Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Cell Mol Med ; 27(1): 127-140, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36528873

RESUMEN

Follistatin (FST) and activin A as gonadal proteins exhibit opposite effects on follicle-stimulating hormone (FSH) release from pituitary gland, and activin A-FST system is involved in regulation of decidualization in reproductive biology. However, the roles of FST and activin A in migration of decidualized endometrial stromal cells are not well characterized. In this study, transwell chambers and microfluidic devices were used to assess the effects of FST and activin A on migration of decidualized mouse endometrial stromal cells (d-MESCs). We found that compared with activin A, FST exerted more significant effects on adhesion, wound healing and migration of d-MESCs. Similar results were also seen in the primary cultured decidual stromal cells (DSCs) from uterus of pregnant mouse. Simultaneously, the results revealed that FST increased calcium influx and upregulated the expression levels of the migration-related proteins MMP9 and Ezrin in d-MESCs. In addition, FST increased the level of phosphorylation of JNK in d-MESCs, and JNK inhibitor AS601245 significantly attenuated FST action on inducing migration of d-MESCs. These data suggest that FST, not activin A in activin A-FST system, is a crucial chemoattractant for migration of d-MESCs by JNK signalling to facilitate the successful uterine decidualization and tissue remodelling during pregnancy.


Asunto(s)
Movimiento Celular , Endometrio , Folistatina , Sistema de Señalización de MAP Quinasas , Animales , Femenino , Ratones , Embarazo , Movimiento Celular/fisiología , Hormona Folículo Estimulante/metabolismo , Folistatina/genética , Folistatina/metabolismo , Células del Estroma/metabolismo , Útero/metabolismo , Endometrio/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología
2.
Genome Res ; 29(9): 1521-1532, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31315906

RESUMEN

Long noncoding RNAs (lncRNAs) can regulate the activity of target genes by participating in the organization of chromatin architecture. We have devised a "chromatin-RNA in situ reverse transcription sequencing" (CRIST-seq) approach to profile the lncRNA interaction network in gene regulatory elements by combining the simplicity of RNA biotin labeling with the specificity of the CRISPR/Cas9 system. Using gene-specific gRNAs, we describe a pluripotency-specific lncRNA interacting network in the promoters of Sox2 and Pou5f1, two critical stem cell factors that are required for the maintenance of pluripotency. The promoter-interacting lncRNAs were specifically activated during reprogramming into pluripotency. Knockdown of these lncRNAs caused the stem cells to exit from pluripotency. In contrast, overexpression of the pluripotency-associated lncRNA activated the promoters of core stem cell factor genes and enhanced fibroblast reprogramming into pluripotency. These CRIST-seq data suggest that the Sox2 and Pou5f1 promoters are organized within a unique lncRNA interaction network that determines the fate of pluripotency during reprogramming. This CRIST approach may be broadly used to map lncRNA interaction networks at target loci across the genome.


Asunto(s)
Cromatina/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , ARN Largo no Codificante/genética , Factores de Transcripción SOXB1/genética , Análisis de Secuencia de ARN/métodos , Animales , Sistemas CRISPR-Cas , Línea Celular , Reprogramación Celular , Fibroblastos/citología , Fibroblastos/metabolismo , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos
3.
Biol Chem ; 401(2): 297-308, 2020 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-31400749

RESUMEN

Interleukin-9 (IL-9) is a cytokine secreted by T-helper (Th)9 cells, and activin A can enhance Th9 cell differentiation. However, whether activin A affects IL-9 production by natural killer (NK) cells remains unclear. Herein, we found that not only Th cells, but also CD3-CD49b+NKp46+ NK cells of Balb/c mice produced IL-9. Although activin A promoted IL-9 expression in CD4+ Th cells, it inhibited IL-9 production by CD49b+NKp46+ NK cells in mice. Furthermore, the enzyme-linked immunosorbent assay (ELISA) results showed that mouse NK cells could secrete mature IL-9 protein, and activin A inhibited IL-9 release by NK cells. Additionally, activin A inhibited interferon (IFN)-γ production in splenic NK cells in mice, but promoted IL-2 production, and did not alter the production of IL-10. Western blotting results showed that levels of activin type IIA receptor (ActRIIA), Smad3 and phosphorylated-Smad3 (p-SMAD3) protein increased in activin A-treated splenic NK cells, compared with that in control NK cells. The inhibitory effects of activin A on IL-9 production by NK cells were attenuated in the presence of activin antagonist follistatin (FST) or Smad3 knockdown to NK cells. These data suggest that although activin A up-regulates IL-9 expression in Th cells, it inhibits IL-9 production in NK cells through Smad3 signaling.


Asunto(s)
Activinas/metabolismo , Interleucina-9/biosíntesis , Células Asesinas Naturales/metabolismo , Proteína smad3/metabolismo , Animales , Interleucina-9/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Transducción de Señal , Proteína smad3/genética
4.
Exp Cell Res ; 374(1): 114-121, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30458178

RESUMEN

Activin A, a multifunctional cytokine of transforming growth factor-ß (TGF-ß) superfamily, can be produced by the diverse immune cells. NK cells in peripheral blood are one of the major immune cells applied to cancer therapy in recent years. However, whether activin A can be produced by natural killer (NK) cells and be involved in regulation of peripheral blood NK cells activities of mouse are not well characterized. Here, we found that activin type IIA and IIB receptors and signaling molecules Smad2, 3 were expressed in peripheral blood NK cells of mouse by flow cytometry and RT-PCR. The cultured blood NK cells of mouse not only produced activin ßA chain protein by intracellular cytokine staining, but also secreted mature activin A protein by enzyme-linked immunosorbent assay (ELISA), and the production was promoted by IL-2. In addition, IL-2 as a positive control obviously promoted IFNγ production of mouse blood NK cells in vitro. However, activin A suppressed IFNγ production, but enhanced IL-2 synthesis and did not alter IL-10 production. Moreover, we found that activin A significantly suppressed the ability of NK cells to lyse target cells. These data revealed that blood NK cells of mouse were not only the target cells in response to activin A, but also the source of activin A, suggesting that activin A may play an important role in regulation of NK cells activities of mouse in an autocrine / paracrine manner.


Asunto(s)
Activinas/farmacología , Comunicación Autocrina , Células Asesinas Naturales/metabolismo , Comunicación Paracrina , Receptores de Activinas Tipo II/sangre , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Animales , Comunicación Autocrina/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Folistatina/farmacología , Subunidades beta de Inhibinas/sangre , Subunidades beta de Inhibinas/genética , Subunidades beta de Inhibinas/metabolismo , Interferón gamma/biosíntesis , Interleucina-10/biosíntesis , Interleucina-2/biosíntesis , Células Asesinas Naturales/efectos de los fármacos , Masculino , Ratones Endogámicos BALB C , Comunicación Paracrina/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Smad/sangre , Proteínas Smad/genética , Proteínas Smad/metabolismo
5.
J Nerv Ment Dis ; 207(8): 693-699, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31356409

RESUMEN

Postoperative cognitive dysfunction is a severe outcome after lung transplantation, especially in the elderly lung transplant recipients. Home-based computerized cognitive training (CCT) is a widely used intervention for cognition improvement, but its efficacy has not been validated in this population. A randomized controlled trial was conducted to analyze the effect of CCT on elderly lung transplant recipients. The participants received either an 8-week CCT intervention or usual care. The changes of cognitive function were assessed between preintervention (T1), postintervention (T2), and 12 weeks postintervention (T3). Among the 46 participants, 91.3% completed the interventions. The CCT group performed better than the control group on Digit-Span Forward Test (T3: p = 0.0044) and Verbal Fluency Test (T3: p = 0.0331), indicating the efficacy of CCT on verbal memory in the elderly lung transplant recipients. Although varied impacts were observed on different cognitive domains, it seems promising to use CCT on the elderly population after lung transplantation.


Asunto(s)
Remediación Cognitiva , Trasplante de Pulmón/efectos adversos , Complicaciones Cognitivas Postoperatorias/terapia , Terapia Asistida por Computador , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Evaluación de Resultado en la Atención de Salud
6.
Exp Cell Res ; 357(1): 107-115, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28479070

RESUMEN

Activin A belongs to the superfamily of transforming growth factor beta (TGFß) and is a critical regulatory cytokine in breast cancer and inflammation. However, the role of activin A in migration of breast cancer cells and immune cells was not well characterized. Here, a microfluidic device was used to examine the effect of activin A on the migration of human breast cancer cell line MDA-MB-231 cells and human blood neutrophils as well as their migratory interaction. We found that activin A promoted the basal migration but impaired epidermal growth factor (EGF)-induced migration of breast cancer cells. By contrast, activin A reduced neutrophil chemotaxis and transendothelial migration to N-Formyl-Met-Leu-Phe (fMLP). Finally, activin A promoted neutrophil chemotaxis to the supernatant from breast cancer cell culture. Collectively, our study revealed the different roles of activin A in regulating the migration of breast cancer cells and neutrophils and their migratory interaction. These findings suggested the potential of activin A as a therapeutic target for inflammation and breast cancers.


Asunto(s)
Activinas/metabolismo , Neoplasias de la Mama/metabolismo , Movimiento Celular/fisiología , Neutrófilos/metabolismo , Línea Celular Tumoral , Humanos , Inflamación/metabolismo , Neutrófilos/citología , Factor de Crecimiento Transformador beta/metabolismo
7.
J Cell Sci ; 125(Pt 6): 1500-7, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22275431

RESUMEN

Activin A, a member of the transforming growth factor ß (TGFß) superfamily, plays an essential role in neuron survival as a neurotrophic and neuroprotective factor in the central nervous system. However, the effects and mechanisms of action of activin A on the neurite outgrowth of dorsal root ganglia (DRG) remain unclear. In the present study, we found that activin A is expressed in DRG collected from chicken embryos on embryonic day 8 (E8). Moreover, activin A induced neurite outgrowth of the primary cultured DRG and maintained the survival of monolayer-cultured DRG neurons throughout the observation period of ten days. Follistatin (FS), an activin-binding protein, significantly inhibited activin A-induced neurite outgrowth of DRG, but failed to influence the effect of nerve growth factor (NGF) on DRG neurite outgrowth. Furthermore, the results showed that activin A significantly upregulated mRNA expression of activin receptor type IIA (ActRIIA) and calcitonin gene-related peptide (CGRP) in DRG, and stimulated serotonin (5-HT) production from DRG, indicating that activin A might induce DRG neurite outgrowth by promoting CGRP expression and stimulating 5-HT release. These data suggest that activin A plays an important role in the development of DRG in an autocrine or paracrine manner.


Asunto(s)
Activinas/fisiología , Ganglios Espinales/citología , Ganglios Espinales/embriología , Neuritas/fisiología , Células Receptoras Sensoriales/fisiología , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Animales , Comunicación Autocrina/fisiología , Péptido Relacionado con Gen de Calcitonina/genética , Péptido Relacionado con Gen de Calcitonina/metabolismo , Embrión de Pollo , Pollos , Medios de Cultivo/farmacología , Folistatina/metabolismo , Ganglios Espinales/fisiología , Factor de Crecimiento Nervioso/metabolismo , Comunicación Paracrina/fisiología , Cultivo Primario de Células , Células Receptoras Sensoriales/citología , Serotonina/metabolismo
8.
Tumour Biol ; 35(12): 11781-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25190016

RESUMEN

To explore the antitumor effect of caffeic acid 3,4-dihydroxy-phenethyl ester (CADPE) on the breast cancer cell lines and illuminate the related mechanism. After treatment with different concentrations of CADPE for 24, 48, and 72 h, cell proliferation ability of the breast cancer cell lines MDA-MB-231 and MDA-MB-435 was analyzed by the MTT. Changes of the cell cycles were evaluated by PI staining. Cell apoptosis was examined by flow cytometry after Annexin V/7AAD double staining. Nuclear morphologic changes were observed under the inverted fluorescence microscope after staining with Hoechst 33342. Mitochondrial membrane potential and reactive oxygen species (ROS) level were estimated by JC-1 and DCFH-DA staining. In addition, the expression level of mitochondrial signaling pathway proteins Bcl-2, Bax, and caspase-3 were evaluated by Western blot. CADPE has the distinct cytotoxic effect to the breast cancer cells, and the effect is dose dependent. It did not change the cell cycles but induced the cell apoptosis of the breast cancer cells. At the same time, after CADPE treatment, the expression levels of caspase-3 and Bax in the breast cancer cells were upregulated and Bcl-2 expression was declined. The ROS level in the breast cancer cells was enhanced, and mitochondrial membrane potential of the cells was downregulated. CADPE has the antitumor functions. It can induce the cell apoptosis through downregulating Bcl-2 expression, enhancing Bax and caspase-3 expression levels, upregulating ROS level and reducing the mitochondrial membrane potential of the breast cancer cells to trigger the mitochondrial signal pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Ácidos Cafeicos/farmacología , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína X Asociada a bcl-2/metabolismo
9.
Oncol Rep ; 51(2)2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38131250

RESUMEN

Activin A, a member of the transforming growth factor­ß (TGF­ß) superfamily, has been implicated in the tumorigenesis and progression of various cancers. However, it remains unclear whether activin A induces apoptosis in human lung adenocarcinoma cells through the endoplasmic reticulum (ER) stress pathway. In the present study, BrdU, flow cytometry and western blotting were used to examine cell proliferation, apoptosis and protein expression, respectively. The present study revealed that activin A inhibited human lung adenocarcinoma A549 cell proliferation, induced apoptosis, and upregulated the protein levels of C/EBP homologous protein (CHOP), growth arrest and DNA damage­inducible protein 34 (GADD34), cleaved­caspase­3 and caspase­12. Furthermore, the administration of activin A did not alter the levels of suppressor of mothers against decapentaplegic 3 (Smad3) or phosphorylated (p)­Smad3 proteins, whereas, it significantly elevated the levels of ActRIIA and p­extracellular signal regulated kinase proteins 1 and 2 (ERK1/2) proteins in A549 cells. The apoptotic effects of activin A on A549 cells were attenuated by the ERK inhibitor FR180204, which also downregulated CHOP and caspase­12 protein levels. Additionally, activin A increased intracellular calcium flux in A549 cells, and the calcium ion chelator BAPTA acetoxymethyl ester (BAPTA­AM) inhibited activin A­induced A549 cell apoptosis, whereas the calcium agonist ionomycin significantly increased apoptosis of A549 cells induced by activin A. These findings indicated that the activation of the ER stress pathway resulting in apoptosis of A549 cells triggered by activin A is facilitated by the ActRIIA­ERK1/2 signaling and calcium signaling. The present findings suggest that the agonists of ERK and calcium signaling exhibit promising clinical therapeutic potential for the induction of apoptosis in lung adenocarcinoma.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Humanos , Células A549 , Calcio/metabolismo , Caspasa 12 , Línea Celular Tumoral , Apoptosis , Adenocarcinoma del Pulmón/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Estrés del Retículo Endoplásmico
10.
Cells ; 13(9)2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38727264

RESUMEN

Natural killer (NK) cells can migrate quickly to the tumor site to exert cytotoxic effects on tumors, and some chemokines, including CXCL8, CXCL10 or and CXCL12, can regulate the migration of NK cells. Activin A, a member of the transforming growth factor ß (TGF-ß) superfamily, is highly expressed in tumor tissues and involved in tumor development and immune cell activation. In this study, we focus on the effects of activin A on NK cell migration. In vitro, activin A induced NK cell migration and invasion, promoted cell polarization and inhibited cell adhesion. Moreover, activin A increased Ca2+, p-SMAD3 and p-AKT levels in NK cells. An AKT inhibitor and Ca2+ chelator partially blocked activin A-induced NK cell migration. In vivo, exogenous activin A increased tumor-infiltrating NK cells in NS-1 cell solid tumors and inhibited tumor growth, and blocking endogenous activin A with anti-activin A antibody reduced tumor-infiltrating NK cells in 4T-1 cell solid tumors. These results suggest that activin A induces NK cell migration through AKT signaling and calcium signaling and may enhance the antitumor effect of NK cells by increasing tumor-infiltrating NK cells.


Asunto(s)
Activinas , Señalización del Calcio , Movimiento Celular , Células Asesinas Naturales , Proteínas Proto-Oncogénicas c-akt , Animales , Ratones , Activinas/metabolismo , Activinas/farmacología , Señalización del Calcio/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/inmunología , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo
11.
Biomicrofluidics ; 18(3): 031506, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38899164

RESUMEN

The global impact of cancer on human health has raised significant concern. In this context, the tumor microenvironment (TME) plays a pivotal role in the tumorigenesis and malignant progression. In order to enhance the accuracy and efficacy of therapeutic outcomes, there is an imminent requirement for in vitro models that can accurately replicate the intricate characteristics and constituents of TME. Microfluidic devices exhibit notable advantages in investigating the progression and treatment of tumors and have the potential to become a novel methodology for evaluating immune cell activities in TME and assist clinicians in assessing the prognosis of patients. In addition, it shows great advantages compared to traditional cell experiments. Therefore, the review first outlines the applications and advantages of microfluidic chips in facilitating tumor cell culture, constructing TME and investigating immune cell activities. Second, the roles of microfluidic devices in the analysis of circulating tumor cells, tumor prognosis, and drug screening have also been mentioned. Moreover, a forward-looking perspective is discussed, anticipating the widespread clinical adoption of microfluidic devices in the future.

12.
Biosens Bioelectron ; 260: 116460, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-38843769

RESUMEN

Neutrophils need to migrate through tight tissue spaces to eliminate pathogens, but their movement is often hindered by their large and stiff nuclei. Neutrophil migration is impaired in sepsis patients, but it is unclear whether this defect is related to the deformability of their nuclei. Herein, we designed microfluidic devices with micron-scale narrow slits to simulate biological barriers. This setup allowed us to observe and record neutrophil movement and nuclear deformation in real-time. We also developed a method for morphological analysis to quantify nucleus deformation in numerous individual cells. Our studies showed that neutrophils from healthy individuals could adjust their nuclear shape to squeeze through these constrictions, whereas those from sepsis patients demonstrated less flexibility. Neutrophils with rigid nuclei struggled to pass through narrow gaps and were more likely to rupture under pressure. These findings suggest that the migration defects of neutrophils observed in sepsis may be attributed to the inability of neutrophils to deform their nuclei, highlighting the crucial role of microfluidic technologies in offering new insights into migration defects under pathological conditions.


Asunto(s)
Movimiento Celular , Dispositivos Laboratorio en un Chip , Neutrófilos , Sepsis , Humanos , Neutrófilos/citología , Técnicas Biosensibles/instrumentación , Diseño de Equipo , Núcleo Celular
13.
Biochem Biophys Res Commun ; 435(2): 222-8, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23665022

RESUMEN

Activin A, a member of TGF-ß superfamily, is involved in either pro-inflammatory or anti-inflammatory responses. Our previous studies have reported that lipopolysaccharide (LPS) can simulate activin A secretion from macrophage, and activin A can induce rest macrophage activation in mice, but inhibit the activities of the activated macrophages. However, the relationship of activin and LPS actions and their mechanism are not well characterized. In the present study, the results showed that both activin A and LPS promoted the phagocytic activities of mouse peritoneal macrophages in vivo and in vitro, but activin A inhibited the phagocytosis of LPS-activated macrophages. Simultaneously, the results revealed that activin A inhibited the Toll-like receptor 4 (TLR4) expression on LPS-activated mouse peritoneal macrophages in vivo and in vitro, whereas there was no obvious change of TLR2 expression. Moreover, the results showed that activin A obviously reduced the TLR4 mRNA and protein expressions in LPS-activated macrophage cell line RAW264.7 cells, and the inhibitory effect of activin A on the TLR4 expression was significantly attenuated in Smad3 knock-down RAW264.7 cells. Interestingly, LPS promoted the expression of activin type IIA receptor (ActRIIA) on mouse peritoneal macrophages in vivo, and also up-regulated ActRIIA and activin signal molecules Smad2, 3 mRNA expressions. These data suggest that activin A inhibits LPS action on macrophages in vivo via suppressing TLR4 expression, and LPS further augments the negative feedback action of activin A via up-regulating activin signaling transduction.


Asunto(s)
Activinas/farmacología , Lipopolisacáridos , Activación de Macrófagos/fisiología , Macrófagos/fisiología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Células Cultivadas , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C
14.
Cells ; 11(23)2022 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-36497076

RESUMEN

Follistatin (FST) as a gonadal protein is central to the establishment and maintenance of pregnancy. Trophoblasts' migration and invasion into the endometrium are critical events in placental development. This study aimed to elucidate the role of FST in the migration and invasion of placental trophoblasts of mice. We found that FST increased the vitality and proliferation of primary cultured trophoblasts of embryonic day 8.5 (E8.5) mice and promoted wound healing of trophoblasts. Moreover, FST significantly induced migration of trophoblasts in a microfluidic device and increased the number of invasive trophoblasts by Matrigel-coated transwell invasion assay. Being treated with FST, the adhesion of trophoblasts was inhibited, but intracellular calcium flux of trophoblasts was increased. Western blotting results showed that FST had no significant effects on the level of p-Smad3 or the ratio of p-Smad3/Smad3 in trophoblasts. Interestingly, FST elevated the level of p-JNK; the ratio of p-JNK/JNK; and expression of migration-related proteins N-cadherin, vimentin, ezrin and MMP2 in trophoblasts. Additionally, the migration of trophoblasts and expression of N-cadherin, vimentin, and MMP2 in trophoblasts induced by FST were attenuated by JNK inhibitor AS601245. These findings suggest that the elevated FST in pregnancy may act as a chemokine to induce trophoblast migration and invasion through the enhanced JNK signaling to maintain trophoblast function and promote placental development.


Asunto(s)
Factores Quimiotácticos , Folistatina , Placenta , Animales , Femenino , Ratones , Embarazo , Cadherinas/metabolismo , Movimiento Celular , Factores Quimiotácticos/metabolismo , Placenta/metabolismo , Placentación , Trofoblastos/metabolismo , Folistatina/metabolismo
15.
J Neuroimmunol ; 371: 577929, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-35939944

RESUMEN

BACKGROUND: Microglia are involved in many neurodegenerative diseases and repairment of traumatic injury to the CNS. Activin A is a neurotrophic and neuroprotective factor that can regulate the activities of macrophages/microglia. However, the effects of activin A on the migration of microglia are still unclear. In this study, the role of activin A in regulation of the microglia migration was investigated with the murine microglial BV2 cell. METHODS: The levels of cytokines were detected by enzyme-linked immunosorbent assay (ELISA). The protein expression was examined by Western blotting. The adhesion of BV2 cells was assayed by real-time cell analysis (RTCA). The migration of BV2 cells was determined by transwell chamber and microfluidics device. Smad3 was overexpressed or knocked down in BV2 cells by transfection of Smad3 or Smad3 shRNA-expressing plasmids. RESULTS: Activin A inhibited the release of nitric oxide (NO) and inflammatory cytokines of TNF-α and IL-6 and the expression of TNF-α and IL-6 mRNA by BV2 cells. In contrast, activin A promoted the production of TGF-ß1. Activin A inhibited adhesion, promoted wound healing and migration which is related to the expression of N-cadherin and E-cadherin expression. Additionally, Smad3 overexpression in BV2 cells decreased the levels of TNF-α and IL-6, and promoted the wound healing, whereas Smad3 knockdown showed the opposite effects. CONCLUSIONS: These findings revealed that activin A regulated the biological behavior of BV2 cells via Smad3 signaling, suggesting that activin A may serve as a potential treatment target for neuroinflammation and glia scar formation in nervous system.


Asunto(s)
Microglía , Factor de Necrosis Tumoral alfa , Activinas , Animales , Factores Quimiotácticos , Citocinas/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Ratones , Microglía/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
16.
Cells ; 11(19)2022 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-36231092

RESUMEN

Recurrent spontaneous abortion (RSA) is a highly heterogeneous complication of pregnancy with the underlying mechanisms remaining uncharacterized. Dysregulated decidualization is a critical contributor to the phenotypic alterations related to pregnancy complications. To understand the molecular factors underlying RSA, we explored the role of longnoncoding RNAs (lncRNAs) in the decidual microenvironment where the crosstalk at the fetal-maternal interface occurs. By exploring RNA-seq data from RSA patients, we identified H19, a noncoding RNA that exhibits maternal monoallelic expression, as one of the most upregulated lncRNAs associated with RSA. The paternally expressed fetal mitogen IGF2, which is reciprocally coregulated with H19 within the same imprinting cluster, was also upregulated. Notably, both genes underwent loss of imprinting, as H19 and IGF2 were actively transcribed from both parental alleles in some decidual tissues. This loss of imprinting in decidual tissues was associated with the loss of the H3K27m3 repressive histone marker in the IGF2 promoter, CpG hypomethylation at the central CTCF binding site in the imprinting control center (ICR), and the loss of CTCF-mediated intrachromosomal looping. These data suggest that dysregulation of the H19/IGF2 imprinting pathway may be an important epigenetic factor in the decidual microenvironment related to poor decidualization.


Asunto(s)
Histonas , ARN Largo no Codificante , Factor de Unión a CCCTC/genética , Metilación de ADN/genética , Femenino , Impresión Genómica , Histonas/metabolismo , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Mitógenos , Embarazo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN no Traducido/genética
17.
iScience ; 24(2): 102080, 2021 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-33604525

RESUMEN

Activin A levels are elevated during multiple severe infections and associated with an increased risk of death. However, the role of activin A in bacterial infection is still unclear. Here, we found that activin A levels were increased during S. aureus skin infection in mice. Administration of activin A increased the bacterial burden and promoted the spread of bacteria in vivo. Moreover, activin A inhibited neutrophil chemotaxis to N-formylmethionine-leucyl-phenylalanine via the type IIA activin receptor (ActRIIA) in vitro and impaired ActRIIA+ neutrophil recruitment to infection foci in vivo. Additionally, we identified a novel subpopulation of neutrophils, ActRIIA+ neutrophils, which exhibit superior phagocytic capacity compared to ActRIIA- neutrophils and possess an N2-like immunoregulatory activity via secreting IL-10 and TGF-ß. Taken together, these findings indicate that activin A inhibits the recruitment of ActRIIA+ neutrophils to infected foci, leading to the impairment of bacterial clearance, and thus may hamper early infection control.

18.
Front Cell Dev Biol ; 9: 660316, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34095123

RESUMEN

Activin A, a member of the transforming growth factor-beta (TGF-ß) superfamily, contributes to tissue healing and fibrosis. As the innate tissue cells, fibroblasts also play an important role in wound healing and fibrosis. Herein, this study was aimed to investigate how activin A exhibited regulatory effects on adhesion and migration of fibroblasts. We found that activin A induced the migration of fibroblast cell line L929 cells in transwell chamber and microfluidic device. Activin A also promoted L929 cells adhesion, but did not affect L929 cells viability or proliferation. In addition, activin A induced α-SMA expression and TGF-ß1 release, which were factors closely related to tissue fibrosis, but had no effect on IL-6 production, a pro-inflammatory cytokine. Furthermore, activin A elevated calcium levels in L929 cells and increased p-ERK protein levels. Activin A-induced migration of L929 cells was attenuated by ERK inhibitor FR180204. To conclude, these data indicated that activin A as a novel chemokine induced the chemotactic migration of L929 cells via ERK signaling and possessed the pro-fibrosis role. These findings provide a new insight into understanding of activin A in tissue fibrosis.

19.
Clin Breast Cancer ; 20(5): 422-430, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32741666

RESUMEN

BACKGROUND: Breast cancer is the most commonly diagnosed cancer and the leading cause of cancer death among females. Circular RNAs (circRNAs) have been implicated in the initiation and development of cancer. Here, we explored the biological role and regulatory mechanism of circCDYL in breast cancer. MATERIALS AND METHODS: The expression and correlation of circCDYL/miR-190a-3p/TP53INP1 axis in breast cancer tissues and cells were determined by quantitative polymerase chain reaction and Western blot. Cell-counting Kit-8, colony formation, cell migration, and invasion assays were applied to investigate the biological roles of circCDYL in breast cancer development and progression. RESULTS: CircCDYL were down-regulated in breast cancer tissues and cells, the expression of which positively correlated with patients' survival rate. CircCDYL worked as a "sponge," binding to miR-190a-3p directly, which inhibited the expression of miR-190a-3p and relieved the inhibition of tumor suppressor gene TP53INP1. CONCLUSION: CircCDYL promotes apoptosis and inhibits proliferation of the malignant phenotype of breast cancer through regulating miR-190a-3p/TP53INP1 axis, which suggests that circCDYL is a potential therapeutic target for breast cancer.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Co-Represoras/metabolismo , Proteínas de Choque Térmico/metabolismo , Hidroliasas/metabolismo , MicroARNs/metabolismo , ARN Circular/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Apoptosis/fisiología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proteínas Portadoras/genética , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Proteínas Co-Represoras/genética , Proteínas de Choque Térmico/genética , Humanos , Hidroliasas/genética , MicroARNs/genética , Pronóstico , ARN Circular/genética , Tasa de Supervivencia , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Células Tumorales Cultivadas
20.
Sci Rep ; 10(1): 20623, 2020 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-33244088

RESUMEN

Fibroblasts play an important role in inflammation and tissue fibrosis. Both activin A and TNF-α can activate immune cells, however, the roles and relationship of them in activating fibroblasts in inflammation remain unclear. Here, this study revealed that TNF-α promoted the release of NO and IL-6 by L929 fibroblast cells, but co-treatment with activin A attenuated these effects. In contrast, activin A induced cell migration and increased the production of tissue fibrosis-related TGF-ß1 and fibronectin, while TNF-α inhibited these function changes of L929 cells induced by activin A. Moreover, this study revealed that activin A and TNF-α regulated the activities of L929 cells via ERK1/2/MAPK pathway, rather than Smad3-dependent signaling pathway. Taken together, these data indicate that activin A and TNF-α exert mutually antagonistic effects on regulating fibroblasts activities, and the balance between their action may determine the process and outcome of fibroblasts-mediated inflammation.


Asunto(s)
Activinas/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína smad3/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Fibronectinas/metabolismo , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA