Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
BMB Rep ; 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38919016

RESUMEN

Early proatherogenic inflammation constitutes a significant risk factor for atherogenesis development. Despite this, the precise molecular mechanisms driving this pathological progression largely remain elusive. Our study unveils a pivotal role for the microRNA miR-328-5p in dampening endothelial inflammation by modulating the stability of JUNB (JunB proto-oncogene). Perturbation of miR-328-5p levels results in heightened monocyte adhesion to endothelial cells and enhanced transendothelial migration, while its overexpression mitigates these inflammatory processes. Furthermore, miR-328-5p hinders macrophage polarization toward the pro-inflammatory M1 phenotype, and exerts a negative influence on atherosclerotic plaque formation in vivo. By pinpointing JUNB as a direct miR-328-5p target, our research underscores the potential of miR-328-5p as a therapeutic target for inflammatory atherosclerosis. Reintroduction of JUNB effectively counteracts the anti-atherosclerotic effects of miR-328-5p, highlighting the promise of pharmacological miR-328-5p targeting in managing inflammatory atherosclerosis.

2.
Mol Biol Rep ; 51(1): 680, 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38796595

RESUMEN

Menstrual blood-derived endometrial stem cells (MenSCs) have attracted increasing interest due to their excellent safety, and lack of ethical dilemma as well as their ability to be periodically obtained in a noninvasive manner. However, although preclinical research as shown the therapeutic potential of MenSCs in several diseases, their poor cell survival and low engraftment at disease sites reduce their clinical efficacy. Flotillins (including Flot1 and Flot2) are implicated in various cellular processes, such as vesicular trafficking, signal transduction, cell proliferation, migration and apoptosis. In this study, we aimed to determine the effects of Flotillins on MenSCs survival, proliferation and migration. Our experimental results show that MenSCs were modified to overexpress Flot1 and/or Flot2 without altering their intrinsic characteristics. Flot1 and Flot2 co-overexpression promoted MenSC viability and proliferation capacity. Moreover, Flot1 or Flot2 overexpression significantly promoted the migration and inhibited the apoptosis of MenSCs compared with the negative control group, and these effects were stronger in the Flot1 and Flot2 gene co-overexpression group. However, these effects were significantly reversed after Flot1 and/or Flot2 knockdown. In conclusion, our results indicate that Flot1 and Flot2 overexpression in MenSCs improved their proliferation and migration and inhibited their apoptosis, and this might be an effective approach to improve the efficiency of cell-based therapies.


Asunto(s)
Apoptosis , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Proteínas de la Membrana , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Femenino , Endometrio/citología , Endometrio/metabolismo , Células Madre/metabolismo , Células Madre/citología , Células Cultivadas , Transducción de Señal
3.
BMC Ophthalmol ; 23(1): 286, 2023 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-37353739

RESUMEN

BACKGROUND: The pathological basis for many retinal diseases, retinal ischemia is also one of the most common causes of visual impairment. Numerous ocular diseases have been linked to Endoplasmic reticulum(ER)stress. However, there is still no clear understanding of the relationship between ER stress and Müller glial cells during retinal ischemia and hypoxia. This study examined the effects of ER stress on autophagy and apoptosis-related proteins, as well as the microtubule-related protein tau in rMC-1 cells. METHODS: rMC-1 cells were cultured in vitro. RT-PCR、immunofluorescence and Western blotting revealed the expression levels of associated mRNAs and proteins, and the CCK-8 and flow cytometry assays detected cell apoptosis. RESULTS: The results showed that under OGD(Oxygen-glucose deprivation) conditions, the number of rMC-1 cells was decreased, the PERK/eIF2a pathway was activated, and the expressions of p-tau, LC3、Beclin1 and Caspase-12 proteins were increased. After the PERK knockout, the expression of the above proteins was decreased, and the apoptosis was also decreased. CONCLUSION: According to the findings of this study, specific downregulation of PERK expression had an anti-apoptotic effect on OGD-conditioned rMC-1 cells. There is a possibility that this is one of the mechanisms of MG cell apoptosis during retinal ischemic injury.


Asunto(s)
Células Ependimogliales , Transducción de Señal , Ratas , Animales , Ratas Sprague-Dawley , Apoptosis , Estrés del Retículo Endoplásmico
4.
Mol Ther ; 31(5): 1365-1382, 2023 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-36733250

RESUMEN

Mesenchymal stem cells regulate remote intercellular signaling communication via their secreted extracellular vesicles. Here, we report that menstrual blood-derived stem cells alleviate acute lung inflammation and injury via their extracellular vesicle-transmitted miR-671-5p. Disruption of this abundantly expressed miR-671-5p dramatically reduced the ameliorative effect of extracellular vesicles released by menstrual blood-derived stem cells on lipopolysaccharide (LPS)-induced pulmonary inflammatory injury. Mechanistically, miR-671-5p directly targets the kinase AAK1 for post-transcriptional degradation. AAK1 is found to positively regulate the activation of nuclear factor κB (NF-κB) signaling by controlling the stability of the inhibitory protein IκBα. This study identifies a potential molecular basis of how extracellular vesicles derived from mesenchymal stem cells improve pulmonary inflammatory injury and highlights the functional importance of the miR-671-5p/AAK1 axis in the progression of pulmonary inflammatory diseases. More importantly, this study provides a promising cell-based approach for the treatment of pulmonary inflammatory disorders through an extracellular vesicle-dependent pathway.


Asunto(s)
Vesículas Extracelulares , Lesión Pulmonar , MicroARNs , Neumonía , Humanos , FN-kappa B/genética , FN-kappa B/metabolismo , Inflamación/genética , Inflamación/terapia , Inflamación/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Neumonía/genética , Neumonía/terapia , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Lipopolisacáridos/farmacología , Proteínas Serina-Treonina Quinasas
5.
Nucleic Acids Res ; 51(5): 2195-2214, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36794705

RESUMEN

NF-κB activates the primary inflammatory response pathway responsible for methicillin-resistant Staphylococcus aureus (MRSA)-induced lung inflammation and injury. Here, we report that the Forkhead box transcription factor FOXN3 ameliorates MRSA-induced pulmonary inflammatory injury by inactivating NF-κB signaling. FOXN3 competes with IκBα for binding to heterogeneous ribonucleoprotein-U (hnRNPU), thereby blocking ß-TrCP-mediated IκBα degradation and leading to NF-κB inactivation. FOXN3 is directly phosphorylated by p38 at S83 and S85 residues, which induces its dissociation from hnRNPU, thus promoting NF-κB activation. After dissociation, the phosphorylated FOXN3 becomes unstable and undergoes proteasomal degradation. Additionally, hnRNPU is essential for p38-mediated FOXN3 phosphorylation and subsequent phosphorylation-dependent degradation. Functionally, genetic ablation of FOXN3 phosphorylation results in strong resistance to MRSA-induced pulmonary inflammatory injury. Importantly, FOXN3 phosphorylation is clinically positively correlated with pulmonary inflammatory disorders. This study uncovers a previously unknown regulatory mechanism underpinning the indispensable role of FOXN3 phosphorylation in the inflammatory response to pulmonary infection.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Neumonía , Humanos , FN-kappa B/genética , FN-kappa B/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , Fosforilación , Proteínas I-kappa B , Staphylococcus aureus Resistente a Meticilina/metabolismo , Transducción de Señal , Neumonía/genética , Proteínas de Ciclo Celular/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo
6.
Diabetes Res Clin Pract ; 195: 110201, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36493913

RESUMEN

Chronic wounds fail to heal through the three normal stages of healing (inflammatory, proliferative, and remodelling), resulting in a chronic tissue injury that is not repaired within the average time limit. Patients suffering from type 1 and type 2 diabetes are prone to develop diabetic foot ulcers (DFUs), which commonly develop into chronic wounds that are non treatable with conventional therapies. DFU develops due to various risk factors, such as peripheral neuropathy, peripheral vascular disease, arterial insufficiency, foot deformities, trauma and impaired resistance to infection. DFUs have gradually become a major problem in the health care system worldwide. In this review, we not only focus on the pathogenesis of DFU but also comprehensively summarize the outcomes of preclinical and clinical studies thus far and the potential therapeutic mechanism of bone marrow-derived mesenchymal stem cells (BMSCs) for the treatment of DFU. Based on the published results, BMSC transplantation can contribute to wound healing through growth factor secretion, anti-inflammation, differentiation into tissue-specific cells, neovascularization, re-epithelialization and angiogenesis in DFUs. Moreover, clinical trials showed that BMSC treatment in patients with diabetic ulcers improved ulcer healing and the ankle-brachial index, ameliorated pain scores, and enhanced claudication walking distances with no reported complications. In conclusion, although BMSC transplantation exhibits promising therapeutic potential in DFU treatment, additional studies should be performed to confirm their efficacy and long-term safety in DFU patients.


Asunto(s)
Diabetes Mellitus Tipo 2 , Pie Diabético , Células Madre Mesenquimatosas , Humanos , Pie Diabético/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/terapia , Médula Ósea/patología , Cicatrización de Heridas
7.
Int J Pharm ; 592: 120037, 2021 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-33161038

RESUMEN

Multiple signaling pathways are usually involved in the development of tumors. Compared with monospecific antibodies, bispecific antibodies can recognize two different antigens at the same time, so they are more suitable for treating tumor diseases with complex etiology. Immunotoxins have good antitumor activity, however, single targeting limits their effectiveness. Herein, we designed a Pseudomonas exotoxin A (PE)-based bispecific immunotoxin IgBD-HER2-PDGFRß-PE38 which could distinguish HER2 and PDGFRß target in tumor. Meanwhile, IgG-affinity could extend the serum retention of immunotoxins after in vivo injection. In this work, we first detected the selective binding of the immunotoxins and antitumor effect in vitro. Compared with control group, IgBD-HER2-PDGFRß-PE38 exhibited improved efficacy against HER2-positive tumors in an NCI-N87 subcutaneous xenograft model. Then, transcriptome sequencing was performed on tumor tissue originating from different treatment groups of mice bearing NCI-N87 tumors. Seven significantly differentially expressed genes were screened based on human genes, and the differential mouse genes were enriched based on the Reactome Pathway Database. At last, the RNA sequencing results were verified by real-time PCR and ELISA. Therefore, the new construct bispecific immunotoxin represents a potentially attractive therapeutic modality, and the proposed strategy make them promising for use in the development of anti-HER2 cancer therapeutics.


Asunto(s)
Inmunotoxinas , Neoplasias , Animales , Semivida , Xenoinjertos , Ratones , Ratones Desnudos , Receptor ErbB-2/genética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Am J Transl Res ; 12(11): 7127-7143, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33312355

RESUMEN

Selaginella tamariscina (ST), a well-known traditional medicinal plant, has been used to treat various cancers, including pancreatic cancer. However, the underlying mechanism by which Selaginellin B, a natural pigment isolated and purified from ST, protects against pancreatic cells has yet to be fully elucidated. In the present study, the biological functions of Selaginellin B were investigated using apoptosis, migration and colony formation assays in ASPC-1 and PANC-1 cells. In addition, apoptosis-associated proteins were detected by Western blotting. Our results demonstrated that Selaginellin B induced apoptosis, as evidenced by the increased cleaved caspase-3 level and Bax/Bcl-2 ratio. Moreover, Selaginellin B led to a marked up-regulation of the ratio of LC3-II/LC3-I in ASPC-1 and PANC-1 cells, respectively. Furthermore, reverse pharmacophore screening, molecular docking and molecular dynamics simulation studies revealed that Janus kinase 2 (JAK2) may be a potential target for Selaginellin B. In summary, the results of the present research have demonstrated that Selaginellin B is an effective anticancer agent against PANC-1 and ASPC-1 cells, and the compound holds great promise for the treatment of pancreatic cancer.

9.
Genes (Basel) ; 11(11)2020 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-33113878

RESUMEN

Lateral root development is a complex process regulated by numerous factors. An important role for sugar in lateral root development has been known for a while, but the underlying molecular basis still remains unclear. In this study, we first showed that WOX7, a sugar-inducible negative regulator of lateral root development, acts downstream of the glucose sensor HXK1. Using a transgenic line homozygous for a transgene expressing GFP under the control of the WOX7 promoter, we next performed a genetic screen to identify additional genes in this development pathway. A number of mutants with altered level of WOX7 expression were recovered, and two with increased WOX7 expression, named ewe-1 and ewe-2 (for Enhanced WOX7 Expression), were further characterized. Both mutants manifest delayed lateral root development, and genetic analysis indicates that single recessive mutations are responsible for the observed phenotypes. The mutations were then located to similar regions on chromosome 2 by marker-assisted analyses, and candidate genes were identified through whole genome sequencing. The significance and limitations of this work are discussed.


Asunto(s)
Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Arabidopsis/genética , Glucosa/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Raíces de Plantas/crecimiento & desarrollo , Arabidopsis/crecimiento & desarrollo , Arabidopsis/metabolismo , Genoma de Planta/genética , Hexoquinasa/genética , Regiones Promotoras Genéticas/genética , Proteínas RGS/genética , Secuenciación Completa del Genoma
10.
Stem Cell Res Ther ; 11(1): 192, 2020 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-32448377

RESUMEN

Acute lung injury (ALI), an increasingly devastating human disorder, is characterized by a multitude of lung changes arising from a wide variety of lung injuries. Viral infection is the main cause of morbidity and mortality in ALI and acute respiratory distress syndrome (ARDS) patients. In particular, influenza virus, coronavirus, and other respiratory viruses circulate in nature in various animal species and can cause severe and rapidly spread human infections. Although scientific advancements have allowed for rapid progress to be made to understand the pathogenesis and develop therapeutics after each viral pandemic, few effective methods to treat virus-induced ALI have been described. Recently, stem cell therapy has been widely used in the treatment of various diseases, including ALI. In this review, we detail the present stem cell-based therapeutics for lung injury caused by influenza virus and the outlook for the future state of stem cell therapy to deal with emerging influenza and coronaviruses.


Asunto(s)
Lesión Pulmonar Aguda/terapia , Coronavirus/patogenicidad , Orthomyxoviridae/patogenicidad , Trasplante de Células Madre , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/virología , Betacoronavirus/patogenicidad , Tratamiento Basado en Trasplante de Células y Tejidos , Citocinas/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , SARS-CoV-2
11.
12.
Mol Cell Biochem ; 463(1-2): 203-210, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31643040

RESUMEN

As a m6A methylation modifier, METTL3 is functionally involved in various biological processes. Nevertheless, the role of METTL3 in osteogenesis is not determined up to date. In the current study, METTL3 is identified as a crucial regulator in the progression of osteogenic differentiation. Loss of METTL3 significantly augments calcium deposition and enhances alkaline phosphatase activity of mesenchymal stem cells, uncovering an inhibitory role of METTL3 in osteogenesis. More importantly, the underlying molecular basis by which METTL3 regulates osteogenesis is illustrated. We find that METTL3 positively regulates expression of MYD88, a critical upstream regulator of NF-κB signaling, by facilitating m6A methylation modification to MYD88-RNA, subsequently inducing the activation of NF-κB which is widely regarded as a repressor of osteogenesis and therefore suppressing osteogenic progression. Moreover, the METTL3-mediated m6A methylation is found to be dynamically reversed by the demethylase ALKBH5. In summary, this study highlights the functional importance of METTL3 in osteogenic differentiation and METTL3 may serve as a promising molecular target in regenerative medicine, as well as in the field of bone tissue engineering.


Asunto(s)
Desmetilasa de ARN, Homólogo 5 de AlkB/metabolismo , Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Metiltransferasas/metabolismo , FN-kappa B/metabolismo , Osteogénesis , Transducción de Señal , Femenino , Humanos , Células Madre Mesenquimatosas/citología
13.
Toxicol Appl Pharmacol ; 381: 114732, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31454633

RESUMEN

Endothelium inflammation has become a major risk factor for pathological development of atherosclerosis. IMM-H007 (H007), a small molecule compound, is previously reported to reduce inflammatory atherosclerosis. However, the regulatory role of H007 in endothelium inflammation is still unclear. Here, we characterize H007 as a critical repressor in regulation of endothelium inflammation. We find that H007 significantly inhibits monocyte adhesion to endothelial cells and its transendothelial migration. Mechanistically, H007 markedly represses TNFα-induced IκBα degradation and NF-κB nuclear translocation, therefore leading to NF-κB-mediated inflammatory suppression. Moreover, another inflammatory signaling JNK/c-Jun, which is always co-activated with NF-κB in response to pro-inflammatory stimuli, is also found to be restrained by H007 through reducing its phosphorylation status. Thus, we conclude that H007 negatively regulates endothelium inflammation through inactivating NF-κB and JNK/AP1 signaling. More importantly, this study provides us a new insight into understanding the molecular basis by which H007 regulates inflammatory atherosclerosis.


Asunto(s)
Adenosina/análogos & derivados , Antiinflamatorios/farmacología , MAP Quinasa Quinasa 4/metabolismo , FN-kappa B/metabolismo , Factor de Transcripción AP-1/metabolismo , Adenosina/farmacología , Aterosclerosis , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Interleucina-1beta/genética , Interleucina-6/genética , Interleucina-6/metabolismo , Transducción de Señal/efectos de los fármacos , Células THP-1 , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
14.
Mol Cell Biol ; 39(19)2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31285272

RESUMEN

The long noncoding RNA HOXA-AS3 has recently been reported to act as a critical regulator in inflammation-linked lung adenocarcinoma. However, the roles of HOXA-AS3 in endothelium inflammation and related vascular disorders remain poorly defined. In the current study, we identified HOXA-AS3 to be a critical activator to promote NF-κB-mediated endothelium inflammation. HOXA-AS3, a chromatin-associated regulator which colocalizes with NF-κB at specific gene promoters, was found to interact with NF-κB and positively regulate its activity through control of the expression of the NF-κB inhibitor protein IκBα and the acetylation status at the K310 site of p65. More importantly, clinicopathological analysis showed that HOXA-AS3 expression has a significant positive correlation with atherosclerosis. Thus, we conclude that HOXA-AS3 may serve as a crucial biomarker for the clinical diagnosis of atherosclerosis, as well as a promising therapeutic target for the treatment of multiple inflammatory vascular diseases. In addition, this study suggests the functional importance of HOXA-AS3 in the regulation of inflammatory disorders.


Asunto(s)
Aterosclerosis/genética , Marcadores Genéticos/genética , ARN Largo no Codificante/genética , Transducción de Señal , Regulación hacia Arriba , Acetilación , Anciano , Diagnóstico Precoz , Femenino , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Persona de Mediana Edad , Inhibidor NF-kappaB alfa/genética , FN-kappa B/metabolismo , Regiones Promotoras Genéticas
15.
Med Microbiol Immunol ; 208(6): 781-792, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31187242

RESUMEN

Staphylococcal enterotoxins (SEs), as typical superantigens, exhibit promising antitumour activity in the clinic, but their unavoidable side effects related to fever and emesis seriously limit their application for the treatment of malignant tumours. Fortunately, the identification of Staphylococcal enterotoxin-like toxins (SEls), which possess amino acid sequences similar to those of classical SEs but exhibit no or low emetic activity, has provided a set of potential immunomodulatory candidates for cancer therapy. The aim of this study was to examine the effect of SElQ on lymphocyte activation and to further demonstrate its antitumour activity both in vitro and in vivo. High-purity SElQ was successfully harvested, and in vitro results confirmed that SElQ can significantly activate mouse- and human-derived lymphocytes in a dose-dependent manner, particularly CD4+ and CD8+ T cells, which showed significant increases in both percentage and absolute number. Further examination revealed that in addition to the originally recognized TCR Vß5 and 21, TCR Vß14, 17 and 18 were activated in SElQ-induced human PBMCs. Moreover, the expression of IL-2 and IFN-γ was significantly upregulated in vitro and in vivo after SElQ treatment. Based on the findings that SElQ induces lymphocyte activation and cytokine release, we then confirmed its antitumour activity both in vitro and in vivo. The data showed that treatment with a low concentration of SElQ (30 µg/mouse) could inhibit the growth of tumours by approximately 30% and no significant toxicity was observed. Taken together, our results demonstrated that SElQ can significantly induce T cell activation and cytokine release and further elicit substantial antitumour activity and thus provide support for the potential application of SElQ in cancer immunotherapy.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Enterotoxinas/metabolismo , Activación de Linfocitos/efectos de los fármacos , Superantígenos/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Relación Dosis-Respuesta Inmunológica , Femenino , Voluntarios Sanos , Humanos , Ratones Endogámicos BALB C , Receptores de Antígenos de Linfocitos T/metabolismo
16.
Atherosclerosis ; 281: 38-46, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30658190

RESUMEN

BACKGROUND AND AIMS: Endothelium inflammation, which can lead to endothelial activation and dysfunction, is widely accepted as the major event in multiple vascular disorders. The lncRNA HOXA-AS2 was previously reported to be involved in multiple inflammation-linked cancers. However, the role of HOXA-AS2 in endothelium inflammation is poorly understood. This study aims to determine the regulatory role of HOXA-AS2 in endothelium inflammation and related vascular diseases. METHODS: High throughput mRNA sequencing was performed to establish expression profiles after HOXA-AS2 depletion. We extracted total RNAs of human peripheral blood mononuclear cells from normal control group and experimental group with carotid artery atherosclerosis, and performed qRT-PCR to assay the correlation between HOXA-AS2 expression and inflammatory vascular diseases. RESULTS: Inhibition of HOXA-AS2 can induce the activation of NF-κB signaling and subsequent inflammatory response. More importantly, HOXA-AS2 is inversely found to be inversely regulated by NF-κB in a negative feedback manner by helping recruit BRD4/P-TEFb complex to HOXA-AS2 promoter region, therefore facilitating release of the promoter-proximal paused RNA polymerase II and activating transcription elongation. CONCLUSIONS: We identify HOXA-AS2 as a critical repressor of endothelium inflammation. Moreover, this study offers us a new way to balance the NF-κB signaling-driven excessive endothelium inflammation by establishing a NF-κB/HOXA-AS2 negative feedback loop. Based on these findings, we conclude that HOXA-AS2 may serve as a crucial therapeutic target for various vascular disorders which are significantly associated with endothelium inflammation.


Asunto(s)
Enfermedades de las Arterias Carótidas/metabolismo , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Mediadores de Inflamación/metabolismo , FN-kappa B/metabolismo , ARN Largo no Codificante/metabolismo , Acetilación , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/patología , Estudios de Casos y Controles , Proteínas de Ciclo Celular/metabolismo , Técnicas de Cocultivo , Endotelio Vascular/patología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Inhibidor NF-kappaB alfa/metabolismo , Factor B de Elongación Transcripcional Positiva/metabolismo , Procesamiento Proteico-Postraduccional , Proteolisis , ARN Largo no Codificante/genética , Transducción de Señal , Células THP-1 , Factor de Transcripción ReIA/metabolismo , Factores de Transcripción/metabolismo , Migración Transendotelial y Transepitelial
17.
J Mol Cell Cardiol ; 126: 60-69, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30419187

RESUMEN

Endothelium inflammation, a key event in vascular pathological process, can lead to endothelial activation and subsequent vascular disorders. Long non-coding RNA NKILA plays an important regulatory role in pro-inflammatory response. However, the underlying molecular basis by which NKILA regulates endothelial inflammation is poorly understood. In this study, we identify NKILA as a critical repressor to protect the endothelium from inflammation. Mechanistically, we show that NKILA is able to positively mediate the expression of KLF4, an anti-inflammatory atheroprotective regulator in endothelial cells (ECs), by a NF-κB-mediated DNA methylation mechanism. Moreover, NF-κB is found to help recruit DNMT3A to the CpG island of KLF4 promoter, facilitating KLF4 promoter DNA methylation and transcriptional repression. More importantly, we find KLF4 can inversely attenuate NF-κB transcriptional activity via establishing a NF-κB/KLF4 positive feedback loop, which is under the control of NKILA. Hence, sustained endothelium inflammation will occur, once the NKILA becomes dysfunctional. These studies revealed that NKILA can function as a vital regulator to protect the endothelium from inflammatory lesions and related vascular diseases.


Asunto(s)
Endotelio Vascular/patología , Retroalimentación Fisiológica , Inflamación/genética , Factores de Transcripción de Tipo Kruppel/genética , FN-kappa B/metabolismo , ARN Largo no Codificante/genética , ADN (Citosina-5-)-Metiltransferasas , Metilación de ADN/genética , ADN Metiltransferasa 3A , Regulación de la Expresión Génica , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Modelos Biológicos , Regiones Promotoras Genéticas/genética , ARN Largo no Codificante/metabolismo , Transcripción Genética
18.
J Cell Mol Med ; 23(2): 1325-1332, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30536618

RESUMEN

As is previously reported, mesenchymal stem cells have potential ability to differentiate into osteocytes. However, the underlying mechanism during this biological process is poorly understood. In the present study, we identify a novel long non-coding RNA named HOXA-AS2 as a critical regulator during the formation of osteogenesis. Attenuation of HOXA-AS2 can reduce the calcium deposition and repress the alkaline phosphatase activity. Moreover, the expressions of osteogenic marker genes are markedly downregulated after HOXA-AS2 depletion. Mechanistically, we found HOXA-AS2 can regulate the transcriptional activity of NF-κB, a critical inhibitor of osteogenesis. More importantly, HOXA-AS2 knockdown could result in the transcriptional repression of the osteogenic master transcription factor SP7 by a NF-κB/HDAC2-coordinated H3K27 deacetylation mechanism. Based on these studies, we conclude that HOXA-AS2 may serve as a promising therapeutic target for bone tissue repair and regeneration in the near future.


Asunto(s)
Diferenciación Celular , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/citología , FN-kappa B/metabolismo , Osteocitos/citología , Osteogénesis , ARN Largo no Codificante/genética , Movimiento Celular , Proliferación Celular , Células Cultivadas , Transición Epitelial-Mesenquimal , Femenino , Humanos , Células Madre Mesenquimatosas/metabolismo , FN-kappa B/genética , Osteocitos/metabolismo
19.
Stem Cell Res Ther ; 9(1): 330, 2018 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-30486857

RESUMEN

BACKGROUND/AIMS: Menstrual blood-derived endometrial stem cells (MenSCs) emerge as an ideal source for cell-based treatment in regenerative medicine and immunotherapy. However, the major obstacle is the low survival rate in tissues and the limited expansion number. Autophagy is an intracellular metabolic self-degradative process which plays important roles in normal cellular division and survival, and the present study aimed to explore the related mechanisms between autophagy and survival of MenSCs in vitro and in vivo. METHODS: The MenSCs were obtained from menstrual blood procured from healthy female donors. In vitro, MenSCs were exposed to rapamycin and Earle's balanced salts solution (EBSS). We evaluated the MenSCs immunophenotypic cell cycle distribution by propidium iodide (PI) staining and cell apoptosis by Annexin V/PI staining as well as their proliferative potential by the MTT assay. We also assessed the expression of genes associated with the cell cycle and Gsk3ß signaling pathway by western blot analysis. We depressed Atg5 and Gsk3ß expression by short hairpin RNA (shRNA) and undertook the experiments. Moreover, the labeled MenSCs were observed and counted with DiI after transplantation into the mice via the tail vein by microscopy in vivo. RESULTS: In vitro, rapamycin and starvation induced autophagy of MenSCs. Hyperactive autophagy significantly induced G0/G1 arrest and slightly promoted apoptosis of MenSCs. Meanwhile, autophagy could stimulate p-GSK3ß expression in MenSCs. Further, knockdown GSK3ß can accelerate the proliferation of MenSCs by shRNA and CHIR99021. Moreover, the shGSK3ß MenSCs showed strong proliferative activity in vitro and in vivo. CONCLUSIONS: Our results indicate that autophagy induced G0/G1 arrest and apoptosis of MenSCs via GSK3ß/ß-catenin pathway. Inhibiting autophagy or reduced GSK3ß levels may improve survival rate in vivo, thus playing roles in MenSCs therapy.


Asunto(s)
Apoptosis , Autofagia , Puntos de Control del Ciclo Celular , Endometrio/patología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Menstruación/sangre , Células Madre/patología , beta Catenina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Inmunofenotipificación , Masculino , Ratones Endogámicos BALB C , Piridinas/farmacología , Pirimidinas/farmacología , Transducción de Señal , Sirolimus/farmacología , Células Madre/efectos de los fármacos , Células Madre/metabolismo
20.
Exp Ther Med ; 16(4): 3611-3616, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30233716

RESUMEN

Gastric cancer (GC) affects the health of 1,000,000 people per year worldwide; however, the biological basis of GC remains largely unknown. The current study aimed to investigate the aberrant expression of miR-744 in GC for the effective treatment of patients with GC. Tumor and adjacent tissues were obtained from 30 patients who underwent tumor resection surgery at Dongying People's Hospital. The results of reverse transcription-quantitative polymerase chain reaction indicated that the expression of miR-744 was significantly decreased in tumor tissues compared with the levels in adjacent tissues. Human gastric cancer cell line SGC-7901 was then randomly divided into three different groups, including the control, miR-negative control (NC) and miR-744 mimic groups. A Cell Counting Kit-8 assay demonstrated that there was a significant decrease in the proliferation rate of SGC-7901 cells in the miR-744 mimics group compared with that observed in the control and miR-NC mimics groups. In addition, flow cytometry demonstrated that apoptosis was significantly increased in the miR-744 mimics group compared with that observed in the control and miR-NC mimics groups. Western blotting indicated that the expression of B cell lymphoma 2 (Bcl-2), B cell lymphoma-2-associated X protein and caspase-3 protein was significantly increased, while the expression of Bcl-2 was significantly decreased in the miR-744 mimics group compared with the levels observed in the control and miR-NC mimics groups. A dual-luciferase assay verified that miR-744 directly targeted the 3'-untranslated region of Bcl-2. Taken together, the present study suggested that miR-744 serves a tumor suppressive role in GC by targeting Bcl-2.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...