Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Exp Cell Res ; 430(1): 113700, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37380010

RESUMEN

Growing evidence suggests a strong association between decreased lipid catabolism and the development of cancer. Solute carrier family 9 member A5 (SLC9A5) plays a regulatory role in colorectal function. However, the specific involvement of SLC9A5 in colorectal cancer (CRC) remains unclear, as well as its potential connection to lipid catabolism. We found that SLC9A5 exhibited significantly higher expression in CRC tumor tissues compared to adjacent paratumor tissues, as confirmed through analysis of the TCGA database and validation on a CRC tissue chip using IHC. Furthermore, in vitro experiments showed that knockdown of SLC9A5 resulted in suppressed cell proliferation, migration, and invasion. Then we performed bioinformatics analysis and found that SLC9A5 was significantly enriched in peroxisomal fatty acid oxidation (FAO) pathway and negatively correlated with its first rate-limiting enzyme acyl-CoA oxidases (ACOX). Interestingly, the expression of ACOX1, as well as FAO process indicated by changes in very long chain fatty acid levels, were enhanced upon SLC9A5 knockdown in CRC cells. Moreover, the attenuated tumor growth, migration, invasion, and increased FAO observed after SLC9A5 knockdown could be reversed by simultaneous knockdown of both SLC9A5 and ACOX1. In summary, these findings reveal the oncogenic role of SLC9A5 in CRC, particularly in relation to ACOX1-mediated peroxidation, and might serve as a promising therapeutic target for inhibiting the progression of colorectal cancer.


Asunto(s)
Neoplasias Colorrectales , Ácidos Grasos , Humanos , Acil-CoA Oxidasa/genética , Acil-CoA Oxidasa/metabolismo , Lípidos , Neoplasias Colorrectales/patología , Movimiento Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética
2.
Cancer Med ; 11(23): 4491-4500, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35616090

RESUMEN

BACKGROUND: Anaplastic lymphoma kinase (ALK) fusion is a prognostic indicator for patients with non-small cell lung cancer (NSCLC) receiving tyrosine kinase inhibitors (TKIs). The real-world data of ALK TKIs remain a major concern. METHODS: Patients with ALK-positive advanced NSCLC, who received crizotinib or alectinib treatment in first line, were retrospectively reviewed. ALK status was detected using immunohistochemistry (IHC) or next-generation sequencing (NGS). Clinical outcomes have been comprehensively analyzed between TKIs, ALK fusions, EML4-ALK variants, and next-generation TKIs after crizotinib failure. RESULTS: One hundred sixty-eight patients were successively enrolled (crizotinib, n = 109; alctinib, n = 59). Alectinib showed consistent superiority in progressive-free survival (PFS) over crizotinib (hazard ratio [HR]: 0.43, 95% confidential interval [CI]: 0.24-0.77, p = 0.004). Multivariate Cox regression showed chemotherapy (CT) prior to TKIs or synchronous chemotherapy seemed not to improve PFS compared to ALK inhibitors alone (p > 0.05). And, alectinib was superior to crizotinib in prolonging intracranial PFS (HR 0.12, 95% CI: 0.03-0.49, p = 0.003). Patients in EML4 group had a better prognosis than those in non-EML4 group after alectinib administration (HR 0.13, 95% CI: 0.03-0.60, p = 0.009). TP53 co-mutations were relatively common (34.0%) and associated with adverse outcome in ALK-positive patients (adjusted HR 2.22, 95% CI: 1.00-4.92, p = 0.049). After crizotinib failure, 33 patients received a sequential application of next-generation ALK TKIs. Compared to ceritinib and brigatinib, alectinib might have better PFS (p = 0.043). CONCLUSION: Our results revealed alectinib had better PFS and higher intracranial efficacy compared to crizotinib in ALK-positive NSCLC, and might improve PFS by comparison with ceritinib and brigatinib after crizotinib failure.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Crizotinib/uso terapéutico , Estudios Retrospectivos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Carbazoles/efectos adversos , Inhibidores de Proteínas Quinasas/efectos adversos
3.
BMC Cancer ; 21(1): 1215, 2021 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-34774017

RESUMEN

BACKGROUND: In the existing next generation sequencing (NGS) system, epidermal growth factor receptor (EGFR) exon 19 deletion-insertion (19delins) is still interpreted into the category of EGFR exon 19 deletion (19del). However, the controversy exists whether the two mutation types have the similar responses and resistant mechanisms to first-generation EGFR tyrosine kinase inhibitor (TKI) in non-small cell lung cancer (NSCLC) patients. METHODS: We successively and retrospectively reviewed the NGS data of 3054 patients diagnosed as advanced NSCLC from November 2017 to September 2020. Finally, 41 patients with EGFR 19delins mutation and 41 patients with EGFR 19del mutation who received first-generation EGFR TKIs as first-line therapy were included in the study. RESULTS: A total of 17 genotypes were identified in this study, including L747_P753delinsS (10/41), L747_A750delinsP (9/41), L747_T751delinsP (6/41) and E746_S752delinsV (3/41). Under the same baseline characteristics, the population of EGFR 19delins respond well to first line EGFR TKIs as well as those of EGFR 19del, with little difference in median progression-free survival (mPFS): 10.4 months vs. 13.1 months, p = 0.1076). Interestingly, patients with L747_T751delinsP seem to have a better mPFS than others (18.7 months vs. 13.1 months, p = 0.035). After the disease progression, both EGFR 19delins and EGFR 19del had similar rates of developing EGFR T790M mutation resistance (45.8% vs. 57.8%), and those receiving osimeritinib as second-line treatment obtain the similar survival benefits (mPFS: 12.0 months vs. 12.2 months (p = 0.97). CONCLUSIONS: This retrospective cohort study furnish the evidence that therapeutic responses and survival of untreated NSCLC population with EGFR 19delins mutation are equal to those with common EGFR 19del mutation after administration of EGFR TKIs therapy.


Asunto(s)
Acrilamidas/uso terapéutico , Compuestos de Anilina/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Eliminación de Gen , Genes erbB-1 , Neoplasias Pulmonares/tratamiento farmacológico , Mutagénesis Insercional , Inhibidores de Proteínas Quinasas/uso terapéutico , Adulto , Anciano , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Progresión de la Enfermedad , Receptores ErbB/antagonistas & inhibidores , Exones , Femenino , Genotipo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Supervivencia sin Progresión , Estudios Retrospectivos
4.
World J Surg Oncol ; 19(1): 185, 2021 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-34162380

RESUMEN

BACKGROUND: The aim of our study was to observe and compare the curative effect of radiotherapy, chemotherapy, and combined radiotherapy and chemotherapy, as well as comprehensive treatment on superior vena cava syndrome (SVCS) caused by malignant etiology. METHODS: A total of 48 patients with malignant SVCS admitted to our hospital from 2015 to 2020 were selected in this study. According to the different treatment methods, they were divided into radiotherapy group (group 1, 10 cases), chemotherapy group (group 2, 8 cases), combined radiotherapy and chemotherapy group (group 3, 22 cases), and comprehensive treatment group (group 4, 8 cases). RESULTS: There were no significant differences in efficacy and side effects among the four groups (all P > 0.05). Group 4 (median survival time of 36 months) could provide longer survival time than groups 1, 2, and 3 (median survival time of 10 months, 13.5 months, and 12 months, respectively). CONCLUSIONS: For patients with severe symptoms or good prognosis, comprehensive treatment could be selected to improve the quality of life and prolong the survival period; for patients with mild symptoms, radiotherapy, chemotherapy, or combined radiotherapy and chemotherapy could also reduce the symptoms of SVCS and treat tumor lesions.


Asunto(s)
Neoplasias Pulmonares , Síndrome de la Vena Cava Superior , Humanos , Pronóstico , Calidad de Vida , Stents , Síndrome de la Vena Cava Superior/etiología , Síndrome de la Vena Cava Superior/terapia
5.
Clin Epigenetics ; 9: 50, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28491151

RESUMEN

BACKGROUND: DIRAS1 is a new member of the Ras gene family. It was described as a potential tumor suppressor in human glioblastomas and esophageal cancer. The role of DIRAS1 in colorectal cancer remains unclear. METHODS: To explore the epigenetic changes and function of DIRAS1 in human colorectal cancer, we studied ten colorectal cancer cell lines and 146 primary colorectal cancer samples and 50 matched adjacent samples using semi-quantitative reverse transcription PCR, immunohistochemistry, methylation-specific PCR and bisulfite sequencing, western blot, flow cytometry, and transwell assays. RESULTS: DIRAS1 expression was found in DKO and HCT116 cells, while reduced expression was detected in LoVo, SW48, LS180, and SW620 cells, and there was no expression detected in DLD1, HT29, RKO, and SW480 cells. Complete methylation was found in the promoter region of DLD1, HT29, RKO, and SW480 cells. Partial methylation was detected in LoVo, LS180, SW48, and SW620 cells, and unmethylation was found in DKO and HCT116 cells. These results indicate that promoter region methylation correlated with loss of/reduced expression of DIRAS1. Re-expression of DIRAS1 was induced by 5-aza-2'-deoxycytidine, suggesting that the expression of DIRAS1 is regulated by promoter region methylation. DIRAS1 was methylated in 47.3% (69/146) of primary colorectal cancer samples, no methylation was found in non-cancerous colonic tissue samples. Methylation of DIRAS1 was significantly associated with TNM stage (P < 0.05) and short survival time (P = 0.0121). DIRAS1 induced apoptosis and inhibited cell proliferation, migration, and invasion in colorectal cancer. Finally, DIRAS1 suppressed colorectal cancer cell xenograft growth in nude mice. CONCLUSIONS: DIRAS1 is frequently methylated in human colorectal cancer and the expression of DIRAS1 is regulated by promoter region methylation. Methylation of DIRAS1 is a marker of poor prognosis in human colorectal cancer.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Metilación de ADN , GTP Fosfohidrolasas/genética , Proteínas Supresoras de Tumor/genética , Animales , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Progresión de la Enfermedad , Epigénesis Genética , Femenino , GTP Fosfohidrolasas/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HT29 , Humanos , Masculino , Ratones , Estadificación de Neoplasias , Trasplante de Neoplasias , Pronóstico , Regiones Promotoras Genéticas , Proteínas Supresoras de Tumor/metabolismo
6.
Epigenomics ; 9(6): 849-862, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28403629

RESUMEN

AIM: The expression of human SLFN11 was reported to sensitize cancer cells to DNA damaging agents. This study is to explore the epigenetic change and the function of SLFN11 in human colorectal cancer (CRC). MATERIALS & METHODS: Six CRC cell lines and 128 primary CRC samples were used. RESULTS: SLFN11 was methylated in 55.47% (71/128) of primary CRC. The expression of SLFN11 was regulated by promoter region methylation. Methylation of SLFN11 was significantly associated with age, poor 5-year overall survival and 5-year relapse-free survival (all p < 0.05). SLFN11 suppressed CRC cell growth both in vitro and in vivo and sensitized CRC cells to cisplatin. CONCLUSION: SLFN11 is frequently methylated in human CRC, and the expression of SLFN11 is regulated by promoter region methylation. Methylation of SLFN11 reduced the sensitivity of CRC cells to cisplatin.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Metilación de ADN , Resistencia a Antineoplásicos , Proteínas Nucleares/genética , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Células Cultivadas , Cisplatino/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas
7.
Pancreas ; 44(6): 925-9, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25931261

RESUMEN

OBJECTIVE: To date, there are no reports on the cellular localization of dopamine receptors in the human pancreas. In our study, we determined the localization and expression of 5 dopamine receptors (D(1), D(2), D(3), D(4), and D(5)) in normal human pancreas tissue. METHODS: Human nonpathological pancreas tissues were fixed with 4% paraformaldehyde, paraffin-embedded, and processed for immunohistochemical analysis to detect dopamine receptors in the human pancreas tissue by using double immunofluorescent labeling and confocal microscopy. RESULTS: We found that the D(1) receptor is present in ß cells; the D(2) receptor is expressed by α, δ, and pancreatic polypeptide cells; the D(4) receptor is expressed by ß and polypeptide cells; whereas the D(5) receptor is expressed only by δ cells. CONCLUSIONS: Our results identify the dopamine receptors (D(1)-D(5)) in normal pancreas tissue and provide a morphological basis for studying the pancreatic endocrine effects of dopamine and suggest a new target for the clinical treatment of diabetes.


Asunto(s)
Islotes Pancreáticos/química , Receptores Dopaminérgicos/análisis , Técnica del Anticuerpo Fluorescente , Células Secretoras de Glucagón/química , Humanos , Células Secretoras de Insulina/química , Islotes Pancreáticos/citología , Microscopía Confocal , Células Secretoras de Polipéptido Pancreático/química , Adhesión en Parafina , Receptores de Dopamina D1/análisis , Receptores de Dopamina D2/análisis , Receptores de Dopamina D3/análisis , Receptores de Dopamina D4/análisis , Receptores de Dopamina D5/análisis , Células Secretoras de Somatostatina/química
8.
J Neurosci Res ; 93(2): 350-60, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25257318

RESUMEN

Weight loss is an important nonmotor symptom associated with Parkinson's disease (PD). However, the cellular factors responsible for PD-induced weight loss remain unclear. Because the sympathetic nervous system plays an important role in lipid metabolism and fat cell differentiation, this study investigates whether PD-induced changes to this system are associated with weight loss in a rat model of PD. Body weight and food intake were measured in control and PD-model rats. After 10 weeks, retroperitoneal white adipose tissues (RWAT) were removed and weighed. Markers of the sympathetic nervous system were measured in the brainstem dorsal medulla and RWAT. Free fat acids (FFA), triglycerides (TG), adipocyte differentiation-related genes, and lipolysis-related molecules in the RWAT and serum were analyzed. Differences in body weight and food intake were insignificant in PD-model rats and control rats; however, relative RWAT weight and adipocyte surface area were significantly reduced in the PD group. Changes in markers of the sympathetic nervous system were observed in the brainstem dorsal medulla and RWAT of PD rats. Decreased mRNA expression levels of genes involved in adipocyte differentiation, decreased TG levels in RWAT, increased FFA in RWAT, and increased lipolysis-related molecules in RWAT and serum FFA were observed in PD rats. This study demonstrates that degenerated dopaminergic neurons in the nigrostriatal system correlate with increases in sympathetic nervous system function, resulting in lipolysis and inhibition of fat cell differentiation. These factors ultimately result in the decrease of RWAT in PD-model rats.


Asunto(s)
Adipocitos Blancos/fisiología , Lipólisis/fisiología , Enfermedad de Parkinson/patología , Sistema Nervioso Simpático/fisiopatología , Animales , Peso Corporal/fisiología , Diferenciación Celular/fisiología , AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Ingestión de Alimentos/fisiología , Ácidos Grasos/sangre , Masculino , Bulbo Raquídeo/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Enfermedad de Parkinson/sangre , Ratas , Ratas Sprague-Dawley , Rotación , Sistema Nervioso Simpático/patología , Triglicéridos/sangre , Tirosina 3-Monooxigenasa/metabolismo
9.
Transl Res ; 163(2): 171-82, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24161354

RESUMEN

Vesicular monoamine transporter 2 (VMAT2) has been exploited as a biomarker of ß-cell mass in human islets. However, a current report suggested no immunoreactivity of VMAT2 in the ß cells of rat islets. To investigate the cellular localization of VMAT2 in islets further, the pancreatic tissues from monkeys and humans were compared with those of rats and mice. The study was performed using among-species comparisons and a type 1 diabetes model (T1DM) for rats by Western blotting, double-label immunofluorescence, and confocal laser scanning microscopy. We found that VMAT2-immunoreactivity (IR) was distributed peripherally in the islets of rodents, but was widely scattered throughout the islets of primates. Consistent with rodent islets, VMAT2-IR did not exist in insulin (INS)-IR cells but was abundantly present in glucagon (GLU)-IR and pancreatic polypeptide (PP)-IR cells in monkey and human islets. VMAT2-IR had no colocalization with INS-IR in any part of the rat pancreas (head, body, and tail). INS-IR cells were reduced dramatically in T1DM rat islets, but no significant alteration in the proportion of VMAT2-IR cells and GLU-IR cells was observed. Furthermore, a strong colocalization of VMAT2-IR with GLU-IR was distributed in the peripheral regions of diabetic islets. For the first time, the current study demonstrates the presence of VMAT2 in α cells and PP cells but not in ß cells in the islets of monkeys and humans. This study provides convinced morphologic evidence that VMAT2 is not present in ß cells. There needs to be studies for new markers for ß cell mass.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Modelos Animales de Enfermedad , Proteínas de Transporte Vesicular de Monoaminas/metabolismo , Animales , Islotes Pancreáticos/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA