Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Carcinogenesis ; 41(7): 927-939, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-31584613

RESUMEN

New chemotherapeutic agents are needed for pancreatic cancer (PC). We have previously shown that phospho-valproic acid (MDC-1112) is effective in cell-line xenografts of PC. Here, we explored whether MDC-1112 is effective in additional clinically relevant animal models of PC and whether MDC-1112 enhances the anticancer effect of clinically used chemotherapeutic agents. MDC-1112 alone strongly reduced patient-derived pancreatic tumor xenograft growth, and extended survival of LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx1-Cre (KPC) mice. In both models, MDC-1112 inhibited STAT3 activation and its downstream signals, including Bcl-xL and cyclin D1. In human PC cell lines, P-V enhanced the growth inhibitory effect of gemcitabine (GEM), Abraxane and 5-FU, but not that of irinotecan. Normal human pancreatic epithelial cells were more resistant to the cytotoxic effects of MDC-1112/GEM combination. Furthermore, MDC-1112 enhanced GEM's effect on colony formation, apoptosis, cell migration, and cell invasion. In vivo, MDC-1112 and GEM, given alone, reduced patient-derived pancreatic tumor xenograft growth by 58% and 87%, respectively; whereas MDC-1112/GEM combination reduced tumor growth by 94%, inducing tumor stasis. In conclusion, MDC-1112 should be further explored as a potential agent to be used in combination with GEM for treating PC.


Asunto(s)
Anomalías Múltiples/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Queratocono/congénito , Organofosfatos/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Ácido Valproico/análogos & derivados , Anomalías Múltiples/patología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Modelos Animales de Enfermedad , Humanos , Queratocono/tratamiento farmacológico , Queratocono/patología , Ratones , Neoplasias Pancreáticas/patología , Transducción de Señal/efectos de los fármacos , Ácido Valproico/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
2.
J Cell Mol Med ; 24(1): 304-316, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31657132

RESUMEN

The role of long non-coding RNAs (lncRNAs) in thyroid carcinoma (TC), the most frequent endocrine malignancy, has been extensively examined. This study investigated effect of interaction among lncRNA TNRC6C-AS1, serine/threonine-protein kinase 4 (STK4) and Hippo signalling pathway on TC. Initially, lncRNA TNRC6C-AS1 expression in TC tissues was detected. To explore roles of lncRNA TNRC6C-AS1, STK4 and Hippo signalling pathway in TC progression, their expressions were altered. Interaction between lncRNA TNRC6C-AS1 and STK4, STK4 promoter methylation, or Hippo signalling pathway was verified. After that, a series of experiments were employed to evaluate in vitro ability of apoptosis, proliferation and autophagy of TC cells and in vivo tumorigenicity, and tumour growth of TC cells. lncRNA TNRC6C-AS1 was highly expressed while STK4 was poorly expressed in TC tissues. LncRNA TNRC6C-AS1 promoted the STK4 methylation and down-regulated STK4 expression, which further activated the Hippo signalling pathway. STK4 silencing was observed to promote the proliferation ability of TC cells, inhibit the apoptosis and autophagy abilities, as well as enhance the tumorigenicity and tumour growth. Moreover, the in vitro proliferation ability as well as the in vivo tumorigenicity and tumour growth of TC cells were inhibited after the blockade of Hippo signalling pathway, while the apoptosis and autophagy abilities were promoted. The results demonstrate that the lncRNA TNRC6C-AS1 increases STK4 promoter methylation to down-regulate STK4 expression, thereby promoting the development of TC through activation of Hippo signalling pathway. It highlights that lncRNA TNRC6C-AS1 may be a novel therapeutic target for the treatment of TC.


Asunto(s)
Apoptosis/genética , Autofagia/genética , Metilación de ADN/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Largo no Codificante/metabolismo , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Secuencia de Bases , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Vía de Señalización Hippo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Modelos Biológicos , Proteínas Serina-Treonina Quinasas/genética , ARN Largo no Codificante/genética , Transducción de Señal
3.
Carcinogenesis ; 40(12): 1480-1491, 2019 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-30994173

RESUMEN

New therapeutic strategies against glioblastoma multiforme (GBM) are urgently needed. Signal transducer and activator of transcription 3 (STAT3), constitutively active in many GBM tumors, plays a major role in GBM tumor growth and represents a potential therapeutic target. We have documented previously that phospho-valproic acid (MDC-1112), which inhibits STAT3 activation, possesses strong anticancer properties in multiple cancer types. In this study, we explored the anticancer efficacy of MDC-1112 in preclinical models of GBM, and evaluated its mode of action. MDC-1112 inhibited the growth of multiple human GBM cell lines in a concentration- and time-dependent manner. Normal human astrocytes were resistant to MDC-1112, indicating selectivity. In vivo, MDC-1112 reduced the growth of subcutaneous GBM xenografts in mice by up to 78.2% (P < 0.01), compared with the controls. Moreover, MDC-1112 extended survival in an intracranial xenograft model. Although all vehicle-treated mice died by 19 days of treatment, 7 of 11 MDC-1112-treated mice were alive and healthy by the end of 5 weeks, with many showing tumor regression. Mechanistically, MDC-1112 inhibited STAT3 phosphorylation at the serine 727 residue, but not at tyrosine 705, in vitro and in vivo. STAT3 overexpression rescued GBM cells from the cell growth inhibition by MDC-1112. In addition, MDC-1112 reduced STAT3 levels in the mitochondria and enhanced mitochondrial levels of reactive oxygen species, which triggered apoptosis. In conclusion, MDC-1112 displays strong efficacy in preclinical models of GBM, with the serine 727 residue of STAT3 being its key molecular target. MDC-1112 merits further evaluation as a drug candidate for GBM. New therapeutic options are needed for glioblastoma. The novel agent MDC-1112 is an effective anticancer agent in multiple animal models of glioblastoma, and its mechanism of action involves the inhibition of STAT3 phosphorylation, primarily at its Serine 727 residue.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/patología , Glioblastoma/patología , Organofosfatos/farmacología , Factor de Transcripción STAT3/metabolismo , Ácido Valproico/análogos & derivados , Animales , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Glioblastoma/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosforilación/efectos de los fármacos , Factor de Transcripción STAT3/efectos de los fármacos , Ácido Valproico/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Surg Innov ; 24(1): 29-34, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27634477

RESUMEN

The study assessed the role of an activated carbon nanoparticle lymphatic tracer in reducing unintentional damage to the parathyroid glands during thyroidectomy for papillary thyroid non-microcarcinoma diagnosed intraoperatively by cryosections. A total of 103 patients with papillary thyroid non-microcarcinomas diagnosed by intraoperative cryosection were randomly assigned to receive routine radical thyroidectomy or radical thyroidectomy following administration of activated carbon nanoparticle lymphatic tracer to the contralateral thyroid, at the department of Thyroid Surgery, Sun Yat-sen Memorial Hospital (Guangzhou, China), between January 2012 and May 2013. The success of level VI lymphadenectomy and postoperative parathyroid function were compared. Administration of the activated carbon nanoparticle lymphatic tracer did not affect the frequency of recovered lymph nodes containing metastases; however, it did significantly reduce the incidence of permanent and transient hypoparathyroidism from 2 to 0 and 18 to 6, and reduced the mean recovery time for transient hypoparathyroidism from 57.0 days to 22.3 days. Administration of activated carbon nanoparticles to the contralateral thyroid after intraoperative cryosections did not contribute to lymphadenectomy for papillary thyroid non-microcarcinoma, but significantly protected parathyroid functions. This approach could decrease the morbidity of radical thyroidectomy and the occurrence of hypoparathyroidism.


Asunto(s)
Carbono , Carcinoma Papilar/cirugía , Hipoparatiroidismo/prevención & control , Nanopartículas , Neoplasias de la Tiroides/cirugía , Tiroidectomía/métodos , Adulto , Carcinoma Papilar/patología , Femenino , Humanos , Hipoparatiroidismo/etiología , Escisión del Ganglio Linfático , Ganglios Linfáticos/patología , Masculino , Persona de Mediana Edad , Glándulas Paratiroides , Neoplasias de la Tiroides/patología , Tiroidectomía/efectos adversos
5.
World J Surg Oncol ; 12: 29, 2014 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-24485040

RESUMEN

BACKGROUND: High expression of aldehyde dehydrogenase 1 (ALDH1) has been confirmed in many tumors. This enzyme plays an important role in tumor proliferation, metastasis, and drug resistance. However, in the case of papillary thyroid carcinoma (PTC), the relationship between ALDH1 expression and prognosis remains unknown. METHOD: We used tissue microarrays to evaluate ALDH1A1 expression in 247 surgically resected PTC specimens by immunochemistry, and correlated the findings with the clinicopathological parameters. RESULT: ALDH1A1 levels were significantly higher than in normal thyroid tissues. Moreover, ALDH1A1 overexpression was significantly associated with extrathyroid extension (P = 0.001), pT status (P < 0.001), pN status (P = 0.016) and TNM stage (P < 0.001). The Kaplan-Meier survival analysis suggested that high ALDH1A1 expression reflects a poorer lymph node recurrence-free survival (LN-RFS) and distant recurrence-free survival (DRFS) in PTC patients, as compared with patients having low ALDH1A1 expression. Multivariate analysis confirmed the ALDH1A1 expression was an independent prognostic factor for LN-RFS and DRFS in PTC patients. CONCLUSION: In conclusion, high ALDH1A1 expression correlates with poor survival in PTC patients.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Biomarcadores de Tumor/metabolismo , Carcinoma Papilar/metabolismo , Recurrencia Local de Neoplasia/metabolismo , Neoplasias de la Tiroides/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Familia de Aldehído Deshidrogenasa 1 , Carcinoma Papilar/mortalidad , Carcinoma Papilar/secundario , Femenino , Estudios de Seguimiento , Humanos , Técnicas para Inmunoenzimas , Metástasis Linfática , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/mortalidad , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Pronóstico , Retinal-Deshidrogenasa , Tasa de Supervivencia , Neoplasias de la Tiroides/mortalidad , Neoplasias de la Tiroides/patología , Análisis de Matrices Tisulares , Adulto Joven
6.
Neoplasia ; 24(2): 133-144, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34968866

RESUMEN

Novel therapeutic strategies are needed in the fight against pancreatic cancer. We have previously documented the chemopreventive effect of MDC-22 in preclinical models of pancreatic cancer. In the present work, we examined the therapeutic effects of MDC-22 in patient-derived tumor xenografts (PDTXs) and in LSL-KrasG12D/+, LSL-Trp53R172H/+, Pdx1-Cre (KPC) genetically engineered mice, two complementary and clinically relevant animal models of pancreatic cancer. In addition, we evaluated whether MDC-22 could synergize with current chemotherapeutic drugs used in the clinic. MDC-22 reduced the growth of various human pancreatic cancer cell lines in a concentration-dependent manner. In vivo, MDC-22 strongly reduced patient-derived pancreatic tumor xenograft growth by 50%, and extended survival of LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx1-Cre (KPC) mice by over a month (5.3 months versus 7.0 months). In both models, MDC-22 inhibited EGFR activation and its downstream signals, including ERK and FAK phosphorylation. In human pancreatic cancer cell lines, MDC-22 enhanced the growth inhibitory effect of irinotecan, and to a lesser degree those of gemcitabine and nab-paclitaxel. Normal human pancreatic epithelial cells were more resistant to the cytotoxic effects of, both, MDC-22 alone or in combination with irinotecan, indicating selectivity. Furthermore, MDC-22 enhanced irinotecan's effect on cell migration, in part, by inhibiting EGFR/FAK signaling. Collectively, our results indicate that MDC-22 is an effective anticancer drug in preclinical models of pancreatic cancer, and suggest that MDC-22 plus irinotecan as drug combination strategy for pancreatic cancer treatment, which warrants further evaluation.


Asunto(s)
Antineoplásicos/farmacología , Aspirina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Aspirina/análogos & derivados , Modelos Animales de Enfermedad , Quimioterapia Combinada , Receptores ErbB/antagonistas & inhibidores , Humanos , Irinotecán/farmacología , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Resultado del Tratamiento
7.
Oxid Med Cell Longev ; 2022: 9974639, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35308166

RESUMEN

Thyroid cancer is the most common malignancy of the endocrine system, and its outcome remains unsatisfactory. In recent years, circular RNAs (circRNAs) have emerged as crucial regulators in cancers. In the current study, we aimed to investigate whether and how circRNA_0057209 functioned in thyroid cancer. Initial results revealed that circRNA_0057209 and STK4 were both reduced, while miR-183 was up-regulated in thyroid cancer tissues and cells. Experiments including RNA pull-down and RIP assays further identified that upregulation of circRNA_0057209 augmented the expression of STK4, a target gene of miR-183, by competitively-binding to miR-183. Furthermore, functional experiments provided evidence that overexpression of circRNA_0057209 not only inhibited the proliferative, migratory, and invasive properties of thyroid cancer cells while facilitating their apoptosis but also delayed tumor growth. Conversely, upregulation of miR-183 or silencing of STK4 reversed the changes induced by circRNA_0057209. Meanwhile, mechanistic experimentation demonstrated that circRNA_0057209 promoted STK4 expression by sponging miR-183, while STK4 enhanced YAP phosphorylation to mediate the Hippo pathway, thereby suppressing tumor progression. Altogether, our findings indicated that circRNA_0057209 may serve as a competing endogenous RNA of miR-183 to increase STK4 expression, thus inhibiting the development of thyroid cancer.


Asunto(s)
MicroARNs , ARN Circular , Neoplasias de la Tiroides , Línea Celular Tumoral , Proliferación Celular/genética , Vía de Señalización Hippo , Humanos , Péptidos y Proteínas de Señalización Intracelular , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Serina-Treonina Quinasas/genética , ARN Circular/genética , Neoplasias de la Tiroides/genética
8.
J Clin Endocrinol Metab ; 107(2): 474-490, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34562008

RESUMEN

CONTEXT: Abnormally high expression of N6-methyladenosine (m6A) methyltransferase-like 3 (METTL3) has been implied to accompany thyroid carcinoma (TC) development. OBJECTIVE: This study aimed to explore the protumorigenic role and downstream signaling axis of METTL3 in TC. METHODS: This study was conducted at the Sun Yat-Sen Memorial Hospital Sun Yat-Sen University. METTL3 and miR-222-3p were overexpressed or downregulated in TC cells. Tumor and adjacent normal tissues were collected from 80 patients (19 men and 60 women, aged 30-70 years) with a pathological diagnosis of TC from January 2012 to January 2015. Cells were classified and subjected to different treatments. The expression of METTL3 was validated in TC tissues and cell lines. In functional studies, METTL3 and miR-222-3p were overexpressed or downregulated in TC cells to evaluate their effects on malignant behaviors, which were subsequently verified by xenografts in nude mice. RESULTS: The expression of METTL3 was elevated in TC, correlating with poor prognosis of TC patients. Heightened METTL3 expression accelerated malignant behaviors of TC cells. Mechanistically, METTL3 stimulated miR-222-3p expression by mediating the m6A modification of pri-miR-222-3p. miR-222-3p targeted and inversely regulated serine/threonine stress kinase 4 (STK4). Knockdown of METTL3 augmented STK4 expression by downregulating miR-222-3p, thereby suppressing the malignant behaviors of TC cells as well as tumor growth and lung metastasis in nude mice. CONCLUSION: Silencing METTL3 suppresses miR-222-3p expression and thus stimulates STK4 expression, thereby repressing the malignancy and metastasis of TC.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Metiltransferasas/metabolismo , MicroARNs/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Neoplasias de la Tiroides/genética , Adenosina/análogos & derivados , Adenosina/metabolismo , Adulto , Anciano , Animales , Transformación Celular Neoplásica/genética , Regulación hacia Abajo , Femenino , Estudios de Seguimiento , Técnicas de Silenciamiento del Gen , Humanos , Estimación de Kaplan-Meier , Masculino , Metiltransferasas/genética , Ratones , MicroARNs/genética , Persona de Mediana Edad , Glándula Tiroides/patología , Glándula Tiroides/cirugía , Neoplasias de la Tiroides/mortalidad , Neoplasias de la Tiroides/patología , Neoplasias de la Tiroides/cirugía , Tiroidectomía , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Clin Endocrinol Metab ; 105(7)2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32303750

RESUMEN

CONTEXT: Lysine-specific demethylase 1 (LSD1) stabilizes hypoxia-inducible factor 1α (HIF1α) to advance tumor progression, while HIF1α functions as a transcription factor to increase the expression of microRNA-146a (miR-146a). OBJECTIVE: We aim to investigate whether LSD1 affects the development of papillary thyroid carcinoma (PTC) via HIF1α and miR-146a. DESIGN: In vitro assays were performed with Nthy-ori 3-1, BHP5-16, BCPAP, K1, and BHP2-7 cell lines. In vivo assays were conducted with established xenograft tumors in nude mice. SETTING: This study was conducted at our lab. PATIENTS AND MATERIALS: PTC tissues and corresponding adjacent normal tissues were obtained from 45 patients hospitalized in Sun Yat-Sen Memorial Hospital. Assays were conducted using Nthy-ori 3-1, BHP5-16, BCPAP, K1, and BHP2-7 cell lines, as well as 50 male BALB/c nude mice. INTERVENTION: Cells were transfected with sh-LSD1, sh-GABPA, oe-LSD1, oe-HIF1α, miR-146a mimic, and miR-146a inhibitor. In addition, K1 cells expressing lv-oe-LSD1, lv-miR-146a inhibitor, lv-oe-LSD1 or miR-146a inhibitor were injected into the right side of the mice. LSD1 gene and protein expression patterns were analyzed in 45 clinical PTC tissue samples. MAIN OUTCOME MEASURE: Expression of LSD1, HIF1α, miR-146a, and GA-binding protein transcription factor alpha (GABPA), as well as their effects on PTC. RESULTS: LSD1 was highly expressed in clinical PTC tissues. LSD1 stabilized HIF1α and inhibited the degradation of its ubiquitin proteasome. HIF1α was enriched in the promoter region of miR-146a, an upregulated miRNA in PTC. HIF1α increased miR-146a expression to promote PTC progression in vitro, which was achieved by inhibiting GABPA, a target gene of miR-146a. LSD1 upregulated miR-146a to enhance the development and metastasis of PTC in nude mice. CONCLUSION: Our results show that LSD1 functions as an oncogene in PTC by upregulating HIF1α and miR-146a, elucidating an understanding of undefined mechanisms associated with tumor progression in PTC.


Asunto(s)
Histona Demetilasas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , MicroARNs/metabolismo , Cáncer Papilar Tiroideo/metabolismo , Glándula Tiroides/metabolismo , Neoplasias de la Tiroides/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Factor de Transcripción de la Proteína de Unión a GA/genética , Factor de Transcripción de la Proteína de Unión a GA/metabolismo , Regulación Neoplásica de la Expresión Génica , Histona Demetilasas/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Ratones , Ratones Desnudos , MicroARNs/genética , Cáncer Papilar Tiroideo/genética , Cáncer Papilar Tiroideo/patología , Glándula Tiroides/patología , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología
10.
Am J Transl Res ; 12(4): 1362-1378, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32355548

RESUMEN

PURPOSE: The present study aimed to identify differentially expressed circRNAs in thyroid cancer and verify their potential functions. METHODS: Next-generation sequencing was used to identify differentially expressed circRNAs between papillary thyroid carcinoma (PTC) tissues and paired pericarcinomatous tissues. Polymerase chain reaction and Sanger sequencing methods successfully identified hsa_circ_0007694. A hsa_circ_0007694 over-expression vector was prepared to determine the effect of this circRNA on proliferation, migration, invasion, apoptosis, and the cell cycle in PTC cells. An in vivo animal assay was conducted by injecting PTC cells into the chests of mice. Further, RNA-seq was performed, followed by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, to verify the regulatory mechanism of hsa_circ_0007694. Western blotting was used to verify the genes thought to be involved in the hsa_circ_0007694 regulatory pathways based on KEGG analysis. RESULTS: We identified a circRNA, hsa_circ_0007694 that was down-regulated in PTC tissues compared to pericarcinomatous tissues. Over-expression of hsa_circ_0007694 promoted apoptosis and inhibited proliferation, migration, and invasion in PTC cells in vitro, and decreased tumor growth in vivo. Transcriptome sequence analysis suggested 129 differentially expressed genes between PTC tissue and paired pericarcinomatous tissue. KEGG analysis and western blotting indicated that the PI3K/AKT/mTOR and Wnt signaling networks are most likely to be related to hsa_circ_0007694 in thyroid cancer. CONCLUSION: The circRNA hsa_circ_0007694 is down-regulated in PTC and is therefore a potential therapeutic target.

11.
Cancer Manag Res ; 11: 8557-8569, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31572010

RESUMEN

PURPOSE: Papillary thyroid carcinoma (PTC) is the most common endocrine malignancy, and its incidence has continuously increased in recent years. Therefore, it is essential to develop more useful therapeutic strategies. METHODS: We collected 56 pairs of PTC tissues and adjacent normal tissues and determined the expression patterns of linc01278, miR-376c-3p and DNM3. In addition, we analyzed the relationship between linc01278 expression and pathological information of PTC patients. Furthermore, the effects of linc01278, miR-376c-3p and DNM3 overexpression on proliferation, clonality, apoptosis, migration and invasion of PTC cell lines TPC1 and BCPAP were evaluated. The dual luciferase reporter assay was used to confirm the direct interaction between miR-376c-3p and linc01278. RESULTS: Linc01278 and DNM3 were remarkably down-regulated in PTC tissues and cell lines, whereas miR-376c-3p was significantly up-regulated. In addition, lower linc01278 expression was associated with increased tumor size, lymph node metastasis and higher clinical stage. Linc01278 inhibited cell proliferation of PTC cells by inducing apoptosis, and demonstrated attenuating effects on migration and invasion abilities of PTC cells by regulating the EMT process. More importantly, dual luciferase reporter experiments demonstrated the direct interaction between miR-376c-3p and linc01278, which revealed that DNM3 was a novel target of miR-376c-3p. The miR-376c-3p mimic significantly promoted the proliferation, migration and invasion of PTC cells, and inhibited cell apoptosis. Overexpression of DNM3 abolished the effects of the miR-376c-3p mimic on PTC cells. DNM3 expression was negatively correlated with miR-376c-3p expression, but was positively correlated with linc01278 expression. CONCLUSION: Overall, we found that linc01278 can act as a competing endogenous RNA (ceRNA) to sponge miR-376c-3p, thereby positively regulating DNM3 expression and ultimately acting as a tumor suppressor gene in PTC progression.

12.
Oncol Rep ; 41(2): 1253-1263, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30535496

RESUMEN

Papillary thyroid carcinoma (PTC) is a type of cancer with one of the fastest increasing incidences worldwide. However, the therapeutic choices for PTC patients are limited and it is critical to further understand the molecular pathology underlying this disease. Squamous cell carcinoma antigens (SCCAs) are overexpressed in many tumors and participate in tumorigenesis. However, their roles in PTC are incompletely understood. Therefore, this study investigated the role of SCCA in PTC, evaluating its expression, its clinical implications and prognostic significance in PTC patient samples, as well as its function in vitro and in vivo, using a thyroid cancer cell line in which SCCA levels have been knocked down or overexpressed. In this study, SCCA expression levels were measured by immunohistochemistry (IHC) in non­cancerous and tumor tissues. Kaplan­Meier analyses assessed the survival in PTC patients. MTT assay, western blot analysis, invasion assay and xenograft tumor assay were used to calculate cell proliferation, migration, invasion and tumor growth. Our results showed that SCCA was overexpressed in PTC tissues and was correlated with the clinical stage of PTC. Patients with high SCCA expression had lower overall survival (OS), disease­free survival (DFS), lymph node recurrence­free survival (LNRFS), and distant recurrence­free survival (DRFS), compared to patients expressing low level of the SCCA protein. SCCA knockdown suppressed thyroid cancer cell proliferation, invasion and reduced xenograft tumor growth, whereas SCCA overexpression increased cell proliferation, invasion and xenograft tumor growth. Mechanistically, the activation of Ras increased SCCA expression, and SCCA expression was positively correlated with Ras levels in the PTC tissues. In conclusion, SCCA protein is overexpressed in PTC and may represent a predictive prognostic factor for PTC patients. Furthermore, activation of the Ras/SCCA pathway plays an important role in promoting tumor growth, invasion and metastasis in PTC.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Metástasis Linfática , Serpinas/metabolismo , Cáncer Papilar Tiroideo/metabolismo , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Proteínas Activadoras de ras GTPasa/metabolismo , Adulto , Animales , Antígenos de Neoplasias/genética , Línea Celular Tumoral , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Serpinas/genética , Transducción de Señal , Cáncer Papilar Tiroideo/genética , Neoplasias de la Tiroides/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven , Proteínas Activadoras de ras GTPasa/genética
13.
Int J Oncol ; 53(5): 2067-2080, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30106127

RESUMEN

Glioblastoma multiforme (GBM) is the most common primary malignant tumor affecting the human brain. Despite improvements in therapeutic technologies, patients with GBM have a poor clinical result and the molecular mechanisms responsible for the development of GBM have not yet been fully elucidated. 3-phosphoinositide dependent protein kinase 1 (PDK1) is upregulated in various tumors and promotes tumor invasion. In glioma, transforming growth factor-ß (TGF­ß) promotes cell invasion; however, whether TGF­ß directly regulates PDK1 protein and promotes proliferation and invasion is not yet clear. In this study, PDK1 levels were measured in glioma tissues using tissue microarray (TMA) by immunohistochemistry (IHC) and RT­qPCR. Kaplan-Meier analyses were used to calculate the survival rate of patients with glioma. In vitro, U251 and U87 glioma cell lines were used for functional analyses. Cell proliferation and invasion were analyzed using siRNA transfection, MTT assay, RT­qPCR, western blot analysis, flow cytometry and invasion assay. In vivo, U251 glioma cell xenografts were established. The results revealed that PDK1 protein was significantly upregulated in glioma tissues compared with non-tumorous tissues. Furthermore, the higher PDK1 levels were associated with a large tumor size (>5.0 cm), a higher WHO grade and a shorter survival of patients with GBM. Univariate and multivariate analyses indicated that PDK1 was an independent prognostic factor. In vivo, PDK1 promoted glioma tumor xenograft growth. In vitro, functional analyses confirmed that TGF­ß upregulated PDK1 protein expression and PDK1 promoted cell migration and invasion, and functioned as an oncogene in GBM, by upregulating c­Jun protein and inducing epithelial-mesenchymal transition (EMT). c­Jun protein were overexpressed in glioma tissues and positively correlated with PDK1 levels. Moreover, our findings were further validated by the online Oncomine database. On the whole, the findings of this study indicate that in GBM, PDK1 functions as an oncogene, promoting proliferation and invasion.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Neoplasias Encefálicas/patología , Glioma/patología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Adulto , Anciano , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Glioblastoma/mortalidad , Glioblastoma/patología , Glioma/metabolismo , Glioma/mortalidad , Humanos , Masculino , Ratones Desnudos , Persona de Mediana Edad , Oncogenes , Proteínas Proto-Oncogénicas c-jun/genética , Tasa de Supervivencia , Factor de Crecimiento Transformador beta/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Onco Targets Ther ; 10: 2173-2181, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28458560

RESUMEN

Glioma is the most common and aggressive human primary tumor in the central nervous system. Despite present clinical advancements, median survival time remains poor in this malignant tumor. Serpin peptidase inhibitor, clade A member 3 (SERPINA3), is a member of the serpin superfamily of protease inhibitors. Its aberrant expression has been observed in various tumors. However, its clinical significance and biological function in glioma remain unclear, especially for the prognosis of glioma patients. In this study, we investigated SERPINA3 expression in glioma tissue samples and its significance in predicting the prognosis of glioma patients. SERPINA3 protein expression was studied by immunohistochemistry, while real-time polymerase chain reaction was used to study SERPINA3 mRNA expression. We found that SERPINA3 was upregulated in glioma tissue at both mRNA and protein levels, compared with noncancerous brain tissues. We also found that high SERPINA3 expression in glioma tissues correlated significantly with advanced World Health Organization grade. Univariate and multivariate analyses revealed that high SERPINA3 expression was an independent prognostic factor for poor overall survival of glioma patients. Moreover, our findings were further validated by online Oncomine database. Taken together, our results suggest that SERPINA3 plays an oncogenic role in glioma progression and provide an insight into the application of SERPINA3 as a novel predictor of clinical outcomes and a potential biomarker of glioma.

15.
Int J Oncol ; 51(4): 1209-1218, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28848996

RESUMEN

Rho-associated protein kinase 1 (ROCK1), a serine/threonine kinase, has previously been shown to be over-expressed in various types of human malignant tumors and to play an important role in cancer development and progression. Although ROCK1 has gained growing prominence as an important protein kinase in cancer biology, its potential as a predictive biomarker and a therapeutic target in papillary thyroid carcinoma (PTC) remains unknown. In the present study, ROCK1 expression was examined in 356 formalin-fixed, paraffin-embedded papillary thyroid carcinoma tissues using immunohistochemistry, and its clinical implications and prognostic significance were analyzed. Our results showed that ROCK1 expression was significantly increased in PTC compared with normal tissues, and was significantly associated with tumor size, lymphatic metastasis, distant organ metastasis, extrathyroid invasion, vascular invasion and tumor, node and metastasis (TNM) stage. Patients with strong ROCK1 expression had lower overall survival, disease-free survival, lymph node recurrence-free survival and distant recurrence-free survival rates than those with weak expression. Furthermore, overexpression of ROCK1 in papillary thyroid carcinoma cells was found to increase their invasiveness. Silencing ROCK1 by siRNA, however, caused an inhibition of cell invasion. Knockdown of ROCK1 decreased the volume and weight of the xenograft tumors, while overexpression of ROCK1 showed a proliferative tendency with significantly greater tumor volume and weight in vivo. Moreover, the upregulation of ROCK1 increased the expression of MMP-9, and levels of MMP-9 positively correlated with the ROCK1 levels in PTC tissues, implicating that MMP-9 may be involved in the mechanism of ROCK1 in the development and progression of PTC. These data suggest that ROCK1 might be a potential prognostic marker and therapeutic target for the treatment of PTC.


Asunto(s)
Carcinoma Papilar/enzimología , Metaloproteinasa 9 de la Matriz/metabolismo , Neoplasias de la Tiroides/enzimología , Quinasas Asociadas a rho/metabolismo , Adolescente , Adulto , Anciano , Animales , Carcinoma Papilar/patología , Femenino , Xenoinjertos , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Pronóstico , Cáncer Papilar Tiroideo , Neoplasias de la Tiroides/patología , Adulto Joven
16.
Cancer Biomark ; 18(3): 273-284, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28009325

RESUMEN

OBJECTIVE: In this study, we examined the relationships between the expression level of CHI3L1 and the clinicopathological characteristics of papillary thyroid carcinoma. METHODS: A total of 322 tissue samples from patients with papillary thyroid carcinoma were collected, and the CHI3L1 expression levels in tumor tissues, matched adjacent noncancerous tissues were detected using immunohistochemistry (IHC) and qRT-PCR. The relationships between CHI3L1 expression levels and the clinical characteristics were evaluated. RESULTS: CHI3L1 expression was significantly increased in papillary thyroid carcinoma compared with matched adjacent noncancerous tissues (P< 0.001), tumor tissues with lymph node metastasis (LNM) compared with tumor tissues without LNM (P< 0.001) and tumor tissues with distant organ metastasis (DOM) compared with tumor tissues without DOM (P< 0.01). CHI3L1 expression was significantly associated with tumor size (P= 0.0001), lymph node metastasis (P< 0.0001), distant organ metastasis (P< 0.0001), extrathyroid invasion (P= 0.0022), vascular invasion (P= 0.0004) and TNM stage (P= 0.0001). CHI3L1 overexpression in papillary thyroid carcinoma tissues correlates with the tumor malignant potential (P< 0.01). More importantly, Cox multifactor analysis indicated that patients with high CHI3L1 expression have lower overall survival, disease-free survival, lymph node recurrence-free survival, and distant recurrence free survival rates than those with low expression (P< 0.05). And our findings were further validated by online Oncomine database. CONCLUSIONS: CHI3L1 is associated with tumor metastasis and might be a prognostic biomarker for papillary thyroid carcinoma.


Asunto(s)
Carcinoma/genética , Carcinoma/mortalidad , Proteína 1 Similar a Quitinasa-3/genética , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/mortalidad , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma/diagnóstico , Carcinoma Papilar , Femenino , Expresión Génica , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Estadificación de Neoplasias , Pronóstico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Cáncer Papilar Tiroideo , Neoplasias de la Tiroides/diagnóstico , Adulto Joven
17.
Oncotarget ; 8(26): 43008-43022, 2017 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-28498808

RESUMEN

Dedifferentiated papillary thyroid cancer (DePTC) is characterized by aggressive growth, recurrence, distant metastasis, and resistance to radioactive iodine (RAI) therapy. DePTC is also accompanied by poor prognosis and high early-mortality. Nevertheless, most DePTC cells show intact p53 downstream functionality. In cells with wild-type p53, the murine double minute2 (MDM2) protein interacts with p53 and abrogates its activity. Inhibition of the MDM2-p53 interaction restores p53 activity and leads to cell cycle arrest and apoptosis. Restoring p53 function by inhibiting its interaction with p53 suppressors such as MDM2 is thus a promising therapeutic strategy for the treatment of DePTC. The novel MDM2-p53 interaction antagonist APG115 is an analogue of SAR405838, and is being tested in a phase I clinical trial. In this study, we evaluated the efficacy of APG115 as a single-agent to treat DePTC. APG115 diminished the viability of p53 wild-type DePTC cells and induced cell cycle arrest and apoptosis. In a human xenograft mouse model, APG115 elicited robust tumor regression and cell apoptosis. These data demonstrate that further research is warranted to determine whether APG115 can be used to effectively treat DePTC patients.


Asunto(s)
Carcinoma Papilar/metabolismo , Indoles/farmacología , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Compuestos de Espiro/farmacología , Neoplasias de la Tiroides/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Carcinoma Papilar/tratamiento farmacológico , Carcinoma Papilar/genética , Carcinoma Papilar/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Expresión Génica , Humanos , Indoles/química , Indoles/farmacocinética , Ratones , Mutación , Clasificación del Tumor , Estabilidad Proteica , Compuestos de Espiro/química , Compuestos de Espiro/farmacocinética , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Kaohsiung J Med Sci ; 30(5): 224-8, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24751384

RESUMEN

This study investigated the expression and role of chemokine receptor-4 (CXCR4) in bone marrow mesenchymal stem cells (BMSCs) from experimental rats with abdominal aortic aneurysms (AAA) for migration of BMSCs. Sprague-Dawley rats were divided into an experimental group and control group (n = 18 each). AAA was induced with 0.75 M solution infiltrate for 30 minutes, after which the abdomen was rinsed and closed. Saline was used in place of CaCl2 in the control group. CD34 and CD29 were detected by flow cytometry, the gene and protein expression of CXCR4 were detected by real-time polymerase chain reaction and western blot, respectively. The migration of BMSCs with stromal-derived factor-1 was detected by Transwell chamber. CD34 expression was negative and CD29 expression was positive. The gene and protein expression of CXCR4 were significantly higher in experimental group than them in control group (p < 0.05), the migration ability of BMSCs from the experimental group was significantly higher than that from the control group (p < 0.05). Stromal-derived factor -1/CXCR4 can enhance the migration of BMSCs in vitro in a rat AAA model.


Asunto(s)
Aneurisma de la Aorta Abdominal/metabolismo , Células de la Médula Ósea/citología , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Receptores CXCR4/metabolismo , Animales , Aneurisma de la Aorta Abdominal/patología , Western Blotting , Forma de la Célula , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptores CXCR4/genética
19.
Otolaryngol Head Neck Surg ; 149(6): 845-50, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24163324

RESUMEN

OBJECTIVE: To investigate the hypothesis that injected carbon nanoparticle (CN) suspension helps identify parathyroid glands (PGs) during thyroid cancer surgery, thereby reducing PG injury. STUDY DESIGN: A prospective, randomized controlled trial. Setting Sun Yet-san Memorial Hospital, Guangzhou, China. SUBJECTS AND METHODS: Thyroid cancer surgeries were performed on 72 consenting patients who were randomized for conventional surgery (control group) or surgery with CN suspension injection (CN group). The primary end point was the prevalence of symptomatic hypocalcemia and serum calcium levels <1.9 mmol/L. RESULTS: From 36 patients diagnosed with thyroid cancer in each group, symptomatic hypocalcemia was found in 10 patients without CN injection and 3 patients with CN suspension injection (P = .032). In total, 5.6% of patients in the CN group presented with muscle cramps compared with 22.2% of the control group (P = .041), which showed a significant difference. CONCLUSION: Our randomized study revealed that CN suspension injection was feasible and appeared to be beneficial for patients undergoing thyroid surgery because the incidence of symptomatic hypocalcemia was lower compared with controls. Therefore, this technology and technique should be more widely considered for thyroid cancer therapy. Additional studies with more patients and longer follow-up times will be needed for a thorough evaluation of this methodology.


Asunto(s)
Calcio/sangre , Carbono/administración & dosificación , Carcinoma/cirugía , Hipoparatiroidismo/prevención & control , Hormona Paratiroidea/sangre , Neoplasias de la Tiroides/cirugía , Tiroidectomía/métodos , Adulto , Biomarcadores/sangre , Carcinoma/complicaciones , Carcinoma/diagnóstico , China , Método Doble Ciego , Estudios de Factibilidad , Femenino , Estudios de Seguimiento , Humanos , Hipocalcemia/tratamiento farmacológico , Hipocalcemia/etiología , Hipoparatiroidismo/sangre , Hipoparatiroidismo/etiología , Bombas de Infusión , Masculino , Nanopartículas/administración & dosificación , Estudios Prospectivos , Servicio de Cirugía en Hospital , Neoplasias de la Tiroides/complicaciones , Neoplasias de la Tiroides/diagnóstico , Tiroidectomía/efectos adversos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA