Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Exp Cell Res ; 431(1): 113739, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37567436

RESUMO

Oral cancer is a common malignant tumor of the oral cavity that affects many countries with a prevalent distribution in the Indian subcontinent, with poor prognosis rate on account of locoregional metastases. Gain-of-function mutations in p53 and overexpression of its related transcription factor, p63 are both widely reported events in oral cancers. However, targeting these alterations remains a far-achieved aim due to lack of knowledge on their downstream signaling pathways. In the present study, we characterize the isoforms of p63 and using knockdown strategy, decipher the functions and oncogenic signaling of p63 in oral cancers. Using Microarray and Chromatin Immunoprecipitation experiments, we decipher a novel transcriptional regulatory axis between p63 and Activin A and establish its functional significance in migration of oral cancer cells. Using an orally bioavailable inhibitor of the Activin A pathway to attenuate oral cancer cell migration and invasion, we further demonstrate the targetability of this signaling axis. Our study highlights the oncogenic role of ΔNp63 - Activin A - SMAD2/3 signaling and provides a basis for targeting this oncogenic pathway in oral cancers.


Assuntos
Ativinas , Neoplasias Bucais , Fatores de Transcrição , Proteínas Supressoras de Tumor , Humanos , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/genética , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Movimento Celular , Transdução de Sinais , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Ativinas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
J Oral Pathol Med ; 52(9): 849-859, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37573872

RESUMO

BACKGROUND: Translationally controlled tumour protein (TCTP) is a multifunctional protein elevated in multiple cancers. However, studies on its role in oral carcinogenesis and prognosis are rare. We recently reported the role of its interacting partner, MCL1, in oral cancer progression and outcome. Hence, the present study aimed to assess TCTP expression in oral tumorigenesis and its association with patient outcomes alone and in combination with MCL1. METHODS: TCTP expression was assessed by immunohistochemistry and immunoblotting in oral tissues and cells, respectively. Cell viability post siRNA/dihydroartemisinin treatment was analysed by tetrazolium salt assay. Cell survival, invasion and tumorigenic potential post TCTP knockdown were assessed by clonogenic, Matrigel and soft-agar assays, respectively. The association of TCTP with patient outcome was analysed by Kaplan-Meier and Cox regression. RESULTS: TCTP was significantly overexpressed in oral premalignant lesions (p < 0.0001), oral tumours (p < 0.0001) and oral dysplastic and cancer cells versus normal oral mucosa and also in recurrent (p < 0.05) versus non-recurrent oral tumours. Further, elevated TCTP was significantly (p < 0.05) associated with poor recurrence free survival (RFS) and poor overall survival (OS; hazard ratio = 2.29; p < 0.05). Intriguingly, the high co-expression of TCTP and MCL1 further reduced the RFS (p < 0.05) and OS (p < 0.05; hazard-ratio = 3.49; p < 0.05). Additionally, TCTP knockdown decreased survival (p < 0.05), invasion (p < 0.01) and in vitro tumorigenic potential (p < 0.0001). Dihydroartemisinin treatment reduced TCTP levels and viability of oral cancer cells. CONCLUSION: Our studies demonstrate an oncogenic role of TCTP in oral cancer progression and poor outcome. Thus, TCTP may be a potential prognostic marker and therapeutic target in oral cancers.


Assuntos
Artemisininas , Neoplasias Bucais , Humanos , Artemisininas/farmacologia , Artemisininas/uso terapêutico , Biomarcadores Tumorais/metabolismo , Neoplasias Bucais/genética , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteína Tumoral 1 Controlada por Tradução
3.
Biochem Biophys Res Commun ; 620: 1-7, 2022 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-35772211

RESUMO

Loss of the desmosomal plaque protein plakophilin3 (PKP3) leads to increased tumor progression and metastasis. As metastatic tumors are often resistant to therapy, we wished to determine whether PKP3 loss led to increased radioresistance. PKP3 knockdown cells showed increased resistance to radiation in vitro and in vivo. The increase in resistance was accompanied by an increased ability to clear reactive oxygen species (ROS) and increased autophagy. The increase in autophagy was required for radioresistance and ROS clearance as inhibiting autophagy using either chloroquine or knocking down ATG3 re-sensitized the PKP3 knockdown clones to radiotherapy. These experiments suggest that autophagy inhibitors could target therapy-resistant PKP3 deficient tumors.


Assuntos
Neoplasias , Placofilinas , Autofagia/genética , Linhagem Celular Tumoral , Células Clonais/metabolismo , Humanos , Neoplasias/metabolismo , Placofilinas/genética , Placofilinas/metabolismo , Espécies Reativas de Oxigênio
4.
Biochem Biophys Res Commun ; 586: 14-19, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34823217

RESUMO

Plakophilin3 (PKP3) loss leads to tumor progression and metastasis of colon cancer cells. The goal of this report was to determine if PKP3 loss led to increased disease progression in mice. We generated a colonocyte-specific knockout of PKP3 in APCmin mice, which led to increased adenoma formation, the formation of rectal prolapse, and a significant decrease in survival. The observed increase in rectal prolapse formation and decrease in survival correlated with an increase in the expression of Lipocalin2 (LCN2). Increased disease progression was observed even upon treatment with 5-fluorouracil (5FU). These results suggest that an increase in LCN2 expression might lead to therapy resistance and that LCN2 might serve as a potential therapeutic target in colorectal cancer.


Assuntos
Adenoma/genética , Neoplasias Colorretais/genética , Resistencia a Medicamentos Antineoplásicos/genética , Lipocalina-2/genética , Placofilinas/genética , Prolapso Retal/genética , Adenoma/tratamento farmacológico , Adenoma/mortalidade , Adenoma/patologia , Animais , Antimetabólitos Antineoplásicos/farmacologia , Colo/efeitos dos fármacos , Colo/metabolismo , Colo/patologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Progressão da Doença , Feminino , Fluoruracila/farmacologia , Regulação Neoplásica da Expressão Gênica , Queratina-8/genética , Queratina-8/metabolismo , Lipocalina-2/metabolismo , Masculino , Metaloproteinase 7 da Matriz/genética , Metaloproteinase 7 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Placofilinas/deficiência , Prolapso Retal/tratamento farmacológico , Prolapso Retal/mortalidade , Prolapso Retal/patologia , Transdução de Sinais , Análise de Sobrevida
5.
Genes Cells ; 26(6): 426-446, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33813791

RESUMO

14-3-3 proteins bind to ligands via phospho-serine containing consensus motifs. However, the molecular mechanisms underlying complex formation and dissociation between 14-3-3 proteins and their ligands remain unclear. We identified two conserved acidic residues in the 14-3-3 peptide-binding pocket (D129 and E136) that potentially regulate complex formation and dissociation. Altering these residues to alanine led to opposing effects on centrosome duplication. D129A inhibited centrosome duplication, whereas E136A stimulated centrosome amplification. These results were due to the differing abilities of these mutant proteins to form a complex with NPM1. Inhibiting complex formation between NPM1 and 14-3-3γ led to an increase in centrosome duplication and over-rode the ability of D129A to inhibit centrosome duplication. We identify a novel role of 14-3-3γ in regulating centrosome licensing and a novel mechanism underlying the formation and dissociation of 14-3-3 ligand complexes dictated by conserved residues in the 14-3-3 family.


Assuntos
Proteínas 14-3-3/metabolismo , Centrossomo/metabolismo , Proteínas Nucleares/metabolismo , Fosfopeptídeos/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Centríolos/metabolismo , Células HCT116 , Células HEK293 , Humanos , Modelos Biológicos , Proteínas Mutantes/metabolismo , Nucleofosmina , Fenótipo , Fosfopeptídeos/química , Fosforilação , Multimerização Proteica , Quinases Associadas a rho/metabolismo
6.
Int J Cancer ; 149(7): 1495-1511, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34146401

RESUMO

Lipocalin 2 is a siderophore-binding protein that regulates iron homeostasis. Lipocalin 2 expression is elevated in multiple tumor types; however, the mechanisms that drive tumor progression upon Lipocalin 2 expression remain unclear. When Lipocalin 2 is over-expressed, it leads to resistance to 5-fluorouracil in colon cancer cell lines in vitro and in vivo by inhibiting ferroptosis. Lipocalin 2 inhibits ferroptosis by decreasing intracellular iron levels and stimulating the expression of glutathione peroxidase4 and a component of the cysteine glutamate antiporter, xCT. The increase in xCT levels is dependent on increased levels of ETS1 in Lipocalin 2 over-expressing cells. Inhibiting Lipocalin 2 function with a monoclonal antibody leads to a decrease in chemo-resistance and transformation in vitro, and a decrease in tumor progression and chemo-resistance in xenograft mouse models. Lipocalin 2 and xCT levels exhibit a positive correlation in human tumor samples suggesting that the pathway we have identified in cell lines is operative in human tumor samples. These results indicate that Lipocalin 2 is a potential therapeutic target and that the monoclonal antibody described in our study can serve as the basis for a potential therapeutic in patients who do not respond to chemotherapy.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Fluoruracila/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Lipocalina-2/metabolismo , Animais , Antimetabólitos Antineoplásicos/farmacologia , Apoptose , Biomarcadores Tumorais/genética , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Humanos , Lipocalina-2/genética , Camundongos , Camundongos Nus , Prognóstico , Espécies Reativas de Oxigênio/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Exp Cell Res ; 369(2): 251-265, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29803740

RESUMO

An increase in tumour formation and metastasis are observed upon plakophilin3 (PKP3) loss. To identify pathways downstream of PKP3 loss that are required for increased tumour formation, a gene expression analysis was performed, which demonstrated that the expression of lipocalin2 (LCN2) was elevated upon PKP3 loss and this is consistent with expression data from human tumour samples suggesting that PKP3 loss correlates with an increase in LCN2 expression. PKP3 loss leads to an increase in invasion, tumour formation and metastasis and these phenotypes were dependent on the increase in LCN2 expression. The increased LCN2 expression was due to an increase in the activation of p38 MAPK in the HCT116 derived PKP3 knockdown clones as LCN2 expression decreased upon inhibition of p38 MAPK. The phosphorylated active form of p38 MAPK is translocated to the nucleus upon PKP3 loss and is dependent on complex formation between p38 MAPK and PKP3. WT PKP3 inhibits LCN2 reporter activity in PKP3 knockdown cells but a PKP3 mutant that fails to form a complex with p38 MAPK cannot suppress LCN2 promoter activity. Further, LCN2 expression is decreased upon loss of p38ß, but not p38α, in the PKP3 knockdown cells. These results suggest that PKP3 loss leads to an increase in the nuclear translocation of p38 MAPK and p38ß MAPK is required for the increase in LCN2 expression.


Assuntos
Lipocalina-2/metabolismo , Neoplasias/metabolismo , Placofilinas/deficiência , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HCT116 , Xenoenxertos , Humanos , Lipocalina-2/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação , Neoplasias/etiologia , Neoplasias/genética , Placofilinas/antagonistas & inibidores , Placofilinas/genética , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Biochem Biophys Res Commun ; 495(2): 1998-2003, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29253567

RESUMO

Desmosomes are cell-cell adhesion junctions that anchor intermediate filaments. Loss of 14-3-3γ in HCT116 cells led to defects in desmosome assembly due to a decrease in the transport of Plakoglobin (PG) to the cell border thus disrupting desmosome formation. Desmosome formation in cells lacking 14-3-3γ was restored by artificially localizing PG to the cell border by fusing it to EGFP-f (PG-EGFP-f). These results suggest that a major role of 14-3-3γ in desmosome assembly is to transport PG to the cell border leading to the initiation of desmosome formation.


Assuntos
Proteínas 14-3-3/metabolismo , Membrana Celular/metabolismo , Neoplasias Colorretais/metabolismo , Desmossomos/metabolismo , Frações Subcelulares/metabolismo , Linhagem Celular Tumoral , Humanos , gama Catenina/metabolismo
9.
Biochem Biophys Res Commun ; 495(1): 768-774, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29146182

RESUMO

Previous reports show that the desmosomal plaque protein plakophilin3 (PKP3) is essential for desmosome formation. Here, we report that PKP3 over-expression decreases calcium dependency for de novo desmosome formation and makes existing cell-cell adhesion junctions more resilient in low calcium medium due to an increase in desmocollin2 expression. PKP3 overexpression increases the stability of other desmosomal proteins independently of the increase in DSC2 levels and regulates desmosome formation and stability by a multimodal mechanism affecting transcription, protein stability and cell border localization of desmosomal proteins.


Assuntos
Adesão Celular/fisiologia , Desmocolinas/metabolismo , Desmossomos/fisiologia , Desmossomos/ultraestrutura , Placofilinas/metabolismo , Linhagem Celular , Humanos , Tamanho da Partícula
10.
J Biol Chem ; 291(31): 16068-81, 2016 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-27261462

RESUMO

Loss of 14-3-3σ has been observed in multiple tumor types; however, the mechanisms by which 14-3-3σ loss leads to tumor progression are not understood. The experiments in this report demonstrate that loss of 14-3-3σ leads to a decrease in the expression of epithelial markers and an increase in the expression of mesenchymal markers, which is indicative of an induction of the epithelial to mesenchymal transition (EMT). The EMT was accompanied by an increase in migration and invasion in the 14-3-3σ(-/-) cells. 14-3-3σ(-/-) cells show increased stabilization of c-Jun, resulting in an increase in the expression of the EMT transcription factor slug. 14-3-3σ induces the ubiquitination and degradation of c-Jun in an FBW7-dependent manner. c-Jun ubiquitination is dependent on the presence of an intact nuclear export pathway as c-Jun is stabilized and localized to the nucleus in the presence of a nuclear export inhibitor. Furthermore, the absence of 14-3-3σ leads to the nuclear accumulation and stabilization of c-Jun, suggesting that 14-3-3σ regulates the subcellular localization of c-Jun. Our results have identified a novel mechanism by which 14-3-3σ maintains the epithelial phenotype by inhibiting EMT and suggest that this property of 14-3-3σ might contribute to its function as a tumor suppressor gene.


Assuntos
Proteínas 14-3-3/metabolismo , Núcleo Celular/metabolismo , Transição Epitelial-Mesenquimal , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitinação , Proteínas 14-3-3/genética , Transporte Ativo do Núcleo Celular/genética , Linhagem Celular , Núcleo Celular/genética , Regulação da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Humanos , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-jun/genética , Fatores de Transcrição da Família Snail/biossíntese , Fatores de Transcrição da Família Snail/genética , Proteínas Supressoras de Tumor/genética
11.
J Cell Sci ; 127(Pt 10): 2174-88, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24610948

RESUMO

The regulation of cell-cell adhesion is important for the processes of tissue formation and morphogenesis. Here, we report that loss of 14-3-3γ leads to a decrease in cell-cell adhesion and a defect in the transport of plakoglobin and other desmosomal proteins to the cell border in HCT116 cells and cells of the mouse testis. 14-3-3γ binds to plakoglobin in a PKCµ-dependent fashion, resulting in microtubule-dependent transport of plakoglobin to cell borders. Transport of plakoglobin to the border is dependent on the KIF5B-KLC1 complex. Knockdown of KIF5B in HCT116 cells, or in the mouse testis, results in a phenotype similar to that observed upon 14-3-3γ knockdown. Our results suggest that loss of 14-3-3γ leads to decreased desmosome formation and a decrease in cell-cell adhesion in vitro, and in the mouse testis in vivo, leading to defects in testis organization and spermatogenesis.


Assuntos
Proteínas 14-3-3/metabolismo , Desmossomos/metabolismo , gama Catenina/metabolismo , Animais , Transporte Biológico , Adesão Celular/fisiologia , Células HCT116 , Humanos , Técnicas In Vitro , Infertilidade Masculina/metabolismo , Cinesinas , Masculino , Camundongos
12.
J Cell Sci ; 124(Pt 12): 2096-106, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21610092

RESUMO

Keratins 8 and 18 (K8 and K18) are predominantly expressed in simple epithelial tissues and perform both mechanical and regulatory functions. Aberrant expression of K8 and K18 is associated with neoplastic progression and invasion in squamous cell carcinomas (SCCs). To understand the molecular basis by which K8 promotes neoplastic progression in oral SCC (OSCC), K8 expression was inhibited in AW13516 cells. The K8-knockdown clones showed a significant reduction in tumorigenic potential, which was accompanied by a reduction in cell motility, cell invasion, decreased fascin levels, alterations in the organization of the actin cytoskeleton and changes in cell shape. Furthermore, K8 knockdown led to a decrease in α6ß4 integrin levels and α6ß4-integrin-dependent signalling events, which have been reported to play an important role in neoplastic progression in epithelial tissues. Therefore, modulation of α6ß4 integrin signalling might be one of the mechanisms by which K8 and K18 promote malignant transformation and/or progression in OSCCs.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Integrina alfa6beta4/metabolismo , Queratina-18/metabolismo , Queratina-8/metabolismo , Animais , Carcinoma de Células Escamosas/patologia , Divisão Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Progressão da Doença , Humanos , Queratina-18/deficiência , Queratina-8/deficiência , Camundongos , Camundongos Nus , Camundongos SCID , Invasividade Neoplásica , Transdução de Sinais
13.
FEBS Lett ; 597(8): 1086-1097, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36650979

RESUMO

Invasive and metastatic tumor cells show an increase in migration and invasion, making the processes contributing to these phenotypes potential therapeutic targets. Lipocalin 2 (LCN2; also known as neutrophil gelatinase-associated lipocalin) is a putative therapeutic target in multiple tumor types and promotes invasion and migration, although the mechanisms underlying these phenotypes are unclear. The data in this report demonstrate that LCN2 promotes actin polymerization, invasion, and migration by inhibiting actin glutathionylation. LCN2 inhibits actin glutathionylation by decreasing the levels of reactive oxygen species (ROS) and by reducing intracellular iron levels. Inhibiting LCN2 function leads to increased actin glutathionylation, decreased migration, and decreased invasion. These results suggest that LCN2 is a potential therapeutic target in invasive tumors.


Assuntos
Actinas , Neoplasias , Humanos , Lipocalina-2 , Lipocalinas , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/metabolismo
14.
BMC Cancer ; 12: 32, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22264292

RESUMO

BACKGROUND: Fascin is a globular actin cross-linking protein, which plays a major role in forming parallel actin bundles in cell protrusions and is found to be associated with tumor cell invasion and metastasis in various type of cancers including oral squamous cell carcinoma (OSCC). Previously, we have demonstrated that fascin regulates actin polymerization and thereby promotes cell motility in K8-depleted OSCC cells. In the present study we have investigated the role of fascin in tumor progression of OSCC. METHODS: To understand the role of fascin in OSCC development and/or progression, fascin was overexpressed along with vector control in OSCC derived cells AW13516. The phenotype was studied using wound healing, Boyden chamber, cell adhesion, Hanging drop, soft agar and tumorigenicity assays. Further, fascin expression was examined in human OSCC samples (N = 131) using immunohistochemistry and level of its expression was correlated with clinico-pathological parameters of the patients. RESULTS: Fascin overexpression in OSCC derived cells led to significant increase in cell migration, cell invasion and MMP-2 activity. In addition these cells demonstrated increased levels of phosphorylated AKT, ERK1/2 and JNK1/2. Our in vitro results were consistent with correlative studies of fascin expression with the clinico-pathological parameters of the OSCC patients. Fascin expression in OSCC showed statistically significant correlation with increased tumor stage (P = 0.041), increased lymph node metastasis (P = 0.001), less differentiation (P = 0.005), increased recurrence (P = 0.038) and shorter survival (P = 0.004) of the patients. CONCLUSION: In conclusion, our results indicate that fascin promotes tumor progression and activates AKT and MAPK pathways in OSCC-derived cells. Further, our correlative studies of fascin expression in OSCC with clinico-pathological parameters of the patients indicate that fascin may prove to be useful in prognostication and treatment of OSCC.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Proteínas de Transporte/metabolismo , Proteínas dos Microfilamentos/metabolismo , Neoplasias Bucais/metabolismo , Proteínas de Neoplasias/metabolismo , Actinas/ultraestrutura , Animais , Western Blotting , Carcinoma de Células Escamosas/patologia , Movimento Celular/fisiologia , Proliferação de Células , Citoesqueleto/ultraestrutura , Progressão da Doença , Humanos , Imuno-Histoquímica , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos SCID , Neoplasias Bucais/patologia , Invasividade Neoplásica/patologia , Células Tumorais Cultivadas , Cicatrização/fisiologia
15.
Cell Mol Life Sci ; 68(8): 1439-54, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20859650

RESUMO

A decrease in the levels of the desmosomal plaque protein, plakophilin3 (PKP3), leads to a decrease in desmosome size and cell-cell adhesion. To test the hypothesis that PKP3 is required for desmosome formation, the recruitment of desmosomal components to the cell surface was studied in the PKP3 knockdown clones. The PKP3 knockdown clones showed decreased cell border staining for multiple desmosomal proteins, when compared to vector controls, and did not form desmosomes in a calcium switch assay. Further analysis demonstrated that PKP3, plakoglobin (PG) and E-cadherin are present at the cell border at low concentrations of calcium. Loss of either PG or E-cadherin led to a decrease in the levels of PKP3 and other desmosomal proteins at the cell border. The results reported here are consistent with the model that PG and E-cadherin recruit PKP3 to the cell border to initiate desmosome formation.


Assuntos
Caderinas/metabolismo , Desmossomos/metabolismo , Placofilinas/metabolismo , gama Catenina/metabolismo , Adesão Celular , Linhagem Celular , Imunofluorescência , Técnicas de Silenciamento de Genes , Células HCT116 , Células HEK293 , Humanos , Microscopia Confocal , Placofilinas/genética
16.
Indian J Exp Biol ; 50(10): 669-76, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23214259

RESUMO

The study of protein-protein interactions, protein localization, protein organization into higher order structures and organelle dynamics in live cells, has greatly enhanced the understanding of various cellular processes. Live cell imaging experiments employ plasmid or viral vectors to express the protein/proteins of interest fused to a fluorescent protein. Unlike plasmid vectors, lentiviral vectors can be introduced into both dividing and non dividing cells, can be pseudotyped to infect a broad or narrow range of cells, and can be used to generate transgenic animals. However, the currently available lentiviral vectors are limited by the choice of fluorescent protein tag, choice of restriction enzyme sites in the Multiple Cloning Sites (MCS) and promoter choice for gene expression. In this report, HIV-1 based bi-cistronic lentiviral vectors have been generated that drive the expression of multiple fluorescent tags (EGFP, mCherry, ECFP, EYFP and dsRed), using two different promoters. The presence of a unique MCS with multiple restriction sites allows the generation of fusion proteins with the fluorescent tag of choice, allowing analysis of multiple fusion proteins in live cell imaging experiments. These novel lentiviral vectors are improved delivery vehicles for gene transfer applications and are important tools for live cell imaging in vivo.


Assuntos
Citomegalovirus/genética , Expressão Gênica , Vetores Genéticos/metabolismo , HIV-1/genética , Rim/citologia , Lentivirus/genética , Ubiquitina C/genética , Células Cultivadas , Imunofluorescência , Vetores Genéticos/genética , Humanos , Processamento de Imagem Assistida por Computador , Rim/metabolismo , Microscopia de Fluorescência
17.
J Cell Commun Signal ; 16(2): 207-222, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34596797

RESUMO

Chronic myeloid leukemia (CML) epitomises successful targeted therapy, where inhibition of tyrosine kinase activity of oncoprotein Bcr-Abl1 by imatinib, induces remission in 86% patients in initial chronic phase (CP). However, in acute phase of blast crisis, 80% patients show resistance, 40% among them despite inhibition of Bcr-Abl1 activity. This implies activation of either Bcr-Abl1- independent signalling pathways or restoration of signalling downstream of inactive Bcr-Abl1. In the present study, mass spectrometry and subsequent in silico pathway analysis of differentiators in resistant CML-CP cells identified key differentiators, 14-3-3ε and p38 MAPK, which belong to Bcr-Abl1 pathway. Their levels and activity respectively, indicated active Bcr-Abl1 pathway in CML-BC resistant cells, though Bcr-Abl1 is inhibited by imatinib. Further, contribution of these components to resistance was demonstrated by inhibition of Bcr-Abl1 down-stream signalling by knocking-out of 14-3-3ε and inhibition of p38 MAPK activity. The observations merit clinical validation to explore their translational potential.

18.
FEBS Lett ; 595(21): 2675-2690, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34626438

RESUMO

14-3-3 proteins are conserved, dimeric, acidic proteins that regulate multiple cellular pathways. Loss of either 14-3-3ε or 14-3-3γ leads to centrosome amplification. However, we find that while the knockout of 14-3-3ε leads to multipolar mitoses, the knockout of 14-3-3γ results in centrosome clustering and pseudo-bipolar mitoses. 14-3-3γ knockouts demonstrate compromised desmosome function and a decrease in keratin levels, leading to decreased cell stiffness and an increase in centrosome clustering. Restoration of desmosome function increased multipolar mitoses, whereas knockdown of either plakoglobin or keratin 5 led to decreased cell stiffness and increased pseudo-bipolar mitoses. These results suggest that the ability of the desmosome to anchor keratin filaments maintains cell stiffness, thus inhibiting centrosome clustering, and that phenotypes observed upon 14-3-3 loss reflect the dysregulation of multiple pathways.


Assuntos
Proteínas 14-3-3 , Centrossomo , Desmossomos , Mitose , Células HCT116 , Humanos , Fuso Acromático
19.
Biochim Biophys Acta Gene Regul Mech ; 1864(4-5): 194703, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33727172

RESUMO

Incorporation of different H3 histone isoforms/variants have been reported to differentially regulate gene expression via alteration in chromatin organization during diverse cellular processes. However, the differential expression of highly conserved histone H3.2 genes, H3C14 and H3C13 in human cancer has not been delineated. In this study, we investigated the expression of H3.2 genes in primary human gastric, brain, breast, colon, liver, and head and neck cancer tissues and tumor cell lines. The data showed overexpression of H3.2 transcripts in tumor samples and cell lines with respect to normal counterparts. Furthermore, TCGA data of individual and TCGA PANCAN cohort also showed significant up-regulation of H3.2 genes. Further, overexpressed H3C14 gene coding for H3.2 protein was regulated by FOXC1 transcription factor and G4-cassette in gastric cancer cell lines. Elevated expression of FOXC1 protein and transcripts were also observed in human gastric cancer samples and cell lines. Further, FOXC1 protein was predominantly localized in the nuclei of neoplastic gastric cells compared to normal counterpart. In continuation, studies with EGF induction, FOXC1 knockdown, and ChIP-qPCR for the first time identified a novel axis, EGFR-FOXC1-H3C14 for regulation of H3C14 gene overexpression in gastric cancer. Therefore, the changes the epigenomic landscape due to incorporation of differential expression H3 variant contributes to change in gene expression pattern and thereby contributing to pathogenesis of cancer.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Regulação Neoplásica da Expressão Gênica , Histonas/biossíntese , Proteínas de Neoplasias/metabolismo , Transdução de Sinais , Neoplasias Gástricas/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Fatores de Transcrição Forkhead/genética , Células Hep G2 , Humanos , Células MCF-7 , Proteínas de Neoplasias/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Células U937
20.
Exp Cell Res ; 315(8): 1448-57, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19331823

RESUMO

Mitotic progression requires the activity of the dual specificity phosphatase, cdc25C. Cdc25C function is inhibited by complex formation with two 14-3-3 isoforms, 14-3-3epsilon and 14-3-3gamma. To understand the molecular basis of specific complex formation between 14-3-3 proteins and their ligands, chimeric 14-3-3 proteins were tested for their ability to form a complex with cdc25C in vivo. Specific complex formation between cdc25C and 14-3-3epsilon in vivo requires a phenylalanine residue at position 135 (F135) in 14-3-3epsilon. Mutation of this residue to the corresponding residue present in other 14-3-3 isoforms (F135V) leads to reduced binding to cdc25C and a decrease in the ability to inhibit cdc25C function in vivo. Similarly, F135V failed to rescue the incomplete S phase and the G2 DNA damage checkpoint defects observed in cells lacking 14-3-3epsilon. A comparative analysis of the 14-3-3 structures present in the database suggested that the F135 in 14-3-3epsilon was required to maintain the integrity of a pocket that might be involved in secondary interactions with cdc25C. These results suggest that the specificity of the 14-3-3 ligand interaction may be dependent on structural motifs present in the individual 14-3-3 isoforms.


Assuntos
Proteínas 14-3-3/genética , Proteínas de Ciclo Celular/metabolismo , Fosfatases cdc25/metabolismo , Proteínas 14-3-3/química , Proteínas 14-3-3/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Filogenia , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Alinhamento de Sequência , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA