Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Carcinogenesis ; 41(6): 723-733, 2020 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-31665243

RESUMO

Nasopharyngeal carcinoma (NPC) is notorious for its aggressiveness and high metastatic potential. NPC patients with distant metastasis have a particularly poor prognosis; however, evaluating metastatic potential by expression profiles of primary tumors is challenging. This study aimed to investigate the association between activation of epidermal growth factor receptor (EGFR) signaling and NPC metastasis and the underlying mechanisms. We found an association between EGFR protein overexpression and intense EGFR immunostaining in NPC samples with advanced tumor node metastasis stage, clinical stage, and distant metastasis in NPC patients. Exogenous EGF stimulates NPC mobility and invasiveness in vitro. Activation of EGFR signaling prompted PKM2 translocation to the nucleus. Silencing either EGFR or PKM2 attenuates NPC cell aggressiveness in vitro and in vivo. Blocking EGFR signaling with cetuximab suppressed NPC cell invasiveness in vitro and metastatic potential in vivo. Comprehensive analyses of transcriptome profiles indicated that the EGFR-PKM2 axis activates a number of novel metastasis promoters, including F3, FOSL1, EPHA2, ANTXR2, and AKR1C2. Finally, we found that the metastasis-promoting function of the EGFR-PKM2 axis is dependent on nuclear PKM2 regulation of the transcription of metastasis-related genes, including FOSL1 and ANTXR2. Our study indicates that EGFR-PKM2 signaling promotes NPC cell invasion and metastasis through induction of FOSL1 and ANTXR2 and identifies EGFR as a promising biomarker for predicting the risk of distant metastasis.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Carcinoma Nasofaríngeo/patologia , Neoplasias Nasofaríngeas/secundário , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de Peptídeos/metabolismo , Hormônios Tireóideos/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/genética , Proteínas de Transporte/genética , Movimento Celular , Proliferação de Células , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Invasividade Neoplásica , Prognóstico , Proteínas Proto-Oncogênicas c-fos/genética , Receptores de Peptídeos/genética , Taxa de Sobrevida , Hormônios Tireóideos/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas de Ligação a Hormônio da Tireoide
2.
Carcinogenesis ; 41(9): 1282-1293, 2020 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31826234

RESUMO

Nasopharyngeal carcinoma (NPC) originates via malignant transformation of the pseudostratified nasopharyngeal epithelium, composed of basal and luminal cells. Super enhancers (SEs) are large clusters of cis-elements involved in the regulation of gene expression through epigenetic regulatory mechanisms. In this study, we demonstrated that basal cell-specific proteins are highly expressed, whereas luminal cell proteins are downregulated in NPC, implying a perturbation of basal-to-luminal differentiation during NPC development. We characterized NPC cell models according to different molecular signatures associated with their differentiation status and found that distinct SE landscapes are tightly associated with basal or luminal-like molecular signatures in NPC cells. Furthermore, the transcription of ΔNP63α, a prominent isoform of TP63, was found to be driven by SEs in NPC cells. Data from chromatin immunoprecipitation (ChIP)-sequencing showed that ΔNP63α largely occupied regions of SEs associated with basal cell-specific genes. Silencing of ΔNP63α led to a loss of H3K27ac occupancy at basal-type SEs and triggered a basal-to-luminal gene expression signature switch, suggesting that ΔNP63α is a master factor contributing to the perturbation of luminal differentiation. Integrative transcriptomics analysis also revealed that ΔNP63α acts as a core factor involved in the dysregulation of gene expression in NPC. Furthermore, ΔNP63α enhanced EGF-stimulated NF-κB activation in NPC cells by activating SE-mediated EGFR transcription. Finally, depletion of ΔNP63α in NPC cells induced robust growth inhibition of NPC cells in vitro and in vivo. Our data revealed that ΔNP63α-dependent SE reprogramming contributes to the blockade of luminal differentiation and uncontrolled proliferation in NPC.


Assuntos
Carcinoma Basocelular/patologia , Diferenciação Celular , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Carcinoma Nasofaríngeo/patologia , Neoplasias Nasofaríngeas/patologia , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma Basocelular/genética , Carcinoma Basocelular/metabolismo , Proliferação de Células , Elementos Facilitadores Genéticos , Epigênese Genética , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Prognóstico , Fatores de Transcrição/genética , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/genética , Ensaios Antitumorais Modelo de Xenoenxerto
3.
J Cell Physiol ; 233(8): 5537-5549, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29266238

RESUMO

Nasopharyngeal carcinoma (NPC) is a unique EBV-associated subtype of head and neck cancer, which has the highest incidence in Southern China and eastern South Asia. The interaction between genetic risk factors and environmental challenge, have been considered to contribute to the development of nasopharyngeal carcinogenesis. Constitutive activation of NF-κB signaling has been seen in NPC tissues and is associated with unfavorable prognosis. Recently, several whole exome sequencing study consistently revealed that high frequency mutations of NF-κB pathway negative regulators is common in nasopharyngeal carcinoma, which reinforce the importance of NF-κB driving oncogenesis. This review focuses on the current state of research in role of NF-κB in NPC carcinogenesis. We summarized the newly identified loss of function (LOF) mutations on NF-κB negative regulators leading to it's activation bypass LMP-1 stimulation. We discussed the critical role of NF-κB activation in immortalization and transformation of nasopharygeal epithelium. We also depicted how NF-κB signaling mediated chronic inflammation contribute to persistent EBV infection, immune evasion of EBV infected cells, metabolic reprogramming, and cancer stem cells (CSCs) formation in NPC. Lastly, we discussed the clinical resonance of targeting NF-κB for NPC precise therapy.


Assuntos
Carcinogênese/genética , Herpesvirus Humano 4/patogenicidade , NF-kappa B/genética , Carcinoma Nasofaríngeo/genética , Neoplasias Nasofaríngeas/genética , Transdução de Sinais/genética , Linhagem Celular Tumoral , Epitélio/virologia , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/virologia , Humanos , Células-Tronco Neoplásicas/virologia
4.
J Cell Physiol ; 233(6): 5002-5013, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29227538

RESUMO

Hepatocellular carcinoma (HCC) is the most common primary tumor of the liver and the sixth most common lethal cancer worldwide. Recent evidences demonstrated that oxidored nitro domain containing protein 1 (NOR1), a putative tumor suppressor gene, is overexpressed in human HCC tissues. However, the role of NOR1 in HCC development remains unclear. Here, we described that NOR1 protein level is elevated in HCC and is associated with poorer clinical outcome. However, ecotopic overexpression of NOR1 protein in human HCC cell line HepG2 cells had no effect on cells proliferation, migration, and clonality. Immunofluoresence assay indicates NOR1 protein is mainly expressed at CD163 positive M2 tumor associated macrophages (TAMs). To explore the role of NOR1 in the development of HCC, we interrogated the susceptibility of mice lacking the NOR1 gene to DEN-induced hepatocarcinogenesis. NOR1 deficient mice displayed resistance to DEN-induced HCC. We also demonstrate that mNOR1 protein is enriched in F4/80 positive Kupffer cells (KCs) infiltrated in DEN induced murine HCC tissues. Loss of NOR1 led to increase of iNOS whereas decrease of Arg1, Ym1 expression in KCs. Overexpression of NOR1 in THP-1 macrophages led to decrease of iNOS but increase of Arg1. Mechanistic investigations showed that inflammatory cytokines IL-6, TNF-α production, and NF-κB activation were also decreased in NOR1 knockout mice exposed to DEN treatment. Our data suggested that NOR1 is overexpressed in HCC associated TAMs and promotes M2 alternative polarization. Genetic deletion of NOR1 in mice leads to impairment of IL-6 production and NF-κB activation, which in turn attenuates DEN-induced HCC development.


Assuntos
Carcinoma Hepatocelular/metabolismo , Dietilnitrosamina , Neoplasias Hepáticas Experimentais/metabolismo , Macrófagos/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Animais , Arginase/metabolismo , Carcinoma Hepatocelular/induzido quimicamente , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Células Hep G2 , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-6/metabolismo , Células de Kupffer/metabolismo , Células de Kupffer/patologia , Lectinas/metabolismo , Neoplasias Hepáticas Experimentais/induzido quimicamente , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/patologia , Macrófagos/patologia , Masculino , Proteínas de Membrana Transportadoras/deficiência , Proteínas de Membrana Transportadoras/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Fenótipo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo
5.
Mol Biol Rep ; 45(5): 1277-1287, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30074127

RESUMO

The RASSF1A, a microtubule associated protein, is a well-known tumor suppressor silenced in various cancer via promoter hypermethylation. RASSF1A is implicated in the regulation of cellular proliferation and apoptosis. However, its role in melanoma A375 cells invasion and metastasis remain unclear. Here, we report an unusual dual function role of ectopic RASSF1A in A375 cells. RASSF1A suppressed A375 cells proliferation but enhanced cells migration, invasiveness and metastatic potential in vivo. We demonstrated RASSF1A simultaneously up-regulated p21 and vimentin expression in A375 cells. Increase of vimentin expression contributes to RASSF1A mediated enhancement of cells mobility and invasion. Transcriptome assay unclosed that RASSF1A promoted IL-6 expression in A375 cells, which in turn activate JAK2/STAT3 signaling. Treatment with recombinant IL-6 enhanced both p21 and vimentin protein level in the empty vector transfected A375 cells to similar level as RASSF1A expressing cells. In contrast, knockdown IL-6 expression by siRNAs decreased p21 and vimentin level in RASSF1A expressing cells. Blockade of JAK2/STAT3 signaling by use of JAK2 inhibitor WP1066 led to decrease of IL-6, p21 and vimentin protein in RASSF1A expressing cells. Our findings unclosed a unusual dual functionality of ectopic RASSF1A overexpression in A375 cells by regulating IL-6/STAT3 regulatory loop, suggesting it should be cautious about the safety of RASSF1A-based gene therapy.


Assuntos
Interleucina-6/metabolismo , Melanoma/metabolismo , Fator de Transcrição STAT3/metabolismo , Proteínas Supressoras de Tumor/biossíntese , Animais , Apoptose/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Xenoenxertos , Humanos , Interleucina-6/genética , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Masculino , Melanoma/genética , Melanoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Metástase Neoplásica , Fator de Transcrição STAT3/genética , Transdução de Sinais , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Regulação para Cima , Vimentina/genética , Vimentina/metabolismo
7.
Genome ; 55(1): 8-14, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22149714

RESUMO

The yellow mustard plant in Northern Shaanxi is a precious germplasm, and the yellow seed trait is controlled by a single recessive gene. In this report, amplified fragment length polymorphism (AFLP) and simple sequence repeat (SSR) techniques were used to identify markers linked to the brown seed locus in an F(2) population consisting of 1258 plants. After screening 256 AFLP primer combinations and 456 pairs of SSR primers, we found 14 AFLP and 2 SSR markers that were closely linked to the brown seed locus. Among these markers, the SSR marker CB1022 showed codominant inheritance. By integrating markers previously found to be linked to the brown seed locus into the genetic map of the F(2) population, 23 markers were linked to the brown seed locus. The two closest markers, EA02MC08 and P03MC08, were located on either side of the brown seed locus at a distance of 0.3 and 0.5 cM, respectively. To use the markers for the breeding of yellow-seeded mustard plants, two AFLP markers (EA06MC11 and EA08MC13) were converted into sequence-characterized amplified region (SCAR) markers, SC1 and SC2, with the latter as the codominant marker. The two SSR markers were subsequently mapped to the A9/N9 linkage group of Brassica napus L. by comparing common SSR markers with the published genetic map of B. napus. A BLAST analysis indicated that the sequences of seven markers showed good colinearity with those of Arabidopsis chromosome 3 and that the homolog of the brown seed locus might exist between At3g14120 and At3g29615 on this same chromosome. To develop closer markers, we could make use of the sequence information of this region to design primers for future studies. Regardless, the close markers obtained in the present study will lay a solid foundation for cloning the yellow seed gene using a map-based cloning strategy.


Assuntos
Arabidopsis/genética , Brassica/genética , Mapeamento Cromossômico/métodos , Ligação Genética , Sinapis/genética , Análise do Polimorfismo de Comprimento de Fragmentos Amplificados , DNA de Plantas/química , Genes de Plantas , Marcadores Genéticos
8.
J Cancer ; 12(11): 3315-3324, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33976741

RESUMO

Background: RNA-binding proteins (RBPs) play essential roles in post-transcriptional control of gene expression. Dysregulation of RBPs is intensively implicated in development and progression of human diseases, including cancers. However, the roles of RBPs in nasopharyngeal carcinoma (NPC), which is a distinct subtype of head and neck cancer, remain elusive. Methods: NPC-related RBPs were explored by analyzing GEO database and high-throughput proteomic data obtained from crosslinking immunoprecipitation. The expression levels of Y box binding protein 1 (YBX1) protein in NPC samples were measured by immunohistochemistry (IHC) staining. The association of YBX1 protein levels with prognosis of NPC patients was analyzed by Kaplan-Meier Plotter. The expression levels of YBX1 in NPC cells were inhibited by RNA interference. Cell growth was measured by CCK-8 assay. Cell mobility and invasiveness were measured by transwell assays. Tumorigenicity was measured by using a xenograft tumor assay. The expression levels of mRNAs or proteins were determined by qPCR or western blot assays, respectively. The mRNAs binding to YBX1 were determined by RNA immunoprecipitation (RIP) and qPCR. The effect of YBX1 on mRNA translation was measured by luciferase reporter assay. Results: In the present study, we demonstrated a differentially expressed RBPs profile between NPC and its normal counterpart. Among these aberrantly expressed RBPs, YBX1 was overexpressed in NPC. We found that YBX1 is mainly localized in the cytoplasm of NPC cells. Loss of YBX1 led to reduced cell proliferation, migration and invasiveness in vitro, and reduced tumorigenicity in vivo. Overexpression of YBX1 associates with high expression of cell cycle G2/M checkpoint modulators. In addition, YBX1 promotes AURKA protein expression by directly binding to its mRNA. Loss of YBX1 leads to reduction of AURKA protein level. Forced expression of AURKA rescues cell proliferation and invasiveness in YBX1-silenced NPC cell. Conclusions: The current study indicated that YBX1 promotes NPC cell proliferation and invasiveness through enhancing protein synthesis of AURKA.

9.
Mol Oncol ; 14(6): 1282-1296, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32216017

RESUMO

Head and neck squamous cell carcinoma (HNSCC) constitute approximately 4% of all cancers worldwide. In this study, we analyzed the expression profile of the long noncoding RNA (lncRNA) of 502 HNSCC patients from The Cancer Genome Atlas database. Among the differentially expressed lncRNAs between HNSCC and normal samples, LNCAROD is overexpressed in HNSCC and associated with advanced T stage and shortened overall survival. The N6-methyladenosine (m6A) modification mediated by METTL3 and METTL14 enhanced the stability of LNCAROD in HNSCC cells. Depletion of LNCAROD attenuated cell proliferation, mobility in vitro, and tumorigenicity in vivo, whereas overexpression of LNCAROD exerted opposite effects. LNCAROD is mainly distributed in nucleus and binds with YBX1 and HSPA1A proteins. Silencing either YBX1 or HSPA1A did not affect the level of LNCAROD. However, loss of LNCAROD led to shortened half-life of YBX1 protein. Mechanistically, LNCAROD protected YBX1 from proteasomal degradation by facilitating YBX1-HSPA1A protein-protein interaction. Depletion of HSPA1A in LNCAROD-overexpressing cells resulted in accelerated proteasomal degradation of YBX1 protein. Moreover, re-expression of Flag-YBX1 in LNCAROD-silenced cells rescued malignant behavior of HNSCC cells. Our study indicates that LNCAROD is an oncogenic lncRNA and dysregulation of m6A modification might account for aberrant expression of LNCAROD in HNSCC. LNCAROD acts as a scaffold for the interaction between YBX1 and HSPA1A, preventing proteasomal degradation of YBX1 in HNSCC cells.


Assuntos
Adenosina/análogos & derivados , Progressão da Doença , Proteínas de Choque Térmico HSP70/metabolismo , Neoplasias de Cabeça e Pescoço/genética , Estabilidade de RNA/genética , RNA Longo não Codificante/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Proteína 1 de Ligação a Y-Box/metabolismo , Adenosina/metabolismo , Carcinogênese/genética , Carcinogênese/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Masculino , Metilação , Metiltransferases/metabolismo , Pessoa de Meia-Idade , Modelos Biológicos , Invasividade Neoplásica , Estadiamento de Neoplasias , Prognóstico , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Resultado do Tratamento
10.
J Cancer ; 11(9): 2485-2495, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32201519

RESUMO

Background: Nasopharyngeal carcinoma (NPC) is a unique subtype of head and neck cancer, within highest incidence in South China and southeastern Asia but rare in other regions worldwide. FOXA1 is a pioneer factor implicated in various human malignancies. Downregulation of FOXA1 promotes NPC cells proliferation, invasiveness in vitro and tumorigenicity in vivo. However, it is remain elusive to determine whether microRNAs (miRNAs) regulated by FOXA1 contribute to NPC progression. Methods: In this study, differentially expressed miRNAs and mRNAs induced by FOXA1 expression were determined by microarray. Integrative miRNA-mRNA regulatory networks mediated by FOXA1 in NPC were established. The expressions of differentially expressed miRNAs in NPC cells were measured by quantitative reverse-transcription PCR. Cell viability was determined by CCK-8 assays. Cell migration and invasiveness were measured by Transwell assays. The correlation between miRNAs and its target mRNAs was analyzed. Results: FOXA1 suppressed the expression of miR-100-5p and miR-125b-5p in NPC cells. Silencing either miR-100-5p or miR-125b-5p inhibited the malignant behaviors of NPC cells, whereas re-expression of miR-100-5p or miR-125b-5p restored the malignancy of NPC cells repressed by FOXA1. Mechanistically, miR-100-5p or miR-125b-5p suppressed RASGRP3 or FOXN3 expression respectively via direct binding to its 3'-UTR. Furthermore, we demonstrated that FOXA1 induced RASGRP3 or FOXN3 expression via inhibiting miR-100-5p or miR-125b-5p. Upregulation of RASGRP3 or FOXN3 contributed to inhibition of NPC by FOXA1. We also demonstrated that the mRNA levels of RASGRP3 and FOXN3 are positively correlated with FOXA1. Conclusion: Our study provided evidence the first time that FOXA1 suppresses NPC cells via downregulation of miR-100-5p or miR-125b-5p.

11.
Mol Metab ; 20: 1-13, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30553771

RESUMO

BACKGROUND: Cancer cells favor the use of less efficient glycolysis rather than mitochondrial oxidative phosphorylation to metabolize glucose, even in oxygen-rich conditions, a distinct metabolic alteration named the Warburg effect or aerobic glycolysis. In adult cells, bifunctional 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase (PFKFB) family members are responsible for controlling the steady-state cytoplasmic levels of fructose-2,6-bisphosphate, which allosterically activates 6-phosphofructo-1-kinase, the key enzyme catalyzing the rate-limiting reaction of glycolysis. PFKFB3 and PFKFB4 are the two main isoenzymes overexpressed in various human cancers. SCOPE OF REVIEW: In this review, we summarize recent findings on the glycolytic and extraglycolytic roles of PFKFB3 and PFKFB4 in cancer progression and discuss potential therapies for targeting of PFKFB3 and PFKFB4. MAJOR CONCLUSIONS: PFKFB3 has the highest kinase activity to shunt glucose toward glycolysis, whereas PFKFB4 has more FBPase-2 activity, redirecting glucose toward the pentose phosphate pathway, providing reducing power for lipid biosynthesis and scavenging reactive oxygen species. Co-expression of PFKFB3 and PFKFB4 provides sufficient glucose metabolism to satisfy the bioenergetics demand and redox homeostasis requirements of cancer cells. Various reversible post-translational modifications of PFKFB3 enable cancer cells to flexibly adapt glucose metabolism in response to diverse stress conditions. In addition to playing important roles in tumor cell glucose metabolism, PFKFB3 and PFKFB4 are widely involved in multiple biological processes, such as cell cycle regulation, autophagy, and transcriptional regulation in a non-glycolysis-dependent manner.


Assuntos
Glicólise , Neoplasias/metabolismo , Fosfofrutoquinase-2/metabolismo , Humanos , Neoplasias/genética , Via de Pentose Fosfato , Fosfofrutoquinase-2/genética
12.
Cancer Lett ; 442: 1-14, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30392786

RESUMO

Nasopharyngeal carcinoma (NPC) is a unique subtype of head and neck squamous carcinoma that is notorious for its high metastatic potential. In this study, we reported that FOXA1 protein was decreased in NPC cells. Loss of FOXA1 is associated with lymph node metastasis and poor prognosis. Silencing FOXA1 in NP69 and C666-1 NPC cells accelerated cell proliferation and migration, while re-expression of FOXA1 has opposite effects. Microarray and RNA-seq analysis revealed that re-expression of FOXA1 in NPC cells reprogrammed the TGF-ß-stimulated transcription program, which is characterized by promotion of TGF-ß-inducible tumor-suppressive targets but repression of TGF-ß-inducible oncogenes expression in NPC cells, leading to restoration of NPC cell sensitivity to TGF-ß's growth-inhibitory effect. BAMBI, a TGF-ß responsive tumor suppressor, was induced by FOXA1 in NPC cells. FOXA1 binding on the BAMBI gene facilitated SMAD2/3 binding to the BAMBI promoter via increasing BAMBI associated H3K4me1 and H3K27ac modification. Enforced expression of BAMBI in NPC cells suppressed cell proliferation and invasiveness. Our data suggested that FOXA1 is a master factor in controlling the TGF-ß-stimulated transcriptome and a regulator of TGF-ß biological functions in NPC oncogenesis.


Assuntos
Reprogramação Celular , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Ativação Transcricional , Fator de Crescimento Transformador beta/metabolismo , Acetilação , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Fator 3-alfa Nuclear de Hepatócito/genética , Histonas/metabolismo , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/secundário , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patologia , Invasividade Neoplásica , Regiões Promotoras Genéticas , Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Exp Clin Cancer Res ; 37(1): 155, 2018 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-30012174

RESUMO

In the publication of this article [1], there is an error in the Figure caption of Figs. 2, 3 and 4. This has now been included in this correction. The authors declare that these corrections do not change the results or conclusions of this paper.

14.
J Exp Clin Cancer Res ; 37(1): 118, 2018 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-29907133

RESUMO

BACKGROUND: Cancer stem cells (CSCs) or tumor-initiating cells (TICs) represent a small population of cancer cells with self-renewal and tumor-initiating properties. Unlike the bulk of tumor cells, CSCs or TICs are refractory to traditional therapy and are responsible for relapse or disease recurrence in cancer patients. Stem cells have distinct metabolic properties compared to differentiated cells, and metabolic rewiring contributes to self-renewal and stemness maintenance in CSCs. MAIN BODY: Recent advances in metabolomic detection, particularly in hyperspectral-stimulated raman scattering microscopy, have expanded our knowledge of the contribution of lipid metabolism to the generation and maintenance of CSCs. Alterations in lipid uptake, de novo lipogenesis, lipid droplets, lipid desaturation, and fatty acid oxidation are all clearly implicated in CSCs regulation. Alterations on lipid metabolism not only satisfies the energy demands and biomass production of CSCs, but also contributes to the activation of several important oncogenic signaling pathways, including Wnt/ß-catenin and Hippo/YAP signaling. In this review, we summarize the current progress in this attractive field and describe some recent therapeutic agents specifically targeting CSCs based on their modulation of lipid metabolism. CONCLUSION: Increased reliance on lipid metabolism makes it a promising therapeutic strategy to eliminate CSCs. Targeting key players of fatty acids metabolism shows promising to anti-CSCs and tumor prevention effects.


Assuntos
Metabolismo dos Lipídeos , Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Autofagia , Biomarcadores , Vias Biossintéticas/efeitos dos fármacos , Metabolismo Energético , Espaço Extracelular/metabolismo , Ácidos Graxos/metabolismo , Humanos , Ferro/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipogênese , Lipólise , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Oxirredução , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA