Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Microbiol Spectr ; 12(8): e0354923, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38916335

RESUMEN

In recent years, most studies on the gut microbiome have primarily focused on feces samples, leaving the microbial communities in the intestinal mucosa relatively unexplored. To address this gap, our study employed shotgun metagenomics to analyze the microbial compositions in normal rectal mucosa and matched feces from 20 patients with colonic polyps. Our findings revealed a pronounced distinction of the microbial communities between these two sample sets. Compared with feces, the mucosal microbiome contains fewer genera, with Burkholderia being the most discriminating genus between feces and mucosa, highlighting its significant influence on the mucosa. Furthermore, based on the microbial classification and KEGG Orthology (KO) annotation results, we explored the association between rectal mucosal microbiota and factors such as age, gender, BMI, and polyp risk level. Notably, we identified novel biomarkers for these phenotypes, such as Clostridium ramosum and Enterobacter cloacae in age. The mucosal microbiota showed an enrichment of KO pathways related to sugar transport and short chain fatty acid metabolism. Our comprehensive approach not only bridges the knowledge gap regarding the microbial community in the rectal mucosa but also underscores the complexity and specificity of microbial interactions within the human gut, particularly in the Chinese population. IMPORTANCE: This study presents a system-level map of the differences between feces and rectal mucosal microbial communities in samples with colorectal cancer risk. It reveals the unique microecological characteristics of rectal mucosa and its potential influence on health. Additionally, it provides novel insights into the role of the gut microbiome in the pathogenesis of colorectal cancer and paves the way for the development of new prevention and treatment strategies.


Asunto(s)
Bacterias , Heces , Microbioma Gastrointestinal , Mucosa Intestinal , Recto , Humanos , Heces/microbiología , Masculino , Mucosa Intestinal/microbiología , Femenino , Microbioma Gastrointestinal/genética , Persona de Mediana Edad , Recto/microbiología , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Anciano , Adulto , Pólipos del Colon/microbiología , Metagenómica , Neoplasias Colorrectales/microbiología
2.
Clin Cancer Res ; 23(8): 2038-2049, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-27683179

RESUMEN

Purpose: Antiproliferative, antiviral, and immunomodulatory activities of endogenous type I IFNs (IFN1) prompt the design of recombinant IFN1 for therapeutic purposes. However, most of the designed IFNs exhibited suboptimal therapeutic efficacies against solid tumors. Here, we report evaluation of the in vitro and in vivo antitumorigenic activities of a novel recombinant IFN termed sIFN-I.Experimental Design: We compared primary and tertiary structures of sIFN-I with its parental human IFNα-2b, as well as affinities of these ligands for IFN1 receptor chains and pharmacokinetics. These IFN1 species were also compared for their ability to induce JAK-STAT signaling and expression of the IFN1-stimulated genes and to elicit antitumorigenic effects. Effects of sIFN-I on tumor angiogenesis and immune infiltration were also tested in transplanted and genetically engineered immunocompetent mouse models.Results: sIFN-I displayed greater affinity for IFNAR1 (over IFNAR2) chain of the IFN1 receptor and elicited a greater extent of IFN1 signaling and expression of IFN-inducible genes in human cells. Unlike IFNα-2b, sIFN-I induced JAK-STAT signaling in mouse cells and exhibited an extended half-life in mice. Treatment with sIFN-I inhibited intratumoral angiogenesis, increased CD8+ T-cell infiltration, and robustly suppressed growth of transplantable and genetically engineered tumors in immunodeficient and immunocompetent mice.Conclusions: These findings define sIFN-I as a novel recombinant IFN1 with potent preclinical antitumorigenic effects against solid tumor, thereby prompting the assessment of sIFN-I clinical efficacy in humans. Clin Cancer Res; 23(8); 2038-49. ©2016 AACR.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Interferón-alfa/química , Interferón-alfa/farmacología , Animales , Femenino , Citometría de Flujo , Humanos , Immunoblotting , Interferón alfa-2 , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Neoplasias Experimentales/tratamiento farmacológico , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología , Resonancia por Plasmón de Superficie , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Biol Chem ; 288(49): 35500-10, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24165128

RESUMEN

Lithium is an effective mood stabilizer that has been clinically used to treat bipolar disorder for several decades. Recent studies have suggested that lithium possesses robust neuroprotective and anti-tumor properties. Thus far, a large number of lithium targets have been discovered. Here, we report for the first time that HDAC1 is a target of lithium. Lithium significantly down-regulated HDAC1 at the translational level by targeting HDAC1 mRNA. We also showed that depletion of HDAC1 is essential for the neuroprotective effects of lithium and for the lithium-mediated degradation of mutant huntingtin through the autophagic pathway. Our studies explain the multiple functions of lithium and reveal a novel mechanism for the function of lithium in neurodegeneration.


Asunto(s)
Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 1/genética , Litio/farmacología , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Autofagia , Proteínas CELF1 , Regulación hacia Abajo/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Células HEK293 , Células HeLa , Histona Desacetilasa 1/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Proteína Huntingtina , Fármacos Neuroprotectores/farmacología , Proteolisis/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo
5.
Oncol Rep ; 30(1): 364-70, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23604401

RESUMEN

Aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor associated with tumor initiation and progression. AhR expression is significantly increased in gastric cancer tissues and gastric cancer cell lines; however, the relationship between AhR and gastric cancer is still unclear. In the present study, we explored the effects of the inhibition of AhR expression by RNA interference on the biological behavior of gastric cancer cells (MKN45 and SGC7901), and elucidated the specific mechanisms of AhR action in the development of gastric cancer. Results showed that small interfering RNA (siRNA) against AhR effectively inhibited the expression of AhR, and decreased the expression of cytochrome P450 (CYP)1A1 and CYP1B1, which are classic target genes of the AhR pathway. Compared to the negative control group, AhR-siRNA-transfected cells showed decreased cellular growth, delayed G1-S cell cycle progression and increased apoptosis rate. Furthermore, inhibition of AhR expression by siRNA in SGC7901 cells led to decreased cell migratory and invasive ability, accompanied by downregulation of expression and activity of matrix metalloproteinase (MMP)-2 and MMP-9. Our results, therefore, suggest that AhR promotes the growth and invasiveness of gastric cancer cells and AhR may serve as a promising therapeutic target for gastric cancer.


Asunto(s)
Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Neoplasias Gástricas/genética , Hidrocarburo de Aril Hidroxilasas/biosíntesis , Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Citocromo P-450 CYP1A1/biosíntesis , Citocromo P-450 CYP1B1 , Regulación hacia Abajo , Humanos , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Invasividad Neoplásica/genética , Interferencia de ARN , ARN Interferente Pequeño , Receptores de Hidrocarburo de Aril/biosíntesis
6.
Turk J Gastroenterol ; 23(3): 212-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22798109

RESUMEN

BACKGROUND/AIMS: Gastrokine-1 is a novel protein that plays an important role in the maintenance of the integrity of the gastric mucosa. However, whether Helicobacter pylori infection and non-steroidal anti-inflammatory drugs, which are known to cause gastric mucosal injuries, affect gastrokine-1 expression in the gastric mucosa is unknown. The aim of the present study was to determine gastric mucosal expression of gastrokine-1 in patients with Helicobacter pylori infection or long-term non-steroidal anti-inflammatory drug administration. MATERIAL AND METHODS: A total of 40 patients with functional dyspepsia (20 with Helicobacter pylori-negative chronic gastritis, and 20 with Helicobacter pylori-positive chronic gastritis), and 37 Helicobacter pylori-negative long-term non-steroidal anti-inflammatory drug users (26 with aspirin, 11 with selective cyclooxygenase-2 inhibitors) were selected. In addition, 20 Helicobacter pylori-negative healthy volunteers were recruited as controls. All subjects underwent endoscopies with biopsies taken from the antrum and the sites with lesions. Gastric mucosal changes were detected endoscopically and histologically, and gastrokine-1 protein expression in the antral mucosa was analyzed by immunohistochemistry. RESULTS: Expression of gastrokine-1 protein was decreased in Helicobacter pylori-positive chronic gastritis compared with Helicobacter pylori-negative subjects and the healthy controls. Similarly, gastrokine-1 expression in non-steroidal anti-inflammatory drug users was also decreased, compared with the healthy controls, but there was no significant difference in gastrokine-1 expression between the aspirin group and selective cyclooxygenase-2 inhibitor group. Moreover, gastrokine-1 expression levels tended to be associated with the severity of chronic gastritis. CONCLUSIONS: Both Helicobacter pylori infection and long-term non-steroidal anti-inflammatory drug administration downregulate gastrokine-1 expression in the gastric mucosa, which may contribute to the gastric mucosal injuries induced by these two factors.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Mucosa Gástrica/efectos de los fármacos , Gastritis/metabolismo , Infecciones por Helicobacter/metabolismo , Helicobacter pylori , Hormonas Peptídicas/metabolismo , Aspirina/administración & dosificación , Estudios de Casos y Controles , Inhibidores de la Ciclooxigenasa 2/administración & dosificación , Regulación hacia Abajo , Dispepsia/metabolismo , Dispepsia/microbiología , Mucosa Gástrica/metabolismo , Gastritis/microbiología , Regulación de la Expresión Génica , Infecciones por Helicobacter/microbiología , Humanos , Inmunohistoquímica , Hormonas Peptídicas/genética
7.
Oncol Rep ; 28(4): 1257-62, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22842654

RESUMEN

Trefoil factor 1 (TFF1) is a small cysteine-rich secreted protein which is principally expressed in the superficial cells of gastric mucosa. In gastric cancer, TFF1 is downregulated and plays an important role. Gastrokine 1 (GKN1) is a secreted protein with similar expression and biological functions to TFF1. This study aimed to determine the expression and biological functions of TFF1 and the relationships between TFF1 and GKN1 in gastric cancer. RT-PCR and immunohistochemistry were performed to detect TFF1 expression in gastric cancer cell lines and tissues. The transfected and co-transfected AGS cells which stably expressed TFF1 or both TFF1 and GKN1 were generated. Phenotypic changes such as cell viability, apoptosis and cell cycle modulation were assayed in the transfected cells. We found that TFF1 expression was significantly downregulated or lost in gastric cancer cell lines, gastric dysplasia and cancer. Restoration of TFF1 expression in AGS cells suppressed tumor cell viability and arrested AGS cells in the G1-S transition phase after olomoucine treatment. However, TFF1 was unable to induce cell apoptosis. In co-transfected cells, we found that TFF1 and GKN1 did not directly interact at the protein level. GKN1 was unable to cooperate with TFF1 on cell viability suppression, cell apoptosis and differentiation. Together, these results indicate that TFF1 expression is significantly downregulated in gastric cancer. TFF1 inhibited cell proliferation by delaying G1-S phase transition but not by inducing apoptosis. TFF1 may not interact or cooperate with GKN1 at the protein and functional level.


Asunto(s)
Hormonas Peptídicas/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adulto , Anciano , Apoptosis/genética , Estudios de Casos y Controles , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Femenino , Mucosa Gástrica/metabolismo , Humanos , Inmunoprecipitación , Masculino , Persona de Mediana Edad , Hormonas Peptídicas/genética , Neoplasias Gástricas/patología , Factor Trefoil-1 , Proteínas Supresoras de Tumor/genética
8.
J Exp Clin Cancer Res ; 31: 46, 2012 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-22592002

RESUMEN

BACKGROUND: Aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor associated with gastric carcinogenesis. 3,3'-Diindolylmethane (DIM) is a relatively non-toxic selective AhR modulator. This study was to detect the effects of DIM on gastric cancer cell growth. METHODS: Gastric cancer cell SGC7901 was treated with DIM at different concentrations (0,10,20,30,40,50 µmol/L) with or without an AhR antagonist, resveratrol. The expression of AhR and Cytochrome P4501A1 (CYP1A1), a classic target gene of AhR pathway, were detected by RT-PCR and Western blot; cell viability was measured by MTT assay, and the changes in cell cycle and apoptosis were analyzed by flow cytometry. RESULTS: RT-PCR and western-blot showed that with the increase of the concentration of DIM, AhR protein gradually decreased and CYP1A1 expression increased, suggesting that DIM activated the AhR pathway and caused the translocation of AhR from cytoplasm to nucleus. MTT assay indicated that the viability of SGC7901 cells was significantly decreased in a concentration- and time-dependent manner after DIM treatment and this could be partially reversed by resveratrol. Flow cytometry analysis showed that DIM arrested cell cycle in G1 phase and induced cell apoptosis. CONCLUSION: Selective aryl hydrocarbon receptor modulator 3,3'-Diindolylmethane inhibits SGC7901 cell proliferation by inducing apoptosis and delaying cell cycle progression. AhR may be a potential therapeutic target for gastric cancer treatment.


Asunto(s)
Indoles/farmacología , Receptores de Hidrocarburo de Aril/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Anticarcinógenos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citocromo P-450 CYP1A1/biosíntesis , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Humanos , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/genética , Resveratrol , Estilbenos/farmacología , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
9.
J Exp Clin Cancer Res ; 31: 49, 2012 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-22621392

RESUMEN

BACKGROUND: Gastrokine-1 (GKN1), a secreted protein, is specifically expressed in gastric mucosa to protect and maintain the integrity of gastric epithelium. The present study investigated differential expression of GKN1 in normal, precancerous, and cancerous gastric tissues, and explored the biological functions of GKN1 protein in gastric cancer cells. METHODS: RT-PCR, Western blot, and immunohistochemistry were performed to detect GKN1 expression in normal, precancerous, cancerous gastric tissues and seven gastric cancer cell lines. Gene transfection was used to restore GKN1 expression in gastric cancer AGS cells. Phenotypic changes (i.e., cell viability, apoptosis, cell cycle modulation, and sensitivity of gastric cancer cells to fluorouracil (5-FU)) were assayed in the transfected cells. DNA microarrays were used to analyze expression changes of apoptosis-related genes. RESULTS: Significant downregulation or absence of GKN1 expression in seven gastric cancer cell lines were detected and progressive decrease of GKN1 expression from normal mucosa, precancerous tissue, to cancer tissues was observed. Moreover, restoration of GKN1 expression suppressed gastric cancer cell viability and induced the cells to undergo apoptosis. GKN1 expression also enhanced tumor cell sensitivity to 5-FU treatment. Moreover, it was found that GKN1 expression in AGS cells modulated expression of 19 apoptosis-related genes. CONCLUSIONS: Expression of GKN1 is progressively lost from normal mucosa, precancerous to cancerous gastric tissues, while restoration of GKN1 expression induces gastric cancer cells to undergo apoptosis, and enhances sensitivity of gastric cancer cells to 5-FU-induced apoptosis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Hormonas Peptídicas , Neoplasias Gástricas , Adulto , Anciano , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular , Epitelio/metabolismo , Femenino , Fluorouracilo/farmacología , Mucosa Gástrica/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Hormonas Peptídicas/genética , Hormonas Peptídicas/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA