Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
FASEB J ; 38(1): e23388, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38145323

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of cells that differentiate from myeloid cells, proliferate in cancer and inflammatory reactions, and mainly exert immunosuppressive functions. Nonetheless, the precise mechanisms that dictate both the accumulation and function of MDSCs remain only partially elucidated. In the course of our investigation, we observed a positive correlation between the content of MDSCs especially G-MDSCs and miR-9 level in the tumor tissues derived from miR-9 knockout MMTV-PyMT mice and 4T1 tumor-bearing mice with miR-9 overexpression. Combined with RNA-seq analysis, we identified SOCS2 and SOCS3 as direct targets of miR-9. Additionally, our research unveiled the pivotal role of the CCL5/CCR5 axis in orchestrating the chemotactic recruitment of G-MDSCs within the tumor microenvironment, a process that is enhanced by miR-9. These findings provide fresh insights into the molecular mechanisms governing the accumulation of MDSCs within the framework of breast cancer development.


Asunto(s)
MicroARNs , Células Supresoras de Origen Mieloide , Neoplasias , Proteína 3 Supresora de la Señalización de Citocinas , Animales , Ratones , Línea Celular Tumoral , Proliferación Celular , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Células Supresoras de Origen Mieloide/patología , Neoplasias/patología , Microambiente Tumoral , Proteína 3 Supresora de la Señalización de Citocinas/genética
2.
J Transl Med ; 22(1): 832, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39256760

RESUMEN

BACKGROUND: The roles of the transcriptional factor SIX2 have been identified in several tumors. However, its roles in gastric cancer (GC) progression have not yet been revealed. Our objective is to explore the impact and underlying mechanisms of SIX2 on the stemness of GC cells. METHODS: Lentivirus infection was employed to establish stable expression SIX2 or PFN2 in GC cells. Gain- and loss-of-function experiments were conducted to detect changes of stemness markers, flow cytometry profiles, tumor spheroid formation, and tumor-initiating ability. ChIP, RNA-sequencing, tissue microarray, and bioinformatics analysis were performed to reveal the correlation between SIX2 and PFN2. The mechanisms underlying the SIX2/PFN2 loop-mediated effects were elucidated through tissue microarray analysis, RNA stability assay, IP-MS, Co-Immunoprecipitation, and inhibition of the JNK signaling pathway. RESULTS: The stemness of GC cells was enhanced by SIX2. Mechanistically, SIX2 directly bound to PFN2's promoter and promoted PFN2 activity. PFN2, in turn, promoted the mRNA stability of SIX2 by recruiting RNA binding protein YBX-1, subsequently activating the downstream MAPK/JNK pathway. CONCLUSION: This study unveils the roles of SIX2 in governing GC cell stemness, defining a novel SIX2/PFN2 regulatory loop responsible for this regulation. This suggests the potential of targeting the SIX2/PFN2 loop for GC treatment (Graphical Abstracts).


Asunto(s)
Retroalimentación Fisiológica , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio , Células Madre Neoplásicas , Profilinas , Neoplasias Gástricas , Neoplasias Gástricas/patología , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Humanos , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/metabolismo , Línea Celular Tumoral , Profilinas/metabolismo , Profilinas/genética , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Animales , Regiones Promotoras Genéticas/genética , Estabilidad del ARN/genética , Sistema de Señalización de MAP Quinasas , Unión Proteica
3.
Mol Cancer ; 22(1): 205, 2023 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-38093288

RESUMEN

BACKGROUND: Targeted drugs are not quite effective for prolonging the survival of patients with gastric cancer due to off-target effects as well as tumor immune escape mechanisms. Circular RNAs widely exist in tumor regions as biomarkers and can be developed as effective drug targets. METHODS: Western blot, QRT-PCR, fluorescence in situ hybridization, and flow cytometry were used to investigate the function of hsa_circ_0136666 in promoting the proliferation of gastric cancer cells. Tissue immunofluorescence, enzyme-linked immunosorbent assay (ELISA), as well as flow cytometric analysis, was conducted to explore the process of tumor immune evasion in tumor-bearing mice. The differences of circRNA expression in clinical samples were analyzed through tissue microarray FISH. The effect of siRNA on improving the efficacy of anti-PDL1 drugs and suppressing the immune microenvironment was evaluated by the coadministration model. RESULTS: We demonstrated that hsa_circ_0136666 was widely and highly expressed in gastric cancer tissues and cells. Functionally, hsa_circ_0136666 promoted gastric cancer tumor proliferation and tumor microenvironment formation, leading to tumorigenesis immune escape, and this effect was dependent on CD8 + T cells. Mechanistically, we confirmed that hsa_circ_0136666 competitively upregulated PRKDC expression by sponging miR-375-3p, regulating immune checkpoint proteins, prompting phosphorylation of PD-L1 to preventing its degradation, driving PD-L1 aggregation and suppressing immune function, thereby impairing cancer immune responses. In terms of application, we found that LNP-siRNA effectively improved anti-PDL1 drug efficacy and inhibited immune escape. CONCLUSION: Our results reveal an oncogenic role played by hsa_circ_0136666 in gastric cancer, driving PD-L1 phosphorylation via the miR-375/PRKDC signaling axis, prompting immune escape. This work proposes a completely new pathogenic mechanism of gastric cancer, uncovers a novel role for hsa_circ_0136666 as an immune target, and provides a rationale for enhancing the efficacy of anti-PD-L1 therapy for gastric cancer.


Asunto(s)
MicroARNs , Neoplasias Gástricas , Humanos , Animales , Ratones , Neoplasias Gástricas/genética , Escape del Tumor/genética , Fosforilación , Antígeno B7-H1/genética , Hibridación Fluorescente in Situ , MicroARNs/genética , ARN Interferente Pequeño , Proliferación Celular , Línea Celular Tumoral , Microambiente Tumoral , Proteína Quinasa Activada por ADN
4.
Cell Mol Life Sci ; 79(7): 360, 2022 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-35690642

RESUMEN

Breast cancer stem cells (BCSCs) are positively correlated with the metastasis, chemoresistance, and recurrence of breast cancer. However, there are still no drugs targeting BCSCs in clinical using for breast cancer treatment. Here, we tried to screen out small-molecule compounds targeting BCSCs from the phenazine library established by us before. We focused on the compounds without affecting cell viability and screened out three potential compounds (CPUL119, CPUL129, CPUL149) that can significantly attenuate the stemness of breast cancer cells, as evident by the decrease of stemness marker expression, CD44+/CD24- subpopulation, mammary spheroid-formation ability, and tumor-initiating capacity. Additionally, these compounds suppressed the metastatic ability of breast cancer cells in vitro and in vivo. Combined with the transcriptome sequencing analysis, ferroptosis was shown on the top of the most upregulated pathways by CPUL119, CPUL129, and CPUL149, respectively. Mechanistically, we found that these three compounds could trigger ferroptosis by accumulating and sequestering iron in lysosomes through interacting with iron, and by regulating the expression of proteins (IRP2, TfR1, ferritin) engaged in iron transport and storage. Furthermore, inhibition of ferroptosis rescued the suppression of these three compounds on breast cancer cell stemness. This study suggests that CPUL119, CPUL129, and CPUL149 can specifically inhibit the stemness of breast cancer cells through triggering ferroptosis and may be the potential compounds for breast cancer treatment.


Asunto(s)
Neoplasias de la Mama , Ferroptosis , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Hierro/metabolismo , Células Madre Neoplásicas/metabolismo , Fenazinas/metabolismo , Fenazinas/farmacología , Fenazinas/uso terapéutico
5.
Molecules ; 28(3)2023 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-36771081

RESUMEN

RING finger protein 168 (RNF168) is an E3 ubiquitin ligase with the RING finger domain. It is an important protein contributing to the DNA double-strand damage repair pathway. Recent studies have found that RNF168 is significantly implicated in the occurrence and development of various cancers. Additionally, RNF168 contributes to the drug resistance of tumor cells by enhancing their DNA repair ability or regulating the degradation of target proteins. This paper summarizes and prospects the research progress of the structure and main functions of RNF168, especially its roles and the underlying mechanisms in tumorigenesis.


Asunto(s)
Reparación del ADN , Ubiquitina-Proteína Ligasas , Humanos , Ubiquitina-Proteína Ligasas/metabolismo , Carcinogénesis/genética , Ubiquitinación , Daño del ADN
6.
Molecules ; 28(3)2023 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-36771004

RESUMEN

The protein ISG15 encoded by interferon-stimulated gene (ISG) 15 is the first identified member of the ubiquitin-like protein family and exists in the form of monomers and conjugated complexes. Like ubiquitin, ISG15 can mediate an ubiquitin-like modification by covalently modifying other proteins, known as ISGylation. There is growing evidence showing that both the free and conjugated ISG15 are involved in multiple key cellular processes, including autophagy, exosome secretion, DNA repair, immune regulation, and cancer occurrence and progression. In this review, we aim to further clarify the function of ISG15 and ISGylation in cancer, demonstrate the important relationship between ISG15/ISGylation and cancer, and emphasize new insights into the different roles of ISG15/ISGylation in cancer progression. This review may contribute to therapeutic intervention in cancer. However, due to the limitations of current research, the regulation of ISG15/ISGylation on cancer progression is not completely clear, thus further comprehensive and sufficient correlation studies are still needed.


Asunto(s)
Citocinas , Neoplasias , Humanos , Citocinas/metabolismo , Interferones , Ubiquitina/metabolismo , Ubiquitinas/genética , Ubiquitinas/metabolismo , Neoplasias/metabolismo
7.
Environ Toxicol ; 37(2): 192-200, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34661962

RESUMEN

Tanshinone IIA is the active constituent extracted from Salvia Miltiorrhza. Numerous studies have shown that Tanshinone IIA could inhibit tumor proliferation and metastasis, including gastric cancer. However, the effect of Tanshinone IIA on gastric cancer cell stemness stays unclear. Here, we found that Tanshinone IIA could reduce gastric cancer cell stemness through detecting spheroid-forming, flow cytometry analysis, and the expression of stemness markers (OCT3/4, ALDH1A1, and CD44). Mechanistically, Tanshinone IIA increased the level of lipid peroxides and decreased glutathione level in gastric cancer cells, both of which are the markers of ferroptosis. Similarly, ferroptosis inducers (erastin, sulfasalazine, and sorafenib) reduced gastric cancer cell stemness. Additionally, the inhibitory effects of Tanshinone IIA on GC cell stemness were reversed by ferroptosis inhibitor (Fer-1) or overexpression of SLC7A11, which is a critical ferroptosis inhibitor. Therefore, we revealed that Tanshinone IIA inhibited the stemness of gastric cancer cells partly through inducing ferroptosis.


Asunto(s)
Antineoplásicos Fitogénicos , Ferroptosis , Neoplasias Gástricas , Abietanos , Antineoplásicos Fitogénicos/farmacología , Línea Celular Tumoral , Proliferación Celular , Humanos , Neoplasias Gástricas/tratamiento farmacológico
8.
Mol Carcinog ; 60(6): 413-426, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33866606

RESUMEN

Ours and other previous studies have shown that CYP4Z1 is specifically and highly expressed in breast cancer, and acts as a promoter for the stemness of breast cancer cells. Here, we explored whether targeting CYP4Z1 could attenuate the stemness of breast cancer cells using HET0016, which has been confirmed to be an inhibitor of CYP4Z1 by us and others. Using the transcriptome-sequencing analysis, we found that HET0016 suppressed the expression of cancer stem cell (CSC) markers and stem cell functions. Additionally, HET0016 indeed reduced the stemness of breast cancer cells, as evident by the decrease of stemness marker expression, CD44+ /CD24- subpopulation with stemness, mammary-spheroid formation, and tumor-initiating ability. Moreover, HET0016 suppressed the metastatic capability through in vitro and in vivo experiments. Furthermore, we confirmed that HET0016 suppressed CYP4Z1 activity, and HET0016-induced inhibition on the stemness and metastasis of breast cancer cells was rescued by CYP4Z1 overexpression. Thus, our results demonstrate that HET0016 can attenuate the stemness of breast cancer cells through targeting CYP4Z1.


Asunto(s)
Amidinas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Familia 4 del Citocromo P450/genética , Células Madre Neoplásicas/efectos de los fármacos , Amidinas/administración & dosificación , Animales , Antinematodos/administración & dosificación , Antinematodos/farmacología , Línea Celular Tumoral , Familia 4 del Citocromo P450/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Ratones Endogámicos BALB C , Células Madre Neoplásicas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Helicobacter ; 26(4): e12813, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33938607

RESUMEN

BACKGROUND: Recent studies and clinical samples have demonstrated that Helicobacter pylori could induce the downregulation of miR-375 in the stomach and promote gastric carcinogenesis. However, whether the immune cells are affected by Helicobacter pylori due to the downregulation of miR-375 is unclear. MATERIALS AND METHODS: In this study, we constructed an overexpression and knockdown of miR-375 and Helicobacter pylori infection cell models in vitro. In addition, the maturity of dendritic cells (DCs) and the expression of IL-6, IL-10, and VEGF at the transcriptional and translational levels were analyzed. Changes in the JAK2-STAT3 signaling pathway were detected. In vivo, the number changes in CD4+ T and CD8+ T cells and the size changes of tumors via models of transplantable subcutaneous tumors were also analyzed. RESULTS: A cell model of Helicobacter pylori and gastric cancer was used to identify the expression of miR-375 and the maturity of dendritic cells. This study found that Helicobacter pylori could downregulate miR-375, which regulates the expression of cytokines IL-6, IL-10, and VEGF in the stomach. MiR-375 regulated the expression of cytokines IL-6, IL-10, and VEGF through the JAK2-STAT3 signaling pathway in vitro. In addition, we found that Helicobacter pylori regulates the maturation of dendritic cells through miR-375. These results were further verified in vivo, and miR-375 diminishes tumor size was also demonstrated. This study showed that immature DCs caused a decrease in the number of CD4+ and CD8+ T cells. CONCLUSIONS: This study demonstrated that Helicobacter pylori can inhibit miRNA-375 expression in the stomach. Downregulated miR-375 activates the JAK2-STAT3 pathway. Activating the JAK2-STAT3 signaling pathway promotes the secretion of IL-6, IL-10, and VEGF, leading to immature differentiation of DCs and induction of gastric cancer.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , MicroARNs/genética , Neoplasias Gástricas , Diferenciación Celular , Células Dendríticas , Regulación hacia Abajo , Mucosa Gástrica , Infecciones por Helicobacter/complicaciones , Humanos , Neoplasias Gástricas/etiología
10.
Breast Cancer Res Treat ; 179(2): 521-522, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31655919

RESUMEN

After publication of this article [1], it came to our attention that there was an error in Fig. 4b and 4c. In Fig.4b, the migration images of vector groups were incorrect based on this error, the authors re-constructed the migration experiments of Fig. 4b, and the consistent results were obtained. In Fig. 4c, the tube formation image of the Z2P-UTR-siRNA&Z1-UTR group (MCF-7) of Fig. 7d was accidentally misused in control group (MCF-7) and vector group (MCF-7) of Fig. 4c. The corrected Fig. 4 is provided below. This error did not impact the conclusions of the article. The authors apologize for this error.

11.
J Cell Sci ; 130(19): 3399-3413, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28818997

RESUMEN

Diverse RNA transcripts acting as competing endogenous RNAs (ceRNAs) can co-regulate each other's expression by competing for shared microRNAs. CCR2 protein, the receptor for CCL2, is implicated in cancer progression. However, we found that a higher CCR2 mRNA level is remarkably associated with prolonged survival of breast cancer patients. These conflicting results prompted us to study the non-coding function of CCR2 mRNA. We found that the CCR2 3' untranslated region (UTR) inhibited MDA-MB-231 and MCF-7 cell metastasis by repressing epithelial-mesenchymal transition (EMT) in vitro, and suppressed breast cancer metastasis in vivo Mechanistically, the CCR2 3'UTR modulated the expression of the RhoGAP protein STARD13 via acting as a STARD13 ceRNA in a microRNA-dependent and protein coding-independent manner. The CCR2 3'UTR blocked the activation of RhoA-ROCK1 pathway, which is the downstream effector of STARD13, and thus decreased the phosphorylation level of myosin light chain 2 (MLC2) and formation of F-actin. Additionally, the function of the CCR2 3'UTR was dependent on STARD13 expression. In conclusion, our results confirmed that the CCR2 3'UTR acts as a metastasis suppressor by acting as a ceRNA for STARD13 and thus inhibiting RhoA-ROCK1-MLC-F-actin pathway in breast cancer cells.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Regiones no Traducidas 3' , Neoplasias de la Mama/metabolismo , ARN Neoplásico/metabolismo , Receptores CCR2/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Células MCF-7 , Metástasis de la Neoplasia , ARN Neoplásico/genética , Receptores CCR2/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
12.
FASEB J ; 32(2): 588-600, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28939591

RESUMEN

Tamoxifen (TAM) is a major adjuvant therapy for patients who are diagnosed with estrogen receptor-α (ER)-positive breast cancer; however, TAM resistance occurs often during treatment and the underlying mechanism is unclear. Here, we report that miR-125a-3p inhibits ERα transcriptional activity and, thus, ER+ breast cancer cell proliferation, which causes cell-cycle arrest at the G1/S stage, inducing apoptosis and suppressing tumor growth by targeting cyclin-dependent kinase 3 (CDK3) in vitro and in vivo. In addition, CDK3 and miR-125a-3p expression levels were measured in 37 cancerous tissues paired with noncancerous samples, and their expression levels were negatively associated with miR-125a-3p level. Of interest, miR-125a-3p level is down-regulated in MCF-7 TAM-resistant (TamR) cells. Of more importance, up-regulation of miR-125a-3p resensitizes MCF-7 TamR cells to TAM, which is dependent on CDK3 expression. These results suggest that miR-125a-3p can function as a novel tumor suppressor in ER+ breast cancer by targeting CDK3, which may be a potential therapeutic approach for TamR breast cancer therapy.-Zheng, L., Meng, X., Li, X., Zhang, Y., Li, C., Xiang, C., Xing, Y., Xia, Y., Xi, T. miR-125a-3p inhibits ERα transactivation and overrides tamoxifen resistance by targeting CDK3 in estrogen receptor-positive breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Quinasa 3 Dependiente de Ciclina/metabolismo , Resistencia a Antineoplásicos , Receptor alfa de Estrógeno/biosíntesis , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , MicroARNs/metabolismo , Proteínas de Neoplasias/metabolismo , ARN Neoplásico/metabolismo , Tamoxifeno , Activación Transcripcional , Adulto , Anciano , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Quinasa 3 Dependiente de Ciclina/genética , Receptor alfa de Estrógeno/genética , Femenino , Humanos , Células MCF-7 , MicroARNs/genética , Persona de Mediana Edad , Proteínas de Neoplasias/genética , ARN Neoplásico/genética
13.
Mol Pharm ; 15(1): 63-71, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29179557

RESUMEN

The balance of pro- and antiapoptotic gene expression programs dominates the apoptotic progress of cancer cells. We previously demonstrated that STARD13 3'UTR suppressed breast cancer metastasis via inhibiting epithelial-mesenchymal transition (EMT). However, the roles of STARD13 3'UTR in breast cancer apoptosis remain elusive. Here, we identified that STARD13 3'UTR promoted cell apoptosis in vitro and in vivo. Mechanistically, STARD13 3'UTR acted as a ceRNA for BMF (Bcl-2 modifying factor), thus increasing BMF expression in an miRNA-dependent manner. Meanwhile, STARD13 3'UTR enhanced the interaction of BMF/Bcl-2 to release Bax (Bcl-2 associated X protein) in breast cancer cells. Finally, we verified the ceRNA relationship between STARD13 and BMF in vivo. Collectively, these findings suggest that STARD13 3'UTR could act as a ceRNA for BMF to promote apoptosis and recognize STARD13 3'UTR as a potential therapeutic target in breast cancer cells.


Asunto(s)
Regiones no Traducidas 3'/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias de la Mama/metabolismo , MicroARNs/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Apoptosis/genética , Apoptosis/fisiología , Neoplasias de la Mama/genética , Línea Celular Tumoral , Femenino , Citometría de Flujo , Proteínas Activadoras de GTPasa , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Inmunoprecipitación , Etiquetado Corte-Fin in Situ , Células MCF-7 , Proteínas Supresoras de Tumor/genética , Proteína X Asociada a bcl-2
14.
Mol Pharm ; 15(6): 2123-2132, 2018 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-29733656

RESUMEN

RNA binding proteins (RBPs) are pivotal post-transcriptional regulators. RNPC1, an RBP, acts as a tumor suppressor through binding and regulating the expression of target genes in cancer cells. This study disclosed that RNPC1 expression was positively correlated with breast cancer patients' relapse-free and overall survival and that RNPC1 suppressed breast cancer cell metastasis. Mechanistically, RNPC1 promotes competing endogenous RNA (ceRNA) network crosstalk among STARD13, CDH5, HOXD10, and HOXD1 (STARD13-correlated ceRNA network), which we previously confirmed in breast cancer cells through stabilizing the transcripts and thus facilitating the expression of these four genes in breast cancer cells. Furthermore, RNPC1 overexpression restrained the promotion of STARD13, CDH5, HOXD10, and HOXD1 knockdown on cell metastasis. Notably, RNPC1 expression was positively correlated with CDH5, HOXD1, and HOXD10 expression in breast cancer tissues and attenuated adriamycin resistance. Taken together, these results identified that RNPC1 could inhibit breast cancer cell metastasis via promoting a STARD13-correlated ceRNA network.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas Activadoras de GTPasa/genética , Regulación Neoplásica de la Expresión Génica , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Supresoras de Tumor/genética , Regiones no Traducidas 3'/genética , Neoplasias de la Mama/patología , Femenino , Proteínas Activadoras de GTPasa/metabolismo , Técnicas de Silenciamiento del Gen , Redes Reguladoras de Genes , Células HEK293 , Humanos , Células MCF-7 , ARN Largo no Codificante/genética , Proteínas de Unión al ARN/genética , Proteínas Supresoras de Tumor/metabolismo
15.
Helicobacter ; 22(5)2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28805287

RESUMEN

BACKGROUND: Gram-positive enhancer matrix particles (GEM) produced by Lactococcus lactis can enhance vaccine-induced immune response. However, the mechanism under which this adjuvant mounts the efficacy of orally administered vaccines remains unexplored. MATERIALS AND METHODS: We used a prophylactic mice model to investigate the mechanism of GEM-adjuvanted vaccination. Helicobacter pylori urease-specific antibody response was monitored and detected in murine serum by ELISA. Urease-specific splenic cytokine profile was examined. Gastric inflammatory responses were measured on day 43 or 71 by quantitative real-time PCR, flow cytometry and histology. RESULTS: We found that GEM enhanced the efficiency of oral H. pylori vaccine by promoting innate immunity. The vaccine CUE-GEM composed of GEM particles and recombinant antigen CTB-UE provided protection of immunized mice against H. pylori insult. The protective response was associated with induction of postimmunization gastritis and local Th1/Th17 cell-medicated immune response. We showed that innate inflammatory responses including neutrophil chemokines CXCL1-2, neutrophils, and antimicrobial proteins S100A8 and MUC1 were significantly elevated. Within all infected mice, S100A8 and MUC1 levels were negatively correlated with H. pylori burden. Strikingly, mice receiving GEM also show reduction of colonization, possibly through natural host response pathways to recruit CD4+ T cells and promote S100A8 expression. CONCLUSIONS: These findings suggest that GEM-based vaccine may impact Th1/Th17 immunity to orchestrate innate immune response against H. pylori infection.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacunas Bacterianas/inmunología , Infecciones por Helicobacter/prevención & control , Helicobacter pylori/inmunología , Inmunidad Innata , Lactococcus lactis/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/aislamiento & purificación , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Mucosa Gástrica/patología , Infecciones por Helicobacter/patología , Leucocitos Mononucleares/inmunología , Masculino , Ratones Endogámicos BALB C , Ureasa/inmunología
16.
Biochem Biophys Res Commun ; 478(2): 900-7, 2016 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-27520371

RESUMEN

To investigate the effects of CYP4Z1 3'UTR in migration of breast cancer cells, a series of assays were used to confirm that overexpression of CYP4Z1 3'UTR could suppress the capacity of migration and adhesion of MCF-7 and MDA-MB-231 cells. EMT (Epithelial-mesenchymal transition)-related proteins were regulated by CYP4Z1 3'UTR. Mesenchyma markers like Vimentin, MMP-2, and MMP-9 were down-regulated, while the expression of E-cadherin was up-regulated with CYP4Z1 3'UTR overexpression. Notably, luciferase reporter and qRT-PCR assays were applied to verify that CYP4Z1 3'UTR was the potential target of miR-9. In addition, our results showed that CYP4Z1 3'UTR repressed the expression of E-cadherin in a miRNA-dependent manner. Combining with our previous study, we have discovered the underlying link between CYP4Z1 and E-cadherin. Therefore, those preliminary data suggest that CYP4Z1 3'UTR could inhibit the migration and EMT of breast cancer cells via acting as a ceRNA for E-cadherin.


Asunto(s)
Regiones no Traducidas 3'/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Movimiento Celular/genética , Familia 4 del Citocromo P450/genética , Antígenos CD , Cadherinas , Adhesión Celular , Línea Celular Tumoral , Familia 4 del Citocromo P450/metabolismo , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , MicroARNs/genética , MicroARNs/metabolismo , ARN Interferente Pequeño/metabolismo , Transfección
17.
Biochem Biophys Res Commun ; 472(1): 262-9, 2016 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-26926567

RESUMEN

Competitive endogenous messenger RNAs (ceRNAs) affect other RNAs transcription through competitively binding common microRNAs (miRNAs). In this study we identified long non-coding RNA (lncRNA) MALAT1 can function as a ceRNA of cell division cycle 42 (cdc42) 3'UTR in inducing migration and invasion of breast cancer cells via miR-1. We found that miR-1 bound both MALAT1 and cdc42 3'UTR directly. Further study showed that MALAT1 induced migration and invasion of breast cancer cells while reduced the level of cdc42. Our results suggest that MALAT1 regulated migration and invasion of breast cancer cells via affecting cdc42 through binding miR-1 competitively.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/genética , Proteína de Unión al GTP cdc42/genética , Regiones no Traducidas 3' , Unión Competitiva , Neoplasias de la Mama/fisiopatología , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/fisiología , Regulación hacia Abajo , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , Células MCF-7 , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Invasividad Neoplásica/fisiopatología , ARN Largo no Codificante/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Regulación hacia Arriba
20.
Breast Cancer Res Treat ; 150(1): 105-18, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25701119

RESUMEN

Pseudogenes are now known to regulate their protein-coding counterparts. Additionally, disturbances of 3'UTRs could increase the risk of cancer susceptibility by acting as modulators of gene expression. The aim of this study was to investigate the roles of the pseudogene CYP4Z2P-3'UTR and functional gene CYP4Z1-3'UTR in breast cancer angiogenesis process. The levels of CYP4Z2P- and CYP4Z1-3'UTR and miRNA of interests were measured in 22 cancerous tissues paired with non-cancerous samples by qRT-PCR. The effects of CYP4Z2P- and CYP4Z1-3'UTR were studied by overexpression and RNA interference approaches in vitro and ex vivo. Insights of the mechanism of competitive endogenous RNAs were gained from bioinformatic analysis, luciferase assays, and western blot. The positive CYP4Z2P/CYP4Z1 interaction and negative interaction between predicted miRNAs and CYP4Z2P or CYP4Z1 were identified via qRT-PCR assay and bivariate correlation analysis. CYP4Z2P- and CYP4Z1-3'UTR share several miRNA-binding sites, including miR-211, miR-125a-3p, miR-197, miR-1226, and miR-204. The CYP4Z2P- and CYP4Z1-3'UTRs arrest the interference caused by of these miRNAs, resulting in increased translation of CYP4Z1. Moreover, ectopic expression of the CYP4Z2P- and CYP4Z1-3'UTRs exhibit tumor angiogenesis-promoting properties in breast cancer collectively by inducing the phosphorylation of ERK1/2 and PI3K/Akt. Co-transfection with Dicer siRNA reversed the CYP4Z2P 3'UTR-mediated changes. Additionally, PI3K or ERK inhibitors reversed CYP4Z2P- and CYP4Z1-3'UTR-mediated changes in VEGF-A expression. Increased CYP4Z2P- and CYP4Z1-3'UTR expression promotes tumor angiogenesis in breast cancer partly via miRNA-dependent activation of PI3K/Akt and ERK1/2. The CYP4Z2P- and CYP4Z1-3'UTRs could thus be used as combinatorial miRNA inhibitors.


Asunto(s)
Regiones no Traducidas 3' , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Sistema Enzimático del Citocromo P-450/genética , Seudogenes , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Embrión de Pollo , Sistema Enzimático del Citocromo P-450/química , Familia 4 del Citocromo P450 , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , MicroARNs/química , MicroARNs/genética , Proteína Quinasa 1 Activada por Mitógenos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neovascularización Patológica/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Mensajero/química , ARN Mensajero/genética , Ratas , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA