Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 18(11): e1010506, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36441670

RESUMEN

Short telomeres induce a DNA damage response (DDR) that evokes apoptosis and senescence in human cells. An extant question is the contribution of telomere dysfunction-induced DDR to the phenotypes observed in aging and telomere biology disorders. One candidate is RAP1, a telomere-associated protein that also controls transcription at extratelomeric regions. To distinguish these roles, we generated a knockin mouse carrying a mutated Rap1, which was incapable of binding telomeres and did not result in eroded telomeres or a DDR. Primary Rap1 knockin embryonic fibroblasts showed decreased RAP1 expression and re-localization away from telomeres, with an increased cytosolic distribution akin to that observed in human fibroblasts undergoing telomere erosion. Rap1 knockin mice were viable, but exhibited transcriptomic alterations, proinflammatory cytokine/chemokine signaling, reduced lifespan, and decreased healthspan with increased body weight/fasting blood glucose levels, spontaneous tumor incidence, and behavioral deficits. Taken together, our data present mechanisms distinct from telomere-induced DDR that underlie age-related phenotypes.


Asunto(s)
Complejo Shelterina , Telómero , Animales , Humanos , Ratones , Longevidad , Fenotipo , Telómero/genética , Acortamiento del Telómero
2.
EMBO J ; 39(21): e103420, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32935380

RESUMEN

Short telomeres are a principal defining feature of telomere biology disorders, such as dyskeratosis congenita (DC), for which there are no effective treatments. Here, we report that primary fibroblasts from DC patients and late generation telomerase knockout mice display lower nicotinamide adenine dinucleotide (NAD) levels, and an imbalance in the NAD metabolome that includes elevated CD38 NADase and reduced poly(ADP-ribose) polymerase and SIRT1 activities, respectively, affecting many associated biological pathways. Supplementation with the NAD precursor, nicotinamide riboside, and CD38 inhibition improved NAD homeostasis, thereby alleviating telomere damage, defective mitochondrial biosynthesis and clearance, cell growth retardation, and cellular senescence of DC fibroblasts. These findings reveal a direct, underlying role of NAD dysregulation when telomeres are short and underscore its relevance to the pathophysiology and interventions of human telomere-driven diseases.


Asunto(s)
Disqueratosis Congénita/genética , Disqueratosis Congénita/metabolismo , Fibroblastos/metabolismo , NAD/metabolismo , Telomerasa/genética , Telómero/metabolismo , ADP-Ribosil Ciclasa 1/genética , Animales , Encéfalo/patología , Línea Celular , Senescencia Celular , Disqueratosis Congénita/patología , Femenino , Homeostasis , Humanos , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Niacinamida/análogos & derivados , Niacinamida/metabolismo , Fenotipo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Compuestos de Piridinio/metabolismo , Telomerasa/metabolismo
3.
Br J Haematol ; 181(3): 372-377, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29693246

RESUMEN

In a previous whole exome sequencing of patients from 41 families with Hodgkin lymphoma, we identified two families with distinct heterozygous rare coding variants in POT1 (D224N and Y36H), both in a highly conserved region of the gene. POT1 D224N mutant did not bind to a single-stranded telomere oligonucleotide in vitro suggesting the mutation perturbs POT1's ability to bind to the telomeric G-rich overhang. Human HT1080 cells expressing POT1 D224N and lymphoblastoid cells carrying Y36H both showed increased telomere length and fragility in comparison to wild type cells. This strongly suggests that mutant POT1 causes chromosome instability and may play a role in lymphomagenesis in these families.


Asunto(s)
Inestabilidad Cromosómica , Familia , Mutación de Línea Germinal , Enfermedad de Hodgkin , Mutación Missense , Proteínas de Unión a Telómeros , Sustitución de Aminoácidos , Línea Celular Tumoral , Femenino , Enfermedad de Hodgkin/genética , Enfermedad de Hodgkin/metabolismo , Enfermedad de Hodgkin/patología , Humanos , Masculino , Complejo Shelterina , Telómero/genética , Telómero/metabolismo , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo
4.
Mol Cancer ; 13: 180, 2014 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-25070141

RESUMEN

BACKGROUND: WRAP53, including α, ß and γ isoforms, plays an important role not only in the stability of p53 mRNA, but also in the assembly and trafficking of the telomerase holoenzyme. It has been considered an oncogene and is thought to promote the survival of cancer cells. The aim of this study was to detect the role of TCAB1 (except WRAP53α) in the occurrence and development of head and neck carcinomas. METHODS: Immunohistochemistry was used to detect the TCAB1 expression in clinical specimen sections and performed western blotting to check the TCAB1 expression levels in cell lines. TCAB1 was depleted using shRNA lentivirus and the knockdown efficiency was assessed using q-PCR and Western blotting. We performed CCK-8 assays and flow cytometry to check the cell proliferation potential and used the trans-well assay to test the invasion ability in vitro. Xenografts were used to detect the tumor formation potential in vivo. Moreover, we performed cDNA microarray to investigate the candidate factors involved in this process. RESULTS: We observed a notable overexpression of TCAB1 in head and neck carcinoma clinical specimens as well as in carcinoma cell lines. Knockdown of TCAB1 decreased the cellular proliferation potential and invasion ability in vitro. cDNA microarray analysis suggested the possible involvement of several pathways and factors associated with tumorigenesis and carcinoma development in the TCAB1-mediated regulation of cancers. Furthermore, the xenograft assay confirmed that the depletion of TCAB1 would inhibit tumor formation in nude mice. The immunohistochemistry results of the mice tumor tissue sections revealed that the cells in shTCAB1 xenografts showed decreased proliferation potential and increased apoptotic trend, meanwhile, the angiogenesis was inhibited in the smaller tumors form shTCAB1 cells. CONCLUSIONS: Our study demonstrated that depletion of TCAB1 decreased cellular proliferation and invasion potential both in vitro and in vivo. The data indicated that TCAB1 might facilitate the occurrence and development of head and neck carcinomas. In future, TCAB1 might be useful as a prognostic biomarker or a potential target for the diagnosis and therapy of head and neck carcinomas.


Asunto(s)
Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/terapia , Neoplasias de Cabeza y Cuello/diagnóstico , Neoplasias de Cabeza y Cuello/terapia , Terapia Molecular Dirigida , Telomerasa/metabolismo , Animales , Apoptosis , Carcinoma de Células Escamosas/irrigación sanguínea , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Femenino , Técnicas de Silenciamiento del Gen , Neoplasias de Cabeza y Cuello/irrigación sanguínea , Humanos , Ratones Endogámicos BALB C , Chaperonas Moleculares , Invasividad Neoplásica , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Biol Pharm Bull ; 37(9): 1450-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25177028

RESUMEN

Diosgenyl saponins possess a variety of biological functions. Herein, we demonstrate a new type of diosgenyl saponin derivatives that inhibit cellular proliferation of oral squamous cell carcinoma (OSCC) cell lines. Thereafter, we analyzed these cells' expression of apoptosis-related proteins. Crucial proteins that participate in apoptosis regulation including caspases 8, 9, and 3, and cleaved Bid were activated and upregulated accompanied by increased concentrations of diosgenyl saponins. Meanwhile, Bcl-2 was downregulated and mitochondrial membrane potential decreased. In our mice model of OSCC, compound 1 showed potent inhibition of solid tumor growth and salient antitumor activity. Diosgenyl ß-D-galactopyranosyl-(1→4)-ß-D-glucopyranoside might induce OSCC cell line apoptosis through extrinsic and intrinsic pathways, and might provide a mechanistic background for the development of this new type of diosgenylsaponin derivatives into anti-oral cancer agents against OSCC.


Asunto(s)
Antineoplásicos/farmacología , Diosgenina/análogos & derivados , Diosgenina/farmacología , Glicósidos/farmacología , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Caspasas/metabolismo , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Diosgenina/uso terapéutico , Femenino , Glicósidos/uso terapéutico , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Endogámicos BALB C , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Carga Tumoral/efectos de los fármacos
6.
Geroscience ; 45(4): 2213-2228, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36826621

RESUMEN

Short telomeres are a defining feature of telomere biology disorders (TBDs), including dyskeratosis congenita (DC), for which there is no effective general cure. Patients with TBDs often experience bone marrow failure. NAD, an essential metabolic coenzyme, is decreased in models of DC. Herein, using telomerase reverse transcriptase null (Tert-/-) mice with critically short telomeres, we investigated the effect of NAD supplementation with the NAD precursor, nicotinamide riboside (NR), on features of health span disrupted by telomere impairment. Our results revealed that NR ameliorated body weight loss in Tert-/- mice and improved telomere integrity and telomere dysfunction-induced systemic inflammation. NR supplementation also mitigated myeloid skewing of Tert-/- hematopoietic stem cells. Furthermore, NR alleviated villous atrophy and inflammation in the small intestine of Tert-/- transplant recipient mice. Altogether, our findings support NAD intervention as a potential therapeutic strategy to enhance aspects of health span compromised by telomere attrition.


Asunto(s)
Disqueratosis Congénita , Trasplante de Células Madre Hematopoyéticas , Humanos , Animales , Ratones , NAD , Telómero/metabolismo , Disqueratosis Congénita/genética , Disqueratosis Congénita/metabolismo , Inflamación
7.
Cell Death Dis ; 13(8): 701, 2022 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-35961969

RESUMEN

Oral submucous fibrosis (OSF) is a chronic and insidious oral potentially malignant disorder associated with a 4-17% risk of oral squamous cell carcinoma (OSCC). Our previous study found that proteasomal activator 28 gamma (PA28γ) is frequently overexpressed in oral squamous cell carcinoma and negatively correlated with poor patient prognosis. However, the role of PA28γ in the occurrence and development of OSF remains unclear. Here, we screened PA28γ-related genes and investigated their function in OSF. We demonstrated that the expression of PA28γ was positively associated with MEK1 and gradually elevated from normal to progressive stages of OSF tissue. Arecoline, a pathogenic component of OSF, could upregulate the protein levels of PA28γ and phosphorylated MEK1 and contribute to epithelial to mesenchymal transition (EMT) in epithelial cells. Notably, PA28γ could interact with MEK1 and upregulate its phosphorylation level. Furthermore, arecoline upregulated BRAF, which can interact with PA28γ and upregulate its protein level. Additionally, BRAF, PA28γ, and MEK1 could form protein complexes and then enhance the MEK1/ERK signaling pathways. The concrete mechanism of the protein stability of PA28γ is that BRAF mediates its degradation by inhibiting its ubiquitination. These findings underscore the instrumental role of PA28γ in the BRAF/MEK1 pathway and enhanced EMT through MEK1/ERK activation in OSF.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Fibrosis de la Submucosa Bucal , Arecolina/farmacología , Autoantígenos , Carcinoma de Células Escamosas/patología , Transición Epitelial-Mesenquimal/genética , Humanos , MAP Quinasa Quinasa 1/metabolismo , Neoplasias de la Boca/patología , Fibrosis de la Submucosa Bucal/genética , Complejo de la Endopetidasa Proteasomal , Proteínas Proto-Oncogénicas B-raf , Carcinoma de Células Escamosas de Cabeza y Cuello
8.
Nat Commun ; 12(1): 3520, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34112784

RESUMEN

The Immunodeficiency Centromeric Instability Facial Anomalies (ICF) 4 syndrome is caused by mutations in LSH/HELLS, a chromatin remodeler promoting incorporation of histone variant macroH2A. Here, we demonstrate that LSH depletion results in degradation of nascent DNA at stalled replication forks and the generation of genomic instability. The protection of stalled forks is mediated by macroH2A, whose knockdown mimics LSH depletion and whose overexpression rescues nascent DNA degradation. LSH or macroH2A deficiency leads to an impairment of RAD51 loading, a factor that prevents MRE11 and EXO1 mediated nascent DNA degradation. The defect in RAD51 loading is linked to a disbalance of BRCA1 and 53BP1 accumulation at stalled forks. This is associated with perturbed histone modifications, including abnormal H4K20 methylation that is critical for BRCA1 enrichment and 53BP1 exclusion. Altogether, our results illuminate the mechanism underlying a human syndrome and reveal a critical role of LSH mediated chromatin remodeling in genomic stability.


Asunto(s)
ADN Helicasas/metabolismo , Replicación del ADN , Inestabilidad Genómica , Histonas/metabolismo , Recombinasa Rad51/metabolismo , Animales , Proteína BRCA1/metabolismo , Línea Celular Tumoral , Supervivencia Celular/genética , Ensamble y Desensamble de Cromatina/genética , Secuenciación de Inmunoprecipitación de Cromatina , ADN Helicasas/deficiencia , ADN Helicasas/genética , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Replicación del ADN/genética , Epigénesis Genética , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Inestabilidad Genómica/genética , Histonas/deficiencia , Histonas/genética , Humanos , Proteína Homóloga de MRE11/genética , Proteína Homóloga de MRE11/metabolismo , Metilación , Ratones , ARN Interferente Pequeño , Recombinasa Rad51/genética , Regulación hacia Arriba
9.
Cell Death Dis ; 11(9): 742, 2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32917854

RESUMEN

Chronic inflammation induced by persistent viruses infection plays an essential role in tumor progression, which influenced on the interaction between the tumor cells and the tumor microenvironment. Our earlier study showed that ATR, a key kinase participant in single-stranded DNA damage response (DDR), was obviously activated by Epstein-Barr virus (EBV) in nasopharyngeal carcinoma (NPC). However, how EBV-induced ATR activation promotes NPC by influencing inflammatory microenvironment, such as tumor-associated macrophages (TAMs), remains elusive. In this study, we showed that EBV could promote the expression of p-ATR and M2-type TAMs transformation in clinical NPC specimens. The expression of p-ATR and M2-type TAMs were closely correlated each other and involved in TNM stage, lymph node metastasis and poor prognosis of the patients. In addition, the expression levels of CD68+CD206+, Arg1, VEGF, and CCL22 were increased in EB+ CNE1 cells, and decreased when ATR was inhibited. In the nude mice, EBV-induced ATR activation promoted subcutaneous transplanted tumor growth, higher expression of Ki67 and lung metastasis via M2-type TAMs recruitment. Experimental data also showed that the polarization of M2, the declined tumor necrosis factor-α (TNF-α) and increased transforming growth factor-ß (TGF-ß) were associated with ATR. Meanwhile, ATR activation could promote PPAR-δ and inhibited c-Jun and p-JNK expression, then downregulate JNK pathway. Collectively, our current study demonstrated the EBV infection could activate the ATR pathway to accelerate the transition of TAMs to M2, suggesting ATR knockdown could be a potential effective treatment strategy for EBV-positive NPC.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Infecciones por Virus de Epstein-Barr/complicaciones , Inmunoterapia/métodos , Activación de Macrófagos/genética , Carcinoma Nasofaríngeo/genética , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Carcinoma Nasofaríngeo/patología , Microambiente Tumoral
10.
Cancer Manag Res ; 11: 573-585, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30666155

RESUMEN

PURPOSE: Epstein-Barr virus (EBV) infection is closely associated with nasopharyngeal carcinoma (NPC) and increases the chemotherapy resistance of tumor cells. Although the mechanism by which EBV manipulates ataxia telangiectasia mutation (ATM)-mediated DNA damage response in NPC has been extensively studied, the relationship between ATR (ATM and Rad-3 related) and EBV infection is largely unexplored, and also the role of ATR in chemotherapy resistance in EBV-positive NPC has not been specifically reported. MATERIALS AND METHODS: Levels of γ-H2AX, latent membrane protein 1 (LMP1), and EBV-encoded RNA in clinical NPC and nasopharyngeal inflammation (NPI) specimens were examined using immunohistochemistry and in situ hybridization. The effects of EBV infection, chemotherapy drugs cisplatin (CDDP) and 5-fluorouracil (5-FU) treatment, and ATR silencing were assessed in NPC cells in vitro using immunofluorescence, Western blot, and flow cytometry. RESULTS: A notable increase of γ-H2AX expression was examined in the EBV-positive NPC clinical specimens. Additionally, we observed that the phosphorylation of ATR/checkpoint kinase 1 (CHK1) pathway protein was gradually activated along with the duration of EBV exposure in NPC cell lines, which was obviously inhibited after ATR depletion. Moreover, EBV infection promoted the resistance of NPC cells to CDDP and 5-FU, whereas the chemosensitivity of cells was significantly enhanced following ATR knockdown. Furthermore, ATR depletion caused both S-phase cell arrest and apoptosis, enhanced p53 phosphorylation, and impaired the formation of Rad51. CONCLUSION: Our data suggest that EBV activation of ATR-mediated DNA damage response might result in chemotherapy resistance to CDDP and 5-FU in NPC. Accordingly, ATR knockdown may serve as an effective treatment strategy for chemotherapy-resistant, EBV-positive NPC.

11.
Nat Struct Mol Biol ; 26(3): 213-219, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30833786

RESUMEN

The growth of telomerase-deficient cancers depends on the alternative lengthening of telomeres (ALT), a homology-directed telomere-maintenance pathway. ALT telomeres exhibit a unique chromatin environment and generally lack the nucleosome remodeler ATRX, pointing to an epigenetic basis for ALT. Recently, we identified a protective role for the ATRX-interacting macroH2A1.2 histone variant during homologous recombination and replication stress (RS). Consistent with an inherent susceptibility to RS, we show that human ALT telomeres are highly enriched for macroH2A1.2. However, in contrast to ATRX-proficient cells, ALT telomeres transiently lose macroH2A1.2 during acute RS to facilitate DNA double-strand break (DSB) formation, a process that is almost completely prevented by ectopic ATRX expression. Telomeric macroH2A1.2 is re-deposited in a DNA damage response (DDR)-dependent manner to promote homologous recombination-associated ALT pathways. Our findings thus identify the dynamic exchange of macroH2A1.2 on chromatin as an epigenetic link among ATRX loss, RS-induced DDR initiation and telomere maintenance via homologous recombination.


Asunto(s)
Cromatina/metabolismo , Reparación del ADN/genética , Histonas/genética , Recombinación Homóloga/genética , Homeostasis del Telómero/genética , Proteína Nuclear Ligada al Cromosoma X/genética , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Células HEK293 , Células HeLa , Humanos , Telomerasa/metabolismo
13.
Sci Rep ; 7(1): 3218, 2017 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-28607398

RESUMEN

Telomerase Cajal body protein 1 (TCAB1), which is involved in Cajal body maintenance, telomere elongation and ribonucleoprotein biogenesis, has been linked to cancer predisposition, including nasopharyngeal carcinoma (NPC), due to its oncogenic properties. However, there are no specific reports to date on the functional relevance of TCAB1 and Epstein-Barr virus (EBV), which is considered to be a risk factor for NPC. In this study, we first examined NPC clinical tissues and found a notable overexpression of TCAB1 in EBV-positive specimens. Secondly, on a cellular level, we also observed that TCAB1 expression rose gradually along with the increased duration of EBV exposure in NPC cell lines. Additionally, EBV infection promoted cell proliferation and telomerase activity, but the activation was significantly inhibited after TCAB1 knockdown. Moreover, depletion of TCAB1 caused both cell cycle arrest and apoptosis, and suppressed the activation of ataxia telangiectasia and Rad3 related protein (ATR) induced by EBV, resulting in accumulation of DNA damage. Taken together, we here demonstrate that up-regulated expression of TCAB1, induced by EBV in the development of NPC, is involved in stimulating telomerase activity and regulating the DNA damage response within the context of EBV infection.


Asunto(s)
Daño del ADN , Infecciones por Virus de Epstein-Barr/metabolismo , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Telomerasa/metabolismo , Regulación hacia Arriba , Animales , Apoptosis/genética , Puntos de Control del Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Proliferación Celular/genética , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/fisiología , Humanos , Chaperonas Moleculares , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/virología , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/virología , Interferencia de ARN , Telomerasa/genética
14.
Oncotarget ; 8(2): 2141-2152, 2017 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-27924064

RESUMEN

Precision medicine is a new strategy that aims at preventing and treating human diseases by focusing on individual variations in people's genes, environment and lifestyle. Precision medicine has been used for cancer diagnosis and treatment and shows evident clinical efficacy. Rapid developments in molecular biology, genetics and sequencing technologies, as well as computational technology, has enabled the establishment of "big data", such as the Human Genome Project, which provides a basis for precision medicine. Head and neck squamous cell carcinoma (HNSCC) is an aggressive cancer with a high incidence rate and low survival rate. Current therapies are often aggressive and carry considerable side effects. Much research now indicates that precision medicine can be used for HNSCC and may achieve improved results. From this perspective, we present an overview of the current status, potential strategies, and challenges of precision medicine in HNSCC. We focus on targeted therapy based on cell the surface signaling receptors epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF) and human epidermal growth factor receptor-2 (HER2), and on the PI3K/AKT/mTOR, JAK/STAT3 and RAS/RAF/MEK/ERK cellular signaling pathways. Gene therapy for the treatment of HNSCC is also discussed.


Asunto(s)
Carcinoma de Células Escamosas/terapia , Neoplasias de Cabeza y Cuello/terapia , Medicina de Precisión/tendencias , Animales , Humanos , Sistema de Señalización de MAP Quinasas , Terapia Molecular Dirigida , Fosfatidilinositol 3-Quinasas/metabolismo , Medicina de Precisión/métodos , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Transcripción STAT3 , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello , Serina-Treonina Quinasas TOR/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Cell Death Dis ; 8(9): e3035, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28880263

RESUMEN

Oral squamous cell carcinoma (OSCC) is a leading cause of cancer-related deaths worldwide, with 500 000 new cases each year. However, the mechanisms underlying OSCC development are relatively unknown. In this study, matrix-assisted laser desorption ionization imaging mass spectrometry (MALDI-IMS)-based proteomic strategy was used to profile the differentially expressed peptides/proteins between OSCC tissues and their adjacent noncancerous tissues. Sixty-seven unique peptide peaks and five distinct proteins were identified with changed expression levels. Among them, LRP6 expression was found to be upregulated in OSCC tissues, and correlated with a cluster of clinicopathologic parameters, including smoking, drinking, tumor differentiation status, lymph node metastasis and survival time. Notably, knockdown of LRP6 inhibited the proliferation ability of OSCC cells. Furthermore, we demonstrated that the expression of LRP6 in OSCC cells is positively correlated with its downstream oncogene, FGF8. The present study suggests that LRP6 could be a potential biomarker for OSCC patients, and might further assist in the therapeutic decisions in OSCC treatment.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/genética , Regulación Neoplásica de la Expresión Génica , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Neoplasias de la Boca/genética , Proteínas de Neoplasias/genética , Consumo de Bebidas Alcohólicas/fisiopatología , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular , Fumar Cigarrillos/fisiopatología , Factor 8 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Metástasis Linfática , Neoplasias de la Boca/diagnóstico , Neoplasias de la Boca/mortalidad , Neoplasias de la Boca/patología , Proteínas de Neoplasias/metabolismo , Estadificación de Neoplasias , Pronóstico , Proteoma/genética , Proteoma/metabolismo , Factores de Riesgo , Transducción de Señal , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Análisis de Supervivencia
16.
Oncol Rep ; 35(5): 2499-515, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26986034

RESUMEN

Oncogene activation and tumor-suppressor gene inactivation are considered as the main causes driving the transformation of normal somatic cells into malignant tumor cells. Cancer cells are the driving force of tumor development and progression. Yet, cancer cells are unable to accomplish this alone. The tumor microenvironment is also considered to play an active role rather than simply acting as a by-stander in tumor progression. Through different pathways, tumor cells efficiently recruit stromal cells, which in turn, provide tumor cell growth signals, intermediate metabolites, and provide a suitable environment for tumor progression as well as metastasis. Through reciprocal communication, cancer cells and the microenvironment act in collusion leading to high proliferation and metastatic capability. Understanding the role of the tumor microenvironment in tumor progression provides us with novel approaches through which to target the tumor microenvironment for efficient anticancer treatment. In this review, we summarize the mechanisms involved in the recruitment of stromal cells by tumor cells to the primary tumor site and highlight the role of the tumor microenvironment in the regulation of tumor progression. We further discuss the potential approaches for cancer therapy.


Asunto(s)
Neoplasias/patología , Animales , Movimiento Celular , Progresión de la Enfermedad , Fibroblastos/patología , Humanos , Invasividad Neoplásica , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Neovascularización Patológica/fisiopatología , Estrés Fisiológico , Microambiente Tumoral
17.
Sci Rep ; 6: 29943, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27416981

RESUMEN

Presently, various studies had investigated the accuracy of autofluorescence in diagnosing oral squamous cell carcinoma (OSCC) and oral potentially malignant disorders (OPMD) with diverse conclusions. This study aimed to assess its accuracy for OSCC and OPMD and to investigate its applicability in general dental practice. After a comprehensive literature search, a meta-analysis was conducted to calculate the pooled diagnostic indexes of autofluorescence for premalignant lesions (PML) and malignant lesions (ML) of the oral cavity, lung, esophagus, stomach and colorectum and to compute indexes regarding the detection of OSCC aided by algorithms. Besides, a u test was performed. Twenty-four studies detecting OSCC and OPMD in 2761 lesions were included. This demonstrated that the overall accuracy of autofluorescence for OSCC and OPMD was superior to PML and ML of the lung, esophagus and stomach, slightly inferior to the colorectum. Additionally, the sensitivity and specificity for OSCC and OPMD were 0.89 and 0.8, respectively. Furthermore, the specificity could be remarkably improved by additional algorithms. With relatively high accuracy, autofluorescence could be potentially applied as an adjunct for early diagnosis of OSCC and OPMD. Moreover, approaches such as algorithms could enhance its specificity to ensure its efficacy in primary care.


Asunto(s)
Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/patología , Sistema Digestivo/patología , Neoplasias de la Boca/diagnóstico , Neoplasias de la Boca/patología , Sistema Respiratorio/patología , Algoritmos , Fluorescencia , Humanos , Sesgo de Publicación
18.
Oncotarget ; 7(37): 60348-60365, 2016 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-27533088

RESUMEN

Oral submucous fibrosis (OSF) is a pre-cancerous lesion, which is characterized by fibrosis of the oral submucosa. Despite large body of studies focusing on this disease, the molecular mechanisms underlying the progression of OSF remained unclear. In this study, 2-DE-based proteomic approaches were employed to identify the differently expressed proteins between OSF and normal tissues. In total, 88 proteins were identified with altered expression levels, including CypA. Upregulation of CypA was further validated through immunohistochemistry staining combined with Q-PCR and western blot by using clinical samples. Statistical analyses reveal that CypA expression level is correlated to the progression of OSF. Finally, functional study reveals a pro-proliferative property of CypA in fibroblast cells by using multiple in vitro models. The present data suggest that CypA might be a potential biomarker and therapeutic target for OSF, and will lead to a better understanding of OSF pathogenesis.


Asunto(s)
Biomarcadores/metabolismo , Ciclofilina A/metabolismo , Fibroblastos/metabolismo , Fibrosis de la Submucosa Bucal/metabolismo , Proteómica/métodos , Apoptosis/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Línea Celular , Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Ciclofilina A/genética , Electroforesis en Gel Bidimensional , Humanos , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Interferencia de ARN , Espectrometría de Masas en Tándem
19.
Oncotarget ; 7(37): 59987-60004, 2016 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-27509054

RESUMEN

Oral squamous cell carcinoma (OSCC) ranks among the most common cancer worldwide, and is associated with severe morbidity and high mortality. Oral submucous fibrosis (OSF), characterized by fibrosis of the mucosa of the upper digestive tract, is a pre-malignant lesion, but the molecular mechanisms underlying this malignant transformation remains to be elucidated. In this study, matrix-assisted laser desorption ionization imaging mass spectrometry (MALDI-IMS)-based proteomic strategy was employed to profile the differentially expressed peptides/proteins between OSCC tissues and the corresponding adjacent non-cancerous OSF tissues. Sixty-five unique peptide peaks and nine proteins were identified with altered expression levels. Of them, expression of NCOA7 was found to be up-regulated in OSCC tissues by immunohistochemistry staining and western blotting, and correlated with a pan of clinicopathologic parameters, including lesion site, tumor differentiation status and lymph node metastasis. Further, we show that overexpression of NCOA7 promotes OSCC cell proliferation in either in vitro or in vivo models. Mechanistic study demonstrates that NCOA7 induces OSCC cell proliferation probably by activating aryl hydrocarbon receptor (AHR). The present study suggests that NCOA7 is a potential biomarker for early diagnosis of OSF malignant transformation, and leads to a better understanding of the molecular mechanisms responsible for OSCC development.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias de la Boca/metabolismo , Coactivadores de Receptor Nuclear/metabolismo , Fibrosis de la Submucosa Bucal/metabolismo , Adulto , Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis Linfática/genética , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/diagnóstico , Neoplasias de la Boca/patología , Coactivadores de Receptor Nuclear/genética , Fibrosis de la Submucosa Bucal/diagnóstico , Fibrosis de la Submucosa Bucal/patología , Embarazo , ARN Interferente Pequeño/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Regulación hacia Arriba
20.
J Exp Clin Cancer Res ; 35: 35, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26892607

RESUMEN

BACKGROUND: Despite recent advances in oral squamous cell carcinoma (OSCC) diagnosis and therapy, disease recurrence remains common and is strongly associated with mortality. It is therefore critical to identify new targets for both treatment and diagnostic purposes. We aimed at investigating the role of PA28α, a proteasomal activator in OSCC. METHODS: The expression of PA28α was examined in a panel of OSCC cell lines and tissues, associated with oncomine analysis. In a large OSCC patient cohort, the prognostic value of PA28α expression was evaluated. Primary clinical end points were recurrence-free and overall survival rate. Functional involvement of PA28α in OSCC was examined in both in vitro and in vivo models upon specific siRNA knockdown. RESULTS: PA28α was found to be overexpressed in OSCC cell lines and tumor tissues. High expression of PA28α was significantly associated with recurrence and poorer overall survival. Specific knockdown of PA28α inhibited OSCC cell proliferation, migration, invasion in vitro and reduced the growth of OSCC xenografts in vivo. Multivariate Cox regression analyses revealed PA28α as independent prognostic predictors. CONCLUSIONS: These results suggest that PA28α is involved in OSCC oncogenesis and may serve as a potential prognostic factor.


Asunto(s)
Carcinoma de Células Escamosas/patología , Neoplasias de la Boca/patología , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Trasplante de Neoplasias , Pronóstico , Análisis de Supervivencia , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA