Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 80(8): 229, 2023 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-37498354

RESUMEN

circACTA2 derived from the smooth muscle α-actin gene plays an important role in the regulation of vascular smooth muscle cell (VSMC) phenotype. The activation of NLRP3 inflammasome is involved in VSMC phenotypic switching. However, the mechanistic relationship between circACTA2 and NLRP3 inflammasome during vascular remodeling remains poorly understood. Here, we showed that circACTA2 was down-regulated in human intimal hyperplasia. circACTA2 overexpression in circACTA2 transgenic mice significantly decreased the neointimal hyperplasia induced by vascular injury, which is concomitant with a decrease in IL-18, IL-1ß, TNF-α, and IL-6 levels. Gain- and loss-of-function studies revealed that circACTA2 alleviated VSMC inflammation by suppressing the activation of NLRP3 inflammasome. Mechanistically, circACTA2 inhibited the expression of NF-κB p65 and p50 subunits and interacted with p50, which impedes the formation of the p50/p65 heterodimer and nuclear translocation induced by TNF-α, thus resulting in the suppression of NLRP3 gene transcription and inflammasome activation. Furthermore, circACTA2 overexpression mitigated inflammation via repressing NLRP3 inflammasome-mediated VSMC pyroptosis. Importantly, employing a decoy oligonucleotide to compete with circACTA2 for binding to p50 could attenuate the expression of NLRP3, ASC, and caspase-1. These findings provide a novel insight into the functional roles of circACTA2 in VSMCs, and targeting the circACTA2-NF-κB-NLRP3 axis represents a promising therapeutic strategy for vascular remodeling.


Asunto(s)
Inflamasomas , FN-kappa B , Ratones , Animales , Humanos , FN-kappa B/metabolismo , Inflamasomas/genética , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Músculo Liso Vascular/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Remodelación Vascular , Hiperplasia/metabolismo , Inflamación/patología
2.
Glia ; 71(7): 1715-1728, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36971019

RESUMEN

Our previous studies indicated that RhoA knockdown or inhibition could alleviate the proliferation, migration, and differentiation of Schwann cells. However, the role of RhoA in Schwann cells during nerve injury and repair is still unknown. Herein, we developed two lines of Schwann cells conditional RhoA knockout (cKO) mice by breeding RhoAflox / flox mice with PlpCre -ERT2 or DhhCre mice. Our results indicate that RhoA cKO in Schwann cells accelerates axonal regrowth and remyelination after sciatic nerve injury, which enhances the recovery of nerve conduction and hindlimb gait, and alleviates the amyotrophy in gastrocnemius muscle. Mechanistic studies in both in vivo and in vitro models revealed that RhoA cKO could facilitate Schwann cell dedifferentiation via JNK pathway. Schwann cell dedifferentiation subsequently promotes Wallerian degeneration by enhancing phagocytosis and myelinophagy, as well as stimulating the production of neurotrophins (NT-3, NGF, BDNF, and GDNF). These findings shed light on the role of RhoA in Schwann cells during nerve injury and repair, indicating that cell type-specific RhoA targeting could serve as a promising molecular therapeutic strategy for peripheral nerve injury.


Asunto(s)
Traumatismos de los Nervios Periféricos , Neuropatía Ciática , Ratones , Animales , Desdiferenciación Celular , Nervio Ciático/metabolismo , Células de Schwann/metabolismo , Neuropatía Ciática/metabolismo , Regeneración Nerviosa/fisiología , Traumatismos de los Nervios Periféricos/metabolismo
3.
FASEB J ; 36(11): e22602, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36250925

RESUMEN

Chronic inflammation is one of the definite factors leading to the occurrence and development of tumors, including prostate cancer (PCa). The androgen receptor (AR) pathway is essential for PCa tumorigenesis and inflammatory response. However, little is known about the AR-regulated NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) inflammasome pathway in human PCa. In this study, we explored the expression of inflammatory cytokine and AR in high-grade PCa and observed that NLRP3 inflammasome-associated genes were upregulated in high-grade PCa compared with that in low-grade PCa and benign prostatic hyperplasia and were associated with AR expression. In addition, we identified circAR-3-a circRNA derived from the AR gene-which is involved in the AR-regulated inflammatory response and cell proliferation by activating the NLRP3 inflammatory pathway. While circAR-3 overexpression promoted cell proliferation and the inflammatory response, its depletion induced opposite effects. Mechanistically, we noted that circAR-3 mediated the acetylation modification of NLRP3 by KAT2B and then promoted NLRP3 inflammasome complex subcellular distribution and assembly. Disturbing NLRP3 acetylation or blocking inflammasome assembly with an inhibitor suppressed the progression of PCa xenograft tumors. Our findings provide the first evidence that targeting NLRP3 acetylation or inflammasome assembly may be effective in inhibiting PCa progression.


Asunto(s)
Neoplasias de la Próstata , Receptores Androgénicos , Acetilación , Citocinas/metabolismo , Humanos , Inflamasomas/genética , Inflamasomas/metabolismo , Masculino , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neoplasias de la Próstata/metabolismo , ARN Circular , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo
4.
PLoS Biol ; 18(8): e3000808, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32817651

RESUMEN

Although dysregulation of mitochondrial dynamics has been linked to cellular senescence, which contributes to advanced age-related disorders, it is unclear how Krüppel-like factor 5 (Klf5), an essential transcriptional factor of cardiovascular remodeling, mediates the link between mitochondrial dynamics and vascular smooth muscle cell (VSMC) senescence. Here, we show that Klf5 down-regulation in VSMCs is correlated with rupture of abdominal aortic aneurysm (AAA), an age-related vascular disease. Mice lacking Klf5 in VSMCs exacerbate vascular senescence and progression of angiotensin II (Ang II)-induced AAA by facilitating reactive oxygen species (ROS) formation. Klf5 knockdown enhances, while Klf5 overexpression suppresses mitochondrial fission. Mechanistically, Klf5 activates eukaryotic translation initiation factor 5a (eIF5a) transcription through binding to the promoter of eIF5a, which in turn preserves mitochondrial integrity by interacting with mitofusin 1 (Mfn1). Accordingly, decreased expression of eIF5a elicited by Klf5 down-regulation leads to mitochondrial fission and excessive ROS production. Inhibition of mitochondrial fission decreases ROS production and VSMC senescence. Our studies provide a potential therapeutic target for age-related vascular disorders.


Asunto(s)
Aneurisma de la Aorta Abdominal/genética , Células Endoteliales/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Mitocondrias/metabolismo , Factores de Iniciación de Péptidos/genética , Proteínas de Unión al ARN/genética , Anciano , Angiotensina II/genética , Angiotensina II/metabolismo , Angiotensina II/farmacología , Animales , Aorta/diagnóstico por imagen , Aorta/metabolismo , Aorta/patología , Aneurisma de la Aorta Abdominal/diagnóstico por imagen , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Senescencia Celular/efectos de los fármacos , Ecocardiografía , Células Endoteliales/patología , Femenino , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/deficiencia , Masculino , Ratones , Ratones Noqueados , Mitocondrias/patología , Dinámicas Mitocondriales/efectos de los fármacos , Factores de Iniciación de Péptidos/deficiencia , Cultivo Primario de Células , Regiones Promotoras Genéticas , Unión Proteica , Especies Reactivas de Oxígeno/metabolismo , Factor 5A Eucariótico de Iniciación de Traducción
5.
Mol Ther ; 30(1): 164-174, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33992807

RESUMEN

Transthyretin (TTR) amyloidosis is a hereditary life-threatening disease characterized by deposition of amyloid fibrils. The main causes of TTR amyloidosis are mutations in the TTR gene that lead to the production of misfolded TTR protein. Reducing the production of toxic protein in the liver is a validated strategy to treat TTR amyloidosis. In this study, we established a humanized mouse model that expresses mutant human TTR (hTTR; V30M) protein in the liver to model TTR amyloidosis. Then, we compared the efficiency of reducing the expression of mutant hTTR by dual adeno-associated virus 8 (AAV8)-mediated split SpCas9 with that by single AAV8-mediated Nme2Cas9 in this model. With two gRNAs targeting different exons, dual AAV-mediated split SpCas9 system achieved efficiencies of 37% and 34% reduction of hTTR mRNA and reporter GFP expression, respectively, in the liver. Surprisingly, single AAV-mediated Nme2Cas9 treatment resulted in 65% and 71% reduction of hTTR mRNA and reporter GFP, respectively. No significant editing was identified in predicted off-target sites in the mouse and human genomes after Nme2Cas9 targeting. Thus, we provide proof of principle for using single AAV-mediated CRISPR-Nme2Cas9 to effectively reduce mutant hTTR expression in vivo, which may translate into gene therapy for TTR amyloidosis.


Asunto(s)
Neuropatías Amiloides Familiares , Amiloide , Neuropatías Amiloides Familiares/genética , Neuropatías Amiloides Familiares/terapia , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Prealbúmina/genética
6.
Mol Ther ; 30(1): 283-294, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-34298129

RESUMEN

Prime editor (PE), a new genome editing tool, can generate all 12 possible base-to-base conversions, insertion, and deletion of short fragment DNA. PE has the potential to correct the majority of known human genetic disease-related mutations. Adeno-associated viruses (AAVs), the safe vector widely used in clinics, are not capable of delivering PE (∼6.3 kb) in a single vector because of the limited loading capacity (∼4.8 kb). To accommodate the loading capacity of AAVs, we constructed four split-PE (split-PE994, split-PE1005, split-PE1024, and split-PE1032) using Rma intein (Rhodothermus marinus). With the use of a GFP-mutated reporter system, PE reconstituting activities were screened, and two efficient split-PEs (split-PE1005 and split-PE1024) were identified. We then demonstrated that split-PEs delivered by dual-AAV1, especially split-PE1024, could mediate base transversion and insertion at four endogenous sites in human cells. To test the performance of split-PE in vivo, split-PE1024 was then delivered into the adult mouse retina by dual-AAV8. We demonstrated successful editing of Dnmt1 in adult mouse retina. Our study provides a new method to deliver PE to adult tissue, paving the way for in vivo gene-editing therapy using PE.


Asunto(s)
Dependovirus , Edición Génica , Animales , ADN , Dependovirus/genética , Edición Génica/métodos , Vectores Genéticos/genética , Inteínas/genética , Ratones , Mutación
7.
J Neuroinflammation ; 18(1): 234, 2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34654444

RESUMEN

BACKGROUND: Plenty of macrophages are recruited to the injured nerve to play key roles in the immunoreaction and engulf the debris of degenerated axons and myelin during Wallerian degeneration, thus creating a conducive microenvironment for nerve regeneration. Recently, drugs targeting the RhoA pathway have been widely used to promote peripheral axonal regeneration. However, the role of RhoA in macrophage during Wallerian degeneration and nerve regeneration after peripheral nerve injury is still unknown. Herein, we come up with the hypothesis that RhoA might influence Wallerian degeneration and nerve regeneration by affecting the migration and phagocytosis of macrophages after peripheral nerve injury. METHODS: Immunohistochemistry, Western blotting, H&E staining, and electrophysiology were performed to access the Wallerian degeneration and axonal regeneration after sciatic nerve transection and crush injury in the LyzCre+/-; RhoAflox/flox (cKO) mice or Lyz2Cre+/- (Cre) mice, regardless of sex. Macrophages' migration and phagocytosis were detected in the injured nerves and the cultured macrophages. Moreover, the expression and potential roles of ROCK and MLCK were also evaluated in the cultured macrophages. RESULTS: 1. RhoA was specifically knocked out in macrophages of the cKO mice; 2. The segmentation of axons and myelin, the axonal regeneration, and nerve conduction in the injured nerve were significantly impeded while the myoatrophy was more severe in the cKO mice compared with those in Cre mice; 3. RhoA knockout attenuated the migration and phagocytosis of macrophages in vivo and in vitro; 4. ROCK and MLCK were downregulated in the cKO macrophages while inhibition of ROCK and MLCK could weaken the migration and phagocytosis of macrophages. CONCLUSIONS: Our findings suggest that RhoA depletion in macrophages exerts a detrimental effect on Wallerian degeneration and nerve regeneration, which is most likely due to the impaired migration and phagocytosis of macrophages resulted from disrupted RhoA/ROCK/MLCK pathway. Since previous research has proved RhoA inhibition in neurons was favoring for axonal regeneration, the present study reminds us of that the cellular specificity of RhoA-targeted drugs is needed to be considered in the future application for treating peripheral nerve injury.


Asunto(s)
Macrófagos/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo , Degeneración Walleriana/metabolismo , Degeneración Walleriana/prevención & control , Proteína de Unión al GTP rhoA/deficiencia , Animales , Movimiento Celular/fisiología , Células Cultivadas , Femenino , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Traumatismos de los Nervios Periféricos/patología , Degeneración Walleriana/patología , Proteína de Unión al GTP rhoA/genética
8.
Transgenic Res ; 30(1): 63-76, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33394315

RESUMEN

Taking advantage of their ability to integrate their genomes into the host genome, lentiviruses have been used to rapidly produce transgenic mice in biomedical research. In most cases, transgenes delivered by lentiviral vectors have resisted silencing mediated by epigenetic modifications in mice. However, some studies revealed that methylation caused decreased transgene expression in mice. Therefore, there is conflicting evidence regarding the methylation-induced silencing of transgenes delivered by lentiviral transduction in mice. In this study, we present evidence that the human TTR transgene was silenced by DNA methylation in the liver of a transgenic mouse model generated by lentiviral transduction. The density of methylation on the transgene was increased during reproduction, and the expression of the transgene was completely silenced in mice of the F2 generation. Interestingly, 5-azacytidine (5-AzaC), a methyltransferase inhibitor, potently reactivated the silenced genes in neonatal mice whose hepatocytes were actively proliferating and led to stable transgene expression during development. However, 5-AzaC did not rescue liver transgene expression when administered to adult mice. Moreover, 5-AzaC at the given dose had low developmental toxicity in the newborn mice. In summary, we demonstrate the methylation-induced silencing of an exogenous gene in the liver of a mouse model generated by lentiviral transduction and show that the silenced transgene can be safely and efficiently reactivated by 5-AzaC treatment, providing an alternative way to obtain progeny with stable transgene expression in the case of the methylation of exogenous genes in transgenic mice generated by lentiviral transduction.


Asunto(s)
Metilación de ADN/genética , Lentivirus/genética , Prealbúmina/genética , Transgenes/genética , Animales , Animales Recién Nacidos , Azacitidina/farmacología , Metilación de ADN/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/efectos de los fármacos , Humanos , Lentivirus/efectos de los fármacos , Ratones , Ratones Transgénicos/genética
10.
Circ Res ; 121(6): 628-635, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28698179

RESUMEN

RATIONALE: Neuregulin-1 (NRG-1) includes an extracellular epidermal growth factor-like domain and an intracellular domain (NRG-1-ICD). In response to transforming growth factor-ß1, its cleavage by proteolytic enzymes releases a bioactive fragment, which suppresses the vascular smooth muscle cell (VSMC) proliferation by activating ErbB (erythroblastic leukemia viral oncogene homolog) receptor. However, NRG-1-ICD function in VSMCs remains unknown. OBJECTIVE: Here, we characterize the function of NRG-1-ICD and underlying mechanisms in VSMCs. METHODS AND RESULTS: Immunofluorescence staining, Western blotting, and quantitative real-time polymerase chain reaction showed that NRG-1 was expressed in rat, mouse, and human VSMCs and was upregulated and cleaved in response to transforming growth factor-ß1. In the cytoplasm of HASMCs (human aortic smooth muscle cells), the NRG-1-ICD participated in filamentous actin formation by interacting with α-SMA (smooth muscle α-actin). In the nucleus, the Nrg-1-ICD induced circular ACTA2 (alpha-actin-2; circACTA2) formation by recruitment of the zinc-finger transcription factor IKZF1 (IKAROS family zinc finger 1) to the first intron of α-SMA gene. We further confirmed that circACTA2, acting as a sponge binding microRNA (miR)-548f-5p, interacted with miR-548f-5p targeting 3' untranslated region of α-SMA mRNA, which in turn relieves miR-548f-5p repression of the α-SMA expression and thus upregulates α-SMA expression, thereby facilitating stress fiber formation and cell contraction in HASMCs. Accordingly, in vivo studies demonstrated that the localization of the interaction of circACTA2 with miR-548f-5p is significantly decreased in human intimal hyperplastic arteries compared with normal arteries, implicating that dysregulation of circACTA2 and miR-548f-5p expression is involved in intimal hyperplasia. CONCLUSIONS: These results suggest that circACTA2 mediates NRG-1-ICD regulation of α-SMA expression in HASMCs via the NRG-1-ICD/circACTA2/miR-548f-5p axis. Our data provide a molecular basis for fine-tuning α-SMA expression and VSMC contraction by transcription factor, circular RNA, and microRNA.


Asunto(s)
Actinas/metabolismo , MicroARNs/genética , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neurregulina-1/metabolismo , Actinas/genética , Animales , Células Cultivadas , Células HEK293 , Humanos , Factor de Transcripción Ikaros/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Ratas
11.
Circ Res ; 120(5): 799-815, 2017 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-28115390

RESUMEN

RATIONALE: Abdominal aortic aneurysms (AAAs) are characterized by pathological remodeling of the aortic wall. Although both increased Krüppel-like factor 5 (KLF5) expression and macrophage infiltration have been implicated in vascular remodeling, the role of KLF5 in macrophage infiltration and AAA formation remains unclear. OBJECTIVE: To determine the role of KLF5 in AAA formation and macrophage infiltration into AAAs. METHODS AND RESULTS: KLF5 expression was significantly increased in human AAA tissues and in 2 mouse models of experimental AAA. Moreover, in myeloid-specific Klf5 knockout mice (myeKlf5-/- mice), macrophage infiltration, medial smooth muscle cell loss, elastin degradation, and AAA formation were markedly decreased. In cell migration and time-lapse imaging analyses, the migration of murine myeKlf5-/- macrophages was impaired, and in luciferase reporter assays, KLF5 activated Myo9b (myosin IXB) transcription by direct binding to the Myo9b promoter. In subsequent coimmunostaining studies, Myo9b was colocalized with filamentous actin, cortactin, vinculin, and Tks5 in the podosomes of phorbol 12,13-dibutyrate-treated macrophages, indicating that Myo9b participates in podosome formation. Gain- and loss-of-function experiments showed that KLF5 promoted podosome formation in macrophages by upregulating Myo9b expression. Furthermore, RhoA-GTP levels increased after KLF5 knockdown in macrophages, suggesting that KLF5 lies upstream of RhoA signaling. Finally, Myo9b expression was increased in human AAA tissues, located in macrophages, and positively correlated with AAA size. CONCLUSIONS: These data are the first to indicate that KLF5-dependent regulation of Myo9b/RhoA is required for podosome formation and macrophage migration during AAA formation, warranting consideration of the KLF5-Myo9b-RhoA pathway as a therapeutic target for AAA treatment.


Asunto(s)
Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/prevención & control , Factores de Transcripción de Tipo Kruppel/biosíntesis , Macrófagos/metabolismo , Miosinas/biosíntesis , Podosomas/metabolismo , Proteína de Unión al GTP rhoA/biosíntesis , Animales , Línea Celular , Células HEK293 , Humanos , Factores de Transcripción de Tipo Kruppel/deficiencia , Masculino , Ratones , Ratones Noqueados , Miosinas/deficiencia , Transducción de Señal/fisiología , Proteína de Unión al GTP rhoA/deficiencia
12.
Cell Physiol Biochem ; 46(4): 1536-1554, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29689560

RESUMEN

BACKGROUND/AIMS: This study determined the role and mechanism of action of transcription factor EB (TFEB) in H2O2-induced neuronal apoptosis. METHODS: SH-SY5Y cells were treated with Akt inhibitor/activator and different concentrations of H2O2. Cell apoptosis was detected by flow cytometric analysis. Akt and TFEB phosphorylation and PARP cleavage were determined by Western blotting. HEK293T cells were transfected with different truncated TFEB mutants and HA-Akt-WT; SH-SY5Y cells were transfected with Flag-vector, Flag-TFEB, Flag-TFEB-S467A or Flag-TFEB-S467D; and TFEB interaction with Akt was determined by co-immunoprecipitation and GST pull-down assays. RESULTS: A low concentration of H2O2 induces TFEB phosphorylation at Ser467 and nuclear translocation, facilitating neuronal survival, whereas a high concentration of H2O2 promotes SH-SY5Y cell apoptosis via suppressing TFEB Ser467 phosphorylation and nuclear translocation. The TFEB-S467D mutant is more easily translocated into the nucleus than the non-phosphorylated TFEB-S467A mutant. Further, Akt physically binds to TFEB via its C-terminal tail interaction with the HLH domain of TFEB and phosphorylates TFEB at Ser467. Mutation of TFEB-Ser467 can prevent the phosphorylation of TFEB by Akt, preventing inhibition of oxidative stress-induced apoptosis. CONCLUSIONS: Oxidative stress induces neuronal apoptosis through suppressing TFEB phosphorylation at Ser467 by Akt, providing a novel therapeutic strategy for neurodegenerative diseases.


Asunto(s)
Apoptosis/efectos de los fármacos , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Peróxido de Hidrógeno/toxicidad , Estrés Oxidativo/efectos de los fármacos , Androstadienos/farmacología , Animales , Línea Celular Tumoral , Flavonoides/farmacología , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Serina/metabolismo , Transducción de Señal/efectos de los fármacos , Wortmanina
13.
Biochim Biophys Acta Mol Basis Dis ; 1864(2): 374-386, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29074464

RESUMEN

Atherogenesis is a chronic inflammatory process that involves complex interactions between endothelial dysfunction, lipid deposition and vascular smooth-muscle cell (VSMC) proliferation. However, the molecular mechanism is still unclear. We found that a pro-atherosclerotic factor (oxLDL) induced the expression of Krüppel-like factor 5 (KLF5), which in turn increased miR-29a expression levels. The increased miR-29a was retained within HASMCs and down-regulated Fbw7/CDC4 expression by targeting the 3´UTR of Fbw7/CDC4, subsequently increasing KLF5 stability by reducing the Fbw7/CDC4-dependent ubiquitination of KLF5, forming a positive feedback loop to enhance VSMC proliferation and promote atherogenesis. These results indicate a potentially important role for the oxLDL-activated feedback mechanism in VSMC proliferation and atherogenesis. Suppression of miR-29a may be an effective way to attenuate atherosclerosis. In conclusion, our data are the first to reveal that the regulatory crosstalk between KLF5, miR-29a, and Fbw7/CDC4 cooperatively promotes atherosclerotic development.


Asunto(s)
Aterosclerosis/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Regulación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , MicroARNs/metabolismo , Regiones no Traducidas 3' , Animales , Aorta/citología , Biomarcadores/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Perfilación de la Expresión Génica , Humanos , Inflamación , Factores de Transcripción de Tipo Kruppel/genética , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Noqueados para ApoE , MicroARNs/genética , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Células 3T3 NIH , Ubiquitinación
14.
Mol Ther ; 25(6): 1279-1294, 2017 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-28408180

RESUMEN

The vascular response to pro-atherosclerotic factors is a multifactorial process involving endothelial cells (ECs), macrophages (MACs), and smooth muscle cells (SMCs), although the mechanism by which these cell types communicate with each other in response to environmental cues is yet to be understood. Here, we show that miR-155, which is significantly expressed and secreted in Krüppel-like factor 5 (KLF5)-overexpressing vascular smooth muscle cells (VSMCs), is a potent regulator of endothelium barrier function through regulating endothelial targeting tight junction protein expression. VSMCs-derived exosomes mediate the transfer of KLF5-induced miR-155 from SMCs to ECs, which, in turn, destroys tight junctions and the integrity of endothelial barriers, leading to an increased endothelial permeability and enhanced atherosclerotic progression. Moreover, overexpression of miR-155 in ECs inhibits endothelial cell proliferation/migration and re-endothelialization in vitro and in vivo and thus increases vascular endothelial permeability. Blockage of the exosome-mediated transfer of miR-155 between these two cells may serve as a therapeutic target for atherosclerosis.


Asunto(s)
Aterosclerosis/genética , Aterosclerosis/metabolismo , Células Endoteliales/metabolismo , Exosomas/metabolismo , Técnicas de Transferencia de Gen , MicroARNs/genética , MicroARNs/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Aterosclerosis/patología , Transporte Biológico , Permeabilidad de la Membrana Celular , Modelos Animales de Enfermedad , Endotelio/metabolismo , Endotelio/patología , Eliminación de Gen , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Mutación
15.
Neural Plast ; 2018: 8561704, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29849572

RESUMEN

Accumulating evidences suggest that peripheral nerve injury (PNI) may initiate astrocytic responses in the central nervous system (CNS). However, the response of astrocytes in the spinal ventral horn and its potential role in nerve regeneration after PNI remain unclear. Herein, we firstly illustrated that astrocytes in the spinal ventral horn were dramatically activated in the early stage following sciatic nerve injury, and these profiles were eliminated in the chronic stage. Additionally, we found that the expression of neurotrophins, including brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), and neurotrophin-3 (NT-3), also accompanied with astrocyte activation. In comparison with the irreversible transected subjects, astrocyte activation and the neurotrophic upregulation in the early stage were more drastic in case the transected nerve was rebridged immediately after injury. Furthermore, administering fluorocitrate to inhibit astrocyte activation resulted in decreased neurotrophin expression in the spinal ventral horn and delayed axonal regeneration in the nerve as well as motor function recovery. Overall, the present study indicates that peripheral nerve injury can initiate astrocyte activation accompanied with neurotrophin upregulation in the spinal ventral horn. The above responses mainly occur in the early stage of PNI and may contribute to nerve regeneration and motor function recovery.


Asunto(s)
Astrocitos/metabolismo , Regeneración Nerviosa , Traumatismos de los Nervios Periféricos/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Asta Ventral de la Médula Espinal/metabolismo , Animales , Femenino , Factores de Crecimiento Nervioso/metabolismo , Traumatismos de los Nervios Periféricos/complicaciones , Ratas Sprague-Dawley , Nervio Ciático/lesiones , Traumatismos de la Médula Espinal/complicaciones , Asta Ventral de la Médula Espinal/fisiopatología
16.
Biochem Biophys Res Commun ; 482(2): 366-374, 2017 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-27856242

RESUMEN

KLF5 and nuclear factor κB (NF-κB) regulate cell proliferation and inflammation. Vitamin D signaling through vitamin D receptor (VDR) exerts anti-proliferative and anti-inflammatory actions. However, an actual relationship between KLF5, NF-κB and VDR in the inflammation and proliferation of macrophages is still unclear. Here, we showed that LPS and proinflammatory cytokines stimulate KLF5 gene expression in macrophages, and that 1, 25(OH)2D3 suppresses LPS-induced KLF5 expression and cell proliferation via upregulation of VDR expression. Mechanistic studies suggested that KLF5 interacts with p50 subunit of NF-κB to cooperatively induce the expressions of positive cell cycle regulators cyclin B1 and Cdk1/Cdc2 in LPS-treated macrophages. Further studies revealed that 1, 25(OH)2D3-induced interaction of VDR with p50 decreases LPS-induced interaction of KLF5 with p50. Collectively, we identify a novel regulatory pathway in which 1, 25(OH)2D3 induces VDR expression and promotes VDR interaction with p50 subunit of NF-κB, which in turn attenuates the association of KLF5 with p50 subunit of NF-κB and thus exerts anti-inflammatory and anti-proliferative effects on macrophages.


Asunto(s)
Proliferación Celular/fisiología , Factores de Transcripción de Tipo Kruppel/metabolismo , Lipopolisacáridos/administración & dosificación , Activación de Macrófagos/fisiología , Receptores de Calcitriol/metabolismo , Vitamina D/análogos & derivados , Animales , Sitios de Unión , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Activación de Macrófagos/efectos de los fármacos , Ratones , FN-kappa B , Unión Proteica , Subunidades de Proteína/metabolismo , Células RAW 264.7 , Vitamina D/administración & dosificación
17.
Biochim Biophys Acta Mol Basis Dis ; 1863(11): 2821-2834, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28711598

RESUMEN

Inducible NO synthase (iNOS) expression and peroxynitrite formation are significantly increased in diabetic vascular tissues. Transcription factor KLF5 activates iNOS gene transcription and is involved in vascular inflammatory injury and remodeling. However, mutual regulation between KLF5, iNOS and peroxynitrite in diabetic vascular inflammation, as well as the underlying mechanisms, remain largely unknown. In this study, we found a marked increase in KLF5 and iNOS expression in vascular smooth muscle cells (VSMC) of diabetic patients. High glucose-induced expression of KLF5 and iNOS was also observed in cultured mouse VSMCs. Further investigation showed that high glucose induced KLF5 nitration by iNOS-mediated peroxynitrite generation, and nitrated KLF5 increased its interaction with NF-κB p50 and thus cooperatively activated the expression of inflammatory cytokines TNF-α and IL-1ß. Furthermore, we showed that the VSMC-specific knockout of KLF5 dramatically reduced inflammatory cytokine expression in the vascular tissues of diabetic mice. Moreover, 17ß-estradiol (E2) inhibited high glucose-mediated effects in VSMCs, and in the response to E2, estrogen receptor (ER) α competed with KLF5 for binding to NF-κB p50, which in turn leads to the suppression of inflammatory gene expression in VSMCs. Together, the present findings were the first to show that KLF5 expression and nitration by iNOS-mediated peroxynitrite are necessary for the induction of TNF-α and IL-1ß expression in VSMCs of diabetic vascular tissues.


Asunto(s)
Angiopatías Diabéticas/metabolismo , Glucosa/farmacología , Factores de Transcripción de Tipo Kruppel/biosíntesis , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ácido Peroxinitroso/metabolismo , Angiopatías Diabéticas/patología , Femenino , Glucosa/efectos adversos , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Masculino , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Subunidad p50 de NF-kappa B/metabolismo
18.
Exp Cell Res ; 342(1): 20-31, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26945917

RESUMEN

The regulation of vascular smooth muscle cell (VSMC) proliferation is an important issue due to its major implications for the prevention of pathological vascular conditions. The objective of this work was to assess the function of small ubiquitin-like modifier (SUMO)ylated Krϋppel-like transcription factor 4 (KLF4) in the regulation of VSMC proliferation in cultured cells and in animal models with balloon injury. We found that under basal conditions, binding of non-SUMOylated KLF4 to p300 activated p21 (p21(WAF1/CIP1))transcription, leading to VSMC growth arrest. PDGF-BB promoted the interaction between Ubc9 and KLF4 and the SUMOylation of KLF4, which in turn recruited transcriptional corepressors to the p21 promoter. The reduction in p21 enhanced VSMC proliferation. Additionally, the SUMOylated KLF4 did not affect the expression of KLF4, thereby forming a positive feedback loop enhancing cell proliferation. These results demonstrated that SUMOylated KLF4 plays an important role in cell proliferation by reversing the transactivation action of KLF4 on p21 induced with PDGF-BB.


Asunto(s)
Proliferación Celular , Regulación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , Miocitos del Músculo Liso/fisiología , Sumoilación , Animales , Becaplermina , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Arteria Femoral/lesiones , Arteria Femoral/patología , Humanos , Factor 4 Similar a Kruppel , Masculino , Ratones Endogámicos C57BL , Músculo Liso Vascular/patología , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-sis/fisiología , Ratas Sprague-Dawley , Transcripción Genética , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Enfermedades Vasculares/metabolismo
19.
Biochim Biophys Acta ; 1852(7): 1477-89, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25892184

RESUMEN

In response to vascular injury, inflammation, oxidative stress, and cell proliferation often occur simultaneously in vascular tissues. We previously observed that microRNA-155 (miR-155), which is implicated in proliferation and inflammation is involved in neointimal hyperplasia; however, the molecular mechanisms by which it regulates these processes remain largely unknown. In this study, we observed that vascular smooth muscle cell (VSMC) proliferation and neointimal formation in wire-injured femoral arteries were reduced by the loss of miR-155 and increased by the gain of miR-155. The proliferative effect of miR-155 was also observed in cultured VSMCs. Notably, expression of the miR-155-target protein mammalian sterile 20-like kinase 2 (MST2) was increased in the injured arteries of miR-155-/- mice. miR-155 directly repressed MST2 and thus activated the extracellular signal-regulated kinase (ERK) pathway by promoting an interaction between RAF proto-oncogene serine/threonine-protein kinase (Raf-1) and mitogen-activated protein kinase kinase (MEK) and stimulating inflammatory and oxidative stress responses; together, these effects lead to VSMC proliferation and vascular remodeling. Our data reveal that MST2 mediates miR-155-promoted inflammatory and oxidative stress responses by altering the interaction of MEK with Raf-1 and MST2 in response to vascular injury. Therefore, suppression of endogenous miR-155 might be a novel therapeutic strategy for vascular injury and remodeling.


Asunto(s)
Proliferación Celular , MicroARNs/genética , Miocitos del Músculo Liso/metabolismo , Neointima/metabolismo , Estrés Oxidativo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Células HEK293 , Humanos , Inflamación/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/fisiología , Neointima/patología , Proteínas Serina-Treonina Quinasas/genética , Proto-Oncogenes Mas , Serina-Treonina Quinasa 3
20.
FASEB J ; 29(9): 4059-70, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26082460

RESUMEN

Transcriptional activation by transcription factors is coupled with histone acetylation and chromatin remodeling. However, the relationship between TGF-ß1-induced gene transcription by Krüppel-like factor (KLF)-4 and histone acetylation remains unknown. In our study, KLF4 overexpression or knockdown, respectively increased or decreased H3 acetylation and p300 occupancy, which is concentrated in the region containing TGF-ß1 control elements (TCEs) of the genes by TGF-ß1 regulation during vascular smooth muscle cell (VSMC) differentiation. Coimmunoprecipitation and glutathione S-transferase pull-down assays showed that phosphatase and tensin homolog (PTEN) formed a complex with KLF4 to inhibit the phosphorylation of the latter in basal conditions. After TGF-ß1 signaling activation, PTEN was phosphorylated by p38 MAPK or PI3K/Akt signaling, phosphorylated PTEN lost its ability to dephosphorylate KLF4, and the cofactors interacting with KLF4 switched from PTEN to p300. Then, KLF4-p300 complexes were recruited to KLF4-binding sites of the gene promoter of VSMCs, to acetylate histone H3 and activate transcription. In addition, phosphorylated KLF4 enhanced p300 histone acetyltransferase (HAT) activity via the p38 MAPK pathway, which may be responsible for H3 acetylation. Taken together, the results of our study reveal a novel mechanism whereby KLF4 mediates the link between TGF-ß1-induced gene transcription activation and H3 acetylation during VSMC differentiation.


Asunto(s)
Histonas/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Transcripción Genética/fisiología , Factor de Crecimiento Transformador beta1/metabolismo , Acetilación , Animales , Diferenciación Celular/fisiología , Histonas/genética , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA