancer patient serum compared to non-cancer controls. Significantly elevated KPNα2 and RAN levels were also identified in oesophageal cancer serum samples. Logistics regression analyses revealed IPO5 and TNPO1 to be the best performing individual candidate biomarkers in discriminating between cancer cases and controls. The combination of KPNß1, CRM1, KPNα2, CAS, RAN, IPO5 and TNPO1 as a panel of biomarkers had the highest diagnostic capacity with an area under the curve of 0.944 and 0.963, for cervical cancer and oesophageal cancer, and sensitivity of 92.5% at 86.8% specificity and 95.3% sensitivity at 87.5% specificity, respectively. These results suggest that nuclear transport proteins have potential as diagnostic biomarkers for cervical and oesophageal cancers, with a combination of protein family members being the best predictor.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteínas de Transporte/metabolismo , Núcleo Celular/metabolismo , Neoplasias Esofágicas/diagnóstico , Proteínas Nucleares/metabolismo , Secretoma/metabolismo , Neoplasias do Colo do Útero/diagnóstico , Transporte Ativo do Núcleo Celular , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/diagnóstico , Carcinoma de Células Escamosas do Esôfago/metabolismo , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/metabolismo , Adulto Jovem
4.
Exp Cell Res ; 404(2): 112637, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34019908

RESUMO

Karyopherin beta 1 (Kpnß1) is a major nuclear import receptor that mediates the import of cellular cargoes into the nucleus. Recently it has been shown that Kpnß1 is highly expressed in several cancers, and its inhibition by siRNA induces apoptotic cancer cell death, while having little effect on non-cancer cells. This study investigated the effect of a novel small molecule, Inhibitor of Nuclear Import-60 (INI-60), on cancer cell biology, as well as nuclear import activities associated with Kpnß1, and cancer progression in vivo using cervical and oesophageal cancer cell lines. INI-60 treatment resulted in the inhibition of cancer cell proliferation, colony formation, migration and invasion, and induced a G1/S cell cycle arrest, followed by cancer cell death via apoptosis. Non-cancer cells were minimally affected by INI-60 at concentrations that inhibited cancer cells. INI-60 treatment altered the localisation of Kpnß1 and its cargoes, NFκB/p65, NFAT and AP-1, and the overexpression of Kpnß1 reduced INI-60 cytotoxicity. INI-60 also inhibited KYSE 30 oesophageal cancer cell line growth in vivo. Taken together, these results show that INI-60 inhibits the nuclear import of Kpnß1 cargoes and interferes with cancer cell biology. INI-60 presents as a potential therapeutic approach for cancers of different tissue origins and warrants further investigation as a novel anti-cancer agent.


Assuntos
Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , beta Carioferinas/antagonistas & inibidores , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/genética , beta Carioferinas/genética
5.
BMC Cancer ; 21(1): 106, 2021 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-33530952

RESUMO

BACKGROUND: Inhibition of nuclear import via Karyopherin beta 1 (Kpnß1) shows potential as an anti-cancer approach. This study investigated the use of nuclear import inhibitor, INI-43, in combination with cisplatin. METHODS: Cervical cancer cells were pre-treated with INI-43 before treatment with cisplatin, and MTT cell viability and apoptosis assays performed. Activity and localisation of p53 and NFκB was determined after co-treatment of cells. RESULTS: Pre-treatment of cervical cancer cells with INI-43 at sublethal concentrations enhanced cisplatin sensitivity, evident through decreased cell viability and enhanced apoptosis. Kpnß1 knock-down cells similarly displayed increased sensitivity to cisplatin. Combination index determination using the Chou-Talalay method revealed that INI-43 and cisplatin engaged in synergistic interactions. p53 was found to be involved in the cell death response to combination treatment as its inhibition abolished the enhanced cell death observed. INI-43 pre-treatment resulted in moderately stabilized p53 and induced p53 reporter activity, which translated to increased p21 and decreased Mcl-1 upon cisplatin combination treatment. Furthermore, cisplatin treatment led to nuclear import of NFκB, which was diminished upon pre-treatment with INI-43. NFκB reporter activity and expression of NFκB transcriptional targets, cyclin D1, c-Myc and XIAP, showed decreased levels after combination treatment compared to single cisplatin treatment and this associated with enhanced DNA damage. CONCLUSIONS: Taken together, this study shows that INI-43 pre-treatment significantly enhances cisplatin sensitivity in cervical cancer cells, mediated through stabilization of p53 and decreased nuclear import of NFκB. Hence this study suggests the possible synergistic use of nuclear import inhibition and cisplatin to treat cervical cancer.


Assuntos
Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Antineoplásicos/farmacologia , Biomarcadores Tumorais/metabolismo , Cisplatino/farmacologia , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Pirróis/farmacologia , Quinoxalinas/farmacologia , Neoplasias do Colo do Útero/tratamento farmacológico , beta Carioferinas/antagonistas & inibidores , Antineoplásicos/uso terapêutico , Apoptose , Biomarcadores Tumorais/genética , Movimento Celular , Proliferação de Células , Cisplatino/uso terapêutico , Quimioterapia Combinada , Feminino , Humanos , Pirróis/uso terapêutico , Quinoxalinas/uso terapêutico , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia
6.
Mol Cancer Res ; 18(9): 1340-1353, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32503923

RESUMO

There is accumulating evidence for a link between circadian clock disruption and cancer progression. In this study, the circadian clock was investigated in cervical and esophageal cancers, to determine whether it is disrupted in these cancer types. Oncomine datamining revealed downregulation of multiple members of the circadian clock gene family in cancer patient tissue compared with matched normal epithelium. Real-time RT-PCR analysis confirmed significant downregulation of CLOCK, PER1, PER2, PER3, CRY1, CRY2, REV-ERBα, and RORα in esophageal tumor tissue. In cell line models, expression of several circadian clock genes was significantly decreased in transformed and cancer cells compared with noncancer controls, and protein levels were dysregulated. These effects were mediated, at least in part, by methylation, where CLOCK, CRY1, and RORα gene promoter regions were found to be methylated in cancer cells. Overexpression of CLOCK and PER2 in cancer cell lines inhibited cell proliferation and activation of RORα and REV-ERBα using agonists resulted in cancer cell death, while having a lesser effect on normal epithelial cells. Despite dysregulated circadian clock gene expression, cervical and esophageal cancer cells maintain functional circadian oscillations after Dexamethasone synchronization, as revealed using real-time bioluminescence imaging, suggesting that their circadian clock mechanisms are intact. IMPLICATIONS: This study is a first to describe dysregulated, yet oscillating, circadian clock gene expression in cervical and esophageal cancer cells, and knowledge of circadian clock functioning in these cancer types has the potential to inform chronotherapy approaches, where the timing of administration of chemotherapy is optimized on the basis of the circadian clock.


Assuntos
Relógios Circadianos/genética , Neoplasias Esofágicas/genética , Genes Supressores de Tumor/fisiologia , Neoplasias do Colo do Útero/genética , Proliferação de Células , Regulação para Baixo , Neoplasias Esofágicas/patologia , Feminino , Humanos , Neoplasias do Colo do Útero/patologia
7.
IUBMB Life ; 71(7): 771-780, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30674076

RESUMO

The circadian clock and the ~24 h rhythms it generates are essential in maintaining regular tissue functioning. At the molecular level, the circadian clock comprises a core set of rhythmically expressed genes and gene products that are able to drive rhythmic expression of other genes to generate overt circadian rhythms. It has recently come to light that perturbations of circadian rhythms contribute to the development of pathological states such as cancer, and altered expression and/or regulation of circadian clock genes has been identified in multiple tumour types. This review summarises the important role the circadian system plays in regulating cellular processes, including the cell cycle, apoptosis, DNA repair, the epithelial-to-mesenchymal transition, metabolism and immunity and how its dysregulation has widespread implications and could be a critical player in the development of cancer. Understanding its role in cancer development is important for the field chronotherapy, where the timing of chemotherapy administration is optimised based on differences in circadian clock functioning in normal and cancer cells. This has been found to influence the patient response, minimising the side effects commonly associated with chemotherapy. © 2019 IUBMB Life, 2019.


Assuntos
Antineoplásicos/uso terapêutico , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/metabolismo , Ritmo Circadiano/fisiologia , Neoplasias/prevenção & controle , Animais , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/genética , Humanos , Neoplasias/genética , Neoplasias/metabolismo
8.
Crit Rev Eukaryot Gene Expr ; 29(5): 413-424, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32421998

RESUMO

Transcription factors control numerous cellular processes, including proliferation, apoptosis, differentiation, and inflammation. Abnormal transcription factor activity has been implicated in a variety of diseases, especially cancer. The correct subcellular localization of transcription factors determines their activation status, implicating the nuclear transport receptors as key players in regulating transcription factor function. Dysregulation of the nuclear transport machinery has been described in numerous cancer types. This review summarizes how altered nuclear transport activity affects transcription factor localization and activity, and contributes to cancer development. Furthermore, the potential of targeting nuclear transporters for cancer therapy is discussed.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Regulação da Expressão Gênica , Neoplasias/metabolismo , Fatores de Transcrição/metabolismo , Apoptose , Diferenciação Celular , Humanos , Inflamação , Fatores de Transcrição/genética
9.
BMC Cancer ; 18(1): 1123, 2018 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-30445944

RESUMO

BACKGROUND: Karyopherin ß1 (Kpnß1) is the main nuclear import protein involved in the transport of cargoes from the cytoplasm into the cell nucleus. Previous research has found Kpnß1 to be significantly overexpressed in cervical cancer and other cancer tissues, and further studies showed that inhibition of Kpnß1 expression by siRNA resulted in cancer cell death, while non-cancer cells were minimally affected. These results suggest that Kpnß1 has potential as an anticancer therapeutic target, thus warranting further research into the association between Kpnß1 expression and cancer progression. Here, the biological effects associated with Kpnß1 overexpression were investigated in order to further elucidate the relationship between Kpnß1 and the cancer phenotype. METHODS: To evaluate the effect of Kpnß1 overexpression on cell biology, cell proliferation, cell cycle, cell morphology and cell adhesion assays were performed. To determine whether Kpnß1 overexpression influences cell sensitivity to chemotherapeutic agents like Cisplatin, cell viability assays were performed. Expression levels of key proteins were analysed by Western blot analysis. RESULTS: Our data revealed that Kpnß1 overexpression, above that which was already detected in cancer cells, resulted in reduced proliferation of cervical cancer cells. Likewise, normal epithelial cells showed reduced proliferation after Kpnß1 overxpression. Reduced cancer cell proliferation was associated with a delay in cell cycle progression, as well as changes in the morphology and adhesion properties of cells. Additionally, Kpnß1 overexpressing HeLa cells exhibited increased sensitivity to cisplatin, as shown by decreased cell viability and increased apoptosis, where p53 and p21 inhibition reduced and enhanced cell sensitivity to Cisplatin, respectively. CONCLUSIONS: Overall, our results suggest that a tight balance of Kpnß1 expression is required for cellular function, and that perturbation of this balance results in negative effects associated with a variety of biological processes.


Assuntos
Neoplasias do Colo do Útero/metabolismo , beta Carioferinas/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Cisplatino/farmacologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/fisiologia , Células HeLa , Humanos , RNA Interferente Pequeno/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Neoplasias do Colo do Útero/tratamento farmacológico
10.
Oncotarget ; 8(20): 32833-32847, 2017 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-28427184

RESUMO

Karyopherin ß1 is a nuclear import protein involved in the transport of proteins containing a nuclear localisation sequence. Elevated Karyopherin ß1 expression has been reported in cancer and transformed cells and is essential for cancer cell proliferation and survival. Transcription factors such as NFĸB and AP-1 contain a nuclear localisation sequence and initiate the expression of multiple factors associated with inflammation and cancer cell biology. Our study investigated the effect of inhibiting nuclear import via Karyopherin ß1 on cancer cell motility and inflammatory signaling using siRNA and the novel small molecule, Inhibitor of Nuclear Import-43, INI-43. Inhibition of Karyopherin ß1 led to reduced migration and invasion of cervical cancer cells. Karyopherin ß1 is essential for the translocation of NFĸB into the nucleus as nuclear import inhibition caused its cytoplasmic retention and decreased transcriptional activity. A similar decrease was seen in AP-1 transcriptional activity upon Karyopherin ß1 inhibition. Consequently reduced interleukin-6, interleukin-1 beta, tumour necrosis factor alpha and granulocyte macrophage colony stimulating factor expression, target genes of NFkB and AP-1, was observed. Migration studies inhibiting individual transcription factors suggested that INI-43 may affect a combination of signaling events. Our study provides further evidence that inhibiting KPNB1 has anti-cancer effects and shows promise as a chemotherapeutic target.


Assuntos
Núcleo Celular/metabolismo , Interleucina-1beta/metabolismo , Fator de Transcrição AP-1/metabolismo , Neoplasias do Colo do Útero/metabolismo , beta Carioferinas/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Interleucina-6/metabolismo , Invasividade Neoplásica , RNA Interferente Pequeno/farmacologia , Fator de Necrose Tumoral alfa , beta Carioferinas/genética
11.
IUBMB Life ; 68(4): 268-80, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26970212

RESUMO

The Karyopherin superfamily is a major class of soluble transport receptors consisting of both import and export proteins. The trafficking of proteins involved in transcription, cell signalling and cell cycle regulation among other functions across the nuclear membrane is essential for normal cellular functioning. However, in cancer cells, the altered expression or localization of nuclear transporters as well as the disruption of endogenous nuclear transport inhibitors are some ways in which the Karyopherin proteins are dysregulated. The value of nuclear transporters in the diagnosis, prognosis and treatment of cancer is currently being elucidated with recent studies highlighting their potential as biomarkers and therapeutic targets.


Assuntos
Antineoplásicos/farmacologia , Biomarcadores Tumorais/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/farmacologia , Acetiltransferases/genética , Acetiltransferases/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Biomarcadores Tumorais/metabolismo , Humanos , Carioferinas/genética , Carioferinas/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Prognóstico , Transporte Proteico/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , alfa Carioferinas/genética , alfa Carioferinas/metabolismo , Proteína Exportina 1
12.
Mol Cancer Ther ; 15(4): 560-73, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26832790

RESUMO

Karyopherin beta 1 (Kpnß1) is a nuclear transport receptor that imports cargoes into the nucleus. Recently, elevated Kpnß1 expression was found in certain cancers and Kpnß1 silencing with siRNA was shown to induce cancer cell death. This study aimed to identify novel small molecule inhibitors of Kpnß1, and determine their anticancer activity. An in silico screen identified molecules that potentially bind Kpnß1 and Inhibitor of Nuclear Import-43, INI-43 (3-(1H-benzimidazol-2-yl)-1-(3-dimethylaminopropyl)pyrrolo[5,4-b]quinoxalin-2-amine) was investigated further as it interfered with the nuclear localization of Kpnß1 and known Kpnß1 cargoes NFAT, NFκB, AP-1, and NFY and inhibited the proliferation of cancer cells of different tissue origins. Minimum effect on the proliferation of noncancer cells was observed at the concentration of INI-43 that showed a significant cytotoxic effect on various cervical and esophageal cancer cell lines. A rescue experiment confirmed that INI-43 exerted its cell killing effects, in part, by targeting Kpnß1. INI-43 treatment elicited a G2-M cell-cycle arrest in cancer cells and induced the intrinsic apoptotic pathway. Intraperitoneal administration of INI-43 significantly inhibited the growth of subcutaneously xenografted esophageal and cervical tumor cells. We propose that Kpnß1 inhibitors could have therapeutic potential for the treatment of cancer. Mol Cancer Ther; 15(4); 560-73. ©2016 AACR.


Assuntos
Antineoplásicos/farmacologia , beta Carioferinas/antagonistas & inibidores , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Simulação por Computador , Computadores Moleculares , Modelos Animais de Doenças , Descoberta de Drogas , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Expressão Gênica , Humanos , Camundongos , Modelos Moleculares , Terapia de Alvo Molecular , Ligação Proteica , Transporte Proteico , Bibliotecas de Moléculas Pequenas , Relação Estrutura-Atividade , Fatores de Transcrição/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , beta Carioferinas/química , beta Carioferinas/genética
13.
Oncol Rep ; 32(2): 730-8, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24898882

RESUMO

The nuclear export receptor, Crm1 (exportin 1), is involved in the nuclear translocation of proteins and certain RNAs from the nucleus to the cytoplasm and is thus crucial for the correct localisation of cellular components. Crm1 has recently been reported to be highly expressed in certain types of cancers, yet its expression in oesophageal cancer has not been investigated to date. We investigated the expression of Crm1 in normal and tumour tissues derived from 56 patients with human oesophageal squamous cell carcinoma and its functional significance in oesophageal cancer cell line models. Immunohistochemistry revealed that Crm1 expression was significantly elevated in oesophageal tumour tissues compared to normal tissues and its localisation shifted from predominantly nuclear to nuclear and cytoplasmic. Real­time RT­PCR revealed that Crm1 expression was elevated at the mRNA level. To determine the functional significance of elevated Crm1 expression in oesophageal cancer, its expression was inhibited using siRNA, and a significant decrease in cell proliferation was observed associated with G1 cell cycle arrest and the induction of apoptosis. Similarly, leptomycin B (LMB) treatment resulted in the effective killing of oesophageal cancer cells at nanomolar concentrations. Normal oesophageal epithelial cells, however, were much less sensitive to Crm1 inhibition with siRNA and LMB. Together, this study reveals that Crm1 expression is increased in oesophageal cancer and is required for the proliferation and survival of oesophageal cancer cells.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/metabolismo , Carioferinas/genética , Carioferinas/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Adulto , Idoso , Antibióticos Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células , Citoplasma/metabolismo , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago , Ácidos Graxos Insaturados/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Proteína Exportina 1
14.
Life Sci ; 103(1): 25-33, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-24637022

RESUMO

AIM: Recent studies have emphasized the importance of the extracellular microenvironment in modulating cell growth, motility, and signalling. In this study we have evaluated the ability of a fibroblast derived-extracellular matrix (fd-ECM) to regulate type I collagen synthesis and degradation in fibroblasts. MAIN METHODS: Fibroblasts were plated on plastic (control) or on fd-ECM and type I collagen synthesis and degradation was evaluated. MTT, western blotting, real time PCR, zymographic analysis and inhibitor assays were utilised to investigate the molecular mechanism of type I collagen regulation by the fd-ECM. KEY FINDINGS: Fibroblasts plated on fd-ECM showed significant downregulation in the production of type I collagen and COL1A2 messenger ribonucleic acid (mRNA) whilst COL1A1 mRNA remained unchanged. Cells grown on fd-ECM exhibited increased matrix metalloproteases (MMPs) and their corresponding mRNAs. The use of transforming growth factor ß (TGF-ß) and MMP inhibitors showed that the excess COL1A1 polypeptide chains were degraded by the combined action of MMP-1, MMP-2, MMP-9 and cathepsins. SIGNIFICANCE: These results show the crucial role played by proteases in regulating extracellular matrix protein levels in the feedback regulation of connective tissue gene expression.


Assuntos
Colágeno Tipo I/biossíntese , Matriz Extracelular/fisiologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Catepsinas/metabolismo , Linhagem Celular , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Retroalimentação Fisiológica , Fibroblastos/ultraestrutura , Humanos , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz/farmacologia , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , RNA Mensageiro/análise , Fator de Crescimento Transformador beta/farmacologia
15.
Carcinogenesis ; 35(5): 1121-31, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24398670

RESUMO

The karyopherin ß proteins are involved in nuclear-cytoplasmic trafficking and are crucial for protein and RNA subcellular localization. We previously showed that Kpnß1, a nuclear importin protein, is overexpressed in cervical cancer and is critical for cervical cancer cell survival and proliferation, whereas non-cancer cells are less dependent on its expression. This study aimed to identify the mechanisms by which inhibition of Kpnß1 results in cervical cancer cell death. We show that the inhibition of Kpnß1 results in the induction of apoptosis and a prolonged mitotic arrest, accompanied by distinct mitotic defects in cervical cancer cells but not non-cancer cells. In cervical cancer cells, Kpnß1 downregulation results in sustained degradation of the antiapoptotic protein, Mcl-1, and elevated Noxa expression, as well as mitochondrial membrane permeabilization resulting in the release of cytochrome C and activation of associated caspases. Although p53 becomes stabilized in Kpnß1 knockdown cervical cancer cells, apoptosis occurs in a p53-independent manner. These results demonstrate that blocking Kpnß1 has potential as an anticancer therapeutic approach.


Assuntos
Apoptose/genética , Pontos de Checagem do Ciclo Celular/genética , Mitose/genética , Transdução de Sinais , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/metabolismo , beta Carioferinas/genética , Linhagem Celular Tumoral , Feminino , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Potencial da Membrana Mitocondrial/genética , Modelos Biológicos , Interferência de RNA , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , beta Carioferinas/metabolismo
16.
Cell Commun Signal ; 11: 75, 2013 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-24090133

RESUMO

BACKGROUND: Recent studies have revealed that interactions between tumour cells and the surrounding stroma play an important role in facilitating tumour growth and invasion. Stromal fibroblasts produce most of the extracellular matrix components found in the stroma. The aim of this study was to investigate mechanisms involved in tumour cell-mediated regulation of extracellular matrix and adhesion molecules in co-cultured fibroblasts. To this end, microarray analysis was performed on CCD-1068SK human fibroblast cells after direct co-culture with MDA-MB-231 human breast tumour cells. RESULTS: We found that the expression of both connective tissue growth factor (CTGF/CCN2) and type I collagen was negatively regulated in CCD-1068SK fibroblast cells under direct co-culture conditions. Further analysis revealed that Smad7, a known negative regulator of the Smad signalling pathway involved in CCN2 promoter regulation, was increased in directly co-cultured fibroblasts. Inhibition of Smad7 expression in CCD-1068SK fibroblasts resulted in increased CCN2 expression, while Smad7 overexpression had the opposite effect. Silencing CCN2 gene expression in fibroblasts led, in turn, to a decrease in type I collagen mRNA and protein levels. ERK signalling was also shown to be impaired in CCD-1068SK fibroblasts after direct co-culture with MDA-MB-231 tumour cells, with Smad7 overexpression in fibroblasts leading to a similar decrease in ERK activity. These effects were not, however, seen in fibroblasts that were indirectly co-cultured with tumour cells. CONCLUSION: We therefore conclude that breast cancer cells require close contact with fibroblasts in order to upregulate Smad7 which, in turn, leads to decreased ERK signalling resulting in diminished expression of the stromal proteins CCN2 and type I collagen.


Assuntos
Neoplasias da Mama/metabolismo , Colágeno Tipo I/metabolismo , Fator de Crescimento do Tecido Conjuntivo/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína Smad7/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Regulação para Baixo , Fibroblastos , Humanos , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 1 da Matriz/metabolismo , Proteína Smad7/genética
17.
PLoS One ; 8(6): e66892, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23818969

RESUMO

Activating Protein 1 (AP-1) plays a vital role in cell proliferation, differentiation and apoptosis. While de-regulation of AP-1 has been linked to many cancers, little is known regarding its downstream transcriptional targets that associate with cellular transformation. Previous studies identified PAK3, a serine/threonine kinase, as a potential AP-1 target gene. PAK3 has been implicated in a variety of pathological disorders and over-expression of other PAK-family members has been linked to cancer. In this study, we investigate AP-1 regulation of PAK3 expression and the role of PAK3 in cJun/AP-1-associated cellular transformation. Our results showed elevated PAK3 expression at both the mRNA and protein level in cJun-over-expressing Rat1a fibroblasts, as well as in transformed human fibroblasts. Elevated PAK3 expression in cJun/AP-1 over-expressing cells associated with a significant increase in PAK3 promoter activation. This increased promoter activity was lost when a single putative Jun binding site, which can bind AP-1 directly both in vitro and in vivo, was mutated. Further, inhibition of PAK3 using siRNA showed a regression in the cell morphology, migratory potential and actin organisation associated with AP-1 transformed cells. Our study is a first to describe a role for AP-1 in regulating PAK3 expression and suggest that PAK3 is an AP-1 target required for actin organization and migration observed in transformed cells.


Assuntos
Actinas/metabolismo , Movimento Celular/fisiologia , Fibroblastos/metabolismo , Fator de Transcrição AP-1/metabolismo , Quinases Ativadas por p21/metabolismo , Animais , Sítios de Ligação/genética , Western Blotting , Linhagem Celular , Linhagem Celular Transformada , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Forma Celular/genética , Forma Celular/fisiologia , Doxiciclina/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Expressão Gênica , Humanos , Microscopia de Fluorescência , Mutação , Regiões Promotoras Genéticas/genética , Ligação Proteica , Interferência de RNA , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição AP-1/genética , Quinases Ativadas por p21/genética
18.
Crit Rev Eukaryot Gene Expr ; 23(1): 1-10, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23557333

RESUMO

Many proteins require transport across the nuclear envelope, the physical barrier separating the nucleus from the cytoplasm. Karyopherin ß (Kpnß1) proteins are the major nuclear receptor proteins in the cell that cargo proteins across the nuclear envelope, allowing them to enter and exit the cell nucleus. Karyopherin ß1, a major nuclear import receptor, plays an integral role in importing transcription factors, cell signaling proteins, cell cycle proteins, and so forth, into the nucleus, thus playing a crucial role in maintaining normal cell homeostasis. However, cancer cells appear to differentially regulate the expression of the Karyopherin ß proteins, presumably in order to maintain increased nuclear transport rates, thus implicating this protein family as a target for cancer therapy. The role of Kpnß1 in cancer is only now being elucidated, and recent work points to its potential usefulness as an anti-cancer target.


Assuntos
Terapia de Alvo Molecular , Neoplasias/genética , Membrana Nuclear , beta Carioferinas/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoplasma/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/patologia , Neoplasias/terapia , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , beta Carioferinas/metabolismo
19.
IUBMB Life ; 64(1): 87-98, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22131293

RESUMO

The extracellular matrix (ECM) provides the microenvironment that is pivotal for cell growth, motility, attachment, and differentiation. Advances in cell culture techniques have led to the development of cell-derived ECM model systems that are more reflective of the in vivo architecture of the ECM in tissue. In this study, a fibroblast-derived ECM (fd-ECM) was used to study the feedback regulation of type I collagen synthesis in fibroblasts. Fibroblasts plated on a preformed fd-ECM showed a significant decrease in the production of type I collagen and pro-α2(1) collagen mRNA compared to cells grown in the absence of a matrix. Function-blocking antibodies showed that this downregulation of type I collagen gene expression is mediated via α2ß1 integrin. The use of several kinase inhibitors and a dominant negative ras construct (N17Ras) showed that the matrix-mediated downregulation of COL1A2 occurs via Ras-dependent activation of the MEK/ERK signaling pathway. Deletion analysis of the COL1A2 promoter implicated the region between -375 and -107 as containing a potential matrix responsive element. The use of Sp1 siRNA demonstrated that Sp1 is an important mediator of this feedback inhibition. This study provides some new insights into the feedback regulation of COL1A2 gene expression.


Assuntos
Colágeno Tipo I/genética , Regulação da Expressão Gênica , Sistema de Sinalização das MAP Quinases , Sítios de Ligação , Linhagem Celular , Colágeno Tipo I/metabolismo , Regulação para Baixo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Humanos , Integrina alfa2beta1/metabolismo , Integrina alfa3beta1/metabolismo , Regiões Promotoras Genéticas , Elementos de Resposta , Fator de Transcrição Sp1/metabolismo , Proteínas ras/metabolismo
20.
PLoS One ; 6(11): e27723, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22125623

RESUMO

The Karyopherin superfamily comprises nuclear transport proteins, involved in the shuttling of certain cargo proteins into and out of the nucleus. Karyopherin ß1 (Kpnß1) and Karyopherin α2 (Kpnα2) are importin proteins, which work in concert to transport their cargo into the nucleus. We previously identified increased expression of Kpnß1 and Kpnα2 in cervical tumours compared to normal epithelium and in transformed cells compared to their normal counterparts. This study therefore aimed to identify the transcription regulatory mechanisms associated with high Kpnß1 and Kpnα2 levels in cancer cells. Kpnß1 (-2013 to +100) and Kpnα2 (-1900 to +69) promoter fragments were separately cloned into the reporter vector, pGL3-basic, and luciferase assays revealed both as significantly more active in cancer and transformed cells compared to normal. A series of deletion constructs identified the -637 to -271 Kpnß1 and -180 to -24 Kpnα2 promoter regions as responsible for the differential promoter activity, and a number of highly conserved E2F binding sites were identified within these regions. Mutation analysis confirmed the requirement of E2F sites for promoter activity, and ChIP analysis confirmed E2F2/Dp1 binding to the Kpnß1 and Kpnα2 promoters in vivo. Dp1 inhibition resulted in decreased levels of the respective proteins, confirming the role of E2F in the overexpression of Kpnß1 and Kpnα2 proteins in cancer. E2F activity is known to be deregulated in cervical cancer cells due to the inhibition of its repressor, Rb, by HPV E7. The inhibition of E7 using siRNA resulted in decreased Kpnß1 and Kpnα2 promoter activities, as did the overexpression of Rb. In conclusion, this study is a first to show that elevated Kpnß1 and Kpnα2 expression in cancer cells correlates with altered transcriptional regulation associated with deregulated E2F/Rb activities.


Assuntos
Fator de Transcrição E2F2/metabolismo , alfa Carioferinas/metabolismo , beta Carioferinas/metabolismo , Sequência de Bases , Sítios de Ligação/genética , Western Blotting , Linhagem Celular , Linhagem Celular Transformada , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Fator de Transcrição E2F2/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Luciferases/genética , Luciferases/metabolismo , Mutagênese Sítio-Dirigida , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Regiões Promotoras Genéticas/genética , Ligação Proteica , Interferência de RNA , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Deleção de Sequência , Fator de Transcrição DP1/genética , Fator de Transcrição DP1/metabolismo , alfa Carioferinas/genética , beta Carioferinas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA