Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Intervalo de año de publicación
1.
BMC Cancer ; 23(1): 882, 2023 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-37726690

RESUMEN

PURPOSE: Thyroid cancer (TC) is one of the most common endocrine malignancies, and its morbidity continues to rise. N6-methyladenosine (m6A) RNA methylation, an epigenetic modification, is an important regulator of gene expression in TC. Therefore, it's worth finding the characteristics and predictive value of the m6A RNA methylation regulators in thyroid cancer (TC). METHOD: RNA-seq data of TC was downloaded from the Cancer Genome Atlas (TCGA) database to screen out the differential expressed regulators. The absolute contraction selection operator (Lasso) Cox regression was used to construct the risk model of m6A methylation regulators. The predictive value of the risk scoring model was evaluated by Kaplan Meier (K-M) analysis and receiver operating characteristic (ROC) curves. The underlying mechanism of m6A methylation regulators in TC was predicted by gene set enrichment analysis (GSEA). Further validation was performed by using immunohistochemistry (IHC) and q-PCR. The correlation between risk-related gene and immune infiltration was evaluated by Tumour Immune Estimation Resource (TIMER). RESULTS: IGF2BP2, YTHDF1 and YTHDF3 were screened out as strong independent prognostic factors of TC. Then a risk score model was established to further screen the predictors. Finally, according to the results of overall survival (OS) and clinical characteristics of TC, YTHDF3 was screened out as a potential predictor. Meanwhile, IHC and qPCR confirmed that YTHDF3 was expressed differential in TC. The expression of YTHDF3 was positively associated with the infiltration level of CD4+ T cells and macrophages. It was strongly correlated with a variety of immune markers in TC. CONCLUSION: We confirmed that YTHDF3 can be used as a potential prognostic biomarker of TC. It not only plays a decisive role in the initiation and development of TC, but also provides a new perspective for understanding the modification of m6A RNA in TC.


Asunto(s)
Neoplasias de la Tiroides , Humanos , Pronóstico , Neoplasias de la Tiroides/genética , Cognición , Bases de Datos Factuales , Epigénesis Genética , Proteínas de Unión al ARN/genética
2.
Front Endocrinol (Lausanne) ; 14: 1131078, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37455914

RESUMEN

Background: Hashimoto's thyroiditis (HT) is an autoimmune thyroid disease. Papillary thyroid carcinoma (PTC) is the most common endocrine cancer. In recent years the rate of coexistence between PTC and HT has increased but the relationship between them remains unclear, meaning it is necessary to find potential biomarkers for PTC coexistence with HT to predict its potential pathways. Method: A co-expression network was constructed using the weighted gene co-expression network analysis (WGCNA) in the R package. The modules of PTC associated with HT (PTC-W) were identified from the GSE138198 dataset. Protein-protein interaction network (PPI) was used to screen the hub genes. Immunohistochemical (IHC) analysis was performed to validate the expression of the hub genes in tissues. Clinical data from The Cancer Genome Atlas (TCGA) datasets were used to analyse the prognosis of the hub genes. Gene set enrichment analysis (GSEA) was used to screen potential pathways of PTC-W. Result: The MEbrown module representing the most significant module, with 958 differentially expressed genes (DEGs), was screened in PTC-W, based on WGCNA analysis. Through PPI, SERPINA1 was identified as a hub gene. Immunostaining validated that SERPINA1 was highly expressed in PTC-W. Moreover, PTC-W expressing SERPINA1 exhibits a better prognosis than PTC without HT (PTC-WO). Conclusion: Our study demonstrates that SERPINA1 promotes the occurrence of PTC-W, and its prognosis is better than PTC-WO. SERPINA1 promotes a better prognosis for PTC-W, possibly through a tumour inhibition signalling pathway.


Asunto(s)
Enfermedad de Hashimoto , Neoplasias de la Tiroides , Humanos , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/complicaciones , Neoplasias de la Tiroides/diagnóstico , Neoplasias de la Tiroides/genética , Enfermedad de Hashimoto/complicaciones , Enfermedad de Hashimoto/genética , Enfermedad de Hashimoto/patología , Pronóstico , Perfilación de la Expresión Génica , alfa 1-Antitripsina/genética
3.
Endocrine ; 78(2): 306-314, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35962894

RESUMEN

BACKGROUND: Graves' disease (GD) is an autoimmune disease, the incidence of which is increasing yearly. GD requires long-life therapy. Therefore, the potential immune-related biomarkers of GD need to be studied. METHOD: In our study, differentially expressed genes (DEGs) were derived from the online Gene Expression Omnibus (GEO) microarray expression dataset GSE71956. Protein‒protein interaction (PPI) network analyses were used to identify hub genes, which were validated by qPCR. GSEA was used to screen potential pathways and related immune cells. Next, CIBERSORT analysis was used to further explore the immune subtype distribution pattern among hub genes. ROC curves were used to analyze the specificity and sensitivity of hub genes. RESULT: 44 DEGs were screened from the GEO dataset. Two hub genes, EEF1A1 and EIF4B, were obtained from the PPI network and validated by qPCR (p < 0.05). GSEA was conducted to identify potential pathways and immune cells related to these the two hub genes. Immune cell subtype analysis revealed that hub genes had extensive associations with many different types of immune cells, particularly resting memory CD4+ T cells. AUCs of ROC analysis were 0.687 and 0.733 for EEF1A1 and EIF4B, respectively. CONCLUSION: Our study revealed two hub genes, EEF1A1 and EIF4B, that are associated with resting memory CD4+ T cells and potential immune-related molecular biomarkers and therapeutic targets of GD.


Asunto(s)
Biología Computacional , Enfermedad de Graves , Humanos , Regulación Neoplásica de la Expresión Génica , Biomarcadores de Tumor/metabolismo , Mapas de Interacción de Proteínas/genética , Biomarcadores/metabolismo , Enfermedad de Graves/diagnóstico , Enfermedad de Graves/genética
4.
Am J Transl Res ; 13(11): 12601-12613, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34956476

RESUMEN

OBJECTIVE: The association of the gut microbiome with bone turnover markers (BTMs) in postmenopausal women is poorly understood. METHODS: Fecal samples were collected from 97 Chinese postmenopausal women, and the serum CTX and P1NP were determined. Individuals with serum CTX lower or higher than the median value were divided into LCTX and P1NP groups; and individuals with serum P1NP lower or higher than the median value were grouped into LP1NP and HP1NP groups. Microbiota profiles were determined by high-throughput 16S rRNA gene sequencing. RESULTS: In postmenopausal women, only Faecalibacterium showed significant alteration in the HCTX group compared with the LCTX group (P=0.004, q=0.143). Linear discriminant analysis effect size (LEfSe) analysis revealed that Clostridiaceae (P=0.015, LDA=2.89), Faecalibacterium (P=0.017, LDA=4.60), Prevotella (P=0.040, LDA=3.61) and Clostridium (P=0.007, LDA=2.79) were abundant in the LCTX group, and Facklamia (P=0.044, LDA=3.10) was enriched in the HCTX group. Peptostreptococcaceae (P=0.048, LDA=2.83) and the SMB53 (P=0.028, LDA=2.05) genus were enriched in the LPINP group, and Veillonellaceae (P=0.025, LDA=4.43) and the S24_7 (P=0.023, LDA=3.08) family were enriched in the HPINP group. Six taxa correlated with BTMs in all subjects, including Clostridium (Clostridiaceae) that was negatively correlated with serum CTX amounts significantly (r=-0.34, P<0.001). CONCLUSION: This study identified taxa-specific differences in the intestinal microflora associated with BTMs, notably CTX. These findings may help in uncovering the roles of gut microbiota on bone metabolism.

5.
Cell Death Dis ; 13(1): 1, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34911928

RESUMEN

Fulminant hepatic failure (FHF) is a potentially fatal liver disease that is associated with intrahepatic infiltration of inflammatory cells. As the receptor of polyunsaturated long chain fatty acids, GPR120 can regulate cell differentiation, proliferation, metabolism, and immune response. However, whether GPR120 is involved in FHF remains unknown. Using Propionibacterium acnes (P. acnes)-primed, LPS-induced FHF in mice, we found that interference with GPR120 activity using pharmacological agonist attenuated the severity of the liver injury and mortality of FHF in mice, while a lack of GPR120 exacerbated the disease. GPR120 activation potently alleviated FHF and led to decreased T helper (Th) 1 cell response and expansion of regulatory T cells (Tregs). Interestingly, GPR120 agonist didn't directly target T cells, but dramatically induced a distinct population of CD11c+MHC IIlowCD80lowCD86low regulatory DCs in the livers of FHF mice. GPR120 was found to restrict HIF-1α-dependent glycolysis. The augmented HIF-1α stabilization caused by GPR120 antagonism or deletion could be attenuated by the inhibition of ERK or by the activation of AMPK. Through the analysis of the clinical FHF, we further confirmed the activation of GPR120 was negatively associated with the severity in patients. Our findings indicated that GPR120 activation has therapeutic potential in FHF. Strategies to target GPR120 using agonists or free fatty acids (FFAs) may represent a novel approach to FHF treatment.


Asunto(s)
Células Dendríticas/metabolismo , Fallo Hepático Agudo/genética , Receptores Acoplados a Proteínas G/metabolismo , Animales , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Femenino , Glucólisis , Humanos , Ratones
6.
Adv Exp Med Biol ; 1207: 435-443, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32671766

RESUMEN

Autophagy is a dynamic process, regulated by a variety of factors, and may play different roles in different thyroid diseases or in different stages of the same thyroid disease. Autophagy can mediate inflammatory response and immunity, which is closely related to the pathogenesis of thyroid autoimmune diseases. Therefore, it is still necessary to further understand the relationship between autophagy and autoimmune thyroid disease and hypothyroidism. With more and more studies on the relationship between autophagy and thyroid cancer, the relationship between the two is becoming more and more complicated. From the perspective of current studies, it is still worth pondering whether inhibition or activation of autophagy can be a valuable targeted therapy for thyroid cancer, and further research and efforts are still needed.


Asunto(s)
Autofagia , Enfermedades de la Tiroides , Humanos , Hipotiroidismo , Neoplasias de la Tiroides
7.
Adv Exp Med Biol ; 1207: 445-461, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32671767

RESUMEN

The prevalence of obesity is increasing rapidly and is closely associated with a variety of metabolic diseases. Recent studies have suggested that autophagy is likely to play an important role in the development of obesity and may be related to insulin sensitivity. Autophagy may be involved in the browning of white adipose tissue and may also affect the metabolic balance of lipids. Autophagy can degrade cytoplasmic lipids by lipophagy in hepatocytes. Furthermore, Autophagy in hepatocytes helps prevent NAFLD. The study of autophagy in glucose metabolism is still in a very preliminary stage. Changes in autophagy activity play an important role in the development of insulin resistance in diabetes and many metabolic diseases. Therefore, it is still worth further exploration on the deeper mechanism of oxidative stress induction of insulin resistance to autophagy and whether there will be corresponding complications to the body.


Asunto(s)
Autofagia , Diabetes Mellitus , Obesidad , Tejido Adiposo Pardo , Tejido Adiposo Blanco , Humanos , Resistencia a la Insulina
8.
Adv Exp Med Biol ; 1207: 463-466, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32671768

RESUMEN

Polycystic Ovary Syndrome (PCOS) is a common obesity-related reproductive disease in women of child-bearing age,which is usually accompanied with endocrine and metabolic abnormalities such as hyperandrogenemia and hyperinsulinemia. The abnormal reproductive function of PCOS is mainly characterized by the morphological and functional changes of ovary. Autophagy is involved in the maintenance of human ovarian physiological function as well as in the process of luteal degeneration, and affects the survival of granulosa cells. This chapter introduces the latest research progress of the relationship between autophagy and PCOS. How autophagy is involved in the occurrence and development of PCOS remains to be further studied.


Asunto(s)
Autofagia , Síndrome del Ovario Poliquístico , Femenino , Células de la Granulosa , Humanos , Hiperandrogenismo/complicaciones , Hiperinsulinismo/complicaciones , Obesidad/complicaciones , Ovario/patología , Ovario/fisiología , Ovario/fisiopatología , Síndrome del Ovario Poliquístico/complicaciones
9.
J Clin Endocrinol Metab ; 105(10)2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32556197

RESUMEN

CONTEXT: Papillary thyroid carcinoma (PTC) is the most common endocrine malignancy. Chaperone-mediated autophagy (CMA), 1 type of autophagy, is thought to promote or suppress cancer development in different cancer types. However, the effect of CMA on PTC development and the underlying mechanisms remain unknown. OBJECTIVE: To determine whether CMA plays implied critical roles in the development of PTC. DESIGN: We investigated the association between CMA and PTC development in PTC tissues and normal thyroid tissues by detecting the key protein of CMA, lysosome-associated membrane protein type 2A (LAMP2A), using quantitative polymerase chain reaction (PCR) and immunohistochemistry, which were further validated in the TGCA dataset. The effect of CMA on PTC development was studied by cell proliferation, migration, and apoptosis assays. The underlying mechanisms of peroxisome proliferator-activated receptor γ (PPARγ)-stromal cell-derived factor 1 (SDF1)/ C-X-C motif chemokine receptor 4 (CXCR4) signaling were clarified by western blotting, quantitative PCR, and rescue experiments. Knockdown and tamoxifen were used to analyze the effect of estrogen receptor (ER) α on CMA. RESULTS: Our study confirmed that CMA, indicated by LAMP2A expression, was significantly increased in PTC tumor tissues and cell lines, and was associated with tumor size and lymph node metastasis of patients. Higher CMA in PTC promoted tumor cell proliferation and migration, thereby promoting tumor growth and metastasis. These effects of CMA on PTC were exerted by decreasing PPARγ protein expression to enhance SDF1 and CXCR4 expression. Furthermore, CMA was found positively regulated by ERα signaling in PTC. CONCLUSION: Our investigation identified CMA regulated by ERα promoting PTC tumor progression that enhanced tumor cell proliferation and migration by targeting PPARγ-SDF1/CXCR4 signaling, representing a potential target for treatment of PTC.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Carcinogénesis/patología , Autofagia Mediada por Chaperones/fisiología , Receptor alfa de Estrógeno/metabolismo , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/patología , Animales , Antineoplásicos Hormonales/uso terapéutico , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Autofagia Mediada por Chaperones/efectos de los fármacos , Autofagia Mediada por Chaperones/genética , Quimiocina CXCL12/metabolismo , Conjuntos de Datos como Asunto , Progresión de la Enfermedad , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Persona de Mediana Edad , PPAR gamma/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico , Cáncer Papilar Tiroideo/tratamiento farmacológico , Cáncer Papilar Tiroideo/cirugía , Glándula Tiroides/patología , Glándula Tiroides/cirugía , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/cirugía , Tiroidectomía , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Pharmacol Res ; 158: 104884, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32428667

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a chronic and irreversible lung disease with limited therapeutic strategies. Lycorine (LYC), an alkaloid isolated from Amaryllidaceae family plants, exhibits effective anti-inflammatory, antiviral, and anti-tumor activities. In this study, we attempted to determine the effect of LYC on bleomycin (BLM)-induced IPF and NLRP3 inflammasome activation. Our results demonstrated that the LYC treatment ameliorated BLM-induced pulmonary fibrosis and inflammation in mice. LYC inhibited active Caspase-1 expression and lactate dehydrogenase (LDH) release during BLM-induced acute lung injury (ALI) in mice. Furthermore, our in vitro assay showed that LYC inhibited LPS/Nigericin- or LPS/ATP-induced NACHT, LRP and PYD domains-containing protein 3 (NLRP3) inflammasome activation, and pyroptosis in bone marrow-derived macrophages (BMDMs). Mechanically, LYC could disturb the interaction of NLRP3 with apoptosis-associated speck-like protein containing a CARD (ASC) by targeting the pyrin domain (PYD) on Leu9, Leu50, and Thr53. Our findings indicate that LYC ameliorated BLM-induced pulmonary fibrosis by inhibiting NLRP3 inflammasome activation and pyroptosis through targeting the PYD domain of ASC. Thus, LYC might be a potential therapeutic agent for pulmonary inflammation and fibrosis.


Asunto(s)
Alcaloides de Amaryllidaceae/uso terapéutico , Bleomicina/toxicidad , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Fenantridinas/uso terapéutico , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Piroptosis/efectos de los fármacos , Alcaloides de Amaryllidaceae/química , Alcaloides de Amaryllidaceae/farmacología , Animales , Antibióticos Antineoplásicos/toxicidad , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular/métodos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Fenantridinas/química , Fenantridinas/farmacología , Estructura Secundaria de Proteína , Fibrosis Pulmonar/metabolismo , Piroptosis/fisiología
11.
BMC Cancer ; 19(1): 1094, 2019 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-31718595

RESUMEN

BACKGROUND: The incidence of papillary thyroid carcinoma (PTC) has been increasing worldwide in recent years. Therefore, novel potential therapeutic targets for PTC are urgently needed. Enhancer of zeste homolog 2 (EZH2), a methyltransferase belonging to PRC2, plays important roles in epigenetic silencing and cell cycle regulation. EZH2 overexpression has been found in several malignant tumor tissues, while its expression and function in PTC are largely unknown. METHODS: Sixty-five cases of PTC tissue confirmed by pathology and 30 cases of normal thyroid tissue adjacent to PTC tissue were collected from patients undergoing surgical treatment, between February 2003 and February 2006. We investigated the clinic pathologic significance of EZH2 expression using Realtime-PCR and IHC in 65 human PTC tissues and 30 normal thyroid tissue samples. The EZH2 expression in human PTC cell lines (K1 and W3) and the normal thyroid follicular epithelial cell line Nthy-ori 3-1 was analyzed by Western blotting and Realtime PCR. The expressions of ERα and ERß in cell lines were analyzed by Realtime PCR.The tumor cell biological behavior was evaluated by CCK8 assay, colony formation assay, transwell migration assay and xenograft tumors model. RESULTS: Higher rate of EZH2 expression was found in PTC tissues than in normal thyroid tissues, EZH2 expression is associated with lymph node metastasis and recurrent. Inhibition of EZH2 in PTC cell lines downregulates cellular proliferation and migration. PTC is a disease with high incidence of female and E2-ERα upregulates EZH2 expression. CONCLUSIONS: These results suggest a potential role of EZH2 for the PTC growth and metastasis. As a novel therapy, a pharmacological therapy targeting EZH2 has full potential in treatment of PTC.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/genética , Receptor alfa de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica , Cáncer Papilar Tiroideo/genética , Cáncer Papilar Tiroideo/metabolismo , Adulto , Anciano , Animales , Estudios de Casos y Controles , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Femenino , Xenoinjertos , Humanos , Inmunohistoquímica , Masculino , Ratones , Persona de Mediana Edad , Metástasis de la Neoplasia , Estadificación de Neoplasias , Cáncer Papilar Tiroideo/patología , Carga Tumoral
12.
Biol Res ; 52(1): 57, 2019 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-31767027

RESUMEN

BACKGROUND: Gastric cancer is a common malignant tumor with high morbidity and mortality worldwide, which seriously affects human health. Gramicidin is a short peptide antibiotic which could be used for treating infection induced by bacteria or fungi. However, the anti-cancer effect of gramicidin on gastric cancer cells and its underlying mechanism remains largely unknown. RESULTS: Gastric cancer cells SGC-7901, BGC-823 and normal gastric mucosal cells GES-1 were treated with different concentrations of gramicidin respectively. The results of CCK-8 experiment revealed cellular toxicity of gramicidin to cancer cells while cell colony formation assay showed that gramicidin significantly inhibited the proliferation of gastric cancer cells, but had little effect on normal gastric mucosal cells. In addition, the wound healing assay showed that gramicidin inhibited the migration of SGC-7901 cell. Meanwhile, apoptosis and cell cycle analysis revealed that gramicidin induced cell apoptosis with G2/M cell cycle inhibition. Furthermore, western blot analysis demonstrated that gramicidin down-regulated the expression of cyclinD1 and Bcl-2 as well as the FoxO1 phosphorylation. CONCLUSIONS: The current study illustrated the anti-tumor activity of gramicidin on gastric cancer cells, providing a possibility for gramicidin to be applied in clinical practice for the treatment of gastric cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Gramicidina/farmacología , Neoplasias Gástricas/patología , Línea Celular Tumoral , Ciclina D1/efectos de los fármacos , Ciclina D1/metabolismo , Regulación hacia Abajo , Proteína Forkhead Box O1/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
13.
EMBO Mol Med ; 11(6)2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31036704

RESUMEN

Mutations to KRAS are recurrent in lung adenocarcinomas (LUAD) and are daunting to treat due to the difficulties in KRAS oncoprotein inhibition. A possible resolution to this problem may lie with co-mutations to other genes that also occur in KRAS-driven LUAD that may provide alternative therapeutic vulnerabilities. Approximately 3% of KRAS-mutant LUADs carry functional mutations in NF1 gene encoding neurofibromin-1, a negative regulator of focal adhesion kinase 1 (FAK1). We evaluated the impact of Nf1 loss on LUAD development using a CRISPR/Cas9 platform in a murine model of Kras-mutant LUAD We discovered that Nf1 deactivation is associated with Fak1 hyperactivation and phosphoserine aminotransferase 1 (Psat1) upregulation in mice. Nf1 loss also accelerates murine Kras-driven LUAD tumorigenesis. Analysis of the transcriptome and metabolome reveals that LUAD cells with mutation to Nf1 are addicted to glutamine metabolism. We also reveal that this metabolic vulnerability can be leveraged as a treatment option by pharmacologically inhibiting glutaminase and/or Psat1. Lastly, the findings advocate that tumor stratification by co-mutations to KRAS/NF1 highlights the LAUD patient population expected to be susceptible to inhibiting PSAT1.


Asunto(s)
Adenocarcinoma del Pulmón , Ácido Glutámico/metabolismo , Neoplasias Pulmonares , Mutación , Neurofibromina 1 , Proteínas Proto-Oncogénicas p21(ras) , Transaminasas , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/patología , Animales , Línea Celular , Quinasa 1 de Adhesión Focal , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Noqueados , Neurofibromina 1/genética , Neurofibromina 1/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transaminasas/genética , Transaminasas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Stem Cell Res Ther ; 10(1): 31, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30646961

RESUMEN

BACKGROUND: Fully understanding the developmental process of hepatic stem cells (HSCs) and the mechanisms of their committed differentiation is essential for optimizing the generation of functional hepatocytes for cell therapy in liver disease. Delta-like 1 homolog (Dlk1), primarily the membrane-bound form (Dlk1M), is generally used as a surface marker for fetal hepatic stem cell isolation, while its soluble form (Dlk1S) and the functional roles of different Dlk1 isoforms in HSC differentiation remain to be investigated. METHODS: Hepatic spheroid-derived cells (HSDCs) were isolated from E12.5 mouse livers to obtain Dlk1+ and Dlk1-subpopulations. Colony formation, BrdU staining, and CCK8 assays were used to evaluate the cell proliferation capacity, and hepatic/cholangiocytic differentiation and osteogenesis/adipogenesis were used to assess the multipotency of the two subpopulations. Transformation of Dlk1+ cells into Dlk1- cells was detected by FACS, and the expression of Dlk1 isoforms were measured by western blot. The distinct roles and regulatory mechanisms of Dlk1 isoforms in HSC differentiation were investigated by overexpressing Dlk1M. RESULTS: HSDCs were capable of differentiating into liver and mesenchymal lineages, comprising Dlk1+ and Dlk1- subpopulations. Dlk1+ cells expressed both Dlk1M and Dlk1S and lost expression of Dlk1M during passaging, thus transforming into Dlk1- cells, which still contained Dlk1S. Dlk1- cells maintained a self-renewal ability similar to that of Dlk1+ cells, but their capacity to differentiate into cholangiocytes was obviously enhanced. Forced expression of Dlk1M in Dlk1- cells restored their ability to differentiate into hepatocytes, with an attenuated ability to differentiate into cholangiocytes, suggesting a functional role of Dlk1 in regulating HSC differentiation in addition to acting as a biomarker. Further experiments illustrated that the regulation of committed HSC differentiation by Dlk1 was mediated by the AKT and MAPK signaling pathways. In addition, bFGF was found to serve as an important inducement for the loss of Dlk1M from Dlk1+ cells, and autophagy might be involved. CONCLUSIONS: Overall, our study uncovered the differential expression and regulatory roles of Dlk1 isoforms in the commitment of HSC differentiation and suggested that Dlk1 functions as a key regulator that instructs cell differentiation rather than only as a marker of HSCs. Thus, our findings expand the current understanding of the differential regulation of bi-potential HSC differentiation and provide a fine-tuning target for cell therapy in liver disease.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Hepatocitos/citología , Células Madre/citología , Animales , Diferenciación Celular/fisiología , Femenino , Hepatocitos/metabolismo , Hígado/citología , Hígado/embriología , Ratones , Ratones Endogámicos C57BL , Embarazo , Isoformas de Proteínas , Células Madre/metabolismo
15.
Diagn Pathol ; 14(1): 3, 2019 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-30636642

RESUMEN

BACKGROUND: Lipopolysaccharide (LPS) from Helicobacter pylori (HP) plays an important role in gastric cancer occurrence and development. Toll-like receptor 4 (TLR4) and myeloid differential protein-2 (MD-2) are also reported to be involved in gastric cancer cell proliferation and invasion. CXC chemokine receptor 7 (CXCR7), a second receptor for CXCL12, has been detected in multiple types of tumor tissues. Nevertheless, the biological function and regulation of CXCR7 and its relationship with TLR4 and MD-2 in gastric cancer are not completely understood and therefore warrant further study. METHODS: CXCR7 expression was examined in 150 gastric cancer tissues using immunohistochemistry (IHC). RT-PCR and western blotting were used to detect CXCR7 expression in several gastric cancer cell lines (SGC7901, AGS, MGC-803, MKN-45 and BGC823). shRNAs were designed using a pGPU6/GFP/Neo vector. A CCK-8 assay was used to assess cell proliferation, and transwell assays were performed to assess cell migration. In addition, a gastric cancer xenograft model was generated. RESULTS: The LPS-TLR4-MD-2 pathway elevates CXCR7 expression in SGC7901 cells, and TLR4/MD-2-mediated increases in CXCR7 levels modulate the proliferation and migration of tumor cells. Knockdown of TLR4 and MD-2 demonstrated that both are essential for LPS-induced CXCR7 expression, which in turn is responsible for LPS-induced SGC7901 cell proliferation and migration. Moreover, higher TLR4, MD-2 and CXCR7 expression was detected in gastric cancer tissues than in paracancerous normal control tissues. The expression levels of TLR4, MD-2 and CXCR7 were closely related to gastric cancer TNM stage and lymph node metastasis. In an animal model, significant differences in CXCR7 expression in tumor masses were observed between the control group and experimental group. CONCLUSIONS: The results of this study indicate that CXCR7 plays an important role in gastric cancer progression via inflammatory mechanisms, suggesting that CXCR7 could provide a basis for the development and clinical application of a targeted drug for gastric cancer.


Asunto(s)
Helicobacter pylori/química , Antígeno 96 de los Linfocitos/metabolismo , Receptores CXCR/metabolismo , Neoplasias Gástricas/patología , Receptor Toll-Like 4/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Lipopolisacáridos/farmacología , Metástasis Linfática , Antígeno 96 de los Linfocitos/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , ARN Interferente Pequeño , Receptores CXCR/genética , Neoplasias Gástricas/metabolismo , Receptor Toll-Like 4/genética , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cancer Lett ; 447: 56-65, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30685413

RESUMEN

L1 cell adhesion molecule (L1CAM) promotes invasiveness and metastasis in non-small cell lung cancer (NSCLC) cells and is upregulated by the p53-regulated transcription factor Slug. p21-activated kinase 4 (PAK4) directly phosphorylates Slug, resulting in pro-malignant Slug stabilization. We hypothesized that microRNA-based negative regulation of PAK4 would reduce L1CAM-induced NSCLC aggressiveness via destabilizing Slug. We found that elevated L1CAM expression was tightly correlated with p53 loss-of-function and reduced NSCLC patient survival. L1CAM suppression reduced NSCLC cell migration and invasiveness in vitro as well as tumor formation and distal metastasis in vivo. Mechanistically, p53 restricts L1CAM expression through the ß-catenin/Slug pathway, with levels of ß-catenin and Slug positively correlating with L1CAM expression in NSCLC tumors. The microRNA miR-193a-3p directly targets PAK4 and suppresses downstream p-Slug and L1CAM expression. Silencing PAK4, Slug, and L1CAM mirrored miR-193a-3p's effects upon the migration and invasiveness of NSCLC cells in vitro. Decreased miR-193a-3p levels correlated with elevated PAK4, p-Slug, and L1CAM levels in NSCLC tumors. Our findings support a model of miR-193a-3p as a suppressor of metastatic disease progression in NSCLC via modulation of the p53/Slug/L1CAM pathway.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , MicroARNs/genética , Transducción de Señal/genética , Quinasas p21 Activadas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Pulmonares/patología , Molécula L1 de Adhesión de Célula Nerviosa/genética , Factores de Transcripción de la Familia Snail/genética , Proteína p53 Supresora de Tumor/genética
17.
Biol. Res ; 52: 57-57, 2019. ilus, graf
Artículo en Inglés | LILACS | ID: biblio-1505777

RESUMEN

BACKGROUND: Gastric cancer is a common malignant tumor with high morbidity and mortality worldwide, which seriously affects human health. Gramicidin is a short peptide antibiotic which could be used for treating infection induced by bacteria or fungi. However, the anti-cancer effect of gramicidin on gastric cancer cells and its underlying mechanism remains largely unknown. RESULTS: Gastric cancer cells SGC-7901, BGC-823 and normal gastric mucosal cells GES-1 were treated with different concentrations of gramicidin respectively. The results of CCK-8 experiment revealed cellular toxicity of gramicidin to cancer cells while cell colony formation assay showed that gramicidin significantly inhibited the proliferation of gastric cancer cells, but had little effect on normal gastric mucosal cells. In addition, the wound healing assay showed that gramicidin inhibited the migration of SGC-7901 cell. Meanwhile, apoptosis and cell cycle analysis revealed that gramicidin induced cell apoptosis with G2/M cell cycle inhibition. Furthermore, western blot analysis demonstrated that gramicidin down-regulated the expression of cyclinD1 and Bcl-2 as well as the FoxO1 phosphorylation. CONCLUSIONS: The current study illustrated the anti-tumor activity of gramicidin on gastric cancer cells, providing a possibility for gramicidin to be applied in clinical practice for the treatment of gastric cancer.


Asunto(s)
Humanos , Neoplasias Gástricas/patología , Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Gramicidina/farmacología , Fosforilación , Regulación hacia Abajo , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ciclina D1/efectos de los fármacos , Ciclina D1/metabolismo , Línea Celular Tumoral , Proteína Forkhead Box O1/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo
18.
Metabolism ; 88: 31-39, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30309516

RESUMEN

OBJECTIVE: Obesity is associated with an increased risk of many metabolic disorders, including non-alcoholic fatty liver disease (NAFLD). However, the underlying mechanisms remain poorly understood. Recent studies have demonstrated that MicroRNA-mediated gene silencing plays an important role in hepatic triglyceride (TG) metabolism. In the present study, we aimed to investigate the pathological function of miR-361-5p in the development of NAFLD. METHODS: Expression levels of miR-361-5p was determined by quantitative real-time PCR in livers of obese mice and NAFLD patients. Liver tissues from mice with miR-361-5p overexpression or inhibition were collected and analyzed by TG contents, gene expression profile. RESULTS: Expression of miR-361-5p was increased in the livers of two obese mouse models and NAFLD subjects. Overexpression of miR-361-5p in C57BL/6 mice led to hepatosteatosis, whereas inhibition of miR-361-5p expression in db/db mice improved TG accumulation and insulin sensitivity. Mechanistically, we identified Sirt1 as a direct target gene of miR-361-5p and re-introduction of Sirt1 largely abolished the metabolic action of miR-361-5p. CONCLUSIONS: Our results demonstrated the role of miR-361-5p in the regulation of hepatic TG homeostasis, which may provide potential therapeutic target for hepatosteatosis.


Asunto(s)
MicroARNs/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/metabolismo , Sirtuina 1/metabolismo , Regulación hacia Arriba , Animales , Humanos , Resistencia a la Insulina , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Triglicéridos/metabolismo
19.
Mol Cell Biochem ; 420(1-2): 195-206, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27514537

RESUMEN

A specialized culture medium termed ciliary neurotrophic factor-treated astrocyte-conditioned medium (CNTF-ACM) allows investigators to assess the peripheral effects of CNTF-induced activated astrocytes upon cultured neurons. CNTF-ACM has been shown to upregulate neuronal L-type calcium channel current activity, which has been previously linked to changes in mitochondrial respiration and oxidative stress. Therefore, the aim of this study was to evaluate CNTF-ACM's effects upon mitochondrial respiration and oxidative stress in rat cortical neurons. Cortical neurons, CNTF-ACM, and untreated control astrocyte-conditioned medium (UC-ACM) were prepared from neonatal Sprague-Dawley rat cortical tissue. Neurons were cultured in either CNTF-ACM or UC-ACM for a 48-h period. Changes in the following parameters before and after treatment with the L-type calcium channel blocker isradipine were assessed: (i) intracellular calcium levels, (ii) mitochondrial membrane potential (ΔΨm), (iii) oxygen consumption rate (OCR) and adenosine triphosphate (ATP) formation, (iv) intracellular nitric oxide (NO) levels, (v) mitochondrial reactive oxygen species (ROS) production, and (vi) susceptibility to the mitochondrial complex I toxin rotenone. CNTF-ACM neurons displayed the following significant changes relative to UC-ACM neurons: (i) increased intracellular calcium levels (p < 0.05), (ii) elevation in ΔΨm (p < 0.05), (iii) increased OCR and ATP formation (p < 0.05), (iv) increased intracellular NO levels (p < 0.05), (v) increased mitochondrial ROS production (p < 0.05), and (vi) increased susceptibility to rotenone (p < 0.05). Treatment with isradipine was able to partially rescue these negative effects of CNTF-ACM (p < 0.05). CNTF-ACM promotes mitochondrial respiration and oxidative stress in cortical neurons through elevating L-type calcium channel activity.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Corteza Cerebral/metabolismo , Medios de Cultivo Condicionados/farmacología , Mitocondrias/metabolismo , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Animales , Corteza Cerebral/citología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Neuronas/citología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
20.
Sci Rep ; 6: 26108, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-27198888

RESUMEN

The genetic regulation of cell cycle progression and cell proliferation plays a role in the growth of non-small cell lung cancer (NSCLC), one of the most common causes of cancer-related mortality. Although DEAD-box RNA helicases are known to play a role in cancer development, including lung cancer, the potential involvement of the novel family member DDX51 has not yet been investigated. In the current study we assessed the role of DDX51 in NSCLC using a siRNA-based approach. DDX51 siRNA-expressing cells exhibited a slower cell proliferation rate and underwent arrest in S-phase of the cell cycle compared with control cells. Microarray analyses revealed that DDX51siRNA expression resulted in the dysregulation of a number of cell signalling pathways. Moreover, injection of DDX51 siRNA into an animal model resulted in the formation of smaller tumours compared with the control group. We also assessed the expression of DDX51 in patients with NSCLC, and the data revealed that the expression was correlated with patient age but no other risk factors. Overall, our data suggest for the first time that DDX51 aids cell cancer proliferation by regulating multiple signalling pathways, and that this protein might be a therapeutic target for NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/patología , ARN Helicasas DEAD-box/metabolismo , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Animales , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , ARN Helicasas DEAD-box/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/mortalidad , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , ARN Interferente Pequeño/genética , Transducción de Señal , Análisis de Supervivencia , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...