Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nutr Neurosci ; : 1-17, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38689541

RESUMEN

PURPOSE: Evidence shows diet promotes brain health. Combining foods and nutrients may have beneficial synergistic effects, but the effects on cognitive function interventions are inconsistent. So, a meta-analysis of RCTs was conducted to examine the specific effects on cognitive function. METHODS: We searched four databases from creation to April 2023. Eligible randomized controlled trials were identified. A random-effects meta-analysis was used to combine standardized mean differences (SMD) (95% confidence intervals [CI]), and homogeneity tests for a variance were calculated. RESULTS: A total of 19 studies involving 12,119 participants were included in this systematic review. The dietary intervention group had a positive effect on overall cognitive functioning compared to the control group (SMD = 0.14, 95% CI [0.08, 0.20], P < 0.00001). The dietary intervention improved executive function, processing speed and language skills (SMD = -0.10, 95% CI [-0.17,-0.04], P = 0.002, I2 = 0%), (SMD = -0.16, 95% CI [-0.23,-0.09], P < 0.00001, I2 = 0%), (SMD = 0.10, 95% CI [0.01, 0.20], P = 0.03, I2 = 0%). The dietary intervention had no effect on delayed memory and spatial ability (SMD = 0.04, 95% CI [-0.02, 0.09], P = 0.20, I2 = 0%), (SMD = 0.08, 95% CI [-0.01, 0.16], P = 0.08, I2 = 0%). CONCLUSION: The Mediterranean diet, a diet with restricted caloric intake, a diet incorporating aerobic exercise, a low-carbohydrate diet, and a healthy lifestyle diet (increased intake of fruits and vegetables, and weight and blood pressure management) appear to have positive effects on cognitively healthy adults, as reflected in their overall cognitive, processing speed, executive, and language functions. PROSPERO REGISTRATION NUMBER: CRD42023414704.

2.
Cell Host Microbe ; 32(2): 191-208.e9, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38237593

RESUMEN

Aspirin-related gastrointestinal damage is of growing concern. Aspirin use modulates the gut microbiota and associated metabolites, such as bile acids (BAs), but how this impacts intestinal homeostasis remains unclear. Herein, using clinical cohorts and aspirin-treated mice, we identified an intestinal microbe, Parabacteroides goldsteinii, whose growth is suppressed by aspirin. Mice supplemented with P. goldsteinii or its BA metabolite, 7-keto-lithocholic acid (7-keto-LCA), showed reduced aspirin-mediated damage of the intestinal niche and gut barrier, effects that were lost with a P. goldsteinii hdhA mutant unable to generate 7-keto-LCA. Specifically, 7-keto-LCA promotes repair of the intestinal epithelium by suppressing signaling by the intestinal BA receptor, farnesoid X receptor (FXR). 7-Keto-LCA was confirmed to be an FXR antagonist that facilitates Wnt signaling and thus self-renewal of intestinal stem cells. These results reveal the impact of oral aspirin on the gut microbiota and intestinal BA metabolism that in turn modulates gastrointestinal homeostasis.


Asunto(s)
Aspirina , Microbioma Gastrointestinal , Ratones , Animales , Aspirina/farmacología , Ácidos y Sales Biliares , Receptores Citoplasmáticos y Nucleares , Homeostasis
3.
J Vis Exp ; (194)2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-37125810

RESUMEN

Contractile forces generated by actin and non-muscle myosin II ("actomyosin contractility") are critical for morphological changes of cells and tissues at multiple length scales, such as cell division, cell migration, epithelial folding, and branching morphogenesis. An in-depth understanding of the role of actomyosin contractility in morphogenesis requires approaches that allow the rapid inactivation of actomyosin, which is difficult to achieve using conventional genetic or pharmacological approaches. The presented protocol demonstrates the use of a CRY2-CIBN based optogenetic dimerization system, Opto-Rho1DN, to inhibit actomyosin contractility in Drosophila embryos with precise temporal and spatial controls. In this system, CRY2 is fused to the dominant negative form of Rho1 (Rho1DN), whereas CIBN is anchored to the plasma membrane. Blue light-mediated dimerization of CRY2 and CIBN results in rapid translocation of Rho1DN from the cytoplasm to the plasma membrane, where it inactivates actomyosin by inhibiting endogenous Rho1. In addition, this article presents a detailed protocol for coupling Opto-Rho1DN-mediated inactivation of actomyosin with laser ablation to investigate the role of actomyosin in generating epithelial tension during Drosophila ventral furrow formation. This protocol can be applied to many other morphological processes that involve actomyosin contractility in Drosophila embryos with minimal modifications. Overall, this optogenetic tool is a powerful approach to dissect the function of actomyosin contractility in controlling tissue mechanics during dynamic tissue remodeling.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/metabolismo , Actomiosina/metabolismo , Optogenética , Embrión no Mamífero , Citoesqueleto de Actina/metabolismo , Proteínas de Drosophila/genética , Morfogénesis , Proteínas de Unión al GTP rho/metabolismo
4.
STAR Protoc ; 4(1): 101972, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36598852

RESUMEN

We describe a protocol for optogenetic inhibition of the small GTPase Rho1 (RhoA) in Drosophila embryos, which allows rapid and spatially confined inactivation of Rho1 and Rho1-mediated actomyosin contractility. We provide step-by-step instruction for optogenetic manipulations of Drosophila embryos using confocal and multiphoton imaging systems. This tool is useful for determining the site- and stage-specific function of Rho1 in Drosophila embryos and for studying the immediate tissue response to acute elimination of cellular contractility. For complete details on the use and execution of this protocol, please refer to Guo et al. (2022).1.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/genética , Proteínas de Drosophila/genética , Optogenética , Embrión no Mamífero , Actomiosina
5.
Microb Pathog ; 174: 105937, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36496058

RESUMEN

Helicobacter pylori (H. pylori), a gram-negative bacterial microbiological carcinogen, has been identified as the leading jeopardy feature for developing human gastric cancer (GC). As a result, inhibiting H. pylori growth has been identified as an effective and critical technique for preventing GC development. In this study, geraniol inhibits H. pylori-induced gastric carcinogen signalling in human gastric epithelial cells (GES-1). Geraniol prevents cytotoxicity, ROS and apoptosis in H. pylori-induced GES-1 cells. Furthermore, geraniol protects against H. -induced antioxidant depletion caused by malondialdehyde, damage of reactive DNA and nuclear fragmentation. Geraniol significantly reduced the expression of phosphorylated mitogen activated protein kinases (MAPKs) proteins such as p38 MAPK, extracellular signal-regulated kinase-1 (ERK1), c-Jun N-terminal kinase (c-JNK), tumour necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and cyclooxygenase-2 (COX-2) in GES-1 infected with H. pylori. Furthermore, geraniol increased the antioxidant protein peroxiredoxin-1 (Prdx-1) in H. pylori-infected cells. Geraniol thus protects H. pylori-concomitant infection, and its resistance may be a possible method in preventing gastric cancer caused by H. pylori.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Neoplasias Gástricas , Humanos , Antioxidantes/farmacología , Antioxidantes/metabolismo , Carcinógenos/metabolismo , Carcinógenos/farmacología , Células Epiteliales , Mucosa Gástrica/patología , Infecciones por Helicobacter/microbiología , Neoplasias Gástricas/patología
6.
Cell Death Dis ; 13(10): 883, 2022 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-36266267

RESUMEN

High-fat diet (HFD) has been implicated to promote colorectal cancer (CRC). Recently, oncogene Cyclophilin B (CypB) is reported to be induced by cholesterol. However, the role of CypB in CRC carcinogenesis and metastasis associated with HFD remains unknown. In the present study, we showed that HFD-induced CypB enhances proliferation and metastasis through an inflammation-driven circuit, including Signal Transducer and Activator of Transcription 3 (STAT3)-triggered transcription of lncRNA-PVT1, and its binding with CypB that promotes activation of STAT3. CypB was found to be upregulated in CRC, which was correlated with elevated body mass index and poor prognosis. HFD induced CypB expression and proinflammatory cytokines in colon of mice. Besides, CypB restoration facilitated growth, invasion and metastasis in CRC cells both in vitro and in vivo. Moreover, RIP sequencing data identified lncRNA-PVT1 as a functional binding partner of CypB. Mechanistically, PVT1 increased the phosphorylation and nuclear translocation of STAT3 in response to IL-6, through directly interaction with CypB, which impedes the binding of Suppressors Of Cytokine Signalling 3 (SOCS3) to STAT3. Furthermore, STAT3 in turn activated PVT1 transcription through binding to its promoter, forming a regulatory loop. Finally, this CypB/STAT3/PVT1 axis was verified in TCGA datasets and CRC tissue arrays. Our data revealed that CypB linked HFD and CRC malignancy by enhancing the CypB/STAT3/PVT1 feedforward axis and activation of STAT3.


Asunto(s)
Neoplasias Colorrectales , ARN Largo no Codificante , Animales , Ratones , Carcinogénesis , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Dieta Alta en Grasa , Interleucina-6/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
7.
Front Cardiovasc Med ; 9: 918167, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35795364

RESUMEN

Background: Contrast-induced acute kidney injury (CI-AKI) is a common complication of patients undergoing percutaneous coronary intervention (PCI). Data regarding the influence of sodium-glucose cotransporter-2 (SGLT2) inhibitor on the CI-AKI incidence and renal outcomes of patients undergoing PCI are limited. This study aimed to examine the real-world risk of CI-AKI in SGLT2 inhibitor users undergoing PCI. Methods: We used longitudinal data from the medical records of the First Affiliated Hospital of Xi'an Jiaotong University. We selected SGLT inhibitor users and nonusers [patients with type 2 diabetes (T2D) without SGLT2 inhibitor prescription] undergoing PCI. We determined CI-AKI by the ESUR (European Society of Urogenital Radiology, AKIESUR) and KDIGO definition (Kidney Disease: Improving Global Outcomes, AKIKDIGO). We performed 1:1 nearest-neighbor propensity matching and calculated unadjusted odds ratios (ORs) and adjusted ORs (aORs; accounting for covariates poorly balanced) for AKI in primary and sensitivity analyses. We compared the renal function indicators in users and nonusers at 24, 48, and 72 h post-PCI. Results: We identified 242 SGLT2 inhibitor users and 242 nonusers in the cohort. The unadjusted ORs of CI-AKIESUR were 63% lower in users [OR: 0.37 (95% CI: 0.18-0.68); P = 0.01], which was unchanged [aOR: 0.37 (95% CI: 0.19-0.67); P < 0.01] post adjustment. These estimates did not qualitatively change across several sensitivity analyses. There was no significant difference in urea nitrogen, creatinine, and estimated glomerular filtration rate (eGFR) values between the two groups before PCI, and at 24 h, while the creatinine (48 and 72 h post-PCI) and CyC (24 and 48 h post-PCI) were significantly lower than those in the nonuser group (P < 0.05). Conclusion: Our findings do not suggest an increased risk of CI-AKI associated with SGLT2 inhibitor use in patients with CAD and T2D undergoing PCI.

8.
Front Cell Dev Biol ; 10: 867438, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35547820

RESUMEN

The folding of two-dimensional epithelial sheets into specific three-dimensional structures is a fundamental tissue construction mechanism in animal development. A common mechanism that mediates epithelial folding is apical constriction, the active shrinking of cell apices driven by actomyosin contractions. It remains unclear whether cells outside of the constriction domain also contribute to folding. During Drosophila mesoderm invagination, ventrally localized mesoderm epithelium undergoes apical constriction and subsequently folds into a furrow. While the critical role of apical constriction in ventral furrow formation has been well demonstrated, it remains unclear whether, and if so, how the laterally localized ectodermal tissue adjacent to the mesoderm contributes to furrow invagination. In this study, we combine experimental and computational approaches to test the potential function of the ectoderm in mesoderm invagination. Through laser-mediated, targeted disruption of cell formation prior to gastrulation, we found that the presence of intact lateral ectoderm is important for the effective transition between apical constriction and furrow invagination in the mesoderm. In addition, using a laser-ablation approach widely used for probing tissue tension, we found that the lateral ectodermal tissues exhibit signatures of tissue compression when ablation was performed shortly before the onset of mesoderm invagination. These observations led to the hypothesis that in-plane compression from the surrounding ectoderm facilitates mesoderm invagination by triggering buckling of the mesoderm epithelium. In support of this notion, we show that the dynamics of tissue flow during mesoderm invagination displays characteristic of elastic buckling, and this tissue dynamics can be recapitulated by combining local apical constriction and global compression in a simulated elastic monolayer. We propose that Drosophila mesoderm invagination is achieved through epithelial buckling jointly mediated by apical constriction in the mesoderm and compression from the neighboring ectoderm.

9.
Elife ; 112022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35195065

RESUMEN

Apical constriction driven by actin and non-muscle myosin II (actomyosin) provides a well-conserved mechanism to mediate epithelial folding. It remains unclear how contractile forces near the apical surface of a cell sheet drive out-of-the-plane bending of the sheet and whether myosin contractility is required throughout folding. By optogenetic-mediated acute inhibition of actomyosin, we find that during Drosophila mesoderm invagination, actomyosin contractility is critical to prevent tissue relaxation during the early, 'priming' stage of folding but is dispensable for the actual folding step after the tissue passes through a stereotyped transitional configuration. This binary response suggests that Drosophila mesoderm is mechanically bistable during gastrulation. Computer modeling analysis demonstrates that the binary tissue response to actomyosin inhibition can be recapitulated in the simulated epithelium that undergoes buckling-like deformation jointly mediated by apical constriction in the mesoderm and in-plane compression generated by apicobasal shrinkage of the surrounding ectoderm. Interestingly, comparison between wild-type and snail mutants that fail to specify the mesoderm demonstrates that the lateral ectoderm undergoes apicobasal shrinkage during gastrulation independently of mesoderm invagination. We propose that Drosophila mesoderm invagination is achieved through an interplay between local apical constriction and mechanical bistability of the epithelium that facilitates epithelial buckling.


Asunto(s)
Proteínas de Drosophila , Drosophila , Actomiosina/metabolismo , Animales , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Epitelio/metabolismo , Mesodermo/metabolismo , Morfogénesis , Optogenética
10.
Pathol Res Pract ; 224: 153532, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34214844

RESUMEN

Due to lack of effective biomarkers for early diagnosis, most patients are diagnosed with advanced gastric cancer and have lower survival rates. 5-Fluorouracil (5-FU) is one of commonly used drugs for chemotherapy of gastric cancer, but drug resistance limits the wide application of agents. Retinoblastoma tumor suppressor gene 1 (RB1) is a key regulator in the progression of various human cancers, including gastric cancer. However, the effects of RB1 on chemosensitivity and the underlying mechanisms in gastric cancer (GC) are not clear. In this study, expressions of RB1 in GC cell lines were evaluated by RT-qPCR and western blot assay. CCK-8 was applied to examine the effect of 5-FU on cell viability. Meanwhile, IC50 values were calculated. The drug-resistance protein MDR1 and autophagy-related proteins were detected by western blot assay. Flow cytometry was used to detect cell cycle. The results showed that RB1 expressions were downregulated in GC cell lines and had significant differences between 5-FU resistance cell lines (SNU-620/5-FU and NUGC-3/5-FU) and non-resistance cell lines (SNU-620 and NUGC-3). Overexpression of RB1 enhanced 5-FU sensitivity of GC cells and caused cell cycle arrest in the S phase. Meanwhile, autophagy-related proteins were downregulated. Mechanistically, SDF-1/CXCR4 participated in the regulation of RB1 on cell autophagy. Autophagy activator, SDF-1 treatment and CXCR4 activation reversed the promoted effects of RB1 on 5-FU sensitivity in GC cells. In conclusion, our data revealed that RB1 was downregulated in GC cell lines. RB1 overexpression enhanced 5-FU chemosensitivity in GC cells by regulating cell autophagy via SDF-1/CXCR4 pathway. RB1 might serve as a promising therapeutic target of GC.


Asunto(s)
Autofagia/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Receptores CXCR4/metabolismo , Retinoblastoma/genética , Neoplasias Gástricas/patología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Quimiocina CXCL12/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Fluorouracilo/farmacología , Humanos , Neoplasias de la Retina/genética , Neoplasias de la Retina/patología , Retinoblastoma/patología , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/tratamiento farmacológico
11.
Life Sci ; 257: 118100, 2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32679149

RESUMEN

AIM: Nexrutine, an herbal extract of Phellodendron amurense, has been found to play a tumor-suppressive role in many cancers. However, its role in the pathogenesis of gastric cancer remains unclear. MATERIALS AND METHODS: Gastric cancer cells (SGC-7901 and MGC-803) were treated with nexrutine, and cell proliferation, invasion and apoptosis were analyzed. And the MGC-803 cells-derived xenograft mouse models were fed pelleted diet containing 600 mg/kg nexrutine for 21 days after inoculation. Mechanically, we focused on the influences of nexrutine on the levels and activation of STAT3 and NF-κB as well as their upstream regulator FAK. Additionally, we further verified whether nexrutine affected the proliferation, invasion and apoptosis of gastric cancer cells via FAK by upregulating FAK expression before nexrutine treatment. KEY FINDINGS: We found nexrutine inhibited the viability, invasion, and expression levels of PCNA, CyclinD1 and Bcl-2, promoted the apoptosis and Bax expression, decreased levels of STAT3, phospho-STAT3, NF-κB p65, phospho-p65, FAK and phospho-FAK in gastric cancer cells. Overexpression of FAK reversed the impacts of nexrutine on the levels of STAT3, phospho-STAT3, NF-κB p65, phospho-p65, as well as the malignant characteristics of gastric cancer cells. Moreover, nexrutine suppressed tumor volumes and weights, and decreased expression and phosphorylation of FAK, STAT3 and NF-κB p65 in vivo. SIGNIFICANCE: Nexrutine inhibited the malignant progression of gastric cancer via negatively regulating STAT3/NF-κB signaling pathway by suppressing FAK expression and activation.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Quinasa 1 de Adhesión Focal/genética , Extractos Vegetales/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , FN-kappa B/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Cell Physiol ; 235(5): 4709-4719, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31650535

RESUMEN

Studies have found that miR-665 acted as a tumor suppressor or an oncogene in different malignancies. miR-665 expression was elevated in gastric adenocarcinoma tissues; however, its role and mechanism in this disease are not fully clarified. The expression of miR-665 and its target gene was detected in human gastric adenocarcinoma tissues and cells. Moreover, we analyzed the effects of miR-665 on the proliferation, migration, and epithelial-mesenchymal transition (EMT) of gastric adenocarcinoma cells as well as tumor growth in vivo. The mechanisms of miR-665 in gastric adenocarcinoma were investigated by using molecular biology techniques. We found miR-665 was upregulated and suppressor of cytokine signaling 3 (SOCS3) was downregulated in gastric adenocarcinoma tissues and cells. Elevated miR-665 was positively correlated with tumor size, lymph node metastasis, invasion depth, TNM stage, and poor differentiation in gastric adenocarcinoma patients. Overexpression of miR-665 promoted, whereas knockdown of miR-665 suppressed the proliferation, migration, and EMT of gastric adenocarcinoma cells. Furthermore, we demonstrated that miR-665 functioned through targeting SOCS3, followed by activation of the FAK/Src signaling pathway in gastric adenocarcinoma cells. miR-665 antagomir inhibited tumor growth as well as the activation of the FAK/Src pathway but increased SOCS3 expression in nude mice. In addition, miR-665 expression was negatively regulated by long noncoding RNA maternally expressed gene 3 (MEG3). In conclusion, miR-665 acted as an oncogene in gastric adenocarcinoma by inhibiting SOCS3 followed by activation of the FAK/Src pathway and it was negatively modulated by MEG3. miR-665 may be a promising therapeutic target for the treatment of gastric adenocarcinoma.


Asunto(s)
Adenocarcinoma/enzimología , Quinasa 1 de Adhesión Focal/metabolismo , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Neoplasias Gástricas/enzimología , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Activación Enzimática , Transición Epitelial-Mesenquimal , Femenino , Quinasa 1 de Adhesión Focal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones Desnudos , MicroARNs/genética , Persona de Mediana Edad , ARN Largo no Codificante/genética , Transducción de Señal , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Proteína 3 Supresora de la Señalización de Citocinas/genética , Carga Tumoral
13.
Gut ; 68(10): 1751-1763, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30635407

RESUMEN

BACKGROUND AND AIMS: Gastric intestinal metaplasia (IM) is common in the gastric epithelium of patients with chronic atrophic gastritis. CDX2 activation in IM is driven by reflux of bile acids and following chronic inflammation. But the mechanism underlying how bile acids activate CDX2 in gastric epithelium has not been fully explored. METHODS: We performed microRNA (miRNA) and messenger RNA (mRNA) profiling using microarray in cells treated with bile acids. Data integration of the miRNA/mRNA profiles with gene ontology (GO) analysis and bioinformatics was performed to detect potential miRNA-mRNA regulatory circuits. Transfection of gastric cancer cell lines with miRNA mimics and inhibitors was used to evaluate their effects on the expression of candidate targets and functions. Immunohistochemistry and in situhybridisation were used to detect the expression of selected miRNAs and their targets in IM tissue microarrays. RESULTS: We demonstrate a bile acids-triggered pathway involving upregulation of miR-92a-1-5p and suppression of its target FOXD1 in gastric cells. We first found that miR-92a-1-5p was increased in IM tissues and induced by bile acids. Moreover, miR-92a-1-5p was found to activate CDX2 and downstream intestinal markers by targeting FOXD1/FOXJ1 axis and modulating activation of nuclear factor kappa B (NF-κB) pathway. Furthermore, these effects were found to be clinical relevant, as high miR-92a-1-5p levels were correlated with low FOXD1 levels and high CDX2 levels in IM tissues. CONCLUSION: These findings suggest a miR-92a-1-5p/FOXD1/NF-κB/CDX2 regulatory axis plays key roles in the generation of IM phenotype from gastric cells. Suppression of miR-92a-1-5p and restoration of FOXD1 may be a preventive approach for gastric IM in patients with bile regurgitation.


Asunto(s)
Factores de Transcripción Forkhead/genética , Mucosa Gástrica/patología , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Neoplasias Gástricas/genética , Ácidos y Sales Biliares/efectos adversos , Línea Celular Tumoral , Factores de Transcripción Forkhead/metabolismo , Mucosa Gástrica/metabolismo , Humanos , Inmunohistoquímica , Hibridación in Situ , Metaplasia/genética , Metaplasia/metabolismo , Metaplasia/patología , MicroARNs/biosíntesis , ARN Neoplásico/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Regulación hacia Arriba
14.
Plant Cell Physiol ; 59(5): 997-1005, 2018 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-29444306

RESUMEN

Plant ammonium transporters in the AMT/MEP/Rh (ammonium transporter/methylammonium and ammonium permease/Rhesus factor) superfamily have only been previously characterized in flowering plants (angiosperms). Plant AMT1s are electrogenic, while plant AMT2s are electroneutral, and MEP and Rh transporters in other organisms are electroneutral. We analyzed the transport function of MpAMT1;2 from the basal land plant Marchantia polymorpha, a liverwort. MpAMT1;2 was shown to localize to the plasma membrane in Marchantia gametophyte thallus by stable transformation using a C-terminal citrine fusion. MpAMT1;2 expression was studied using quantitative real-time PCR and shown to be higher when plants were N deficient and lower when plants were grown on media containing ammonium, nitrate or the amino acid glutamine. Expression in Xenopus oocytes and analysis by electrophysiology revealed that MpAMT1;2 is an electrogenic ammonium transporter with a very high affinity for ammonium (7 µM at pH 5.6 and a membrane potential of -137 mV). A conserved inhibitory phosphorylation site identified in angiosperm AMT1s is also present in all AMT1s in Marchantia. Here we show that a phosphomimetic mutation T475D in MpAMT1;2 completely inhibits ammonium transport activity. The results indicate that MpAMT1;2 may be important for ammonium uptake into cells in the Marchantia thallus.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Membrana Celular/metabolismo , Marchantia/metabolismo , Proteínas de Plantas/metabolismo , Secuencia de Aminoácidos , Compuestos de Amonio/metabolismo , Proteínas de Transporte de Catión/química , Proteínas de Transporte de Catión/genética , Regulación de la Expresión Génica de las Plantas , Concentración de Iones de Hidrógeno , Cinética , Potenciales de la Membrana , Mutación/genética , Fosforilación , Proteínas de Plantas/química , Proteínas de Plantas/genética , Transporte de Proteínas , Especificidad por Sustrato
15.
Int J Oncol ; 50(6): 2091-2100, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28498440

RESUMEN

Multidrug resistance (MDR) is one of the major reasons for the failure of chemotherapy-based gastric carcinoma (GC) treatments, hence, biologically based therapies are urgently needed. Gastrin (GAS), a key gastrointestinal (GI) hormone, was found to be involved in tumor formation, progression, and metastasis. In this study, quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemical staining analysis revealed a high level of expression of GAS in drug-insensitive GC tissues (P<0.01) and similar results were revealed in GC cell lines SGC7901 and its multidrug-resistant variants SGC7901/VCR and SGC7901/ADR. We constructed a eukaryotic expression vector pCDNA3.1(+)/GAS for GAS overexpression and recombinant lentiviral vectors for specific siRNA (siGAS). Transfection of pCDNA3.1(+)/GAS increased (P<0.05) while transfection of siGAS (P<0.05) and co-treated with paclitaxel (TAX) and vincristine (VCR) combination (TAX-VCR) decreased (P<0.01) the cell viability of SGC7901, SGC7901/VCR and SGC7901/ADR. Apoptosis rates of SGC7901/VCR and SGC7901/ADR were reduced by pCDNA3.1(+)/GAS and increased by siGAS (P<0.05). The apoptosis rates of SGC7901/VCR, SGC7901/ADR and SGC7901 were all upregulated (P<0.01) when cells were co-treated with a combination of siGAS and TAX-VCR. Additionally, siGAS significantly downregulated the expression of Bcl-2 and multidrug-resistant associate protein (MRP1) and P-glycoprotein (Pgp) (P<0.05) in SGC7901/VCR and SGC7901/ADR cells. Moreover, GAS overexpression in SGC7901 cells significantly inhibited p27Kip1 expression but increased phosphorylation levels of p27Kip1 on Thr (187) and Ser (10) sites (P<0.05), as well as increasing nuclear accumulation of S-phase kinase-associated protein 2 (Skp2) and cytoplasmic accumulation of the Kip1 ubiquitination-promoting complex (KPC) (P<0.05). Silencing of Skp2 blocked the promoting effects of pCDNA3.1(+)/GAS on viability, the expression of MRP1 and Pgp and the inhibitory effects of pCDNA3.1(+)/GAS on apoptosis. In conclusion, we suggest that GAS contributes to the emergence of MDR of SGC7901 cells via the degradation of p27Kip1.


Asunto(s)
Carcinoma/tratamiento farmacológico , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Gastrinas/genética , Neoplasias Gástricas/tratamiento farmacológico , Anciano , Camptotecina/administración & dosificación , Camptotecina/análogos & derivados , Carcinoma/genética , Carcinoma/patología , Línea Celular Tumoral , Resistencia a Múltiples Medicamentos/genética , Resistencia a Antineoplásicos/genética , Femenino , Fluorouracilo/administración & dosificación , Regulación Neoplásica de la Expresión Génica , Humanos , Irinotecán , Masculino , Persona de Mediana Edad , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Fosforilación , Proteolisis , ARN Interferente Pequeño/genética , Proteínas Quinasas Asociadas a Fase-S/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología
16.
Cancer Res ; 77(5): 1227-1240, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28011625

RESUMEN

Molecular links between inflammation and cancer remain obscure despite their great pathogenic significance. The JAK2/STAT3 pathway activated by IL6 and other proinflammatory cytokines has garnered attention as a pivotal link in cancer pathogenesis, but the basis for its activation in cancer cells is not understood. Here we report that an IL6-triggered feedback loop involving STAT3-mediated suppression of miR-520d-5p and upregulation of its downstream target cyclophilin B (CypB) regulate the growth and survival of gastric cancer cells. In clinical specimens of gastric cancer, we documented increased expression of CypB and activation of STAT3. Mechanistic investigations identified miR-520d-5p as a regulator of CypB mRNA levels. This signaling axis regulated gastric cancer growth by modulating phosphorylation of STAT3. Furthermore, miR-520d-5p was identified as a direct STAT3 target and IL6-mediated inhibition of miR-520d-5p relied upon STAT3 activity. Our findings define a positive feedback loop that drives gastric carcinogenesis as influenced by H. pylori infections that involve proinflammatory IL6 stimulation. Cancer Res; 77(5); 1227-40. ©2016 AACR.


Asunto(s)
Ciclofilinas/metabolismo , MicroARNs/metabolismo , Factor de Transcripción STAT3/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Humanos , Transducción de Señal , Neoplasias Gástricas/genética , Transfección
17.
Mol Cancer ; 13: 263, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25496021

RESUMEN

BACKGROUND: Loss of cell-cell adhesion is important for the development of cancer invasion and metastasis. Vinculin, a key adhesion-related protein, can affect metastasis and prognosis in several tumours. Here, we determined the biological roles of vinculin in the metastasis of colorectal cancer (CRC) and evaluated its clinical significance as a potential disease biomarker. METHODS: The expression level of vinculin in CRC cell lines and tissues was measured using Real-Time PCR and western blotting. Moreover, vinculin function was analysed using Transwell assays and in vivo metastasis assays in gain- and loss-of-function experiments. Furthermore, the impact of vinculin together with membrane-bound ß-catenin on the prognosis of 228 CRC patients was investigated by immunohistochemistry. Additionally, the expression of epithelial-mesenchymal transition (EMT) indicators was verified by immunohistochemistry in CRC tissues obtained from these patients. RESULT: Vinculin expression was found to be significantly downregulated in highly metastatic CRC cell lines and metastatic tissues. Both in vitro and in vivo experiments showed that vinculin suppressed invasion, migration and metastasis in CRC cells and that this suppression could be attenuated by silencing ß-catenin. Moreover, the expression of vinculin and membrane-bound ß-catenin were positively correlated in CRC tissues, and lack of vinculin expression emerged as an independent prognostic factor in patients with CRC. Finally, the loss of vinculin and membrane-bound ß-catenin was associated with node metastasis, organ metastasis and expression of EMT indicators. CONCLUSION: Our results suggest that vinculin may play specific roles in the EMT and metastasis of CRC and that loss of vinculin could be used as a prognostic factor for CRC.


Asunto(s)
Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/metabolismo , Metástasis de la Neoplasia/patología , Vinculina/metabolismo , beta Catenina/metabolismo , Animales , Células CACO-2 , Línea Celular Tumoral , Movimiento Celular/fisiología , Neoplasias Colorrectales/patología , Regulación hacia Abajo , Transición Epitelial-Mesenquimal/fisiología , Femenino , Células HCT116 , Células HT29 , Humanos , Ratones , Ratones Desnudos , Persona de Mediana Edad , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA