Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Pharmazie ; 74(8): 462-466, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31526437

RESUMO

The potential uses of lyophilized cell free supernatant (CFS) of human oral derived Lactobacillus paracasei SD1 and Lactobacillus rhamnosus SD11 as cosmeceutical ingredients were investigated in the present study. Lyophilized CFS of both strains showed the antioxidant activity in concentration dependent manner. They also exhibited antimicrobial activity against P. acne, S. aureus and S. epidermidis. In combination, these two strains produced synergistic responses, not only on antioxidant activity but also on antimicrobial activity. A liposomal delivery system was employed to mask the unpleasant colour and odour of CFS. The optimal liposome formulation was characterized by a particle size of 344 nm, PDI of 0.19, zeta value of -48.05 mV and %EE of 69.45. The cytotoxicity results showed that the lyophilized CFS, which was toxic, became non-toxic after encapsulating into liposomes. Altogether, current findings demonstrate the worthiness of development of liposomes of probiotic's lyophilized CFS for cosmeceutical applications.


Assuntos
Cosmecêuticos/administração & dosagem , Cosmecêuticos/química , Lipossomos/administração & dosagem , Lipossomos/química , Probióticos/administração & dosagem , Probióticos/química , Anti-Infecciosos/administração & dosagem , Anti-Infecciosos/química , Antioxidantes/administração & dosagem , Antioxidantes/química , Liofilização , Humanos , Lacticaseibacillus paracasei/química , Lacticaseibacillus paracasei/metabolismo , Lacticaseibacillus rhamnosus/química , Lacticaseibacillus rhamnosus/metabolismo , Staphylococcus aureus/efeitos dos fármacos
2.
Oncogene ; 35(8): 990-1002, 2016 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-25961928

RESUMO

FOXM1 has been implicated in taxane resistance, but the molecular mechanism involved remains elusive. In here, we show that FOXM1 depletion can sensitize breast cancer cells and mouse embryonic fibroblasts into entering paclitaxel-induced senescence, with the loss of clonogenic ability, and the induction of senescence-associated ß-galactosidase activity and flat cell morphology. We also demonstrate that FOXM1 regulates the expression of the microtubulin-associated kinesin KIF20A at the transcriptional level directly through a Forkhead response element (FHRE) in its promoter. Similar to FOXM1, KIF20A expression is downregulated by paclitaxel in the sensitive MCF-7 breast cancer cells and deregulated in the paclitaxel-resistant MCF-7Tax(R) cells. KIF20A depletion also renders MCF-7 and MCF-7Tax(R) cells more sensitive to paclitaxel-induced cellular senescence. Crucially, resembling paclitaxel treatment, silencing of FOXM1 and KIF20A similarly promotes abnormal mitotic spindle morphology and chromosome alignment, which have been shown to induce mitotic catastrophe-dependent senescence. The physiological relevance of the regulation of KIF20A by FOXM1 is further highlighted by the strong and significant correlations between FOXM1 and KIF20A expression in breast cancer patient samples. Statistical analysis reveals that both FOXM1 and KIF20A protein and mRNA expression significantly associates with poor survival, consistent with a role of FOXM1 and KIF20A in paclitaxel action and resistance. Collectively, our findings suggest that paclitaxel targets the FOXM1-KIF20A axis to drive abnormal mitotic spindle formation and mitotic catastrophe and that deregulated FOXM1 and KIF20A expression may confer paclitaxel resistance. These findings provide insights into the underlying mechanisms of paclitaxel resistance and have implications for the development of predictive biomarkers and novel chemotherapeutic strategies for paclitaxel resistance.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos , Fatores de Transcrição Forkhead/fisiologia , Cinesinas/genética , Mitose , Paclitaxel/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Feminino , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Humanos , Cinesinas/metabolismo , Camundongos , Mitose/efeitos dos fármacos , Regiões Promotoras Genéticas , Fuso Acromático/fisiologia , Células Tumorais Cultivadas
3.
Oncogene ; 35(11): 1433-44, 2016 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-26148240

RESUMO

The forkhead transcription factor FOXM1 has a key role in DNA damage response, and its deregulated overexpression is associated with genotoxic drug resistance in breast cancer. However, little is known about the posttranslational mechanisms by which FOXM1 expression is regulated by genotoxic agents and how they are deregulated in resistant cells. Initial co-immunoprecipitation studies verified previous proteomic analysis finding that the OTUB1 is a novel FOXM1-interacting protein. Western blot analysis showed that both OTUB1 and FOXM1 expression reduced upon genotoxic agent treatment in MCF-7 cells, but remained relatively constant in resistant cells. FOXM1 expression reduced upon OTUB1 depletion by siRNA and increased with OTUB1 overexpression in MCF-7 cells, arguing that OTUB1 positively regulates FOXM1 expression. In agreement, co-immunoprecipitation experiments demonstrated that FOXM1 expression is associated with OTUB1 binding but inversely correlates with conjugation to the protein degradation-associated Lys-48-linked ubiquitin-chains. Overexpression of wild-type (WT) OTUB1, but not the OTUB1(C91S) mutant, disrupted the formation of Lys48-linked ubiquitin-conjugates on FOXM1. Importantly, knockdown of OTUB1 by siRNA resulted in an increase in turnover of FOXM1 in MCF-7 cells treated with the protein synthesis inhibitor cycloheximide, whereas overexpression of WT OTUB1, but not the OTUB1(C91S) mutant, significantly enhances the half-life of FOXM1. In addition, proliferative and clonogenic assays also show that OTUB1 can enhance the proliferative rate and epirubicin resistance through targeting FOXM1, as OTUB1 has little effect on FOXM1-deficient cells. The physiological relevance of the regulation of FOXM1 by OTUB1 is further underscored by the significant correlations between FOXM1 and OTUB1 expression in breast cancer patient samples. Cox-regression survival analysis indicates that OTUB1 overexpression is linked to poorer outcome in particular in patients treated with chemotherapy. Collectively, these data suggest that OTUB1 limits the ubiquitination and degradation of FOXM1 in breast cancer and has a key role in genotoxic agent resistance.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Neoplasias da Mama/genética , Cisteína Endopeptidases/genética , Resistencia a Medicamentos Antineoplásicos/genética , Epirubicina/farmacologia , Fatores de Transcrição Forkhead/metabolismo , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cicloeximida/farmacologia , Dano ao DNA/genética , Reparo do DNA/genética , Enzimas Desubiquitinantes , Feminino , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Células MCF-7 , Inibidores da Síntese de Proteínas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/genética , Ubiquitinação/genética
4.
Oncogenesis ; 4: e167, 2015 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-26344694

RESUMO

The forkhead transcription factor FOXK2 has recently been implicated in cancer cell proliferation and survival, but a role in cancer chemotherapeutic drug resistance has hitherto not been explored. Here we demonstrate that FOXK2 has a central role in mediating the cytotoxic drug response in breast cancer. Clonogenic and cell viability assays showed that enhanced FOXK2 expression sensitizes MCF-7 breast cancer cells to paclitaxel or epirubicin treatment, whereas FOXK2 depletion by small interfering RNAs (siRNAs) confers drug resistance. Our data also showed that the activation of the tumour suppressor FOXO3a by paclitaxel and epirubicin is mediated through the induction of FOXK2, as depletion of FOXK2 by siRNA limits the induction of FOXO3a by these drugs in MCF-7 cells. Chromatin immunoprecipitation (ChIP) analysis showed that in response to drug treatment, FOXK2 accumulates and binds to the proximal FOXO3a promoter region in MCF-7 cells. Furthermore, we also uncovered that FOXK2 is deregulated and, therefore, can express at high levels in the nucleus of both the paclitaxel and epirubicin drug-resistant MCF-7 cells. Our results showed that ectopically overexpressed FOXK2 accumulates in the nuclei of drug-resistant MCF-7 cells but failed to be recruited to target genes, including FOXO3a. Crucially, we found that FOXO3a is required for the anti-proliferative and epirubicin-induced cytotoxic function of FOXK2 in MCF-7 cells by sulphorhodamine and clonogenic assays. The physiological importance of the regulation of FOXO3a by FOXK2 is further confirmed by the significant correlations between FOXO3a and FOXK2 expression in breast carcinoma patient samples. Further survival analysis also reveals that high nuclear FOXK2 expression significantly associates with poorer clinical outcome, particularly in patients who have received conventional chemotherapy, consistent with our finding that FOXK2 is deregulated in drug-resistant cells. In summary, our results suggest that paclitaxel and epirubicin target the FOXK2 to modulate their cytotoxicity and deregulated FOXK2 confers drug resistance.

5.
Oncogene ; 33(32): 4144-55, 2014 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-24141789

RESUMO

FOXM1 is implicated in genotoxic drug resistance but its mechanism of action remains elusive. We show here that FOXM1-depletion can sensitize breast cancer cells and mouse embryonic fibroblasts (MEFs) into entering epirubicin-induced senescence, with the loss of long-term cell proliferation ability, the accumulation of γH2AX foci, and the induction of senescence-associated ß-galactosidase activity and cell morphology. Conversely, reconstitution of FOXM1 in FOXM1-deficient MEFs alleviates the accumulation of senescence-associated γH2AX foci. We also demonstrate that FOXM1 regulates NBS1 at the transcriptional level through an forkhead response element on its promoter. Like FOXM1, NBS1 is overexpressed in the epirubicin-resistant MCF-7Epi(R) cells and its expression level is low but inducible by epirubicin in MCF-7 cells. Consistently, overexpression of FOXM1 augmented and FOXM1 depletion reduced NBS1 expression and epirubicin-induced ataxia-telangiectasia mutated (ATM)phosphorylation in breast cancer cells. Together these findings suggest that FOXM1 increases NBS1 expression and ATM phosphorylation, possibly through increasing the levels of the MRN(MRE11/RAD50/NBS1) complex. Consistent with this idea, the loss of P-ATM induction by epirubicin in the NBS1-deficient NBS1-LBI fibroblasts can be rescued by NBS1 reconstitution. Resembling FOXM1, NBS1 depletion also rendered MCF-7 and MCF-7Epi(R) cells more sensitive to epirubicin-induced cellular senescence. In agreement, the DNA repair-defective and senescence phenotypes in FOXM1-deficent cells can be effectively rescued by overexpression of NBS1. Moreover, overexpression of NBS1 and FOXM1 similarly enhanced and their depletion downregulated homologous recombination (HR) DNA repair activity. Crucially, overexpression of FOXM1 failed to augment HR activity in the background of NBS1 depletion, demonstrating that NBS1 is indispensable for the HR function of FOXM1. The physiological relevance of the regulation of NBS1 expression by FOXM1 is further underscored by the strong and significant correlation between nuclear FOXM1 and total NBS1 expression in breast cancer patient samples, further suggesting that NBS1 as a key FOXM1 target gene involved in DNA damage response, genotoxic drug resistance and DNA damage-induced senescence.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Senescência Celular , Dano ao DNA , Resistencia a Medicamentos Antineoplásicos , Epirubicina/química , Fatores de Transcrição Forkhead/fisiologia , Proteínas Nucleares/fisiologia , Animais , Antibióticos Antineoplásicos/química , Proteínas de Ciclo Celular/genética , Reparo do DNA , Proteínas de Ligação a DNA , Fibroblastos/citologia , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/genética , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Células MCF-7 , Camundongos , Proteínas Nucleares/genética , Fenótipo , Fosforilação , Regiões Promotoras Genéticas , Transdução de Sinais
6.
Oncogene ; 33(34): 4316-29, 2014 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-24362530

RESUMO

The forkhead box transcription factor FOXM1 is an essential effector of G2/M-phase transition, mitosis and the DNA damage response. As such, it is frequently deregulated during tumorigenesis. Here we report that FOXM1 is dynamically modified by SUMO1 but not by SUMO2/3 at multiple sites. We show that FOXM1 SUMOylation is enhanced in MCF-7 breast cancer cells in response to treatment with epirubicin and mitotic inhibitors. Mutation of five consensus conjugation motifs yielded a SUMOylation-deficient mutant FOXM1. Conversely, fusion of the E2 ligase Ubc9 to FOXM1 generated an auto-SUMOylating mutant (FOXM1-Ubc9). Analysis of wild-type FOXM1 and mutants revealed that SUMOylation inhibits FOXM1 activity, promotes translocation to the cytoplasm and enhances APC/Cdh1-mediated ubiquitination and degradation. Further, expression of the SUMOylation-deficient mutant enhanced cell proliferation compared with wild-type FOXM1, whereas the FOXM1-Ubc9 fusion protein resulted in persistent cyclin B1 expression and slowed the time from mitotic entry to exit. In summary, our findings suggest that SUMOylation attenuates FOXM1 activity and causes mitotic delay in cytotoxic drug response.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Mitose , Proteína SUMO-1/metabolismo , Sumoilação , Antibióticos Antineoplásicos/farmacologia , Antígenos CD , Sítios de Ligação , Caderinas/metabolismo , Proliferação de Células/efeitos dos fármacos , Citoplasma/metabolismo , Resistencia a Medicamentos Antineoplásicos , Epirubicina/farmacologia , Proteína Forkhead Box M1 , Pontos de Checagem da Fase G2 do Ciclo Celular , Células HeLa , Humanos , Células MCF-7 , Nocodazol/farmacologia , Transporte Proteico , Proteólise
7.
Oncogene ; 32(39): 4634-45, 2013 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-23108394

RESUMO

FOXM1 is implicated in genotoxic drug resistance but its role and mechanism of action remain unclear. Here, we establish that γH2AX foci, indicative of DNA double-strand breaks (DSBs), accumulate in a time-dependent manner in the drug-sensitive MCF-7 cells but not in the resistant counterparts in response to epirubicin. We find that FOXM1 expression is associated with epirubicin sensitivity and DSB repair. Ectopic expression of FOXM1 can increase cell viability and abrogate DSBs sustained by MCF-7 cells following epirubicin, owing to an enhancement in repair efficiency. Conversely, alkaline comet and γH2AX foci formation assays show that Foxm1-null cells are hypersensitive to DNA damage, epirubicin and γ-irradiation. Furthermore, we find that FOXM1 is required for DNA repair by homologous recombination (HR) but not non-homologous end joining (NHEJ), using HeLa cell lines harbouring an integrated direct repeat green fluorescent protein reporter for DSB repair. We also identify BRIP1 as a direct transcription target of FOXM1 by promoter analysis and chromatin-immunoprecipitation assay. In agreement, depletion of FOXM1 expression by small interfering RNA downregulates BRIP1 expression at the protein and mRNA levels in MCF-7 and the epirubicin-resistant MCF-7 Epi(R) cells. Remarkably, the requirement for FOXM1 for DSB repair can be circumvented by reintroduction of BRIP1, suggesting that BRIP1 is an important target of FOXM1 in DSB repair. Indeed, like FOXM1, BRIP1 is needed for HR. These data suggest that FOXM1 regulates BRIP1 expression to modulate epirubicin-induced DNA damage repair and drug resistance.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/fisiologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Epirubicina/farmacologia , Fatores de Transcrição Forkhead/fisiologia , Proteínas de Neoplasias/fisiologia , RNA Helicases/fisiologia , Reparo de DNA por Recombinação/fisiologia , Animais , Dano ao DNA , DNA de Neoplasias/efeitos dos fármacos , DNA de Neoplasias/genética , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi , Feminino , Fibroblastos , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/antagonistas & inibidores , Raios gama , Histonas/análise , Humanos , Células MCF-7/efeitos dos fármacos , Células MCF-7/metabolismo , Células MCF-7/efeitos da radiação , Camundongos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , RNA Helicases/biossíntese , RNA Helicases/genética , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Neoplásico/biossíntese , RNA Interferente Pequeno/farmacologia , Tolerância a Radiação , Proteínas Recombinantes de Fusão/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...