Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Brain Pathol ; 34(1): e13212, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37721122

RESUMEN

Sonic Hedgehog (SHH) subgroup of medulloblastoma (MB) accounts for about 25% of all subgroups of MB. Tumor microenvironment (TME) may play a key role in the tumor progression and therapeutic resistance. Tumor-associated astrocytes (TAAs) are reshaped to drive tumor progression through multiple paracrine signals. However, the mechanism by which TAAs modulate MB cells remains elusive. Here, we illuminated that TAAs showed a specific and dynamic pattern during SHH-MB development. Most TAAs gathered to the tumor margin during the tumor progression, rather than evenly distributed in the early-stage tumors. We further demonstrated that lipocalin-2 (LCN2) secreted by TAAs could promote the tumor growth and was correlated with the poor prognosis of MB patients. Knocking down LCN2 in TAAs in vitro impeded the proliferation and migration abilities of MB cells. In addition, we identified that TAAs accelerated the tumor growth by secreting LCN2 via STAT3 signaling pathway. Accordingly, blockade of STAT3 signaling by its inhibitor WP1066 and AAV-Lcn2 shRNA, respectively, in TAAs abrogated the effects of LCN2 on tumor progression in vitro and in vivo. In summary, we for the first time clarified that LCN2, secreted by TAAs, could promote MB tumor progression via STAT3 pathway and has potential prognostic value. Our findings unveiled a new sight in reprogramming the TME of SHH-MB and provided a potential therapeutic strategy targeting TAAs.


Asunto(s)
Neoplasias Cerebelosas , Lipocalina 2 , Meduloblastoma , Humanos , Astrocitos/patología , Neoplasias Cerebelosas/patología , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/uso terapéutico , Lipocalina 2/genética , Lipocalina 2/metabolismo , Meduloblastoma/genética , Meduloblastoma/patología , Microambiente Tumoral
2.
Membranes (Basel) ; 13(1)2023 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-36676885

RESUMEN

Reverse osmosis (RO) membranes are susceptible to silica scaling, resulting in irreversible degradation of membrane performance. This work covered the fabrication of MIL-101(Fe) for silicic acid adsorption to alleviate the silica scaling of RO membranes. The effect of pH, mixing time and initial concentration on silicic acid adsorption of MIL-101(Fe) was appraised in detail. The adsorption experiments demonstrated that MIL-101(Fe) possessed an excellent adsorption ability for silicic acid with the maximum adsorption capacity reaching 220.1 mgSiO2·g-1. Data fitting confirmed the pseudo-second-order equation and Freundlich equation were consistent with silicic acid adsorption on MIL-101(Fe). Finally, a simulated anti-scaling experiment was carried out using a feed solution pretreated by MIL-101(Fe) adsorption, and the permeance exhibited a much lower decline after 24 h filtration, confirming that MIL-101(Fe) exhibits an excellent application potential for silica-scale mitigation in RO systems.

3.
Sci Adv ; 8(23): eabn3509, 2022 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-35687692

RESUMEN

Most genome-wide association study (GWAS)-identified breast cancer-associated causal variants remain uncharacterized. To provide a framework of understanding GWAS-identified variants to function, we performed a comprehensive study of noncoding regulatory variants at the NTN4 locus (12q22) and NTN4 gene in breast cancer etiology. We find that rs11836367 is the more likely causal variant, disrupting enhancer activity in both enhancer reporter assays and endogenous genome editing experiments. The protective T allele of rs11837367 increases the binding of GATA3 to the distal enhancer and up-regulates NTN4 expression. In addition, we demonstrate that loss of NTN4 gene in mice leads to tumor earlier onset, progression, and metastasis. We discover that NTN4, as a tumor suppressor, can attenuate the Wnt signaling pathway by directly binding to Wnt ligands. Our findings bridge the gaps among breast cancer-associated single-nucleotide polymorphisms, transcriptional regulation of NTN4, and breast cancer biology, which provides previously unidentified insights into breast cancer prediction and prevention.


Asunto(s)
Estudio de Asociación del Genoma Completo , Neoplasias , Netrinas/metabolismo , Alelos , Animales , Predisposición Genética a la Enfermedad , Ratones , Neoplasias/genética , Netrinas/genética , Polimorfismo de Nucleótido Simple , Vía de Señalización Wnt/genética
4.
Cancer Sci ; 113(7): 2457-2471, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35441763

RESUMEN

Our previous works have indicated that extracellular ATP is an important prometastasis factor. However, the molecular mechanism involved needs to be further studied. We demonstrated that extracellular ATP treatment could upregulate the expression of connective tissue growth factor (CTGF) in both triple-negative breast cancer (TNBC) cells and endothelial cells (ECs). Extracellular ATP stimulated the migration of TNBC cells and ECs, and angiogenesis of ECs via the P2Y2--YAP-CTGF axis. Furthermore, we demonstrated that adenosine triphosphate (ATP) stimulated TNBC cell adhesion to ECs and transmigration through the EC layer via CTGF by upregulation of integrin ß1 on TNBC cells and VCAM-1 on ECs. Both apyrase (ATP-diphosphohydrolase) and CTGF shRNA treatments could inhibit the metastasis of inoculated tumors to lung and liver in a mouse model, and these treated tumors had fewer blood vessels. Collectively, our data indicated that extracellular ATP promotes tumor angiogenesis and the interactions between TNBC cells and ECs through upregulation of CTGF, thereby stimulating TNBC metastasis. The pleiotropic effects of ATP in angiogenesis and cell adhesion suggest that extracellular ATP or CTGF could be an effective target for TNBC therapy.


Asunto(s)
Adenosina Trifosfato , Factor de Crecimiento del Tejido Conjuntivo , Neoplasias de la Mama Triple Negativas , Adenosina Trifosfato/metabolismo , Animales , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Células Endoteliales/metabolismo , Humanos , Ratones , Neovascularización Patológica/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Regulación hacia Arriba
5.
Cell Death Dis ; 13(3): 199, 2022 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-35236823

RESUMEN

We have previously demonstrated that extracellular adenosine 5'-triphosphate (ATP) promotes breast cancer cell chemoresistance. However, the underlying mechanism remains unclear. Using a cDNA microarray, we demonstrated that extracellular ATP can stimulate hypoxia-inducible factor (HIF) signaling. In this study, we report that hypoxia-inducible factor 1α (HIF-1α) was upregulated after ATP treatment and mediated the ATP-driven chemoresistance process. We aimed to investigate the mechanisms and identify potential clinically relevant targets that are involved. Using mass spectrometry, we found that aldolase A (ALDOA) interacts with HIF-1α and increases HIF-1α expression. We then demonstrated that STAT3-ALDOA mediates ATP-HIF-1α signaling and upregulates the HIF-1 target genes adrenomedullin (ADM) and phosphoinositide-dependent kinase-1 (PDK1). Moreover, we show that PI3K/AKT acts upstream of HIF-1α in ATP signaling and contributes to chemoresistance in breast cancer cells. In addition, HIF-1α-knockdown or treatment with direct HIF inhibitors combined with the ATP hydrolase apyrase in MDA-MB-231 cells induced enhanced drug sensitivity in nude BALB/c mice. We then used in vitro spheroid formation assays to demonstrate the significance of ATP-HIF-1α in mediating chemoresistance. Furthermore, considering that indirect HIF inhibitors are effective in clinical cancer therapy, we treated tumor-bearing BALB/c mice with STAT3 and PI3K/AKT inhibitors and found that the dual-targeting strategy sensitized breast cancer to cisplatin. Finally, using breast cancer tissue microarrays, we found that ATP-HIF-1α signaling is associated with cancer progression, poor prognosis, and resistance to chemotherapy. Taken together, we suggest that HIF-1α signaling is vital in ATP-driven chemoresistance and may serve as a potential target for breast cancer therapies.


Asunto(s)
Neoplasias de la Mama , Adenosina Trifosfato/metabolismo , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Humanos , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/metabolismo
6.
Sci China Life Sci ; 64(4): 563-574, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32737855

RESUMEN

Slug, a member of the Snail family of transcriptional repressors, plays a key role in cancer progression, cellular plasticity, and epithelial to mesenchymal transition (EMT). Slug is a fast-turnover protein and its stability is controlled by post-translational modifications. Here, we identified that Slug is acetylated by acetyltransferase CREB-binding protein (CBP) in breast cancer cells. CBP directly interacts with the C-terminal domain of Slug through its catalytic histone acetyltransferase (HAT) domain, leading to acetylation of Slug at lysines 166 and 211. Analysis with acetylation-specific antibodies revealed that Slug is highly acetylated in metastatic breast cancer cells. Notably, Slug acetylation, mediated by CBP at lysines 166 and 211, doubles its half-life and increases its stability. Further, acetylated Slug downregulates the expression of E-cadherin, the epithelial marker, and upregulates the expression of N-cadherin and vimentin, thereby promoting breast cancer cell migration. In conclusion, the present study demonstrates that CBP-mediated Slug acetylation increases its stability, promoting EMT and migration of breast cancer cells.


Asunto(s)
Neoplasias de la Mama/genética , Proteína de Unión a CREB/genética , Movimiento Celular/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción de la Familia Snail/genética , Acetilación , Secuencia de Aminoácidos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteína de Unión a CREB/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Células MCF-7 , Estabilidad Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Factores de Transcripción de la Familia Snail/metabolismo
7.
Breast Cancer Res Treat ; 184(1): 237-248, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32757134

RESUMEN

PURPOSE: This study aimed to investigate whether genetic polymorphisms in TGFB1 contribute to breast cancer (BC) susceptibility, and explore the mechanism of action. METHODS: A total of 7 tagging SNPs (tSNPs) were genotyped in 1161 BC cases and 1337 age-matched controls among Chinese Han population. Bioinformatics analysis was used to predict functional SNP closely linked to tSNPs. Luciferase gene reporter assay was performed to determine the effect of genetic variants on promoter activity. DNA pull-down assay and mass spectrometry were used to identify the differentially binding proteins to genetic variants. RESULTS: Genotyping analysis showed that rs1800469 (C>T) in the 5' regulatory region of TGFB1 was associated with reduced BC risk. Bioinformatics analysis predicted that rs11466313 (-2389_-2391 Del/AGG) in the 5' regulatory region of TGFB1, was closely linked to tSNP rs1800469 and could be functional. The genotyping of rs11466313 by PCR-SSCP showed that rs11466313 also conferred decreased BC risk. Luciferase assays demonstrated that rs11466313 minor allele reduced over ninefold of promoter activity compared with its major allele (p < 0.001). DNA pull-down assay and mass spectrometry revealed that rs11466313 minor allele lost the binding ability with FAM98B and HSP90B. Knocking down FAM98B but not HSP90B, the enhanced promoter activity driven by TGFB1 rs11466313 major allele was attenuated. CONCLUSIONS: This study elucidates the impact of functional polymorphism rs11466313 in the regulatory region of TGFB1 on breast cancer susceptibility and gene expression, and could be helpful for future research to determine the value of this TGFB1 variant in the clinical setting.


Asunto(s)
Neoplasias de la Mama , Alelos , Neoplasias de la Mama/genética , Estudios de Casos y Controles , Femenino , Predisposición Genética a la Enfermedad , Haplotipos , Humanos , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Factor de Crecimiento Transformador beta1/genética
8.
Oncogene ; 39(35): 5795-5810, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32724162

RESUMEN

Our previous research demonstrated that extracellular adenosine 5'-triphosphate (ATP) could promote breast cancer cell invasion. However, the impact of extracellular ATP on chemoresistance and the mechanisms behind ATP pro-invasion and pro-chemoresistance remain unclear. Here we aimed to determine the molecules or signaling pathways involved. cDNA microarray was performed to identify the differentially expressed genes before and after ATP treatment. As a result, Sex-determining region Y-box 9 (SOX9) was up-regulated after ATP treatment in breast cancer cells. In vitro invasion and migration assays demonstrated that knocking down SOX9 attenuated ATP-driven invasive capability. Mass spectrometry and co-IP revealed that SOX9 interacted with Janus kinase 1 (JAK1). Afterward, IL-6-JAK1-STAT3 signaling was demonstrated to promote SOX9 expression and invasion following ATP treatment. Notably, ATP-IL-6-SOX9 signaling was shown to stimulate chemoresistance in breast cancer cells. ChIP assays identified some potential SOX9 target genes, among which carcinoembryonic antigen-related cell adhesion molecule 5/6 (CEACAM5/6) was demonstrated to mediate ATP pro-invasive function, while ATP-binding cassette subfamily B member 1 (ABCB1) and ATP-binding cassette subfamily G member 2 (ABCG2) mediated ATP-driven chemoresistance. In addition, SOX9-knockdown and apyrase (an ATP hydrolase)-treated MDA-MB-231 cells illustrated decreased tumor growth and enhanced drug sensitivity in nude mice. In vitro spheroid formation assays also proved the significance of ATP-SOX9 in mediating chemoresistance. Moreover, molecules involved in ATP-SOX9 signaling were up-regulated in human breast carcinoma specimens and were associated with poor prognosis. Altogether, SOX9 signaling is vital in ATP-driven invasion and chemoresistance, which may serve as a potential target for breast cancer therapies.


Asunto(s)
Adenosina Trifosfato/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Factor de Transcripción SOX9/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Humanos , Transducción de Señal , Transfección
9.
RSC Adv ; 10(10): 5648-5655, 2020 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-35497469

RESUMEN

Thin film nanocomposite (TFN) reverse osmosis (RO) membranes were prepared by dispersing 3-aminopropyltriethoxysilane (APTES) modified hydrotalcite (HT), designated as A-HT, in aqueous solution and incorporating the nanoparticles in polyamide layers during the interfacial polymerization process. Results of Fourier transform infrared spectroscopy and zeta potential characterization showed the successful modification of nanoparticles by APTES. In addition, Fourier transform infrared spectroscopy suggested that amidation would take place between the aminosilane on APTES and trimesoyl chloride in organic solution, providing firm covalent interaction between the nanoparticles and polyamide matrix. Dynamic light scattering and transmission electron microscopy indicated that aminosilane modification improved dispersibility of the nanoparticles in aqueous solution and obtained membranes, which suppressed the aggregation. Both the covalent interaction and aggregation suppression were beneficial to compatibility between nanoparticles and the polyamide matrix. TFN RO membranes incorporated with A-HT demonstrated excellent performance. Compared with the pristine RO membrane, the water flux of A-HT-0.050 prepared with an optimum A-HT concentration of 0.050 wt% was enhanced by 18.6% without sacrificing the salt rejection. Moreover, the selectivity of A-HT-0.050 was superior to that of HT-0.050 prepared with HT of 0.050 wt%, which proved aminosilane modification of hydrotalcite was beneficial to high membrane performance especially to selectivity.

10.
Cancer Sci ; 110(8): 2456-2470, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31148343

RESUMEN

Extracellular ATP has been shown to play an important role in invasion and the epithelial-mesenchymal transition (EMT) process in breast cancer; however, the mechanism is unclear. Here, by using a cDNA microarray, we demonstrated that extracellular ATP could stimulate hypoxia-inducible factor (HIF) signaling and upregulate hypoxia-inducible factor 1/2α (HIF-1/2α) expression. After knocking down HIF-1/2α using siRNA, we found that ATP-driven invasion and EMT were significantly attenuated via HIF2A-siRNA in breast cancer cells. By using ChIP assays, we revealed that the biological function of extracellular ATP in invasion and EMT process depended on HIF-2α direct targets, among which lysyl oxidase-like 2 (LOXL2) and matrix metalloproteinase-9 (MMP-9) mediated ATP-driven invasion, and E-cadherin and Snail mediated ATP-driven EMT, respectively. In addition, using silver staining and mass spectrometry, we found that phosphoglycerate kinase 1 (PGK1) could interact with HIF-2α and mediate ATP-driven HIF-2α upregulation. Furthermore, we demonstrated that expressions of HIF-2α and its target proteins could be regulated via ATP by AKT-PGK1 pathway. Using a Balb/c mice model, we illustrated the function of HIF-2α in promoting tumor growth and metastasis in vivo. Moreover, by exploring online databases, we found that molecules involved in ATP-HIF-2α signaling were highly expressed in human breast carcinoma tissues and were associated with poor prognosis. Altogether, these findings suggest that extracellular ATP could promote breast carcinoma invasion and EMT via HIF-2α signaling, which may be a potential target for future anti-metastasis therapy.


Asunto(s)
Adenosina Trifosfato/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Transición Epitelial-Mesenquimal/fisiología , Hipoxia/patología , Invasividad Neoplásica/patología , Aminoácido Oxidorreductasas/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Células MCF-7 , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
11.
Oncol Lett ; 16(5): 6868-6878, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30405831

RESUMEN

The cadherin 1 (CDH1) gene plays critical roles in the epithelial-mesenchymal transition process, potentially offering us a glimpse into the development of endometrial carcinoma (EC). The present study aimed to identify whether genetic variants in CDH1 affect EC susceptibility in Chinese Han women, using a strategy combining haplotype-tagging single nucleotide polymorphisms (htSNPs) association analysis with fine-scale mapping. A total of 9 htSNPs in CDH1 were genotyped among 516 cases and 706 age-matched cancer-free controls. Logistic regression analyses revealed 3 htSNPs (rs17715799, rs6499199 and rs13689) to be associated with increased EC risk and 3 htSNPs (rs12185157, rs10431923 and rs4783689) with decreased EC risk. Furthermore, 14 newly imputed SNPs of CDH1 were identified to be associated with EC risk (P<0.05) using genotype imputation analysis. Notably, multivariate logistic analysis demonstrated that rs13689, rs10431923 and rs10431924 could affect EC susceptibility independently (P≤0.001). Subsequent Generalized Multifactor Dimensionality Reduction analysis revealed several best fitting models for predicting EC risk, including SNP-SNP interactions among rs7100190, rs12185157, rs10431923, rs7186053, rs6499199, rs4783689, rs13689, rs6499197 and rs10431924, and SNP-environment interactions between related SNPs and number of childbirth. Moreover, functional annotations suggest that the majority of these susceptible variants may carry potential biological functions that affect certain gene regulatory elements. In summary, this study suggested that the genetic polymorphisms of CDH1 were indeed associated with EC susceptibility on several levels. If further additional functional studies could verify these findings, these genetic variants may serve as future personalized markers for the early prediction of endometrial cancer in Chinese Han women.

12.
Oncol Rep ; 40(6): 3573-3584, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30272327

RESUMEN

Extensive evidence suggests that the genetic etiologies of breast cancer (BC) and ovarian cancer (OC) show a certain degree of similarity. This study aimed to find out whether the single nucleotide polymorphisms (SNPs) of genes SNAI1 and TWIST1 may affect BC and OC susceptibility. A total of 7 tagging­SNPs (tSNPs) were directly genotyped in 1,161 BC cases, 286 OC cases and 1,273 cancer­free controls among Chinese Han women. Twenty­eight variants in these 2 genes were genotyped by 'in silico' genotype imputation. Logistic regression (LR) revealed that tSNPs SNAI1 rs6125849, TWIST1 rs4721746 and TWIST1 rs4721745 were protective genetic variants for BC/OC. Allelic association tests of gene­wide SNPs demonstrated that the minor alleles of SNAI1 rs6125849, TWIST1 rs4721745 and TWIST1 rs11973396 were strongly associated with BC/OC susceptibility. Multivariate LR presented that SNAI1 rs6125849, TWIST1 rs4721745, rs4721746 and rs11973396 affected BC/OC susceptibility independently, and women harboring all four protective genoytpes had the lowest risk. Multifactor dimensionality reduction analysis further showed that SNAI1 rs6125849 and TWIST1 rs4721745 had the strongest synergistic interaction. Functional annotation predicted that the minor alleles of SNAI1 rs6125849 and TWIST1 rs4721745 altered their binding affinities with transcription factors E2F6 and TCF11­MafG respectively. These results indicate that genetic variants in SNAI1 and TWIST1, most probably SNAI1 rs6125849 and TWIST1 rs4721745, may modulate BC and OC susceptibility.


Asunto(s)
Pueblo Asiatico/genética , Neoplasias de la Mama/genética , Proteínas Nucleares/genética , Neoplasias Ováricas/genética , Polimorfismo de Nucleótido Simple , Factores de Transcripción de la Familia Snail/genética , Proteína 1 Relacionada con Twist/genética , Adulto , Pueblo Asiatico/etnología , Estudios de Casos y Controles , China/etnología , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Persona de Mediana Edad , Proteínas Nucleares/metabolismo , Unión Proteica , Factores de Transcripción de la Familia Snail/metabolismo , Proteína 1 Relacionada con Twist/metabolismo
13.
Cancer Lett ; 430: 1-10, 2018 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-29733962

RESUMEN

Our previous work has demonstrated that extracellular ATP is an important pro-invasive factor, and in this study, we tapped into a possible mechanism involved. We discovered that ATP could upregulate both the intracellular expression and secretion of S100A4 in breast cancer cells and fibroblasts. Apart from stimulating breast cancer cell motility via intracellular S100A4, ATP enhanced the ability of breast cancer cells to transform fibroblasts into cancer-associated fibroblast (CAF)-like cells, which in turn secreted S100A4 to further promote cancer cell motility. Both apyrase and niclosamide treatments could inhibit metastasis of inoculated tumors to lung, liver and kidney in mice model, and CAFs from these treated tumors exhibited weakened migration-stimulating capacity for breast cancer cells. Collectively, our data indicate that extracellular ATP promotes the interactions between breast cancer cells and fibroblasts, which work collaboratively via production of S100A4 to exacerbate breast cancer metastasis.


Asunto(s)
Adenosina Trifosfato/metabolismo , Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/metabolismo , Movimiento Celular/efectos de los fármacos , Proteína de Unión al Calcio S100A4/metabolismo , Animales , Apirasa/farmacología , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , Niclosamida/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Cultivo Primario de Células , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Proteína de Unión al Calcio S100A4/genética , Regulación hacia Arriba
14.
RSC Adv ; 8(22): 12439-12448, 2018 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-35539378

RESUMEN

Thin film nanocomposite (TFN) reverse osmosis (RO) membranes were prepared by incorporating hydrotalcite (HT) in polyamide layer during interfacial polymerization process using two methods: (1) dispersing HT in aqueous solution directly; (2) preparing layered double oxide (LDO) via calcination of HT and then dispersing the obtained LDO in aqueous solution to reconstruct HT. The results demonstrated that TFN RO membranes exhibited higher water flux compared with the pristine RO membrane, which could be mainly attributed to water channels constructed by HT as well as the enhancement of hydrophilicity and the increase of relative surface area. In addition, TFN RO membranes prepared using LDO showed higher water flux than those prepared using HT, which is contributed to the uniform distribution of water channels in polyamide layer resulting from the process of calcination and reconstruction reaction. Compared with pristine RO membrane, water flux of TFN RO membranes prepared with 0.075 wt% HT and those prepared with 0.050 wt% LDO was enhanced by 19.1% and 22.8% without sacrificing the salt rejection.

15.
Oncol Rep ; 39(1): 138-150, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29115551

RESUMEN

Adenosine 5'-triphosphate (ATP) is one of the most abundant biochemical constituents within the tumor microenvironment and is postulated to play critical roles in the progression of a number of types of tumors via interaction with the P2Y2 receptor. In the present study, we demonstrated that the P2Y2 receptor was highly expressed in MCF7 and Hs578T breast cancer cells. Downregulation of the P2Y2 receptor by small interfering RNA (siRNA) significantly attenuated ATP- or UTP-driven migration and invasion of the breast cancer cells as well as expression of EMT-related genes Snail and E-cadherin. Consistent with the observations in vitro, the P2Y2 receptor was found to be abundantly expressed at the invasive edge of the tumor, in infiltrating tumor cells in breast adipose tissues and/or the cancer embolus in the lymphatic sinuses compared with the tumor core areas. Furthermore, high Snail expression and weak or negative expression of E-cadherin were observed at the invasive edge of tumors. Taken together, these data indicate that the P2Y2 receptor promoted cell migration and invasion in breast cancer cells via EMT-related genes Snail and E-cadherin.


Asunto(s)
Neoplasias de la Mama/patología , Cadherinas/metabolismo , Receptores Purinérgicos P2Y2/genética , Receptores Purinérgicos P2Y2/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Adulto , Anciano , Antígenos CD , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Cadherinas/genética , Línea Celular Tumoral , Movimiento Celular , Transición Epitelial-Mesenquimal , Femenino , Humanos , Células MCF-7 , Persona de Mediana Edad , Clasificación del Tumor , Invasividad Neoplásica , Estadificación de Neoplasias , Factores de Transcripción de la Familia Snail/genética , Regulación hacia Arriba , Adulto Joven
16.
Cancer Sci ; 108(7): 1318-1327, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28474758

RESUMEN

Extracellular adenosine 5'-triphosphate (ATP), secreted by living cancer cells or released by necrotic tumor cells, plays an important role in tumor invasion and metastasis. Our previous study demonstrated that ATP treatment in vitro could promote invasion in human prostate cancer cells via P2Y2, a preferred receptor for ATP, by enhancing EMT process. However, the pro-invasion mechanisms of ATP and P2Y2 are still poorly studied in breast cancer. In this study, we found that P2Y2 was highly expressed in breast cancer cells and associated with human breast cancer metastasis. ATP could promote the in vitro invasion of breast cancer cells and enhance the expression of ß-catenin as well as its downstream target genes CD44, c-Myc and cyclin D1, while P2Y2 knockdown attenuated above ATP-driven events in vitro and in vivo. Furthermore, iCRT14, a ß-catenin/TCF complex inhibitor, could also suppress ATP-driven migration and invasion in vitro. These results suggest that ATP promoted breast cancer cell invasion via P2Y2-ß-catenin axis. Thus blockade of the ATP-P2Y2-ß-catenin axis could suppress the invasive and metastatic potential of breast cancer cells and may serve as potential targets for therapeutic interventions of breast cancer.


Asunto(s)
Adenosina Trifosfato/metabolismo , Neoplasias de la Mama/patología , Invasividad Neoplásica/patología , Receptores Purinérgicos P2Y2/metabolismo , Transducción de Señal/fisiología , beta Catenina/metabolismo , Animales , Western Blotting , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Movimiento Celular/fisiología , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Reacción en Cadena en Tiempo Real de la Polimerasa
17.
PLoS One ; 11(5): e0155270, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27171242

RESUMEN

Endometrial cancer (EC) is a complex disease involving multiple gene-gene and gene-environment interactions. TGF-ß signaling plays pivotal roles in EC development. This study aimed to investigate whether the genetic polymorphisms of TGF-ß signaling related genes TGFB1, TGFBR1, SNAI1 and TWIST1 contribute to EC susceptibility. Using the TaqMan Genotyping Assay, 19 tagging-SNPs of these four genes were genotyped in 516 EC cases and 707 controls among Chinese Han women. Logistic regression (LR) showed that the genetic variants of TGFB1 rs1800469, TGFBR1 rs6478974 and rs10733710, TWIST1 rs4721745 were associated with decreased EC risk, and these four loci showed a dose-dependent effect (Ptrend < 0.0001). Classification and regression tree (CART) demonstrated that women carrying both the genotypes of TGFBR1 rs6478974 TT and rs10512263 TC/CC had the highest risk of EC (aOR = 7.86, 95% CI = 3.42-18.07, P<0.0001). Multifactor dimensionality reduction (MDR) revealed that TGFB1 rs1800469 plus TGFBR1 rs6478974 was the best interactional model to detect EC risk. LR, CART and MDR all revealed that TGFBR1 rs6478974 was the most important protective locus for EC. In haplotype association study, TGFBR1 haplotype CACGA carrier showed the lowest EC risk among women with longer menarche-first full term pregnancy intervals (˃11 years) and BMI˂24 (aOR = 0.39, 95% CI = 0.17-0.90, P = 0.0275). These results suggest that polymorphisms in TGFB1, TGFBR1, SNAI1 and TWIST1 may modulate EC susceptibility, both separately and corporately.


Asunto(s)
Pueblo Asiatico/genética , Neoplasias Endometriales/genética , Predisposición Genética a la Enfermedad , Proteínas Nucleares/genética , Polimorfismo de Nucleótido Simple/genética , Proteínas Serina-Treonina Quinasas/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Factores de Transcripción de la Familia Snail/genética , Factor de Crecimiento Transformador beta1/genética , Proteína 1 Relacionada con Twist/genética , Índice de Masa Corporal , Estudios de Casos y Controles , Epistasis Genética , Etnicidad/genética , Femenino , Genes Dominantes , Estudios de Asociación Genética , Haplotipos/genética , Humanos , Desequilibrio de Ligamiento/genética , Modelos Logísticos , Modelos Genéticos , Reducción de Dimensionalidad Multifactorial , Análisis Multivariante , Filogenia , Receptor Tipo I de Factor de Crecimiento Transformador beta , Factores de Riesgo
18.
PLoS One ; 10(8): e0135865, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26285011

RESUMEN

This study aims to investigate whether the germline variants in CDH1 and CTNNB1 would affect breast cancer susceptibility and patients' prognosis among Chinese Han women using a haplotype-based association analysis. We genotyped 12 haplotype-tagging single nucleotide polymorphisms (htSNPs) in CDH1 and CTNNB1 among 1,160 BC cases and 1,336 age-matched cancer-free controls using the TaqMan® Genotyping Assay. For association analyses of germline variants with breast cancer susceptibility, the results showed that rs7200690, rs7198799, rs17715799, rs13689 and diplotype CGC/TGC (rs7200690 + rs12185157 + rs7198799) in CDH1 as well as rs2293303 in CTNNB1 were associated with increased breast cancer risk. In addition, the Generalized Multifactor Dimensionality Reduction (GMDR) and logistic regression analysis predicted an interaction on breast cancer risk between rs17715799 and rs13689 as well as rs13689 and menarche-FFTP (First Full-Term Pregnancy) interval. For survival analyses, the results demonstrated that the minor allele homozygotes of rs13689 and haplotype TGC in CDH1 were linked with unfavorable event-free survival of breast cancer, whereas, rs4783689 of CDH1 showed the opposite effect under dominant model. Notably, the stratified analysis revealed that rs7186053 was associated with favorable event-free survival among patients with estrogen receptor (ER)-positive, progesterone receptor (PR)-positive or lymph node metastasis negative patients. Moreover, rs7200690 and rs7198799 in CDH1 as well as rs4533622 in CTNNB1 were associated with worse event-free survival among patients with clinical stage 0-I tumors. This study indicated that the genetic polymorphisms of CDH1 and CTNNB1 were associated with breast cancer susceptibility and patients' prognosis.


Asunto(s)
Pueblo Asiatico/etnología , Neoplasias de la Mama/genética , Cadherinas/genética , Etnicidad/genética , Predisposición Genética a la Enfermedad/genética , Polimorfismo de Nucleótido Simple , beta Catenina/genética , Antígenos CD , Pueblo Asiatico/genética , Neoplasias de la Mama/diagnóstico , Supervivencia sin Enfermedad , Femenino , Humanos
19.
PLoS One ; 10(7): e0133165, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26182292

RESUMEN

As one member of G protein-coupled P2Y receptors, P2Y2 receptor can be equally activated by extracellular ATP and UTP. Our previous studies have proved that activation of P2Y2 receptor by extracellular ATP could promote prostate cancer cell invasion and metastasis in vitro and in vivo via regulating the expressions of some epithelial-mesenchymal transition/invasion-related genes (including IL-8, E-cadherin, Snail and Claudin-1), and the most significant change in expression of IL-8 was observed after P2Y2 receptor activation. However, the signaling pathway downstream of P2Y2 receptor and the role of IL-8 in P2Y2-mediated prostate cancer cell invasion remain unclear. Here, we found that extracellular ATP/UTP induced activation of EGFR and ERK1/2. After knockdown of P2Y2 receptor, the ATP -stimulated phosphorylation of EGFR and ERK1/2 was significantly suppressed. Further experiments showed that inactivation of EGFR and ERK1/2 attenuated ATP-induced invasion and migration, and suppressed ATP-mediated IL-8 production. In addition, knockdown of IL-8 inhibited ATP-mediated invasion and migration of prostate cancer cells. These findings suggest that P2Y2 receptor and EGFR cooperate to upregulate IL-8 production via ERK1/2 pathway, thereby promoting prostate cancer cell invasion and migration. Thus blocking of the P2Y2-EGFR-ERK1/2 pathway may provide effective therapeutic interventions for prostate cancer.


Asunto(s)
Receptores ErbB/genética , Regulación Neoplásica de la Expresión Génica , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Próstata/metabolismo , Receptores Purinérgicos P2Y2/genética , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Línea Celular Tumoral , Movimiento Celular , Transición Epitelial-Mesenquimal , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Humanos , Interleucina-8/agonistas , Interleucina-8/antagonistas & inhibidores , Interleucina-8/genética , Interleucina-8/metabolismo , Masculino , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Próstata/efectos de los fármacos , Próstata/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptor Cross-Talk , Receptores Purinérgicos P2Y2/metabolismo , Transducción de Señal , Uridina Trifosfato/metabolismo , Uridina Trifosfato/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...