Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 23(2): 229-236, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34949832

RESUMEN

Aging is characterized by an increased vulnerability to infection and the development of inflammatory diseases, such as atherosclerosis, frailty, cancer and neurodegeneration. Here, we find that aging is associated with the loss of diurnally rhythmic innate immune responses, including monocyte trafficking from bone marrow to blood, response to lipopolysaccharide and phagocytosis. This decline in homeostatic immune responses was associated with a striking disappearance of circadian gene transcription in aged compared to young tissue macrophages. Chromatin accessibility was significantly greater in young macrophages than in aged macrophages; however, this difference did not explain the loss of rhythmic gene transcription in aged macrophages. Rather, diurnal expression of Kruppel-like factor 4 (Klf4), a transcription factor (TF) well established in regulating cell differentiation and reprogramming, was selectively diminished in aged macrophages. Ablation of Klf4 expression abolished diurnal rhythms in phagocytic activity, recapitulating the effect of aging on macrophage phagocytosis. Examination of individuals harboring genetic variants of KLF4 revealed an association with age-dependent susceptibility to death caused by bacterial infection. Our results indicate that loss of rhythmic Klf4 expression in aged macrophages is associated with disruption of circadian innate immune homeostasis, a mechanism that may underlie age-associated loss of protective immune responses.


Asunto(s)
Relojes Circadianos/genética , Macrófagos/fisiología , Envejecimiento , Animales , Aterosclerosis/genética , Diferenciación Celular/genética , Regulación de la Expresión Génica/genética , Inmunidad Innata/genética , Inflamación/genética , Factor 4 Similar a Kruppel/genética , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/fisiología , Fagocitosis/genética
2.
Xenobiotica ; 54(9): 730-748, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38568505

RESUMEN

1. Occupational exposure to 4,4'-methylene diphenyl diisocyanate (MDI) is associated with occupational asthma (OA) development. Alveolar macrophage-induced recruitment of immune cells to the lung microenvironment plays an important role during asthma pathogenesis. Previous studies identified that MDI/MDI-glutathione (GSH)-exposure downregulates endogenous hsa-miR-206-3p/hsa-miR-381-3p. Our prior report shows that alternatively activated (M2) macrophage-associated markers/chemokines are induced by MDI/MDI-GSH-mediated Krüppel-Like Factor 4 (KLF4) upregulation in macrophages and stimulates immune cell chemotaxis. However, the underlying molecular mechanism(s) by which MDI/MDI-GSH upregulates KLF4 remain unclear.2. Following MDI-GSH exposure, microRNA(miR)-inhibitors/mimics or plasmid transfection, endogenous hsa-miR-206-3p/hsa-miR-381-3p, KLF4, or M2 macrophage-associated markers (CD206, TGM2), and chemokines (CCL17, CCL22, CCL24) were measured by either RT-qPCR, western blot, or luciferase assay.3. MDI-GSH exposure downregulates hsa-miR-206-3p/hsa-miR-381-3p by 1.46- to 9.75-fold whereas upregulates KLF4 by 1.68- to 1.99-fold, respectively. In silico analysis predicts binding between hsa-miR-206-3p/hsa-miR-381-3p and KLF4. Gain- and loss-of-function, luciferase reporter assays and RNA-induced silencing complex-immunoprecipitation (RISC-IP) studies confirm the posttranscriptional regulatory roles of hsa-miR-206-3p/hsa-miR-381-3p and KLF4 in macrophages. Furthermore, hsa-miR-206-3p/hsa-miR-381-3p regulate the expression of M2 macrophage-associated markers and chemokines via KLF4.4. In conclusion, hsa-miR-206-3p/hsa-miR-381-3p play a major role in regulation of MDI/MDI-GSH-induced M2 macrophage-associated markers and chemokines by targeting the KLF4 transcript, and KLF4-mediated regulation in macrophages.


Asunto(s)
Isocianatos , Factor 4 Similar a Kruppel , Macrófagos , MicroARNs , Humanos , Quimiocinas/metabolismo , Isocianatos/farmacología , Factor 4 Similar a Kruppel/genética , Factor 4 Similar a Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Activación de Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , MicroARNs/metabolismo , MicroARNs/genética , Regulación hacia Arriba/efectos de los fármacos
3.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34001623

RESUMEN

Vascular endothelial cells (ECs) sense and respond to hemodynamic forces such as pulsatile shear stress (PS) and oscillatory shear stress (OS). Among the metabolic pathways, glycolysis is differentially regulated by atheroprone OS and atheroprotective PS. Studying the molecular mechanisms by which PS suppresses glycolytic flux at the epigenetic, transcriptomic, and kinomic levels, we have demonstrated that glucokinase regulatory protein (GCKR) was markedly induced by PS in vitro and in vivo, although PS down-regulates other glycolysis enzymes such as hexokinase (HK1). Using next-generation sequencing data, we identified the binding of PS-induced Krüppel-like factor 4 (KLF4), which functions as a pioneer transcription factor, binding to the GCKR promoter to change the chromatin structure for transactivation of GCKR. At the posttranslational level, PS-activated AMP-activated protein kinase (AMPK) phosphorylates GCKR at Ser-481, thereby enhancing the interaction between GCKR and HK1 in ECs. In vivo, the level of phosphorylated GCKR Ser-481 and the interaction between GCKR and HK1 were increased in the thoracic aorta of wild-type AMPKα2+/+ mice in comparison with littermates with EC ablation of AMPKα2 (AMPKα2-/-). In addition, the level of GCKR was elevated in the aortas of mice with a high level of voluntary wheel running. The underlying mechanisms for the PS induction of GCKR involve regulation at the epigenetic level by KLF4 and at the posttranslational level by AMPK.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Aorta Torácica/metabolismo , Epigénesis Genética , Glucólisis/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Aorta Torácica/citología , Fenómenos Biomecánicos , Hexoquinasa/genética , Hexoquinasa/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Factor 4 Similar a Kruppel/genética , Factor 4 Similar a Kruppel/metabolismo , Masculino , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Unión Proteica , Reología , Transcriptoma
4.
Am J Pathol ; 192(9): 1250-1258, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35750256

RESUMEN

Along with a recent remarkable decrease in Helicobacter pylori-infected individuals, reports of gastric neoplasms such as sporadic foveolar-type gastric adenoma (FGA) in H. pylori-naive patients have been increasing. This tumor, with its raspberry-like appearance, is common in H. pylori-naive gastric mucosa. The current study investigated the genomic features of sporadic FGA. Fresh-frozen sporadic FGA tissue samples from H. pylori-naive patients were subjected to whole genome analysis using a next-generation sequencer. Proliferation ability and apoptotic profiles of human gastric epithelial cells, along with plasmid transfection of candidate variants, were examined. A mean of 6.65 × 108 total reads were obtained for each sample. Common genetic abnormalities in well-known proliferation driver genes of conventional gastric dysplasia/cancer were not found. However, a common single-nucleotide variation (SNV) was noted within the DNA-binding domain of the tumor suppressor gene KLF4. This novel SNV was located in the zinc finger 2 region. Additional experiments showed that it significantly suppressed proliferation of gastric epithelial cells compared with wild-type KLF4 plasmid-transfected cells, although suppression was reduced in early apoptotic phase-related genes. A novel SNV in the KLF4 zinc finger 2 region was commonly found in sporadic FGA tissue samples, which may explain the slow-growing properties of this neoplasm.


Asunto(s)
Adenoma , Neoplasias Gástricas , Adenoma/genética , Adenoma/patología , Pólipos Adenomatosos , Mucosa Gástrica/patología , Infecciones por Helicobacter , Helicobacter pylori , Humanos , Factor 4 Similar a Kruppel/genética , Mutación , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología
5.
Cell Mol Biol Lett ; 28(1): 105, 2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38105235

RESUMEN

BACKGROUND: Pulmonary fibrosis is a growing clinical problem that develops as a result of abnormal wound healing, leading to breathlessness, pulmonary dysfunction and ultimately death. However, therapeutic options for pulmonary fibrosis are limited because the underlying pathogenesis remains incompletely understood. Circular RNAs, as key regulators in various diseases, remain poorly understood in pulmonary fibrosis induced by silica. METHODS: We performed studies with fibroblast cell lines and silica-induced mouse pulmonary fibrosis models. The expression of circZNF609, miR-145-5p, and KLF4 was determined by quantitative real-time polymerase chain reaction (qRT-PCR) analysis. RNA immunoprecipitation (RIP) assays and m6A RNA immunoprecipitation assays (MeRIP), Western blotting, immunofluorescence assays, and CCK8 were performed to investigate the role of the circZNF609/miR-145-5p/KLF4 axis and circZNF609-encoded peptides in fibroblast activation. RESULTS: Our data showed that circZNF609 was downregulated in activated fibroblasts and silica-induced fibrotic mouse lung tissues. Overexpression of circZNF609 could inhibit fibroblast activation induced by transforming growth factor-ß1 (TGF-ß1). Mechanically, we revealed that circZNF609 regulates pulmonary fibrosis via miR-145-5p/KLF4 axis and circZNF609-encoded peptides. Furthermore, circZNF609 was highly methylated and its expression was controlled by N6-methyladenosine (m6A) modification. Lastly, in vivo studies revealed that overexpression of circZNF609 attenuates silica-induced lung fibrosis in mice. CONCLUSIONS: Our data indicate that circZNF609 is a critical regulator of fibroblast activation and silica-induced lung fibrosis. The circZNF609 and its derived peptides may represent novel promising targets for the treatment of pulmonary fibrosis.


Asunto(s)
MicroARNs , Fibrosis Pulmonar , ARN Circular , Animales , Ratones , Pulmón/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Dióxido de Silicio/efectos adversos , Factor de Crecimiento Transformador beta1/metabolismo , Factor 4 Similar a Kruppel/genética , Factor 4 Similar a Kruppel/metabolismo , ARN Circular/genética
6.
Mol Cell Proteomics ; 20: 100064, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33640491

RESUMEN

Prostate cancer (PCa) is the most frequently diagnosed cancer in men and the third cause of cancer mortality. PCa initiation and growth are driven by the androgen receptor (AR). The AR is activated by androgens such as testosterone and controls prostatic cell proliferation and survival. Here, we report an AR signaling network generated using BioID proximity labeling proteomics in androgen-dependent LAPC4 cells. We identified 31 AR-associated proteins in nonstimulated cells. Strikingly, the AR signaling network increased to 182 and 200 proteins, upon 24 h or 72 h of androgenic stimulation, respectively, for a total of 267 nonredundant AR-associated candidates. Among the latter group, we identified 213 proteins that were not previously reported in databases. Many of these new AR-associated proteins are involved in DNA metabolism, RNA processing, and RNA polymerase II transcription. Moreover, we identified 44 transcription factors, including the Kru¨ppel-like factor 4 (KLF4), which were found interacting in androgen-stimulated cells. Interestingly, KLF4 repressed the well-characterized AR-dependent transcription of the KLK3 (PSA) gene; AR and KLF4 also colocalized genome-wide. Taken together, our data report an expanded high-confidence proximity network for AR, which will be instrumental to further dissect the molecular mechanisms underlying androgen signaling in PCa cells.


Asunto(s)
Receptores Androgénicos/metabolismo , Línea Celular , Humanos , Calicreínas/genética , Factor 4 Similar a Kruppel/genética , Factor 4 Similar a Kruppel/metabolismo , Antígeno Prostático Específico/genética , Receptores Androgénicos/genética
7.
Kidney Int ; 102(1): 14-16, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35738827

RESUMEN

Krüppel-like factors (KLFs) are transcription factors with important roles in tissue homeostasis. KLF4 possesses antithrombotic and anti-inflammatory properties. In this issue, Estrada et al. show that endothelial KLF4 prevents complement deposition in glomeruli and in its absence the cell-bound complement regulator CD55 was reduced. The study included endothelial-specific KLF4 knockdown mice that mimic thrombotic microangiopathy and thrombotic microangiopathy patient biopsies showing decreased KLF4 and CD55. The results suggest that KLF4 is involved in the regulation of glomerular complement deposition.


Asunto(s)
Activación de Complemento , Enfermedades Renales , Factor 4 Similar a Kruppel , Microangiopatías Trombóticas , Animales , Humanos , Enfermedades Renales/etiología , Glomérulos Renales , Factor 4 Similar a Kruppel/genética , Ratones , Factores de Transcripción
8.
Biochem Biophys Res Commun ; 587: 29-35, 2022 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-34864392

RESUMEN

During reprogramming of somatic cells, heightened proliferation is one of the earliest changes observed. While other early events such as mesenchymal-to-epithelial transition have been well studied, the mechanisms by which the cell cycle switches from a slow cycling state to a faster cycling state are still incompletely understood. To investigate the role of Oct-3/4 in this early transition, we created a 4-Hydroxytamoxifen (OHT) dependent Oct-3/4 Estrogen Receptor fusion (OctER). We confirmed that OctER can substitute for Oct-3/4 to reprogram mouse embryonic fibroblasts to a pluripotent state. During the early stages of reprograming, Oct-3/4 and Klf4 individually did not affect cell proliferation but in combination hastened the cell cycle. Using OctER + Klf4, we found that proliferative enhancement is OHT dose-dependent, suggesting that OctER is the driver of this transition. We identified Cyclin A2 as a likely target of Oct-3/4 + Klf4. In mESC, Klf4 and Oct-3/4 bind ∼100bp upstream of Cyclin A2 CCRE, suggesting a potential regulatory role. Using inducible OctER, we show a dose-dependent induction of Cyclin A2 promoter-reporter activity. Taken together, our results suggest that Cyclin A2 is a key early target during reprogramming, and support the view that a rapid cell cycle assists the transition to pluripotency.


Asunto(s)
Ciclo Celular/genética , Reprogramación Celular/genética , Ciclina A2/genética , Fibroblastos/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Animales , Ciclo Celular/efectos de los fármacos , Diferenciación Celular , Proliferación Celular , Ciclina A2/metabolismo , Embrión de Mamíferos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel/genética , Factor 4 Similar a Kruppel/metabolismo , Ratones , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Plásmidos/química , Plásmidos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Factores de Tiempo , Transducción Genética
9.
Ann Rheum Dis ; 81(2): 268-277, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34750102

RESUMEN

OBJECTIVES: Systemic sclerosis (SSc) is a complex disease of unknown aetiology in which inflammation and fibrosis lead to multiple organ damage. There is currently no effective therapy that can halt the progression of fibrosis or reverse it, thus studies that provide novel insights into disease pathogenesis and identify novel potential therapeutic targets are critically needed. METHODS: We used global gene expression and genome-wide DNA methylation analyses of dermal fibroblasts (dFBs) from a unique cohort of twins discordant for SSc to identify molecular features of this pathology. We validated the findings using in vitro, ex vivo and in vivo models. RESULTS: Our results revealed distinct differentially expressed and methylated genes, including several transcription factors involved in stem cell differentiation and developmental programmes (KLF4, TBX5, TFAP2A and homeobox genes) and the microRNAs miR-10a and miR-10b which target several of these deregulated genes. We show that KLF4 expression is reduced in SSc dFBs and its expression is repressed by TBX5 and TFAP2A. We also show that KLF4 is antifibrotic, and its conditional knockout in fibroblasts promotes a fibrotic phenotype. CONCLUSIONS: Our data support a role for epigenetic dysregulation in mediating SSc susceptibility in dFBs, illustrating the intricate interplay between CpG methylation, miRNAs and transcription factors in SSc pathogenesis, and highlighting the potential for future use of epigenetic modifiers as therapies.


Asunto(s)
Fibroblastos/patología , Regulación de la Expresión Génica/fisiología , Factor 4 Similar a Kruppel/metabolismo , Esclerodermia Sistémica , Piel/patología , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Factor 4 Similar a Kruppel/genética , MicroARNs/metabolismo , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Piel/metabolismo , Proteínas de Dominio T Box/metabolismo , Factor de Transcripción AP-2/metabolismo , Transcriptoma
10.
Respir Res ; 23(1): 340, 2022 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-36496404

RESUMEN

BACKGROUND: Premature infants, subjected to supplemental oxygen and mechanical ventilation, may develop bronchopulmonary dysplasia, a chronic lung disease characterized by alveolar dysplasia and impaired vascularization. We and others have shown that hyperoxia causes senescence in cultured lung epithelial cells and fibroblasts. Although miR-34a modulates senescence, it is unclear whether it contributes to hyperoxia-induced senescence. We hypothesized that hyperoxia increases miR-34a levels, leading to cellular senescence. METHODS: We exposed mouse lung epithelial (MLE-12) cells and primary human small airway epithelial cells to hyperoxia (95% O2/5% CO2) or air (21% O2/5% CO2) for 24 h. Newborn mice (< 12 h old) were exposed to hyperoxia (> 95% O2) for 3 days and allowed to recover in room air until postnatal day 7. Lung samples from premature human infants requiring mechanical ventilation and control subjects who were not mechanically ventilated were employed. RESULTS: Hyperoxia caused senescence as indicated by loss of nuclear lamin B1, increased p21 gene expression, and senescence-associated secretory phenotype factors. Expression of miR-34a-5p was increased in epithelial cells and newborn mice exposed to hyperoxia, and in premature infants requiring mechanical ventilation. Transfection with a miR-34a-5p inhibitor reduced hyperoxia-induced senescence in MLE-12 cells. Additionally, hyperoxia increased protein levels of the oncogene and tumor-suppressor Krüppel-like factor 4 (KLF4), which were inhibited by a miR-34a-5p inhibitor. Furthermore, KLF4 knockdown by siRNA transfection reduced hyperoxia-induced senescence. CONCLUSION: Hyperoxia increases miR-34a-5p, leading to senescence in lung epithelial cells. This is dictated in part by upregulation of KLF4 signaling. Therefore, inhibiting hyperoxia-induced senescence via miR-34a-5p or KLF4 suppression may provide a novel therapeutic strategy to mitigate the detrimental consequences of hyperoxia in the neonatal lung.


Asunto(s)
Displasia Broncopulmonar , Hiperoxia , Factor 4 Similar a Kruppel , MicroARNs , Animales , Humanos , Ratones , Animales Recién Nacidos , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/tratamiento farmacológico , Dióxido de Carbono , Senescencia Celular , Células Epiteliales/metabolismo , Hiperoxia/genética , Hiperoxia/metabolismo , Factor 4 Similar a Kruppel/genética , Factor 4 Similar a Kruppel/metabolismo , Pulmón/metabolismo , MicroARNs/metabolismo
11.
Int Arch Allergy Immunol ; 183(7): 785-795, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35390783

RESUMEN

INTRODUCTION: Childhood asthma is a common chronic inflammatory lung disease in children, among which airway inflammation is the main driving factor of asthma symptoms. Follistatin-like protein 1 (FSTL1) is involved in multiple inflammatory processes, but its role in airway inflammation has not been fully elucidated. METHODS: We used lipopolysaccharide (LPS) to stimulate human primary bronchial epithelial (BEAS-2B) cells to establish an in vitro airway inflammation model. The expression of FSTL1 was detected by qPCR. Cell Counting Kit-8 and Annexin V-PI double staining was used to analyze the viability and apoptosis of BEAS-2B. The content of IL-6, IL-8 and TNF-α was determined by ELISA kit. Western blot was used to detect the protein expression level of the bone morphogenetic protein 4 (BMP4) and KLF4. The levels of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), and malondialdehyde were measured to assess oxidative stress. RESULTS: The mRNA expression of FSTL1 was significantly increased in LPS-treated BEAS-2B cells. Silencing of FSTL1 inhibited the release of IL-6, IL-8, TNF-α, and cell apoptosis as well as enhanced the activities of SOD, CAT, and GSH-Px. Silencing of FSTL1 reversed the inflammatory state of cells by upregulating BMP4 and increasing the expression level of KLF4. CONCLUSION: Silencing of FSTL1 reduced LPS-induced BEAS-2B cell damage by regulating the BMP4/KLF4 axis. FSTL1 may be a potential target for the treatment of asthma.


Asunto(s)
Asma , Proteína Morfogenética Ósea 4 , Proteínas Relacionadas con la Folistatina , Asma/genética , Asma/metabolismo , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Niño , Células Epiteliales/metabolismo , Proteínas Relacionadas con la Folistatina/genética , Proteínas Relacionadas con la Folistatina/metabolismo , Silenciador del Gen , Humanos , Inflamación/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Factor 4 Similar a Kruppel/genética , Factor 4 Similar a Kruppel/metabolismo , Lipopolisacáridos/farmacología , Estrés Oxidativo , Superóxido Dismutasa , Factor de Necrosis Tumoral alfa/metabolismo
12.
Pharmacol Res ; 180: 106244, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35550167

RESUMEN

Sorafenib, a multikinase inhibitor, has been widely used as a first-line anticancer drug for advanced hepatocellular carcinoma (HCC). However, the development of drug resistance to sorafenib is frequently observed in clinical applications. Potential nonkinase targets of sorafenib have not been well documented and may provide insights into reversing drug resistance and enhancing drug efficacy. Herein, we report that sorafenib exerts its anticancer effects by activating metallothionein 1 G (MT1G) expression. MT1G is a novel marker in HCC that correlates well with patient survival. MT1G overexpression suppressed the cellular proliferation, migration, invasion, and tumour formation of HCC and sensitised cells to sorafenib treatment. However, the disruption of MT1G attenuated the anticancer effects of sorafenib. Mechanistically, sorafenib upregulated MT1G expression via hypomethylation of its promoter region by binding and inhibiting DNA methyltransferase 1 (DNMT1) and increasing its promoter accessibility in HCC cells. Activation of MT1G also inhibited CA9 transcription through the suppression of HIF1A as mediated by KLF4. Our collective data revealed that sorafenib exerts its anticancer effects through epigenetic regulation of the DNMT1/MT1G/KLF4/CA9 axis in HCC and the activation of MT1G might constitute a strategy for enhancing the effect of sorafenib to suppress HCC cells.


Asunto(s)
Antineoplásicos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Sorafenib , Antígenos de Neoplasias , Antineoplásicos/farmacología , Anhidrasa Carbónica IX/genética , Anhidrasa Carbónica IX/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Resistencia a Antineoplásicos , Epigénesis Genética , Humanos , Factor 4 Similar a Kruppel/genética , Factor 4 Similar a Kruppel/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Metalotioneína/genética , Metalotioneína/metabolismo , Transducción de Señal/efectos de los fármacos , Sorafenib/farmacología
13.
Exp Lung Res ; 48(7-8): 213-223, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35950640

RESUMEN

PURPOSE: Bronchopulmonary dysplasia (BPD) is a long-term respiratory condition. More than a quarter of extremely premature newborns are harmed by BPD. At present, there are no apparent effective drugs or treatments for the condition. In this study, we aimed to investigate the functional role and mechanism of lymphoid enhancer-binding factor 1 (Lef1) in BPD in vitro. MATERIALS AND METHODS: Blood samples from BPD patients and healthy volunteers were gathered, and an in vitro model of BPD was developed in alveolar epithelial cells (AECs) MLE-12 induced by hyperoxia. Then expression of krüppel-like factor 4 (KLF4/Klf4) and LEF1/Lef1 were evaluated. After Lef1 overexpressing plasmid and the vector were transfected into hyperoxia-induced MLE-12 cells, cell proliferation assays were carried out. Cell apoptosis was investigated by a flow cytometry assay, and apoptosis related proteins Bcl-2, cleaved-caspase 3 and 9 were analyzed by a western blot assay. The binding between Klf4 and Lef1 promoter predicted on the JASPAR website was verified using luciferase and ChIP assays. For further study of the mechanism of Klf4 and Lef1 in BPD, gain-of-function experiments were performed. RESULTS: The mRNA levels of KLF4/Klf4 and LEF1/Lef1 were diminished in clinical BPD serum samples and hyperoxia-induced MLE-12 cells. Overexpression of Lef1 stimulated AEC proliferation and suppressed AEC apoptosis induced by hyperoxia. Mechanically, Klf4 bound to Lef1's promoter region and aids transcription. Moreover, the results of gain-of-function experiments supported that Klf4 could impede AEC damage induced by hyperoxia via stimulating Lef1. CONCLUSION: Klf4 and Lef1 expression levels were declined in hyperoxia-induced AECs, and Lef1 could be transcriptionally activated by Klf4 and protect against hyperoxia-induced AEC injury in BPD. As a result, Lef1 might become a prospective therapeutic target for BPD.


Asunto(s)
Hipoxia de la Célula , Factor de Unión 1 al Potenciador Linfoide , Células Epiteliales Alveolares/metabolismo , Displasia Broncopulmonar/metabolismo , Humanos , Recién Nacido , Factor 4 Similar a Kruppel/genética , Factor 4 Similar a Kruppel/metabolismo , Factor de Unión 1 al Potenciador Linfoide/genética , Factor de Unión 1 al Potenciador Linfoide/metabolismo
14.
Mol Biol Rep ; 49(9): 8495-8505, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35802277

RESUMEN

BACKGROUND: The present study aimed to investigate the mechanisms through which long non-coding RNA (lncRNA) maternally expressed 3 (MEG3) affected the endothelial differentiation of mouse derived adipose-derived stem cells (ADSCs). MATERIALS AND METHODS: ADSCs were isolated and identified by specific surface marker detection. The effects of lncRNA MEG3 on endothelial differentiation of ADSCs were also detected via quantitative PCR, western blotting, immunofluorescence and Matrigel angiogenesis assays. In addition, using target gene prediction tools and luciferase reporter assays, the downstream target gene was demonstrated. RESULTS: LncRNA MEG3 targeted and reduced the expression levels of microRNA-145-5p (miR-145-5p), which upregulated the expression levels of Krüppel like factor 4 (KLF4), promoting endothelial differentiation of ADSCs. CONCLUSION: LncRNA MEG3 induced endothelial differentiation of ADSCs by targeting miR-145-5p/KLF4, which may provide novel insights to illustrate the mechanism of endothelial differentiation of ADSCs.


Asunto(s)
Endotelio , Factor 4 Similar a Kruppel , MicroARNs , ARN Largo no Codificante , Células Madre , Tejido Adiposo/citología , Animales , Diferenciación Celular/genética , Endotelio/citología , Factor 4 Similar a Kruppel/genética , Ratones , MicroARNs/genética , ARN Largo no Codificante/genética , Células Madre/metabolismo
15.
Ann Vasc Surg ; 78: 328-335, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34182114

RESUMEN

BACKGROUND: Inflammatory dysregulation of KLF4 is related to atheromatosis. In the present study, we explored the impact of colchicine-based regimens on the development of thoracic aortic atheromatosis and KLF4 expression. METHODS: Twenty-eight New Zealand White rabbits were divided to 4 groups. The control group (n = 6) was fed standard chow, group A (n = 6) was fed chow enriched with 1% w/w cholesterol, group B (n = 8) was fed the same cholesterol-enriched diet plus 2 mg/kg body weight/day colchicine and 250 mg/kg body weight/day fenofibrate, while group C (n = 8) was also fed the same diet plus 2 mg/kg body weight/day colchicine and 15 mg/kg body weight/day N-acetylcysteine. After 7 weeks, all animals were euthanized, and their thoracic aortas were isolated. Atherosclerotic plaque area was estimated with morphometric analysis. KLF4 expression was quantified with quantitative RT-PCR. RESULTS: Group A developed significantly more atherosclerosis compared to group B (MD: 13.67, 95% CI: 7.49-19.84) and C (MD: 20.29, 95% CI: 14.12-26.47). Colchicine with N-acetylcysteine resulted in more pronounced reduction in the extent of atherosclerotic plaques compared to colchicine/fibrate (MD: 6.62, 95% CI: 0.90-12.34). Group A exhibited significantly greater KLF4 expression compared to group B (MD: 4.94, 95% CI: 1.11-8.77) and C (MD: 9.94, 95% CI: 6.11-13.77). Combining colchicine with N-acetylcysteine instead of fenofibrate (MD: 5.00, 95% CI: 1.45-8.54) led to a more robust reduction in KLF4 expression. CONCLUSIONS: In the present hyperlipidemic animal model, colchicine-based regimens curtailed de novo atherogenesis and KLF4 overexpression in thoracic aortas.


Asunto(s)
Antiinflamatorios/farmacología , Aorta Torácica/efectos de los fármacos , Enfermedades de la Aorta/tratamiento farmacológico , Aterosclerosis/tratamiento farmacológico , Colchicina/farmacología , Hiperlipidemias/complicaciones , Factor 4 Similar a Kruppel/metabolismo , Placa Aterosclerótica , Acetilcisteína/farmacología , Animales , Aorta Torácica/metabolismo , Aorta Torácica/patología , Enfermedades de la Aorta/etiología , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Ácidos Fíbricos/farmacología , Factor 4 Similar a Kruppel/genética , Masculino , Conejos , Regulación hacia Arriba
16.
Int J Mol Sci ; 23(2)2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-35054772

RESUMEN

Platelet factor 4 (CXCL4) is a chemokine abundantly stored in platelets. Upon injury and during atherosclerosis, CXCL4 is transported through the vessel wall where it modulates the function of vascular smooth muscle cells (VSMCs) by affecting proliferation, migration, gene expression and cytokine release. Variant CXCL4L1 is distinct from CXCL4 in function and expression pattern, despite a minor three-amino acid difference. Here, the effects of CXCL4 and CXCL4L1 on the phenotype and function of human VSMCs were compared in vitro. VSMCs were found to constitutively express CXCL4L1 and only exogenously added CXCL4 was internalized by VSMCs. Pre-treatment with heparin completely blocked CXCL4 uptake. A role of the putative CXCL4 receptors CXCR3 and DARC in endocytosis was excluded, but LDL receptor family members appeared to be involved in the uptake of CXCL4. Incubation of VSMCs with both CXCL4 and CXCL4L1 resulted in decreased expression of contractile marker genes and increased mRNA levels of KLF4 and NLRP3 transcription factors, yet only CXCL4 stimulated proliferation and calcification of VSMCs. In conclusion, CXCL4 and CXCL4L1 both modulate gene expression, yet only CXCL4 increases the division rate and formation of calcium-phosphate crystals in VSMCs. CXCL4 and CXCL4L1 may play distinct roles during vascular remodeling in which CXCL4 induces proliferation and calcification while endogenously expressed CXCL4L1 governs cellular homeostasis. The latter notion remains a subject for future investigation.


Asunto(s)
Calcinosis , Proliferación Celular , Contracción Muscular , Músculo Liso Vascular/metabolismo , Factor Plaquetario 4/fisiología , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Factor 4 Similar a Kruppel/genética , Músculo Liso Vascular/fisiología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Factor Plaquetario 4/metabolismo
17.
Br Poult Sci ; 63(1): 39-45, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34287083

RESUMEN

1. MicroRNAs (miRNAs) play a vital role in the proliferation, differentiation, and apoptosis of myoblasts. However, the effect of miR-7 on the apoptosis of chicken primary myoblasts (CPMs) and its mechanism is still unclear.2. In this study, the expression of apoptosis marker genes (RAF1, Caspase3, Caspase9, Cytc, Fas) in CPMs was significantly increased after transfection of miR-7 mimic. The expression of the apoptosis marker genes in CPMs was significantly reduced after transfection with miR-7 inhibitor. Flow cytometry showed that the late apoptosis rate of the mimic group was significantly higher than the negative control (NC). The viable cells of the mimic group were significantly lower than the NC. In contrast, inhibition of miR-7 had the opposite effect.3. The dual-luciferase assay showed that the KLF4 was a target gene of miR-7. The rescue experiment showed that the KLF4 gene could attenuate the effect of miR-7 on the expression of apoptosis marker genes in CPMs.4. Determination of the function the KLF4 gene showed that the expression of the apoptosis marker genes in CPMs decreased significantly compared with the NC after its overexpression. Inhibition of KLF4 gene had the opposite effect. Flow cytometry showed that overexpression of the KLF4 gene inhibited early apoptosis of myoblasts (P ≤ 0.01), while interference with the KLF4 gene could promote early apoptosis of myoblasts (P ≤ 0.001).5. The results demonstrated, for the first time, that miR-7 promotes apoptosis in chicken primary myoblasts by regulating the expression of the KLF4 gene.


Asunto(s)
Apoptosis , Pollos , Factor 4 Similar a Kruppel/genética , MicroARNs , Mioblastos/citología , Animales , Proliferación Celular , Pollos/genética , MicroARNs/genética
18.
Gut ; 70(5): 928-939, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33028669

RESUMEN

OBJECTIVE: Intraductal papillary mucinous neoplasms (IPMNs) are non-invasive precursor lesions that can progress to invasive pancreatic cancer and are classified as low-grade or high-grade based on the morphology of the neoplastic epithelium. We aimed to compare genetic alterations in low-grade and high-grade regions of the same IPMN in order to identify molecular alterations underlying neoplastic progression. DESIGN: We performed multiregion whole exome sequencing on tissue samples from 17 IPMNs with both low-grade and high-grade dysplasia (76 IPMN regions, including 49 from low-grade dysplasia and 27 from high-grade dysplasia). We reconstructed the phylogeny for each case, and we assessed mutations in a novel driver gene in an independent cohort of 63 IPMN cyst fluid samples. RESULTS: Our multiregion whole exome sequencing identified KLF4, a previously unreported genetic driver of IPMN tumorigenesis, with hotspot mutations in one of two codons identified in >50% of the analyzed IPMNs. Mutations in KLF4 were significantly more prevalent in low-grade regions in our sequenced cases. Phylogenetic analyses of whole exome sequencing data demonstrated diverse patterns of IPMN initiation and progression. Hotspot mutations in KLF4 were also identified in an independent cohort of IPMN cyst fluid samples, again with a significantly higher prevalence in low-grade IPMNs. CONCLUSION: Hotspot mutations in KLF4 occur at high prevalence in IPMNs. Unique among pancreatic driver genes, KLF4 mutations are enriched in low-grade IPMNs. These data highlight distinct molecular features of low-grade and high-grade dysplasia and suggest diverse pathways to high-grade dysplasia via the IPMN pathway.


Asunto(s)
Adenocarcinoma Mucinoso/genética , Carcinoma Papilar/genética , Secuenciación del Exoma , Neoplasias Intraductales Pancreáticas/genética , Adenocarcinoma Mucinoso/patología , Biomarcadores de Tumor/genética , Carcinoma Papilar/patología , Humanos , Factor 4 Similar a Kruppel/genética , Mutación , Clasificación del Tumor , Neoplasias Intraductales Pancreáticas/patología , Estudios Retrospectivos
19.
J Cell Mol Med ; 25(20): 9697-9709, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34562312

RESUMEN

Telocytes (TCs), a novel interstitial cell entity promoting tissue regeneration, have been described in various tissues. Their role in inter-cellular signalling and tissue remodelling has been reported in almost all human tissues. This study hypothesizes that TC also contributes to tissue remodelling and regeneration of the human thoracic aorta (HTA). The understanding of tissue homeostasis and regenerative potential of the HTA is of high clinical interest as it plays a crucial role in pathogenesis from aortic dilatation to lethal dissection. Therefore, we obtained twenty-five aortic specimens of heart donors during transplantation. The presence of TCs was detected in different layers of aortic tissue and characterized by immunofluorescence and transmission electron microscopy. Further, we cultivated and isolated TCs in highly differentiated form identified by positive staining for CD34 and c-kit. Aortic-derived TC was characterized by the expression of PDGFR-α, PDGFR-ß, CD29/integrin ß-1 and αSMA and the stem cell markers Nanog and KLF-4. Moreover, TC exosomes were isolated and characterized for soluble angiogenic factors by Western blot. CD34+ /c-kit+ TCs shed exosomes containing the soluble factors VEGF-A, KLF-4 and PDGF-A. In summary, TC occurs in the aortic wall. Correspondingly, exosomes, derived from aortic TCs, contain vasculogenesis-relevant proteins. Understanding the regulation of TC-mediated aortic remodelling may be a crucial step towards designing strategies to promote aortic repair and prevent adverse remodelling.


Asunto(s)
Aorta/citología , Exosomas/metabolismo , Expresión Génica , Telocitos/citología , Telocitos/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Biomarcadores , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/ultraestructura , Exosomas/ultraestructura , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Inmunofenotipificación , Factor 4 Similar a Kruppel/genética , Factor 4 Similar a Kruppel/metabolismo , Miocitos del Músculo Liso/metabolismo , Telocitos/ultraestructura , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
BMC Cancer ; 21(1): 1218, 2021 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-34774019

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) have been reported to play significant roles in non-small-cell lung cancer (NSCLC). However, the roles of microRNA (miR)-1915-3p in NSCLC remain unclear. In this study, we aimed to explore the biological functions of miR-1915-3p in NSCLC. METHODS: The expression of miR-1915-3p and SET nuclear proto-oncogene (SET) in NSCLC tissues were examined by quantitative real-time PCR (qRT-PCR). Migratory and invasive abilities of lung cancer were tested by wound healing and transwell invasion assay. The direct target genes of miR-1915-3p were measured by dual-luciferase reporter assay and western blot. Finally, the regulation between METTL3/YTHDF2/KLF4 axis and miR-1915-3p were evaluated by qRT-PCR, promoter reporter assay and chromatin immunoprecipitation (CHIP). RESULTS: miR-1915-3p was downregulated in NSCLC tissues and cell lines, and inversely associated with clinical TNM stage and overall survival. Functional assays showed that miR-1915-3p significantly suppressed migration, invasion and epithelial-mesenchymal transition (EMT) in NSCLC cells. Furthermore, miR-1915-3p directly bound to the 3'untranslated region (3'UTR) of SET and modulated the expression of SET. SET inhibition could recapitulate the inhibitory effects on cell migration, invasion and EMT of miR-1915-3p, and restoration of SET expression could abrogate these effects induced by miR-1915-3p through JNK/Jun and NF-κB signaling pathways. What's more, miR-1915-3p expression was regulated by METTL3/YTHDF2 m6A axis through transcription factor KLF4. CONCLUSIONS: These findings demonstrate that miR-1915-3p function as a tumor suppressor by targeting SET and may have an anti-metastatic therapeutic potential for lung cancer treatment.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Proteínas de Unión al ADN/genética , Expresión Génica , Chaperonas de Histonas/genética , Neoplasias Pulmonares/genética , MicroARNs/fisiología , Células A549 , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Movimiento Celular/genética , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Transición Epitelial-Mesenquimal/genética , Femenino , Genes Reporteros , Genes Supresores de Tumor/fisiología , Chaperonas de Histonas/antagonistas & inhibidores , Chaperonas de Histonas/metabolismo , Humanos , Factor 4 Similar a Kruppel/genética , Factor 4 Similar a Kruppel/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Metiltransferasas/genética , Metiltransferasas/metabolismo , MicroARNs/genética , Persona de Mediana Edad , Invasividad Neoplásica/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA