Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 139(4): 679-92, 2009 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-19914164

RESUMEN

Signaling proteins driving the proliferation of stem and progenitor cells are often encoded by proto-oncogenes. EphB receptors represent a rare exception; they promote cell proliferation in the intestinal epithelium and function as tumor suppressors by controlling cell migration and inhibiting invasive growth. We show that cell migration and proliferation are controlled independently by the receptor EphB2. EphB2 regulated cell positioning is kinase-independent and mediated via phosphatidylinositol 3-kinase, whereas EphB2 tyrosine kinase activity regulates cell proliferation through an Abl-cyclin D1 pathway. Cyclin D1 regulation becomes uncoupled from EphB signaling during the progression from adenoma to colon carcinoma in humans, allowing continued proliferation with invasive growth. The dissociation of EphB2 signaling pathways enables the selective inhibition of the mitogenic effect without affecting the tumor suppressor function and identifies a pharmacological strategy to suppress adenoma growth.


Asunto(s)
Receptor EphB2/metabolismo , Transducción de Señal , Animales , Movimiento Celular , Proliferación Celular , Ciclina D1/metabolismo , Epitelio , Humanos , Intestino Delgado/citología , Intestino Delgado/metabolismo , Masculino , Ratones , Células Madre/citología
2.
Environ Toxicol ; 39(5): 2908-2926, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38299230

RESUMEN

BACKGROUND: Colorectal cancer (CRC) presents a significant global health burden, characterized by a heterogeneous molecular landscape and various genetic and epigenetic alterations. Programmed cell death (PCD) plays a critical role in CRC, offering potential targets for therapy by regulating cell elimination processes that can suppress tumor growth or trigger cancer cell resistance. Understanding the complex interplay between PCD mechanisms and CRC pathogenesis is crucial. This study aims to construct a PCD-related prognostic signature in CRC using machine learning integration, enhancing the precision of CRC prognosis prediction. METHOD: We retrieved expression data and clinical information from the Cancer Genome Atlas and Gene Expression Omnibus (GEO) datasets. Fifteen forms of PCD were identified, and corresponding gene sets were compiled. Machine learning algorithms, including Lasso, Ridge, Enet, StepCox, survivalSVM, CoxBoost, SuperPC, plsRcox, random survival forest (RSF), and gradient boosting machine, were integrated for model construction. The models were validated using six GEO datasets, and the programmed cell death score (PCDS) was established. Further, the model's effectiveness was compared with 109 transcriptome-based CRC prognostic models. RESULT: Our integrated model successfully identified differentially expressed PCD-related genes and stratified CRC samples into four subtypes with distinct prognostic implications. The optimal combination of machine learning models, RSF + Ridge, showed superior performance compared with traditional methods. The PCDS effectively stratified patients into high-risk and low-risk groups, with significant survival differences. Further analysis revealed the prognostic relevance of immune cell types and pathways associated with CRC subtypes. The model also identified hub genes and drug sensitivities relevant to CRC prognosis. CONCLUSION: The current study highlights the potential of integrating machine learning models to enhance the prediction of CRC prognosis. The developed prognostic signature, which is related to PCD, holds promise for personalized and effective therapeutic interventions in CRC.


Asunto(s)
Apoptosis , Neoplasias Colorrectales , Humanos , Pronóstico , Aprendizaje Automático , Neoplasias Colorrectales/genética
3.
J Transl Med ; 20(1): 267, 2022 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-35690861

RESUMEN

Heart failure (HF), as the leading cause of death, is continuing to increase along with the aging of the general population all over the world. Identification of diagnostic biomarkers for early detection of HF is considered as the most effective way to reduce the risk and mortality. Herein, we collected plasma samples from HF patients (n = 40) before and after medical therapy to determine the change of circulating miRNAs through a quantitative real-time PCR (QRT-PCR)-based miRNA screening analysis. miR-30a-5p and miR-654-5p were identified as the most significantly changed miRNAs in the plasma of patients upon treatment. In consistence, miR-30a-5p showed upregulation and miR-654-5p showed downregulation in the circulation of 30 HF patients, compared to 15 normal controls in the training phase, from which a two-circulating miRNA model was developed for HF diagnosis. Next, we performed the model validation using an independent cohort including 50 HF patients and 30 controls. As high as 98.75% of sensitivity and 95.00% of specificity were achieved. A comparison between the miRNA model and NT-pro BNP in diagnostic accuracy of HF indicated an upward trend of the miRNA model. Moreover, change of the two miRNAs was further verified in association with the therapeutic effect of HF patients, in which miR-30a-5p showed decrease while miR-654-5p showed increase in the plasma of patients after LVAD implantation. In conclusion, the current study not only identified circulating miR-654-5p for the first time as a novel biomarker of HF, but also developed a novel 2-circulating miRNA model with promising potentials for diagnosis and prognosis of HF patients, and in association with therapeutic effects as well.


Asunto(s)
MicroARN Circulante , Insuficiencia Cardíaca , MicroARNs , Biomarcadores , Biomarcadores de Tumor/genética , MicroARN Circulante/genética , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/terapia , Humanos , MicroARNs/genética , Pronóstico
4.
FASEB J ; 35(5): e21442, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33774850

RESUMEN

Cold atmospheric plasma (CAP) is an emerging technology for biomedical applications, exemplified by its antimicrobial and antineoplastic potentials. On the contrary, acidic fibroblast growth factor (aFGF) has been a long-standing potent mitogen for cells from various origins. In this study, we are the first to develop a multimodal treatment combining the aforementioned physicochemical and pharmacological treatments and investigated their individual and combined effects on wound healing, angiogenesis, neurogenesis, and osteogenesis. This work was performed at the tissue, cellular, protein, and gene levels, using histochemical staining, flow cytometry, ELISA, and PCR, respectively. Depending on the type of target tissue, various combinations of aforementioned methods were used. The results showed that the enhancement on would healing and angiogenesis by CAP and aFGF were synergistic. The former was manifested by increased murine fibroblast proliferation and reduced cutaneous tissue inflammation, whereas the latter by upregulated proangiogenic markers in vivo, for example, CD31, VEGF, and TGF-ß, and downregulated antiangiogenic proteins in vitro, for example, angiostatin and angiopoietin-2, respectively. In addition, aFGF outperformed CAP during neurogenesis, which was evidenced by superior neurite outgrowth, while CAP exceeded aFGF in osteogenesis which was demonstrated by more substantial bone nodule formation. These novel findings not only support the fact that CAP and aFGF are both multipotent agents during tissue regeneration, but also highlight the potential of our multimodal treatment combining the individual advantages of CAP and aFGF. The versatile administration route, that is, topical and/or systemic, might further broaden its applications.


Asunto(s)
Factor 1 de Crecimiento de Fibroblastos/farmacología , Neovascularización Fisiológica , Neurogénesis , Gases em Plasma/farmacología , Regeneración , Cicatrización de Heridas , Animales , Atmósfera , Terapia Combinada , Humanos , Ratones
5.
Circ Res ; 125(6): 590-605, 2019 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-31318658

RESUMEN

RATIONALE: Endothelial dysfunction results in sustained and chronic vascular inflammation, which is central to atherosclerotic diseases. However, transcriptional regulation of vascular endothelial inflammation has not been well clarified. OBJECTIVE: This study aims to explore Foxp (forkhead box P) transcription factor 1 in regulation of endothelial homeostasis, atherogenesis, and its mechanisms. METHODS AND RESULTS: To assess the importance of Foxp1 in atherosclerosis, Foxp1 expression was analyzed in human coronary artery and mouse artery, and we observed significant downregulation of Foxp1 in atherosclerotic and atherosusceptible endothelium. Endothelial-specific Foxp1 knockout mice (Foxp1ECKO) were bred onto ApoeKO mice to generate endothelial Foxp1-deletion hyperlipidemic model Foxp1ECKO;ApoeKO, which displayed significant increases in atherosclerotic lesion formation in aortas and aortic roots with enhanced monocyte adhesion, migration, and infiltration into the vascular wall and formation of inflammatory lipid-laden macrophages. In contrast, endothelial-specific Foxp1 overexpression mice Foxp1ECTg;ApoeKO exhibited reduced atherosclerotic lesion formation with less monocyte infiltration. Foxp1 was further identified as a gatekeeper of vessel inflammation by direct regulation of endothelial inflammasome components, including Nlrp3 (NLR [nucleotide-binding and leucine-rich repeat immune receptors] family pyrin domain containing 3), caspase-1, and IL (interleukin)-1ß. Moreover, endothelial Foxp1 was found to be regulated by Klf2 (Kruppel-like factor 2). Oscillatory shear stress downregulated Foxp1 expression via repressing Klf2 expression in endothelium, and, therefore, promoted endothelial inflammasome activation, leading to atherosclerotic lesion formation. Simvastatin upregulated the reduced expression of Klf2 and Foxp1 in atherosusceptible vascular endothelium and alleviated vascular inflammation contributing to its inhibitory effect in atherosclerosis. CONCLUSIONS: These data are the first in vivo experimental validation of an atheroprotective role of endothelial Klf2 and Foxp1, which reveals a Klf2-Foxp1 transcriptional network in endothelial cells as a novel regulator of endothelial inflammasome activation for atherogenesis, therefore, provides opportunities for therapeutic intervention of atherosclerotic diseases and uncovers a novel atheroprotective mechanism for simvastatin.


Asunto(s)
Aterosclerosis/metabolismo , Células Endoteliales/metabolismo , Factores de Transcripción Forkhead/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas Represoras/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Células Endoteliales/patología , Factores de Transcripción Forkhead/genética , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Inflamasomas/genética , Inflamasomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteínas Represoras/genética
6.
Circulation ; 140(8): 665-680, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31177814

RESUMEN

BACKGROUND: Pathological cardiac fibrosis and hypertrophy, the common features of left ventricular remodeling, often progress to heart failure. Forkhead box transcription factor P1 (Foxp1) in endothelial cells (ECs) has been shown to play an important role in heart development. However, the effect of EC-Foxp1 on pathological cardiac remodeling has not been well clarified. This study aims to determine the role of EC-Foxp1 in pathological cardiac remodeling and the underlying mechanisms. METHODS: Foxp1 EC-specific loss-of-function and gain-of-function mice were generated, and an angiotensin II infusion or a transverse aortic constriction operation mouse model was used to study the cardiac remodeling mechanisms. Foxp1 downstream target gene transforming growth factor-ß1 (TGF-ß1) was confirmed by chromatin immunoprecipitation and luciferase assays. Finally, the effects of TGF-ß1 blockade on EC-Foxp1 deletion-mediated profibrotic and prohypertrophic phenotypic changes were further confirmed by pharmacological inhibition, more specifically by RGD-peptide magnetic nanoparticle target delivery of TGF-ß1-siRNA to ECs. RESULTS: Foxp1 expression is significantly downregulated in cardiac ECs during angiotensin II-induced cardiac remodeling. EC-Foxp1 deletion results in severe cardiac remodeling, including more cardiac fibrosis with myofibroblast formation and extracellular matrix protein production, as well as decompensated cardiac hypertrophy and further exacerbation of cardiac dysfunction on angiotensin II infusion or transverse aortic constriction operation. In contrast, EC-Foxp1 gain of function protects against pathological cardiac remodeling and improves cardiac dysfunction. TGF-ß1 signals are identified as Foxp1 direct target genes, and EC-Foxp1 deletion upregulates TGF-ß1 signals to promote myofibroblast formation through fibroblast proliferation and transformation, resulting in severe cardiac fibrosis. Moreover, EC-Foxp1 deletion enhances TGF-ß1-promoted endothelin-1 expression, which significantly increases cardiomyocyte size and reactivates cardiac fetal genes, leading to pathological cardiac hypertrophy. Correspondingly, these EC-Foxp1 deletion-mediated profibrotic and prohypertrophic phenotypic changes and cardiac dysfunction are normalized by the blockade of TGF-ß1 signals through pharmacological inhibition and RGD-peptide magnetic nanoparticle target delivery of TGF-ß1-siRNA to ECs. CONCLUSIONS: EC-Foxp1 regulates the TGF-ß1-endothelin-1 pathway to control pathological cardiac fibrosis and hypertrophy, resulting in cardiac dysfunction. Therefore, targeting the EC-Foxp1-TGF-ß1-endothelin-1 pathway might provide a future novel therapy for heart failure.


Asunto(s)
Endotelio Vascular/fisiología , Factores de Transcripción Forkhead/metabolismo , Insuficiencia Cardíaca/metabolismo , Miocardio/patología , Proteínas Represoras/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Angiotensina II/metabolismo , Animales , Aorta/cirugía , Modelos Animales de Enfermedad , Endotelina-1/metabolismo , Fibrosis , Factores de Transcripción Forkhead/genética , Insuficiencia Cardíaca/genética , Humanos , Ratones , Ratones Noqueados , Nanotubos de Péptidos , ARN Interferente Pequeño/genética , Proteínas Represoras/genética , Transducción de Señal , Factor de Crecimiento Transformador beta1/genética , Remodelación Ventricular
7.
BMC Cancer ; 20(1): 627, 2020 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-32631271

RESUMEN

BACKGROUND: Emerging evidence has demonstrated the limited access to metabolic substrates as an effective approach to block cancer cell growth. The mechanisms remain unclear. Our previous work has revealed that miR-221/222 plays important role in regulating breast cancer development and progression through interaction with target gene p27. RESULTS: Herein, we determined the miRNA-mRNA interaction in breast cancer cells under induced stress status of starvation. Starvation stimulation attenuated the miR-221/222-p27 interaction in MDA-MB-231 cells, thereby increased p27 expression and suppressed cell proliferation. Through overexpression or knockdown of miR-221/222, we found that starvation-induced stress attenuated the negative regulation of p27 expression by miR-221/222. Similar patterns for miRNA-target mRNA interaction were observed between miR-17-5p and CyclinD1, and between mR-155 and Socs1. Expression of Ago2, one of the key components of RNA-induced silencing complex (RISC), was decreased under starvation-induced stress status, which took responsibility for the impaired miRNA-target interaction since addition of exogenous Ago2 into MDA-MB-231 cells restored the miR-221/222-p27 interaction in starvation condition. CONCLUSIONS: We demonstrated the attenuated interaction between miR-221/222 and p27 by starvation-induced stress in MDA-MB-231 breast cancer cells. The findings add a new page to the general knowledge of negative regulation of gene expression by miRNAs, also demonstrate a novel mechanism through which limited access to nutrients suppresses cancer cell proliferation. These insights provide a basis for development of novel therapeutic options for breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Ayuno/fisiología , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Estrés Fisiológico/genética , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Proliferación Celular/genética , Medios de Cultivo/metabolismo , Ciclina D1/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , MicroARNs/genética , Proteína 1 Supresora de la Señalización de Citocinas/genética
8.
Clin Sci (Lond) ; 134(7): 791-805, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32219337

RESUMEN

The molecular mechanisms governing the secretion of the non-coding genome are poorly understood. We show herein that cyclin D1, the regulatory subunit of the cyclin-dependent kinase that drives cell-cycle progression, governs the secretion and relative proportion of secreted non-coding RNA subtypes (miRNA, rRNA, tRNA, CDBox, scRNA, HAcaBox. scaRNA, piRNA) in human breast cancer. Cyclin D1 induced the secretion of miRNA governing the tumor immune response and oncogenic miRNAs. miR-21 and miR-93, which bind Toll-Like Receptor 8 to trigger a pro-metastatic inflammatory response, represented >85% of the cyclin D1-induced secreted miRNA transcripts. Furthermore, cyclin D1 regulated secretion of the P-element Induced WImpy testis (PIWI)-interacting RNAs (piRNAs) including piR-016658 and piR-016975 that governed stem cell expansion, and increased the abundance of the PIWI member of the Argonaute family, piwil2 in ERα positive breast cancer. The cyclin D1-mediated secretion of pro-tumorigenic immuno-miRs and piRNAs may contribute to tumor initiation and progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Ciclina D1/metabolismo , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , ARN Interferente Pequeño/metabolismo , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Microambiente Celular , Ciclina D1/genética , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Ratones Transgénicos , MicroARNs/genética , MicroARNs/inmunología , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/inmunología , Transducción de Señal
9.
Pharmacol Res ; 152: 104628, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31904506

RESUMEN

Triple-negative breast cancers (TNBC) are more aggressive due to lacking receptors for hormone therapy and maintaining stemness features in cancer cells. Herein we found long non-coding RNA CCAT2 overexpressed specially in TNBC, and in breast cancer stem cells (BCSC) as well. Enforced overexpression and targeted knockdown demonstrated the oncogenic function of CCAT2 both in vitro and in vivo. CCAT2 promoted the expression of stemness markers including OCT4, Nanog and KLF4, increased mammosphere formation and induced ALDH+ cancer stem cell population in TNBC. A chromosomally adjacent gene OCT4-PG1, as a pseudogene of OCT4, was upregulated by CCAT2, and positively regulated the stemness features of TNBC cells. miR-205 was identified as a target gene of CCAT2 in TNBC. Point-mutation in CCAT2 impaired the sponge inhibition of miR-205. Overexpression of miR-205 rescued the oncogenic phenotypes induced by CCAT2. In addition, Notch2, as a target gene of miR-205, was downregulated by miR-205 and upregulated by CCAT2 in TNBC. Collectively, the current study revealed a novel function of CCAT2 in promoting tumor initiation and progression in TNBC through upregulating OCT4-PG1 expression and activating Notch signaling. These findings not only demonstrated a lncRNA-based therapeutic strategy in treatment of TNBC, but also added a node to the regulatory network of CCAT2 that controls aggressiveness of breast cancer stem cells.


Asunto(s)
Carcinogénesis/genética , Neoplasias Mamarias Experimentales/genética , ARN Largo no Codificante , Neoplasias de la Mama Triple Negativas/genética , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Factor 4 Similar a Kruppel , Ratones Desnudos , Células Madre Neoplásicas/fisiología
10.
Arterioscler Thromb Vasc Biol ; 39(5): 888-901, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30943773

RESUMEN

Objective- Transcription factor GATA (GATA zinc finger transcription factor family)-6 is highly expressed in vessels and rapidly downregulated in balloon-injured carotid arteries and viral delivery of GATA-6 to the vessels limited the neointimal formation, however, little is known about its cell-specific regulation of in vivo vascular smooth muscle cell (VSMC) phenotypic state contributing to neointimal formation. This study aims to determine the role of vascular cell-specific GATA-6 in ligation- or injury-induced neointimal hyperplasia in vivo. Approach and Results- Endothelial cell and VSMC-specific GATA-6 deletion mice are generated, and the results indicate that endothelial cell-specific GATA-6 deletion mice exhibit significant decrease of VSMC proliferation and attenuation of neointimal formation after artery ligation and injury compared with the wild-type littermate control mice. PDGF (platelet-derived growth factor)-B is identified as a direct target gene, and endothelial cell-GATA-6-PDGF-B pathway regulates VSMC proliferation and migration in a paracrine manner which controls the neointimal formation. In contrast, VSMC-specific GATA-6 deletion promotes injury-induced VSMC transformation from contractile to proliferative synthetic phenotype leading to increased neointimal formation. CCN (cysteine-rich 61/connective tissue growth factor/nephroblastoma overexpressed family)-5 is identified as a novel target gene, and VSMC-specific CCN-5 overexpression in mice reverses the VSMC-GATA-6 deletion-mediated increased cell proliferation and migration and finally attenuates the neointimal formation. Conclusions- This study gives us a direct in vivo evidence of GATA-6 cell lineage-specific regulation of PDGF-B and CCN-5 on VSMC phenotypic state, proliferation and migration contributing to neointimal formation, which advances our understanding of in vivo neointimal hyperplasia, meanwhile also provides opportunities for future therapeutic interventions.


Asunto(s)
Regulación de la Expresión Génica , Músculo Liso Vascular/metabolismo , Neointima/patología , Lesiones del Sistema Vascular/patología , Dedos de Zinc/genética , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Modelos Animales de Enfermedad , Femenino , Factor de Transcripción GATA6/genética , Hiperplasia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Sensibilidad y Especificidad , Factores de Transcripción/metabolismo
11.
J Cell Mol Med ; 23(1): 476-486, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30334371

RESUMEN

BACKGROUND: Circulating endothelial-derived microparticles (EMPs) are reported to be increased in acute coronary syndrome (ACS). However, it remains unclear whether EMPs from dysfunctional endothelium participate in the initiation and progression of ACS and what the underlying mechanisms might be. METHODS: Plasma EMPs were measured in 22 patients with ACS and 20 control patients without coronary artery diseases. EMPs from dysfunctional human umbilical vein endothelial cells (HUVECs) stressed by serum-starvation or hypoxia were compared to the EMPs from healthy HUVECs. Confocal and fluorescent microscopy was used to visualize the incorporation of EMPs into monocytes and the translocation of NF-кB. Monocyte adhesion, cell proliferation, and phagocytosis were detected by PKH26 red fluorescent labelling, Ki67 immunostaining, and Sudan IV staining for uptake of oxidized low-density lipoprotein, respectively. RESULTS: Plasma EMPs was significantly increased in ACS patients compared to controls. EMPs were incorporated into monocytes and EMPs from stressed HUVECs produced more pro-inflammatory cytokines compared to vehicle control, which was depended on NF-кB and IL-1ß signal pathways. EMPs from dysfunctional endothelium promoted monocyte adherence via NF-кB and IL-1ß-mediated MCP-1 and CCR-5 signals, as well as proliferation via the NF-кB and IL-1ß-mediated Cyclin D1 signals. Finally, EMPs from dysfunctional endothelium showed greater promotion of macrophage phagocytosis forming foam cells to produce more pro-inflammatory cytokines. CONCLUSION: MPs might be involved in the inflammatory process in patients with ACS via NF-κB and IL-1ß-dependent signals. Targeting EMP-mediated inflammatory responses may be a promising therapeutic strategy to limit the progression of disease in ACS.


Asunto(s)
Micropartículas Derivadas de Células/metabolismo , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Anciano , Estudios de Casos y Controles , Adhesión Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Femenino , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Antígeno Ki-67/metabolismo , Lipoproteínas LDL/metabolismo , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Fagocitosis/fisiología
12.
Respir Res ; 20(1): 249, 2019 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-31703732

RESUMEN

Pulmonary fibrosis is a chronic, progressive lung disease associated with lung damage and scarring. The pathological mechanism causing pulmonary fibrosis remains unknown. Emerging evidence suggests prominent roles of epithelial-mesenchymal transition (EMT) of alveolar epithelial cells (AECs) in myofibroblast formation and progressive pulmonary fibrosis. Our previous work has demonstrated the regulation of YY1 in idiopathic pulmonary fibrosis and pathogenesis of fibroid lung. However, the specific function of YY1 in AECs during the pathogenesis of pulmonary fibrosis is yet to be determined. Herein, we found the higher level of YY1 in primary fibroblasts than that in primary epithelial cells from the lung of mouse. A549 and BEAS-2B cells, serving as models for type II alveolar pulmonary epithelium in vitro, were used to determine the function of YY1 during EMT of AECs. TGF-ß-induced activation of the pro-fibrotic program was applied to determine the role YY1 may play in pro-fibrogenesis of type II alveolar epithelial cells. Upregulation of YY1 was associated with EMT and pro-fibrotic phenotype induced by TGF-ß treatment. Targeted knockdown of YY1 abrogated the EMT induction by TGF-ß treatment. Enforced expression of YY1 can partly mimic the TGF-ß-induced pro-fibrotic change in either A549 cell line or primary alveolar epithelial cells, indicating the induction of YY1 expression may mediate the TGF-ß-induced EMT and pro-fibrosis. In addition, the translocation of NF-κB p65 from the cytoplasm to the nucleus was demonstrated in A549 cells after TGF-ß treatment and/or YY1 overexpression, suggesting that NF-κB-YY1 signaling pathway regulates pulmonary fibrotic progression in lung epithelial cells. These findings will shed light on the better understanding of mechanisms regulating pro-fibrogenesis in AECs and pathogenesis of lung fibrosis.


Asunto(s)
Células Epiteliales Alveolares/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibrosis Pulmonar/inducido químicamente , Factor de Crecimiento Transformador beta1/toxicidad , Factor de Transcripción YY1/metabolismo , Células A549 , Transporte Activo de Núcleo Celular , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Animales , Comunicación Celular , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Humanos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción YY1/genética
13.
Circ Res ; 120(1): 85-98, 2017 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-27756792

RESUMEN

RATIONALE: Angiogenic hypersprouting and leaky vessels are essential for tumor growth. MicroRNAs have unique therapeutic advantages by targeting multiple pathways of tumor-associated angiogenesis, but the function of individual miRNAs of miR302-367 cluster in angiogenesis and tumors has not yet been fully evaluated. OBJECTIVE: To investigate the functions of miR302-367 in developmental angiogenesis and tumor angiogenesis and explore the molecular mechanisms of microRNA for the treatment of pathological neovascularization-related diseases. METHODS AND RESULTS: Here, we show that miR302-367 elevation in endothelial cells reduces retinal sprouting angiogenesis and promotes vascular stability in vivo, ex vivo, and in vitro. Erk1/2 is identified as direct target of miR302-367, and downregulation of Erk1/2 on miR302-367 elevation in endothelial cells increases the expression of Klf2 and in turn S1pr1 and its downstream target VE-cadherin, suppressing angiogenesis and improving vascular stability. Conversely, both pharmacological blockade and genetic deletion of S1pr1 in endothelial cells reverse the antiangiogenic and vascular stabilizing effect of miR302-367 in mice. Tumor angiogenesis shares features of developmental angiogenesis, and endothelial specific elevation of miR302-367 reduces tumor growth by restricting sprout angiogenesis and decreasing vascular permeability via the same Erk1/2-Klf2-S1pr1 pathways. CONCLUSIONS: MiR302-367 regulation of an Erk1/2-Klf2-S1pr1 pathway in the endothelium advances our understanding of angiogenesis, meanwhile also provides opportunities for therapeutic intervention of tumor growth.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/biosíntesis , Sistema de Señalización de MAP Quinasas/fisiología , MicroARNs/biosíntesis , Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Receptores de Lisoesfingolípidos/biosíntesis , Inhibidores de la Angiogénesis/biosíntesis , Animales , Carcinoma Pulmonar de Lewis , Técnicas de Cocultivo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Melanoma Experimental , Ratones , Ratones Transgénicos , Neoplasias/patología , Neoplasias/prevención & control , Neovascularización Patológica/patología , Neovascularización Patológica/prevención & control , Receptores de Esfingosina-1-Fosfato , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
14.
Reprod Fertil Dev ; 31(5): 962-971, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30811962

RESUMEN

In this study we identified Elongin B, a regulatory subunit of the trimeric elongation factor Elongin ABC, which increases the overall rate of elongation by RNA polymerase II, as a major binding partner of sperm-associated antigen 16S (SPAG16S), a component of nuclear speckles. Nuclear speckles are nuclear subcompartments involved in RNA maturation. Previously, we showed that SPAG16S is essential for spermatogenesis. In the present study, a specific antibody against mouse Elongin B was generated and reacted with a protein with the predicted size of Elongin B in the testis; immunofluorescence staining revealed that the Elongin B was located in the nuclei and residual bodies. In round spermatids, Elongin B was colocalised with splicing factor SC35 (SC35), a marker of nuclear speckles. During the first wave of spermatogenesis, Elongin B transcripts were initially detected at Postnatal Day (PND) 8, and levels were greatly increased afterwards. However, Elongin B protein was only found from PND30, when germ cells progressed through spermiogenesis. Polysomal gradient analysis of Elongin B transcripts isolated from adult mouse testes revealed that most of the Elongin B mRNA was associated with translationally inactive, non-polysomal ribonucleoproteins. An RNA electrophoretic mobility shift assay demonstrated that the 3' untranslated region of the Elongin B transcript was bound by proteins present in testis but not liver extracts. These findings suggest that post-transcriptional regulation of Elongin B occurs in the testis, which is a common phenomenon during male germ cell development. As a major binding partner of SPAG16S, Elongin B may play an important role in spermatogenesis by modulating RNA maturation.


Asunto(s)
Elonguina/metabolismo , Espermatogénesis/fisiología , Testículo/metabolismo , Animales , Regulación de la Expresión Génica , Masculino , Ratones , Espermatozoides/metabolismo , Transcripción Genética
15.
Future Oncol ; 14(30): 3145-3161, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30220214

RESUMEN

AIM: The present study aimed to identify microRNA (miRNA) that can be used for not only detecting early-stage breast cancer (BC) but also diagnosing atypical hyperplasia (AH). MATERIALS & METHODS: RT-qPCR detected the expression levels of miRNAs and receiver operating characteristic curves were constructed to evaluate sensitivity and specificity of the assay. RESULTS: miR-24 and miR-103a were expressed in an upward trend in serum of benign proliferative tumor subjects, while they were downregulated significantly in serum of AH (p < 0.005) and early-stage BC subjects (p < 0.005) with high sensitivity and specificity as compared with controls. Bioinformatics analysis also revealed the potential molecular mechanism through which miR-24 and miR-103a regulate tumorigenesis in BC. CONCLUSION: miR-24 and miR-103a were valuable biomarkers for distinguishing AH and early-stage BC from healthy individuals/benign proliferative tumor patients.


Asunto(s)
Enfermedades de la Mama/diagnóstico , Enfermedades de la Mama/genética , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , MicroARN Circulante/genética , Hiperplasia/genética , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores , Enfermedades de la Mama/sangre , Neoplasias de la Mama/sangre , MicroARN Circulante/sangre , Diagnóstico Diferencial , Femenino , Humanos , Persona de Mediana Edad , Metástasis de la Neoplasia , Estadificación de Neoplasias , Pronóstico , Curva ROC , Reproducibilidad de los Resultados , Transcriptoma , Flujo de Trabajo
16.
J Card Fail ; 23(5): 403-415, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28284757

RESUMEN

BACKGROUND: Stem cell transplantation offers a promising treatment for heart failure. Recent studies show that both c-kit positive cardiac stem cells (CSCs) and bone marrow-derived mesenchymal stem cells (BM-MSCs) are good candidates for stem cell therapy to treat heart failure; however, the exact mechanism of stem cell therapy in improving cardiac function of ischemic cardiomyopathy is not fully known. Our objective was to test our hypothesis that CSCs and/or BM-MSCs repair the damaged heart by boosting post-myocardial infarction (MI) angiogenesis in a paracrine manner. METHODS AND RESULTS: We isolated and purified CSCs and BM-MSCs from rats. Intramyocardial injections of CSCs and/or BM-MSCs were performed at 28 days after MI. We applied cardiac ultrasound and histological analysis to evaluate the effect of cell therapy on cardiac function and cardiac remodeling. In vivo donor cell transplantation experiments showed that CSCs and/or BM-MSCs improved cardiac function after MI and reduced infarct size. However, in vivo cell tracking experiments showed that minimal donor cells remained in the myocardium after cell transplantation. Our further in vitro and in vivo experiments showed that transplantation of CSCs enhanced the expression of pro-angiogenic factors and boosted post-MI angiogenesis in the myocardium in a paracrine manner, which in part contributed to the effect of CSCs on cardiac recovery after MI. CSCs and BM-MSCs synergistically inhibited CSC/BM-MSC apoptosis and enhanced their proliferation in a paracrine manner. This resulted in a larger number of transplanted cells remaining in the post-MI myocardium after coinjection of CSCs and BM-MSCs, and therefore the accumulation of more pro-angiogenic factors in the heart tissue compared to transplantation of CSCs or MSCs alone. Consequently, transplantation of both CSCs and BM-MSCs was superior to transplantation of either CSCs or BM-MSCs alone to boost post-MI angiogenesis and improve cardiac function after MI. CONCLUSION: C-kit+ CSC and/or BM-MSC transplantation can improve cardiac function after MI in a paracrine manner. Coinjection of both CSCs and BM-MSCs improves cardiac function more significantly than CSC or BM-MSC transplantation alone in a paracrine manner by improving the engraftment of donor cells and boosting the expression of multiple pro-angiogenic factors.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/métodos , Infarto del Miocardio/terapia , Neovascularización Fisiológica/fisiología , Comunicación Paracrina/fisiología , Proteínas Proto-Oncogénicas c-kit , Animales , Médula Ósea/química , Médula Ósea/fisiología , Proliferación Celular/fisiología , Femenino , Inyecciones Intraarteriales , Masculino , Infarto del Miocardio/diagnóstico por imagen , Proteínas Proto-Oncogénicas c-kit/análisis , Ratas , Ratas Endogámicas F344
17.
Int J Mol Sci ; 17(6)2016 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-27338347

RESUMEN

Cardiovascular disease is becoming the leading cause of death throughout the world. However, adult hearts have limited potential for regeneration after pathological injury, partly due to the quiescent status of stem/progenitor cells. Reactivation of cardiac stem/progenitor cells to create more myocyte progeny is one of the key steps in the regeneration of a damaged heart. In this study, miR-708 was identified to be enriched in the neonatal cardiomyocytes of rats, but this has not yet been proven in adult humans. A lower level of miR-708 in c-kit(+) stem/progenitor cells was detected compared to non-progenitors. Overexpression of miR-708 induced cardiomyocyte differentiation of cardiac stem/progenitor cells. This finding strengthened the potential of applying miRNAs in the regeneration of injured hearts, and this indicates that miR-708 could be a novel candidate for treatment of heart diseases.


Asunto(s)
Diferenciación Celular/genética , MicroARNs/genética , Mioblastos Cardíacos/citología , Mioblastos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Análisis por Conglomerados , Perfilación de la Expresión Génica , Miocardio/metabolismo , Miocitos Cardíacos/citología , Ratas , Regeneración , Transcriptoma
18.
J Cell Mol Med ; 18(10): 1992-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25092467

RESUMEN

Ischaemic cardiac arrhythmias cause a large proportion of sudden cardiac deaths worldwide. The ischaemic arrhythmogenesis is primarily because of the dysfunction and adverse remodelling of sarcolemma ion channels. However, the potential regulators of sarcolemma ion channel turnover and function in ischaemic cardiac arrhythmias remains unknown. Our previous studies indicate that dynamin-2 (DNM2), a cardiac membrane-remodelling GTPase, modulates ion channels membrane trafficking in the cardiomyocytes. Here, we have found that DNM2 plays an important role in acute ischaemic arrhythmias. In rat ventricular tissues and primary cardiomyocytes subjected to acute ischaemic stress, the DNM2 protein and transcription levels were markedly down-regulated. This DNM2 reduction was coupled with severe ventricular arrhythmias. Moreover, we identified that the down-regulation of DNM2 within cardiomyocytes increases the action potential amplitude and prolongs the re-polarization duration by depressing the retrograde trafficking of Nav1.5 and Kir2.1 channels. These effects are likely to account for the DNM2 defect-induced arrhythmogenic potentials. These results suggest that DNM2, with its multi-ion channel targeting properties, could be a promising target for novel antiarrhythmic therapies.


Asunto(s)
Arritmias Cardíacas/metabolismo , Dinamina II/metabolismo , Isquemia/metabolismo , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/patología , Biotinilación , Western Blotting , Células Cultivadas , Dinamina II/genética , Electrocardiografía , Electrofisiología , Técnicas para Inmunoenzimas , Isquemia/genética , Isquemia/patología , Masculino , Miocitos Cardíacos/patología , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canales de Potasio de Rectificación Interna/genética , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Molecules ; 19(6): 7122-37, 2014 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-24886939

RESUMEN

The miR-221/222 cluster has been demonstrated to function as oncomiR in human cancers. miR-221/222 promotes epithelial-to-mesenchymal transition (EMT) and confers tamoxifen resistance in breast cancer. However, the effects and mechanisms by which miR-221/222 regulates breast cancer aggressiveness remain unclear. Here we detected a much higher expression of miR-221/222 in highly invasive basal-like breast cancer (BLBC) cells than that in non-invasive luminal cells. A microRNA dataset from breast cancer patients indicated an elevated expression of miR-221/222 in BLBC subtype. S-phase entry of the cell cycle was associated with the induction of miR-221/222 expression. miRNA inhibitors specially targeting miR-221 or miR-222 both significantly suppressed cellular migration, invasion and G1/S transition of the cell cycle in BLBC cell types. Proteomic analysis demonstrated the down-regulation of two tumor suppressor genes, suppressor of cytokine signaling 1 (SOCS1) and cyclin-dependent kinase inhibit 1B (CDKN1B), by miR-221/222. This is the first report to reveal miR-221/222 regulation of G1/S transition of the cell cycle. These findings demonstrate that miR-221/222 contribute to the aggressiveness in control of BLBC.


Asunto(s)
Neoplasias de la Mama/metabolismo , Ciclo Celular/fisiología , Movimiento Celular/fisiología , MicroARNs/metabolismo , Fase S/fisiología , Neoplasias de la Mama/genética , Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , MicroARNs/genética , Fase S/genética
20.
Int Immunopharmacol ; 130: 111797, 2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38442582

RESUMEN

Cancer stem cells (CSCs) are known for their potent ability to drive tumor initiation and recurrence, yet the molecular mechanisms regulating CSCs are still unclear. Our study found a positive correlation between increased levels of miR-29a and better survival rates in early-stage breast cancer patients, but a negative correlation in late-stage patients, suggesting a dual function of miR-29a in regulating breast cancer. Furthermore, miR-29a showed significant downregulation in the ALDH+ breast cancer stem cell population compared to non-stem cancer cells. Overexpression of miR-29a in human breast cancer cells reduced the proportion of CSCs, suppressed their ability to form mammospheres, and inhibited the expression of stemness genes SOX2, KLF4, and hTERT in vitro. Conversely, knockdown of miR-29a in breast cancer cells showed opposite effects. Tumor xenograft experiments revealed that miR-29a overexpression significantly inhibited tumorigenesis initiated by MDA-MB-231 cell transplantation in nude mice. We further demonstrated that Krüppel-like factor 4 (KLF4), a key gene that regulates cell stemness, was a direct target of miR-29a in breast cancer cells. miR-29a suppressed the expression of KLF4 at both mRNA and protein levels. Reintroduction of KLF4 into breast cancer cells rescued the miR-29a-induced CSC suppression phenotype. In summary, our study is the first to demonstrate that miR-29a-KLF4 signaling inhibits breast tumor initiation by regulating CSCs, which provides novel therapeutic targets for preventing breast tumor initiation.


Asunto(s)
Neoplasias de la Mama , Factor 4 Similar a Kruppel , MicroARNs , Células Madre Neoplásicas , Animales , Femenino , Humanos , Ratones , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Factor 4 Similar a Kruppel/metabolismo , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA