Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Commun Biol ; 7(1): 26, 2024 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-38182748

RESUMO

Malignant melanoma is the most aggressive and deadly skin cancer with an increasing incidence worldwide whereas SCC is the second most common non-melanoma human skin cancer with limited treatment options. Here we show that the development and metastasis of melanoma and SCC cancers can be blocked by a combined opposite targeting of RhoA and p110δ PI3K. We found that a targeted induction of RhoA activity into tumours by deletion of p190RhoGAP-a potent inhibitor of RhoA GTPase-in tumour cells together with adoptive macrophages transfer from δD910A/D910A mice in mice bearing tumours with active RhoA abrogated growth progression of melanoma and SCC tumours. Τhe efficacy of this combined treatment is the same in tumours lacking activating mutations in BRAF and in tumours harbouring the most frequent BRAF(V600E) mutation. Furthermore, the efficiency of this combined treatment is associated with decreased ATX expression in tumour cells and tumour stroma bypassing a positive feedback expression of ATX induced by direct ATX pharmacological inactivation. Together, our findings highlight the importance of targeting cancer cells and macrophages for skin cancer therapy, emerge a reverse link between ATX and RhoA and illustrate the benefit of p110δ PI3K inhibition as a combinatorial regimen for the treatment of skin cancers.


Assuntos
Melanoma , Neoplasias Cutâneas , Humanos , Animais , Camundongos , Melanoma/genética , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas B-raf , Neoplasias Cutâneas/genética , Pele
2.
Protein J ; 39(3): 232-239, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32270414

RESUMO

BACKGROUND: Lysine specific demethylase 2B, KDM2B, regulates genes that participate in cellular development, morphogenesis, differentiation and metabolism as a component of the polycomb repressive complex 1 (PRC1). The CxxC finger of KDM2B is responsible for the DNA binding capacity of this epigenetic regulator, acting as a sampling mechanism across chromatin for gene repression OBJECTIVES: The molecular determinants of the CxxC-DNA interaction remain largely unknown, revealing a significant knowledge gap to be explored. Our goal was to elucidate the key residues of the CxxC domain that contribute to its function as well as to further elaborate on the significance of this domain in the KDM2B role METHODS: By using electrophoresis mobility swift assay, we identified structural elements of CxxC domain that participate in the DNA recognition. We created mouse embryonic fibroblasts overexpressing different truncated and point-mutated mouse KDM2B variants to examine the contribution of the KDM2B domains in replicative senescence bypass RESULTS: In this study, we show that only the CxxC finger is essential for the ability of mKDM2B to bypass replicative senescence in primary cells by ink4A-Arf-ink4B locus repression, and that this is mediated by specific interactions of residues R585, K608 and K616 with non-methylated CpG containing DNA CONCLUSIONS: These results provide new structural insights into the molecular interactions of CxxC and could serve as a stepping-stone for developing domain-specific inhibitors for KDM2B.


Assuntos
Fator 1 de Ribosilação do ADP/metabolismo , Senescência Celular/genética , Inibidor de Quinase Dependente de Ciclina p15/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , DNA/química , Proteínas F-Box/química , Histona Desmetilases com o Domínio Jumonji/química , Fator 1 de Ribosilação do ADP/genética , Animais , Sítios de Ligação , Diferenciação Celular , Ilhas de CpG , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , DNA/genética , DNA/metabolismo , Embrião de Mamíferos , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Camundongos , Mutação Puntual , Cultura Primária de Células , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transdução de Sinais
3.
Cancer Biol Ther ; 21(6): 533-540, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32175798

RESUMO

Recent studies revealed that the histone demethylase KDM2B regulates the epithelial markers E-Cadherin and ZO-1, the RhoA/B/C-small-GTPases and actin cytoskeleton organization, in DU-145 prostate- and HCT-116 colon-tumor cells. Here we addressed the role of KDM2B in the activation of Focal Adhesion Kinase (FAK)-signaling and its involvement in regulating tumor cell motility. We used RT-PCR for gene transcriptional analysis, Western blotting for the assessment of protein expression and activity and wound-healing assay for the study of cell migration. KDM2B overexpression or silencing controls the activity of FAK in DU-145 prostate- and HCT-116 colon-tumor cells without affecting gene transcription and protein expression of this kinase. Upon KDM2B overexpression in DU-145 cells, significantly enhanced migration was observed, which was abolished in cells pretreated by the specific phosphoinositide-3 kinase (PI3 K) inhibitor LY294002, implying involvement of FAK/PI3 K signaling in the migration process. In line with this, the p85-PI3 K-subunit was downregulated upon knockdown of KDM2B in DU-145 cells, while the opposite effect became evident in KDM2B-overexpressing cells. These results revealed a novel functional role of KDM2B in regulating the activation of the FAK/PI3 K signaling in prostate cancer cells that participates in the control of cell motility.


Assuntos
Movimento Celular , Neoplasias do Colo/patologia , Proteínas F-Box/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias da Próstata/patologia , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Proteínas F-Box/genética , Quinase 2 de Adesão Focal/genética , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Masculino , Fosfatidilinositol 3-Quinases/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Células Tumorais Cultivadas
4.
Cell Physiol Biochem ; 47(1): 368-377, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29772566

RESUMO

BACKGROUND/AIMS: The epigenetic factor KDM2B is a histone demethylase expressed in various tumors. Recently, we have shown that KDM2B regulates actin cytoskeleton organization, small Rho GTPases signaling, cell-cell adhesion and migration of prostate tumor cells. In the present study, we addressed its role in regulating EMT and small GTPases expression in colon tumor cells. METHODS: We used RT-PCR for the transcriptional analysis of various genes, Western blotting for the assessment of protein expression and immunofluorescence microscopy for visualization of fluorescently labeled proteins. RESULTS: We report here that KDM2B regulates EZH2 and BMI1 in HCT116 colon tumor cells. Knockdown of this epigenetic factor induced potent up-regulation of the protein levels of the epithelial markers E-cadherin and ZO-1, while the mesenchymal marker N-cadherin was downregulated. On the other hand, KDM2B overexpression downregulated the levels of both epithelial markers and upregulated the mesenchymal marker, suggesting control of EMT by KDM2B. In addition, RhoA, RhoB and RhoC protein levels diminished upon KDM2B-knockdown, while all three small GTPases became upregulated in KDM2B-overexpressing HCT116 cell clones. Interestingly, Rac1 GTPase level increased upon KDM2B-knockdown and diminished in KDM2B-overexpressing HCT116 colon tumor- and DU-145 prostate cancer cells. CONCLUSIONS: These results establish a clear functional role of the epigenetic factor KDM2B in the regulation of EMT and small-GTPases expression in colon tumor cells and further support the recently postulated oncogenic role of this histone demethylase in various tumors.


Assuntos
Neoplasias do Colo/genética , Transição Epitelial-Mesenquimal , Proteínas F-Box/genética , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/genética , Proteínas Monoméricas de Ligação ao GTP/genética , Neoplasias do Colo/patologia , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Epigênese Genética , Células HCT116 , Humanos
5.
Biochim Biophys Acta Mol Cell Res ; 1865(4): 587-597, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29408056

RESUMO

The histone demethylase KDM2B is an epigenetic factor with oncogenic properties that is regulated by the basic fibroblasts growth factor (FGF-2). It has recently been shown that KDM2B co-operates with Polycomb Group proteins to promote cell migration and angiogenesis in tumors. In the present study we addressed the role of KDM2B in regulating actin cytoskeleton signaling, cell-cell adhesion and migration of prostate tumor cells. We report here that KDM2B is functionally expressed in DU-145 prostate cancer cells, activated by FGF-2 and regulates EZH2. KDM2B knockdown induced potent up-regulation of gene transcription and protein expression of the epithelial markers E-cadherin and ZO-1, while KDM2B overexpression down-regulated the levels of both markers, suggesting control of cell adhesion by KDM2B. RhoA and RhoB protein expression and activity were diminished upon KDM2B-knockdown and upregulated in KDM2B-overexpressing cell clones. In accordance, actin reorganization with formation of stress fibers became evident in KDM2B-overexpressing cells and abolished in the presence of the Rho inhibitor C3 transferase. DU-145 cell migration was significantly enhanced in KDM2B overexpressing cells and abolished in C3-pretreated cells. Conversely, the retardation of cell migration observed in KDM2B knockdown cells was enhanced in C3-pretreated cells. These results establish a clear functional link between the epigenetic factor KDM2B and the regulation of cell adhesion and Rho-GTPases signaling that controls actin reorganization and cell migration.


Assuntos
Citoesqueleto de Actina/metabolismo , Movimento Celular , Epigênese Genética , Proteínas F-Box/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas rho de Ligação ao GTP/metabolismo , Antígenos CD , Biomarcadores Tumorais/metabolismo , Caderinas/metabolismo , Adesão Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Proteínas F-Box/genética , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Masculino , Modelos Biológicos , Neoplasias da Próstata/genética , Proteína da Zônula de Oclusão-1/metabolismo
6.
Cell Physiol Biochem ; 42(4): 1366-1376, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28704809

RESUMO

BACKGROUND/AIMS: Istaroxime is a validated inotropic Na+/K+ ATPase inhibitor currently in development for the treatment of various cardiac conditions. Recent findings established that this steroidal drug exhibits potent apoptotic responses in prostate tumors in vitro and in vivo, by affecting key signaling orchestrating proliferation and apoptosis, such as c-Myc and caspase 3, Rho GTPases and actin cytoskeleton dynamics. In the present study we examined whether istaroxime is affecting cell motility and analyzed the underlying mechanism in prostate tumor cells. METHODS: Migration was assessed by transwell and wound healing assays, Orai1 and Stim1 abundance by RT-PCR and confocal immunofluorescence microscopy, Fura-2 fluorescence was utilized to determine intracellular Ca2+ and Western blotting for FAK/pFAK measurements. RESULTS: We observed strong inhibition of cell migration in istaroxime treated DU-145 prostate cancer cells. Istaroxime further decreased Orai1 and Stim1 transcript levels and downregulated Orai1 protein expression. Moreover, SOCE was significantly decreased upon istaroxime treatment. Furthermore, istaroxime strikingly diminished phosphorylated FAK levels. Interestingly, the efficacy of istaroxime on the inhibition of DU-145 cell migration was further enhanced by blocking Orai1 with 2-APB and FAK with the specific inhibitor PF-00562271. These results provide strong evidence that istaroxime prevents cell migration and motility of DU-145 prostate tumor cells, an effect at least partially attributed to Orai1 downregulation and FAK de-activation. CONCLUSION: Collectively our results indicate that this enzyme inhibitor, besides its pro-apoptotic action, affects motility of cancer cells, supporting its potential role as a strong candidate for further clinical cancer drug development.


Assuntos
Movimento Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Etiocolanolona/análogos & derivados , Quinase 1 de Adesão Focal/genética , Regulação Neoplásica da Expressão Gênica , Proteína ORAI1/genética , Bloqueadores dos Canais de Sódio/farmacologia , Cálcio/metabolismo , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Linhagem Celular Tumoral , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Etiocolanolona/farmacologia , Corantes Fluorescentes/química , Quinase 1 de Adesão Focal/antagonistas & inibidores , Quinase 1 de Adesão Focal/metabolismo , Fura-2/química , Humanos , Masculino , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/antagonistas & inibidores , Proteína ORAI1/metabolismo , Fosforilação/efeitos dos fármacos , Próstata/efeitos dos fármacos , Próstata/metabolismo , Próstata/patologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Transdução de Sinais , Molécula 1 de Interação Estromal/antagonistas & inibidores , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo , Sulfonamidas/farmacologia
7.
Dev Cell ; 37(5): 473-83, 2016 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-27270042

RESUMO

Membrane contact sites between the ER and multivesicular endosomes/bodies (MVBs) play important roles in endosome positioning and fission and in neurite outgrowth. ER-MVB contacts additionally function in epidermal growth factor receptor (EGFR) tyrosine kinase downregulation by providing sites where the ER-localized phosphatase, PTP1B, interacts with endocytosed EGFR before the receptor is sorted onto intraluminal vesicles (ILVs). Here we show that these contacts are tethered by annexin A1 and its Ca(2+)-dependent ligand, S100A11, and form a subpopulation of differentially regulated contact sites between the ER and endocytic organelles. Annexin A1-regulated contacts function in the transfer of ER-derived cholesterol to the MVB when low-density lipoprotein-cholesterol in endosomes is low. This sterol traffic depends on interaction between ER-localized VAP and endosomal oxysterol-binding protein ORP1L, and is required for the formation of ILVs within the MVB and thus for the spatial regulation of EGFR signaling.


Assuntos
Anexina A1/metabolismo , Colesterol/metabolismo , Retículo Endoplasmático/metabolismo , Endossomos/metabolismo , Transporte Biológico/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/ultraestrutura , Endossomos/efeitos dos fármacos , Endossomos/ultraestrutura , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/metabolismo , Células HeLa , Humanos , Lipoproteínas LDL/farmacologia , Corpos Multivesiculares/efeitos dos fármacos , Corpos Multivesiculares/metabolismo , Corpos Multivesiculares/ultraestrutura , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Receptores de Esteroides/metabolismo , Vesículas Transportadoras/efeitos dos fármacos , Vesículas Transportadoras/metabolismo , Proteínas de Transporte Vesicular/metabolismo
8.
Oncotarget ; 7(17): 24415-28, 2016 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-27027435

RESUMO

Sodium potassium pump (Na+/K+ ATPase) is a validated pharmacological target for the treatment of various cardiac conditions. Recent published data with Na+/K+ ATPase inhibitors suggest a potent anti-cancer action of these agents in multiple indications. In the present study, we focus on istaroxime, a Na+/K+ ATPase inhibitor that has shown favorable safety and efficacy properties in cardiac phase II clinical trials. Our experiments in 22 cancer cell lines and in prostate tumors in vivo proved the strong anti-cancer action of this compound. Istaroxime induced apoptosis, affected the key proliferative and apoptotic mediators c-Myc and caspase-3 and modified actin cystoskeleton dynamics and RhoA activity in prostate cancer cells. Interestingly, istaroxime was capable of binding to mAR, a membrane receptor mediating rapid, non-genomic actions of steroids in prostate and other cells. These results support a multi-level action of Na+/K+ ATPase inhibitors in cancer cells and collectively validate istaroxime as a strong re-purposing candidate for further cancer drug development.


Assuntos
Etiocolanolona/análogos & derivados , Neoplasias da Próstata/tratamento farmacológico , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios Clínicos Fase II como Assunto , Etiocolanolona/metabolismo , Etiocolanolona/farmacologia , Feminino , Células HCT116 , Humanos , Células MCF-7 , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias da Próstata/metabolismo , Ligação Proteica , Receptores Androgênicos/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Carga Tumoral/efeitos dos fármacos
9.
Cell Physiol Biochem ; 37(1): 399-408, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26316086

RESUMO

BACKGROUND/AIMS: Chorein is a protein expressed in various cell types. Loss of function mutations of the chorein encoding gene VPS13A lead to chorea-acanthocytosis, an autosomal recessive genetic disease characterized by movement disorder and behavioral abnormalities. Recent observations revealed that chorein is a powerful regulator of actin cytoskeleton in erythrocytes, platelets, K562 and endothelial HUVEC cells. METHODS: In the present study we have used Western blotting to study actin polymerization dynamics, laser scanning microscopy to evaluate in detail the role of chorein in microfilaments, microtubules and intermediate filaments cytoskeleton architecture and RT-PCR to assess gene transcription of the cytoskeletal proteins. RESULTS: We report here powerful depolymerization of actin microfilaments both, in erythrocytes and fibroblasts isolated from chorea-acanthocytosis patients. Along those lines, morphological analysis of fibroblasts from chorea-acanthocytosis patients showed disarranged microtubular network, when compared to fibroblasts from healthy donors. Similarly, the intermediate filament networks of desmin and cytokeratins showed significantly disordered organization with clearly diminished staining in patient's fibroblasts. In line with this, RT-PCR analysis revealed significant downregulation of desmin and cytokeratin gene transcripts. CONCLUSION: Our results provide for the first time evidence that defective chorein is accompanied by significant structural disorganization of all cytoskeletal structures in human fibroblasts from chorea-acanthocytosis patients.


Assuntos
Citoesqueleto/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Actinas/genética , Actinas/metabolismo , Plaquetas/metabolismo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Desmina/genética , Desmina/metabolismo , Regulação para Baixo/genética , Eritrócitos/metabolismo , Fibroblastos/metabolismo , Humanos , Neuroacantocitose/genética , Neuroacantocitose/metabolismo , Transcrição Gênica/genética
10.
Oncotarget ; 6(12): 10309-19, 2015 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-25871399

RESUMO

Chorein encoded by VPS13A (vacuolar protein sorting-associated protein 13A) is defective in chorea-acanthocytosis. Chorein fosters neuronal cell survival, cortical actin polymerization and cell stiffness. In view of its anti-apoptotic effect in neurons, we explored whether chorein is expressed in cancer cells and influences cancer cell survival. RT-PCR was employed to determine transcript levels, specific siRNA to silence chorein, FACS analysis to follow apoptosis and Western blotting to quantify protein abundance. Chorein transcripts were detected in various cancer cell types. The mRNA coding for chorein and chorein protein were most abundant in drug resistant, poorly differentiated human rhabdomyosarcoma cells. Chorein silencing significantly reduced the ratio of phosphorylated (and thus activated) to total phosphoinositide 3 kinase (PI-3K), pointing to inactivation of this crucial pro-survival signaling molecule. Moreover, chorein silencing diminished transcript levels and protein expression of anti-apoptotic BCL-2 and enhanced transcript levels of pro-apoptotic Bax. Silencing of chorein in rhabdomyosarcoma cells was followed by mitochondrial depolarization, caspase 3 activation and stimulation of early and late apoptosis. In conclusion, chorein is expressed in various cancer cells. In cells with high chorein expression levels chorein silencing promotes apoptotic cell death, an effect paralleled by down-regulation of PI-3K activity and BCL-2/Bax expression ratio.


Assuntos
Neuroacantocitose/metabolismo , Rabdomiossarcoma/metabolismo , Proteínas de Transporte Vesicular/biossíntese , Células CACO-2 , Criança , Feminino , Humanos , Neuroacantocitose/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Rabdomiossarcoma/genética , Transfecção , Células Tumorais Cultivadas , Proteínas de Transporte Vesicular/genética
11.
Nat Cell Biol ; 12(3): 267-72, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20118922

RESUMO

The epidermal growth factor receptor (EGFR) is a critical determinator of cell fate. Signalling from this receptor tyrosine kinase is spatially regulated by progression through the endocytic pathway, governing receptor half-life and accessibility to signalling proteins and phosphatases. Endocytosis of EGFR is required for interaction with the protein tyrosine phosphatase PTP1B (ref. 1), which localizes to the cytoplasmic face of the endoplasmic reticulum (ER), raising the question of how PTP1B comes into contact with endosomal EGFR. We show that EGFR-PTP1B interaction occurs by means of direct membrane contacts between the perimeter membrane of multivesicular bodies (MVBs) and the ER. The population of EGFR interacting with PTP1B is the same population that undergo ESCRT-mediated (endosomal sorting complex required for transport) sorting within MVBs, and PTP1B activity promotes the sequestration of EGFR on to MVB internal vesicles. Membrane contacts between endosomes and the ER form in both the presence and absence of stimulation by EGF. Thus membrane contacts between endosomes and the ER may represent a global mechanism for direct interaction between proteins on these two organelles.


Assuntos
Retículo Endoplasmático/metabolismo , Endossomos/metabolismo , Receptores ErbB/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Retículo Endoplasmático/ultraestrutura , Retículo Endoplasmático Liso/metabolismo , Retículo Endoplasmático Liso/ultraestrutura , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Endossomos/ultraestrutura , Fator de Crescimento Epidérmico/metabolismo , Células HeLa , Humanos , Leupeptinas/farmacologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Microscopia Eletrônica , Corpos Multivesiculares/metabolismo , Corpos Multivesiculares/ultraestrutura , Mutação/fisiologia , Fosfoproteínas/metabolismo , Fosforilação/fisiologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 1/genética , RNA Interferente Pequeno/genética , Transfecção
12.
Mol Biol Cell ; 21(6): 860-70, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20089843

RESUMO

Maintenance of the epithelial phenotype is crucial for tissue homeostasis. In the retina, dedifferentiation and loss of integrity of the retinal pigment epithelium (RPE) leads to retinal dysfunction and fibrosis. Transforming growth factor (TGF)-beta critically contributes to RPE dedifferentiation and induces various responses, including increased Rho signaling, up-regulation of alpha-smooth muscle actin (SMA), and cell migration and dedifferentiation. Cellular TGF-beta responses are stimulated by different signal transduction pathways: some are Smad dependent and others Smad independent. Alterations in Rho signaling are crucial to both types of TGF-beta signaling, but how TGF-beta-stimulates Rho signaling is poorly understood. Here, we show that primary RPE cells up-regulated GEF-H1 in response to TGF-beta. GEF-H1 was the only detectable Rho exchange factor increased by TGF-beta1 in a genome-wide expression analysis. GEF-H1 induction was Smad4-dependant and led to Rho activation. GEF-H1 inhibition counteracted alpha-SMA up-regulation and cell migration. In patients with retinal detachments and fibrosis, migratory RPE cells exhibited increased GEF-H1 expression, indicating that induction occurs in diseased RPE in vivo. Our data indicate that GEF-H1 is a target and functional effector of TGF-beta by orchestrating Rho signaling to regulate gene expression and cell migration, suggesting that it represents a new marker and possible therapeutic target for degenerative and fibrotic diseases.


Assuntos
Actinas/metabolismo , Movimento Celular/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Actinas/genética , Animais , Adesão Celular/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Regulação da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Análise em Microsséries , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/fisiologia , Fatores de Troca de Nucleotídeo Guanina Rho , Transdução de Sinais/fisiologia , Proteína Smad4/metabolismo , Suínos , Proteína rhoA de Ligação ao GTP/genética
13.
J Cell Sci ; 119(Pt 24): 5098-105, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17158914

RESUMO

Epithelial tight junctions participate in the regulation of gene expression by controlling the activity of transcription factors that can interact with junctional components. One such protein is the Y-box transcription factor ZONAB/DbpA that binds to ZO-1, a component of the junctional plaque. Symplekin, another nuclear protein that can associate with tight junctions, functions in the regulation of polyadenylation and thereby promotes gene expression. Here, we addressed the question of whether these two proteins interact and whether this is of functional relevance. We demonstrate that ZONAB/DbpA and symplekin form a complex in kidney and intestinal epithelial cells that can be immunoprecipitated and that exists in the nucleus. The interaction between ZONAB/DbpA and symplekin can be reconstituted with recombinant proteins. In reporter gene assays in which ZONAB/DbpA functions as a repressor, symplekin functionally interacts with ZONAB/DbpA, indicating that symplekin can also promote transcriptional repression. RNAi experiments indicate that symplekin depletion reduces the nuclear accumulation and the transcriptional activity of ZONAB/DbpA in colon adenocarcinoma cells, resulting in inhibition of proliferation and reduced expression of the ZONAB/DbpA-target gene cyclin D1. Our data thus indicate that symplekin and ZONAB/DbpA cooperate in the regulation of transcription, and that they promote epithelial proliferation and cyclin D1 expression.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células CACO-2 , Linhagem Celular , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Cães , Células Epiteliais/metabolismo , Imunofluorescência , Humanos , Imunoprecipitação , Proteínas de Membrana/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Ligação Proteica , Interferência de RNA , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Junções Íntimas/genética , Junções Íntimas/metabolismo , Fatores de Transcrição/genética , Proteína da Zônula de Oclusão-1
14.
Mol Cell Biol ; 26(6): 2387-98, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16508013

RESUMO

The tight junction protein ZO-1 inhibits G1/S-phase transition by cytoplasmic sequestration of a complex formed by CDK4 and the transcription factor ZONAB. Canine ZONAB is the homologue of human DbpA, an E2F target gene that is overexpressed in different carcinomas. Since the ZONAB target genes that are involved in G1/S-phase transition are unknown, we employed the mammary epithelial cell line MCF-10A and cDNA arrays to screen for such genes. We identified genes encoding cell cycle and replication proteins whose expression was altered due to increased ZONAB expression. For proliferative cell nuclear antigen and cyclin D1 genes, we show that increased mRNA levels resulted in increased protein levels and we identified ZONAB-responsive elements in their promoters by using different approaches, including chromatin immunoprecipitation assays. RNA interference and overexpression of ZONAB affected the proliferation of both MCF-10A and MDCK cells as well as the differentiation of MDCK cells into polarized cysts in three-dimensional cultures. These results indicate that ZONAB regulates the transcription of genes that are important for G1/S-phase progression and links tight junctions to the transcriptional control of key cell cycle regulators and epithelial cell differentiation.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Ciclina D1/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais/citologia , Proteínas de Choque Térmico/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proteínas Estimuladoras de Ligação a CCAAT/genética , Técnicas de Cultura de Células , Ciclo Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Ciclina D1/genética , Proteínas de Ligação a DNA/genética , Cães , Células Epiteliais/metabolismo , Feminino , Proteínas de Choque Térmico/genética , Humanos , Glândulas Mamárias Humanas/citologia , Proteínas de Membrana/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fosfoproteínas/metabolismo , Antígeno Nuclear de Célula em Proliferação/genética , Regiões Promotoras Genéticas , Elementos de Resposta , Transdução de Sinais , Fatores de Transcrição/genética , Regulação para Cima , Proteína da Zônula de Oclusão-1
15.
Mol Biol Cell ; 17(3): 1322-30, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16407410

RESUMO

The tight junction adaptor protein ZO-1 regulates intracellular signaling and cell proliferation. Its Src homology 3 (SH3) domain is required for the regulation of proliferation and binds to the Y-box transcription factor ZO-1-associated nucleic acid binding protein (ZONAB). Binding of ZO-1 to ZONAB results in cytoplasmic sequestration and hence inhibition of ZONAB's transcriptional activity. Here, we identify a new binding partner of the SH3 domain that modulates ZO-1-ZONAB signaling. Expression screening of a cDNA library with a fusion protein containing the SH3 domain yielded a cDNA coding for Apg-2, a member of the heat-shock protein 110 (Hsp 110) subfamily of Hsp70 heat-shock proteins, which is overexpressed in carcinomas. Regulated depletion of Apg-2 in Madin-Darby canine kidney cells inhibits G(1)/S phase progression. Apg-2 coimmunoprecipitates with ZO-1 and partially localizes to intercellular junctions. Junctional recruitment and coimmunoprecipitation with ZO-1 are stimulated by heat shock. Apg-2 competes with ZONAB for binding to the SH3 domain in vitro and regulates ZONAB's transcriptional activity in reporter gene assays. Our data hence support a model in which Apg-2 regulates ZONAB function by competing for binding to the SH3 domain of ZO-1 and suggest that Apg-2 functions as a regulator of ZO-1-ZONAB signaling in epithelial cells in response to cellular stress.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Proteínas de Choque Térmico HSP110/metabolismo , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Junções Íntimas/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica/genética , Animais , Células Cultivadas , Cães , Células Epiteliais/citologia , Fase G1/fisiologia , Proteínas de Choque Térmico HSP110/química , Proteínas de Choque Térmico HSP110/deficiência , Hipertermia Induzida , Ligação Proteica , Fase S/fisiologia , Transdução de Sinais , Proteína da Zônula de Oclusão-1
16.
Curr Opin Cell Biol ; 17(5): 453-8, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16098725

RESUMO

Tight junctions are important for the permeability properties of epithelial and endothelial barriers as they restrict diffusion along the paracellular space. Recent observations have revealed that tight junctions also function in the regulation of epithelial proliferation and differentiation. They harbour evolutionarily conserved protein complexes that regulate polarisation and junction assembly. Tight junctions also recruit signalling proteins that participate in the regulation of cell proliferation and differentiation. These signalling proteins include components that affect established signalling cascades and dual localisation proteins that can associate with junctions as well as travel to the nucleus where they regulate gene expression.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células , Células Epiteliais/fisiologia , Junções Íntimas/fisiologia , Animais , Permeabilidade da Membrana Celular/fisiologia , Células Epiteliais/metabolismo , Membranas Intracelulares/metabolismo , Membranas Intracelulares/fisiologia , Mamíferos , Modelos Biológicos , Transdução de Sinais/fisiologia , Junções Íntimas/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia
17.
Exp Cell Res ; 288(1): 94-109, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12878162

RESUMO

The mechanisms through which opioids regulate the activity of malignant breast epithelial cells are currently unknown. In the present study we report the differential actin cytoskeleton reorganization induced by opioids in malignant (MCF7) and nonmalignant (MCF12A) breast epithelial cells expressing functional opioid receptors. Exposure of MCF7 cells to the opioid agonist alpha(s1) casomorphin induced important actin assembly and reorganization, including the formation of filopodia and lamellipodia. In contrast, incubation of MCF12A cells with alpha(s1) casomorphin revealed a partial but transient disassembly of actin microfilaments. Immunoprecipitation and immunoblot analyses showed rapid phosphorylation of focal adhesion kinase (FAK) and vinculin in opioid-treated MCF7 cells. Moreover, FAK associates with phosphatidylinositol-3 (PI-3 kinase), the latter being subsequently phosphorylated and activated. In addition, a substantial activation of the small GTPase Rac1 was observed. Pretreatment of MCF7 cells with the specific PI-3 kinase inhibitor wortmannin abolished both the activation of Rac1 and actin reorganization, while the opioid-induced phosphorylation of FAK and vinculin remained unaffected. Interestingly, in opioid-treated MCF12A cells this signaling cascade remained inactive, while we identified rapid phosphorylation of actin regulating the protein villin. Finally, opioids differentially inhibited cell motility in each cell line. Our data suggest a distinct, opioid-induced, signaling pathway activated in malignant breast epithelial cells, leading to important actin reorganization. These findings may indicate a potential antineoplastic role of opiates, based on the activation of differential signaling mechanisms.


Assuntos
Actinas/efeitos dos fármacos , Neoplasias da Mama/patologia , Citoesqueleto/efeitos dos fármacos , Entorpecentes/farmacologia , Transdução de Sinais/fisiologia , Antineoplásicos/farmacologia , Caseínas/farmacologia , Divisão Celular/efeitos dos fármacos , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Feminino , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Microscopia Confocal , Fragmentos de Peptídeos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Células Tumorais Cultivadas , Vinculina/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...