Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nature ; 604(7907): 749-756, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35444283

RESUMO

Amplification of the CCNE1 locus on chromosome 19q12 is prevalent in multiple tumour types, particularly in high-grade serous ovarian cancer, uterine tumours and gastro-oesophageal cancers, where high cyclin E levels are associated with genome instability, whole-genome doubling and resistance to cytotoxic and targeted therapies1-4. To uncover therapeutic targets for tumours with CCNE1 amplification, we undertook genome-scale CRISPR-Cas9-based synthetic lethality screens in cellular models of CCNE1 amplification. Here we report that increasing CCNE1 dosage engenders a vulnerability to the inhibition of the PKMYT1 kinase, a negative regulator of CDK1. To inhibit PKMYT1, we developed RP-6306, an orally bioavailable and selective inhibitor that shows single-agent activity and durable tumour regressions when combined with gemcitabine in models of CCNE1 amplification. RP-6306 treatment causes unscheduled activation of CDK1 selectively in CCNE1-overexpressing cells, promoting early mitosis in cells undergoing DNA synthesis. CCNE1 overexpression disrupts CDK1 homeostasis at least in part through an early activation of the MMB-FOXM1 mitotic transcriptional program. We conclude that PKMYT1 inhibition is a promising therapeutic strategy for CCNE1-amplified cancers.


Assuntos
Ciclina E , Proteínas de Membrana , Neoplasias Ovarianas , Proteínas Serina-Treonina Quinases , Proteínas Tirosina Quinases , Proteína Quinase CDC2 , Ciclina E/genética , Feminino , Amplificação de Genes , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas de Membrana/genética , Neoplasias/genética , Neoplasias Ovarianas/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Mutações Sintéticas Letais
2.
Mol Cell ; 70(5): 825-841.e6, 2018 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-29861161

RESUMO

Super-enhancers are large clusters of enhancers that activate gene expression. Broad trimethyl histone H3 lysine 4 (H3K4me3) often defines active tumor suppressor genes. However, how these epigenomic signatures are regulated for tumor suppression is little understood. Here we show that brain-specific knockout of the H3K4 methyltransferase MLL4 (a COMPASS-like enzyme, also known as KMT2D) in mice spontaneously induces medulloblastoma. Mll4 loss upregulates oncogenic Ras and Notch pathways while downregulating neuronal gene expression programs. MLL4 enhances DNMT3A-catalyzed DNA methylation and SIRT1/BCL6-mediated H4K16 deacetylation, which antagonize expression of Ras activators and Notch pathway components, respectively. Notably, Mll4 loss downregulates tumor suppressor genes (e.g., Dnmt3a and Bcl6) by diminishing broad H3K4me3 and super-enhancers and also causes widespread impairment of these epigenomic signatures during medulloblastoma genesis. These findings suggest an anti-tumor role for super-enhancers and provide a unique tumor-suppressive mechanism in which MLL4 is necessary to maintain broad H3K4me3 and super-enhancers at tumor suppressor genes.


Assuntos
Neoplasias Cerebelares/genética , Metilação de DNA , Genes Supressores de Tumor , Histona-Lisina N-Metiltransferase/genética , Meduloblastoma/genética , Oncogenes , Processamento de Proteína Pós-Traducional , Acetilação , Animais , Proliferação de Células , Neoplasias Cerebelares/metabolismo , Neoplasias Cerebelares/patologia , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A , Regulação Neoplásica da Expressão Gênica , Genes ras , Histona-Lisina N-Metiltransferase/deficiência , Lisina , Meduloblastoma/metabolismo , Meduloblastoma/patologia , Camundongos Knockout , Proteínas Proto-Oncogênicas c-bcl-6/genética , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais , Sirtuína 1/genética , Sirtuína 1/metabolismo
3.
Cell Biol Int ; 45(8): 1720-1732, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33847415

RESUMO

Keratin 8/18, the predominant keratin pair of simple epithelia, is known to be aberrantly expressed in several squamous cell carcinomas (SCCs), where its expression is often correlated with increased invasion, neoplastic progression, and poor prognosis. The majority of keratin 8/18 structural and regulatory functions are governed by posttranslational modifications, particularly phosphorylation. Apart from filament reorganization, cellular processes including cell cycle, cell growth, cellular stress, and apoptosis are known to be orchestrated by K8 phosphorylation at specific residues in the head and tail domains. Even though deregulation of K8 phosphorylation at two significant sites (Serine73 /Serine431 ) has been implicated in neoplastic progression of SCCs by various in vitro studies, including ours, it is reported to be highly context-dependent. Therefore, to delineate the precise role of Kereatin 8 phosphorylation in cancer initiation and progression, we have developed the tissue-specific transgenic mouse model expressing Keratin 8 wild type and phosphodead mutants under Keratin 14 promoter. Subjecting these mice to 7,12-dimethylbenz(a)anthracene/12-O-tetradecanoylphorbol-13-acetate-mediated skin carcinogenesis revealed that Keratin 8 phosphorylation may lead to an early onset of tumors compared to Keratin 8 wild-type expressing mice. Conclusively, the transgenic mouse model developed in the present study ascertained a positive impact of Keratin 8 phosphorylation on the neoplastic transformation of skin-squamous cells.


Assuntos
Carcinogênese/metabolismo , Queratina-8/metabolismo , Mutação/fisiologia , Neoplasias Cutâneas/metabolismo , Animais , Carcinogênese/genética , Carcinogênese/patologia , Eletroporação/métodos , Células HEK293 , Humanos , Queratina-8/genética , Masculino , Camundongos , Camundongos Transgênicos , Fosforilação/fisiologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia
4.
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
5.
Cell Mol Life Sci ; 72(23): 4577-92, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26305020

RESUMO

Histone methyltransferases and demethylases epigenetically regulate gene expression by modifying histone methylation status in numerous cellular processes, including cell differentiation and proliferation. These modifiers also control methylation levels of various non-histone proteins, such as effector proteins that play critical roles in cellular signaling networks. Dysregulated histone methylation modifiers alter expression of oncogenes and tumor suppressor genes and change methylation states of effector proteins, frequently resulting in aberrant cellular signaling cascades and cellular transformation. In this review, we summarize the role of histone methylation modifiers in regulating the following signaling pathways: NF-κB, RAS/RAF/MEK/MAPK, PI3K/Akt, Wnt/ß-catenin, p53, and ERα.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , NF-kappa B/metabolismo , Via de Sinalização Wnt , Sequência de Aminoácidos , Arginina/metabolismo , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/química , Humanos , Lisina/metabolismo , Sistema de Sinalização das MAP Quinases , Metilação , Dados de Sequência Molecular , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Transdução de Sinais
6.
J Biol Chem ; 289(11): 7483-96, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24482232

RESUMO

Dysregulated expression of histone methyltransferases and demethylases is an emerging epigenetic mechanism underlying cancer development and metastasis. We recently showed that the histone H3 lysine 36 (H3K36) demethylase KDM2A (also called FBXL11 and JHDM1A) is necessary for tumorigenic and metastatic capabilities of KDM2A-overexpressing non-small cell lung cancer (NSCLC) cells. Here, we report that KDM2A transcriptionally represses the histone deacetylase 3 (HDAC3) gene by removing methyl groups from dimethylated H3K36 at the HDAC3 promoter in KDM2A-overexpressing NSCLC cells. KDM2A depletion reduced expression levels of cell cycle-associated genes (e.g. CDK6) and cell invasion-related genes (e.g. NANOS1); these levels were rescued by ectopic expression of KDM2A but not its catalytic mutant. These genes were occupied and down-regulated by HDAC3. HDAC3 knockdown significantly recovered the proliferation and invasiveness of KDM2A-depleted NSCLC cells as well as the levels of CDK6 and NANOS1 expression in these cells. Similar to their previously reported functions in other cell types, CDK6 and NANOS1 were required for the proliferation and invasion, respectively, of KDM2A-overexpressing NSCLC cells. In a mouse xenograft model, HDAC3 depletion substantially restored the tumorigenic ability of KDM2A knockdown cells. These findings reveal a novel cancer-epigenetic pathway in which the antagonistic effect of KDM2A on HDAC3 expression releases cell cycle-associated genes and cell invasion-related genes from HDAC3 repression and indicate the importance of this pathway for tumorigenicity and invasiveness of KDM2A-overexpressing NSCLC cells.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Epigênese Genética , Proteínas F-Box/metabolismo , Regulação Neoplásica da Expressão Gênica , Histona Desacetilases/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Neoplasias Pulmonares/metabolismo , Transcrição Gênica , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Quinase 6 Dependente de Ciclina/metabolismo , Proteínas F-Box/genética , Técnicas de Silenciamento de Genes , Inativação Gênica , Histona Desacetilases/genética , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Neoplasias Pulmonares/genética , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Regiões Promotoras Genéticas , Proteínas de Ligação a RNA/metabolismo
7.
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
8.
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
9.
Mol Cancer Ther ; 21(2): 245-256, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34911817

RESUMO

Ataxia telangiectasia and Rad3-related (ATR) kinase protects genome integrity during DNA replication. RP-3500 is a novel, orally bioavailable clinical-stage ATR kinase inhibitor (NCT04497116). RP-3500 is highly potent with IC50 values of 1.0 and 0.33 nmol/L in biochemical and cell-based assays, respectively. RP-3500 is highly selective for ATR with 30-fold selectivity over mammalian target of rapamycin (mTOR) and more than 2,000-fold selectivity over ataxia telangiectasia mutated (ATM), DNA-dependent protein kinase (DNA-PK), and phosphatidylinositol 3-kinase alpha (PI3Kα) kinases. In vivo, RP-3500 treatment results in potent single-agent efficacy and/or tumor regression in multiple xenograft models at minimum effective doses (MED) of 5 to 7 mg/kg once daily. Pharmacodynamic assessments validate target engagement, with dose-proportional tumor inhibition of phosphorylated checkpoint kinase 1 (pCHK1) (IC80 = 18.6 nmol/L) and induction of phosphorylated H2A.X variant histone (γH2AX), phosphorylated DNA-PK catalytic subunit (pDNA-PKcs), and phosphorylated KRAB-associated protein 1 (pKAP1). RP-3500 exposure at MED indicates that circulating free plasma levels above the in vivo tumor IC80 for 10 to 12 hours are sufficient for efficacy on a continuous schedule. However, short-duration intermittent (weekly 3 days on/4 days off) dosing schedules as monotherapy or given concomitantly with reduced doses of olaparib or niraparib, maximize tumor growth inhibition while minimizing the impact on red blood cell depletion, emphasizing the reversible nature of erythroid toxicity with RP-3500 and demonstrating superior efficacy compared with sequential treatment. These results provide a strong preclinical rationale to support ongoing clinical investigation of the novel ATR inhibitor, RP-3500, on an intermittent schedule as a monotherapy and in combination with PARP inhibitors as a potential means of maximizing clinical benefit.


Assuntos
Ataxia Telangiectasia , Inibidores de Poli(ADP-Ribose) Polimerases , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Proteína Quinase Ativada por DNA/metabolismo , Humanos , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico
10.
Cancer Cell ; 37(4): 599-617.e7, 2020 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-32243837

RESUMO

Epigenetic modifiers frequently harbor loss-of-function mutations in lung cancer, but their tumor-suppressive roles are poorly characterized. Histone methyltransferase KMT2D (a COMPASS-like enzyme, also called MLL4) is among the most highly inactivated epigenetic modifiers in lung cancer. Here, we show that lung-specific loss of Kmt2d promotes lung tumorigenesis in mice and upregulates pro-tumorigenic programs, including glycolysis. Pharmacological inhibition of glycolysis preferentially impedes tumorigenicity of human lung cancer cells bearing KMT2D-inactivating mutations. Mechanistically, Kmt2d loss widely impairs epigenomic signals for super-enhancers/enhancers, including the super-enhancer for the circadian rhythm repressor Per2. Loss of Kmt2d decreases expression of PER2, which regulates multiple glycolytic genes. These findings indicate that KMT2D is a lung tumor suppressor and that KMT2D deficiency confers a therapeutic vulnerability to glycolytic inhibitors.


Assuntos
Adenocarcinoma de Pulmão/patologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Desoxiglucose/farmacologia , Elementos Facilitadores Genéticos , Regulação Neoplásica da Expressão Gênica , Glicólise , Histona-Lisina N-Metiltransferase/fisiologia , Proteína de Leucina Linfoide-Mieloide/fisiologia , Proteínas de Neoplasias/antagonistas & inibidores , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/metabolismo , Animais , Antimetabólitos/farmacologia , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Knockout , Camundongos Nus , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Prognóstico , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Cell Rep ; 33(3): 108293, 2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33086062

RESUMO

Histone methyltransferase KMT2D harbors frequent loss-of-function somatic point mutations in several tumor types, including melanoma. Here, we identify KMT2D as a potent tumor suppressor in melanoma through an in vivo epigenome-focused pooled RNAi screen and confirm the finding by using a genetically engineered mouse model (GEMM) based on conditional and melanocyte-specific deletion of KMT2D. KMT2D-deficient tumors show substantial reprogramming of key metabolic pathways, including glycolysis. KMT2D deficiency aberrantly upregulates glycolysis enzymes, intermediate metabolites, and glucose consumption rates. Mechanistically, KMT2D loss causes genome-wide reduction of H3K4me1-marked active enhancer chromatin states. Enhancer loss and subsequent repression of IGFBP5 activates IGF1R-AKT to increase glycolysis in KMT2D-deficient cells. Pharmacological inhibition of glycolysis and insulin growth factor (IGF) signaling reduce proliferation and tumorigenesis preferentially in KMT2D-deficient cells. We conclude that KMT2D loss promotes tumorigenesis by facilitating an increased use of the glycolysis pathway for enhanced biomass needs via enhancer reprogramming, thus presenting an opportunity for therapeutic intervention through glycolysis or IGF pathway inhibitors.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , Melanoma/genética , Proteína de Leucina Linfoide-Mieloide/metabolismo , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Genes Supressores de Tumor , Glucose/metabolismo , Glicólise/genética , Histona Metiltransferases/genética , Histona Metiltransferases/metabolismo , Histona-Lisina N-Metiltransferase/genética , Humanos , Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Receptor IGF Tipo 1/metabolismo , Sequências Reguladoras de Ácido Nucleico , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
12.
Oncol Rep ; 39(5): 2393-2401, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29512781

RESUMO

Keratins 5/14 (K5/14) are intermediate filament proteins expressed in the basal layer of stratified epithelial cells and are known targets of p63. Previous research in our laboratory showed that upon K5/14 downregulation in oral squamous cell carcinoma (OSCC)­derived cells, there was an increase in intracellular Notch­1 levels and differentiation markers such as involucrin, keratin 1 and a decrease in tumorigenic potential in vivo. However, the molecules involved in the K14 regulated cell differentiation and transformation are not known to date. In order to understand the possible role of TAp63, we downregulated TAp63 in a K14­knockdown background. We observed that there was a decrease in the expression of Notch­1. Expression levels of differentiation markers such as involucrin, K1, loricrin and filaggrin were also decreased. Furthermore, TAp63 downregulation led to an increase in invasion, migration and in vivo tumorigenic potential of these cells. We observed a decrease in ß­catenin signaling in K14­downregulated cells. Notably, when TAp63 was downregulated in K14­knockdown cells, there was increase in non­phospho ß­catenin levels. Hence, this study indicates that TAp63 plays an important role in K14­downregulated cells possibly by regulating the Notch­1 expression. K14 regulates the expression of TAp63 which in turn regulates expression of Notch­1. The present study is a step forward in our quest to understand the functional significance of molecules that regulate the process of differentiation and tumorigenesis in stratified epithelial cells.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Queratina-14/metabolismo , Queratina-5/metabolismo , Neoplasias Bucais/metabolismo , Receptor Notch1/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Regulação para Baixo , Proteínas Filagrinas , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Transplante de Neoplasias
13.
Cancer Res ; 78(14): 3834-3848, 2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29764865

RESUMO

Lung adenocarcinoma is a major form of lung cancer, which is the leading cause of cancer death. Histone methylation reader proteins mediate the effect of histone methylation, a hallmark of epigenetic and transcriptional regulation of gene expression. However, their roles in lung adenocarcinoma are poorly understood. Here, our bioinformatic screening and analysis in search of a lung adenocarcinoma-promoting histone methylation reader protein show that heterochromatin protein 1γ (HP1γ; also called CBX3) is among the most frequently overexpressed and amplified histone reader proteins in human lung adenocarcinoma, and that high HP1γ mRNA levels are associated with poor prognosis in patients with lung adenocarcinoma. In vivo depletion of HP1γ reduced K-RasG12D-driven lung adenocarcinoma and lengthened survival of mice bearing K-RasG12D-induced lung adenocarcinoma. HP1γ and its binding activity to methylated histone H3 lysine 9 were required for the proliferation, colony formation, and migration of lung adenocarcinoma cells. HP1γ directly repressed expression of the transcription-repressive regulators NCOR2 and ZBTB7A. Knockdown of NCOR2 or ZBTB7A significantly restored defects in proliferation, colony formation, and migration in HP1γ-depleted lung adenocarcinoma cells. Low NCOR2 or ZBTB7A mRNA levels were associated with poor prognosis in patients with lung adenocarcinoma and correlated with high HP1γ mRNA levels in lung adenocarcinoma samples. NCOR2 and ZBTB7A downregulated expression of tumor-promoting factors such as ELK1 and AXL, respectively. These findings highlight the importance of HP1γ and its reader activity in lung adenocarcinoma tumorigenesis and reveal a unique lung adenocarcinoma-promoting mechanism in which HP1γ downregulates NCOR2 and ZBTB7A to enhance expression of protumorigenic genes.Significance: Direct epigenetic repression of the transcription-repressive regulators NCOR2 and ZBTB7A by the histone reader protein HP1γ leads to activation of protumorigenic genes in lung adenocarcinoma. Cancer Res; 78(14); 3834-48. ©2018 AACR.


Assuntos
Adenocarcinoma de Pulmão/genética , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Regulação para Baixo/genética , Repressão Epigenética/genética , Correpressor 2 de Receptor Nuclear/genética , Fatores de Transcrição/genética , Adenocarcinoma de Pulmão/patologia , Animais , Carcinogênese/genética , Carcinogênese/patologia , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação da Expressão Gênica/genética , Humanos , Camundongos , Prognóstico , RNA Mensageiro/genética
14.
J Biophotonics ; 10(10): 1377-1384, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28067994

RESUMO

Accurate understanding of cellular processes and responses to stimuli is of paramount importance in biomedical research and diagnosis. Raman spectroscopy (RS), a label-free and nondestructive spectroscopic method has the potential to serve as a novel 'theranostics' tool. Both fiber-optic and micro-Raman studies have demonstrated efficacy in diagnostics and therapeutic response monitoring. In the present study, we have evaluated the potential of micro-Raman spectroscopic maps in identifying changes induced by loss of K8/18 proteins in a tongue cancer cell line. Furthermore, we also evaluated the efficacy of less expensive and commercially available fiber probes to identify K8/18 wild and knock-down cell pellets, in view of the utility of cell pellet-based studies. The findings suggest that major differences in the cellular morphology and biochemical composition can be objectively identified and can be utilized for classification using both micro-Raman and fiber-probe-based RS. These findings highlight the potential of fiber-optic probe-based RS in noninvasive cellular phenotyping for diagnosis and therapeutic response monitoring, especially in low-resource settings.


Assuntos
Técnicas de Silenciamento de Genes , Queratina-18/deficiência , Queratina-18/genética , Queratina-8/deficiência , Queratina-8/genética , Análise Espectral Raman , Linhagem Celular Tumoral , Humanos , Neoplasias Bucais/patologia
15.
PLoS One ; 12(2): e0172559, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28225793

RESUMO

Vimentin is an intermediate filament protein, predominantly expressed in cells of mesenchymal origin, although its aberrant expression is seen in many carcinomas during epithelial mesenchymal transition. In cancer, vimentin expression is associated with the transition from a more differentiated epithelial phenotype to a dedifferentiated state. In view of the perceived role of keratins (Ks) as regulators of differentiation in epithelia, it was important to understand whether vimentin modulates differentiation through the reprogramming of keratins, in transformed cells. To address this, vimentin was stably downregulated in oral cancer derived cells. Further, global keratin profiling was performed after high salt keratin extraction. K5/K14 pair was found to be significantly downregulated, both at protein and mRNA levels upon vimentin downregulation. The previous study from our laboratory has shown a role of the K5/K14 pair in proliferation and differentiation of squamous epithelial cells. Vimentin depleted cells showed an increase in the differentiation state, marked by an increase in the levels of differentiation specific markers K1, involucrin, filaggrin and loricrin while its proliferation status remained unchanged. Rescue experiments with the K5/K14 pair overexpressed in vimentin knockdown background resulted in decreased differentiation state. ΔNp63 emerged as one of the indirect targets of vimentin, through which it modulates the expression levels of K5/K14. Further, immunohistochemistry showed a significant correlation between high vimentin-K14 expression and recurrence/poor survival in oral cancer patients. Thus, in conclusion, vimentin regulates the differentiation switch via modulation of K5/K14 expression. Moreover, vimentin-K14 together may prove to be the novel markers for the prognostication of human oral cancer.


Assuntos
Diferenciação Celular/genética , Regulação Neoplásica da Expressão Gênica , Queratina-14/genética , Neoplasias Bucais/genética , Recidiva Local de Neoplasia/genética , Vimentina/genética , Animais , Linhagem Celular Tumoral , Regulação para Baixo , Feminino , Proteínas Filagrinas , Humanos , Queratina-14/metabolismo , Queratina-5/genética , Queratina-5/metabolismo , Masculino , Camundongos , Neoplasias Bucais/metabolismo , Neoplasias Bucais/mortalidade , Neoplasias Bucais/patologia , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/mortalidade , Recidiva Local de Neoplasia/patologia , Prognóstico , Receptores Notch/genética , Receptores Notch/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Vimentina/metabolismo
16.
Int J Biochem Cell Biol ; 70: 161-72, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26646105

RESUMO

Vimentin expression correlates well with migratory and invasive potential of the carcinoma cells. The molecular mechanism by which vimentin regulates cell motility is not yet clear. Here, we addressed this issue by depleting vimentin in oral squamous cell carcinoma derived cell line. Vimentin knockdown cells showed enhanced adhesion and spreading to laminin-5. However, we found that they were less invasive as compared to the vector control cells. In addition, signaling associated with adhesion behavior of the cell was increased in vimentin knockdown clones. These findings suggest that the normal function of ß4 integrin as mechanical adhesive device is enhanced upon vimentin downregulation. As a proof of principle, the compromised invasive potential of vimentin depleted cells could be rescued upon blocking with ß4 integrin adhesion-blocking (ASC-8) antibody or downregulation of ß4 integrin in vimentin knockdown background. Interestingly, plectin which associates with α6ß4 integrin in the hemidesmosomes, was also found to be upregulated in vimentin knockdown clones. Furthermore, experiments on lysosome and proteasome inhibition revealed that perhaps vimentin regulates the turnover of ß4 integrin and plectin. Moreover, an inverse association was observed between vimentin expression and ß4 integrin in oral squamous cell carcinoma (OSCC). Collectively, our results show a novel role of vimentin in modulating cell motility by destabilizing ß4 integrin-mediated adhesive interactions. Further, vimentin-ß4 integrin together may prove to be useful markers for prognostication of human oral cancer.


Assuntos
Carcinoma de Células Escamosas/genética , Regulação Neoplásica da Expressão Gênica , Integrina beta4/genética , Neoplasias Bucais/genética , Vimentina/genética , Anticorpos Neutralizantes/farmacologia , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/mortalidade , Adesão Celular , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Feminino , Hemidesmossomos/efeitos dos fármacos , Hemidesmossomos/metabolismo , Hemidesmossomos/ultraestrutura , Humanos , Integrina beta4/metabolismo , Filamentos Intermediários/efeitos dos fármacos , Filamentos Intermediários/metabolismo , Filamentos Intermediários/ultraestrutura , Masculino , Pessoa de Meia-Idade , Neoplasias Bucais/diagnóstico , Neoplasias Bucais/metabolismo , Neoplasias Bucais/mortalidade , Invasividade Neoplásica , Estadiamento de Neoplasias , Plectina/genética , Plectina/metabolismo , Cultura Primária de Células , Prognóstico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Análise de Sobrevida , Vimentina/antagonistas & inibidores , Vimentina/metabolismo , Calinina
17.
PLoS One ; 8(1): e53532, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23341946

RESUMO

BACKGROUND: Breast cancer is a complex disease which cannot be defined merely by clinical parameters like lymph node involvement and histological grade, or by routinely used biomarkers like estrogen receptor (ER), progesterone receptor (PGR) and epidermal growth factor receptor 2 (HER2) in diagnosis and prognosis. Breast cancer originates from the epithelial cells. Keratins (K) are cytoplasmic intermediate filament proteins of epithelial cells and changes in the expression pattern of keratins have been seen during malignant transformation in the breast. Expression of the K8/18 pair is seen in the luminal cells of the breast epithelium, and its role in prognostication of breast cancer is not well understood. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we have modulated K8 expression to understand the role of the K8/18 pair in three different breast epithelium derived cell lines: non-transformed MCF10A, transformed but poorly invasive MDA MB 468 and highly invasive MDA MB 435. The up-regulation of K8 in the invasive MDA MB 435 cell line resulted in a significant decrease in proliferation, motility, in-vitro invasion, tumor volume and lung metastasis. The down-regulation of K8 in MDA MB 468 resulted in a significant increase in transformation potential, motility and invasion in-vitro, while MCF10A did not show any changes in cell transformation assays. CONCLUSIONS/SIGNIFICANCE: These results indicate the role of K8/18 in modulating invasion in breast cancer -its presence correlating with less invasive phenotype and absence correlating with highly invasive, dedifferentiated phenotype. These data may have important implications for prognostication of breast cancer.


Assuntos
Neoplasias da Mama/genética , Transformação Celular Neoplásica/genética , Queratina-18/metabolismo , Queratina-8/metabolismo , Animais , Neoplasias da Mama/patologia , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Transformação Celular Neoplásica/patologia , Células Clonais , Regulação para Baixo/genética , Feminino , Humanos , Queratina-18/genética , Queratina-7/metabolismo , Queratina-8/genética , Camundongos , Camundongos SCID , Invasividade Neoplásica , Metástase Neoplásica , Ensaio Tumoral de Célula-Tronco , Regulação para Cima/genética , Vimentina/metabolismo
18.
J Clin Invest ; 123(12): 5231-46, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24200691

RESUMO

Epigenetic dysregulation has emerged as a major contributor to tumorigenesis. Histone methylation is a well-established mechanism of epigenetic regulation that is dynamically modulated by histone methyltransferases and demethylases. The pathogenic role of histone methylation modifiers in non-small cell lung cancer (NSCLC), which is the leading cause of cancer deaths worldwide, remains largely unknown. Here, we found that the histone H3 lysine 36 (H3K36) demethylase KDM2A (also called FBXL11 and JHDM1A) is frequently overexpressed in NSCLC tumors and cell lines. KDM2A and its catalytic activity were required for in vitro proliferation and invasion of KDM2A-overexpressing NSCLC cells. KDM2A overexpression in NSCLC cells with low KDM2A levels increased cell proliferation and invasiveness. KDM2A knockdown abrogated tumor growth and invasive abilities of NSCLC cells in mouse xenograft models. We identified dual-specificity phosphatase 3 (DUSP3) as a key KDM2A target gene and found that DUSP3 dephosphorylates ERK1/2 in NSCLC cells. KDM2A activated ERK1/2 through epigenetic repression of DUSP3 expression via demethylation of dimethylated H3K36 at the DUSP3 locus. High KDM2A levels correlated with poor prognosis in NSCLC patients. These findings uncover an unexpected role for a histone methylation modifier in activating ERK1/2 in lung tumorigenesis and metastasis, suggesting that KDM2A may be a promising therapeutic target in NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Epigênese Genética/fisiologia , Proteínas F-Box/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/fisiologia , Neoplasias Pulmonares/patologia , Sistema de Sinalização das MAP Quinases , Proteínas de Neoplasias/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Divisão Celular , Linhagem Celular Tumoral , Fosfatase 3 de Especificidade Dupla/biossíntese , Fosfatase 3 de Especificidade Dupla/genética , Fosfatase 3 de Especificidade Dupla/fisiologia , Epigênese Genética/genética , Proteínas F-Box/antagonistas & inibidores , Proteínas F-Box/biossíntese , Proteínas F-Box/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Xenoenxertos , Humanos , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Histona Desmetilases com o Domínio Jumonji/biossíntese , Histona Desmetilases com o Domínio Jumonji/genética , Neoplasias Pulmonares/genética , Masculino , Metilação , Camundongos , Camundongos Nus , Invasividade Neoplásica , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Prognóstico , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional/genética , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética
19.
PLoS One ; 7(6): e38561, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22701666

RESUMO

The desmosome anchors keratin filaments in epithelial cells leading to the formation of a tissue wide IF network. Loss of the desmosomal plaque protein plakophilin3 (PKP3) in HCT116 cells, leads to an increase in neoplastic progression and metastasis, which was accompanied by an increase in K8 levels. The increase in levels was due to an increase in the protein levels of the Phosphatase of Regenerating Liver 3 (PRL3), which results in a decrease in phosphorylation on K8. The increase in PRL3 and K8 protein levels could be reversed by introduction of an shRNA resistant PKP3 cDNA. Inhibition of K8 expression in the PKP3 knockdown clone S10, led to a decrease in cell migration and lamellipodia formation. Further, the K8 PKP3 double knockdown clones showed a decrease in colony formation in soft agar and decreased tumorigenesis and metastasis in nude mice. These results suggest that a stabilisation of K8 filaments leading to an increase in migration and transformation may be one mechanism by which PKP3 loss leads to tumor progression and metastasis.


Assuntos
Transformação Celular Neoplásica/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Queratina-8/metabolismo , Metástase Neoplásica/fisiopatologia , Neoplasias/metabolismo , Placofilinas/deficiência , Proteínas Tirosina Fosfatases/metabolismo , Animais , Western Blotting , Desmossomos/metabolismo , Eletroforese em Gel Bidimensional , Transferência Ressonante de Energia de Fluorescência , Técnicas de Silenciamento de Genes , Células HCT116 , Humanos , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Camundongos Nus , Microscopia Confocal , Microscopia de Fluorescência , Oligonucleotídeos/genética , Fosforilação , Placofilinas/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Mol Biol Cell ; 22(21): 4068-78, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21900500

RESUMO

Keratins are cytoplasmic intermediate filament proteins preferentially expressed by epithelial tissues in a site-specific and differentiation-dependent manner. The complex network of keratin filaments in stratified epithelia is tightly regulated during squamous cell differentiation. Keratin 14 (K14) is expressed in mitotically active basal layer cells, along with its partner keratin 5 (K5), and their expression is down-regulated as cells differentiate. Apart from the cytoprotective functions of K14, very little is known about K14 regulatory functions, since the K14 knockout mice show postnatal lethality. In this study, K14 expression was inhibited using RNA interference in cell lines derived from stratified epithelia to study the K14 functions in epithelial homeostasis. The K14 knockdown clones demonstrated substantial decreases in the levels of the K14 partner K5. These cells showed reduction in cell proliferation and delay in cell cycle progression, along with decreased phosphorylated Akt levels. K14 knockdown cells also exhibited enhanced levels of activated Notch1, involucrin, and K1. In addition, K14 knockdown AW13516 cells showed significant reduction in tumorigenicity. Our results suggest that K5 and K14 may have a role in maintenance of cell proliferation potential in the basal layer of stratified epithelia, modulating phosphatidylinositol 3-kinase/Akt-mediated cell proliferation and/or Notch1-dependent cell differentiation.


Assuntos
Diferenciação Celular , Proliferação de Células , Células Epiteliais/fisiologia , Queratina-14/metabolismo , Queratina-5/metabolismo , Animais , Antígenos de Diferenciação/metabolismo , Adesão Celular , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Ativação Enzimática , Células Epiteliais/metabolismo , Matriz Extracelular/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Interfase , Queratina-14/genética , Queratina-5/genética , Camundongos , Camundongos Nus , Microscopia de Fluorescência , Transplante de Neoplasias , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Receptor Notch1/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA