Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 27
Filtrer
1.
Int J Mol Sci ; 24(3)2023 Jan 25.
Article de Anglais | MEDLINE | ID: mdl-36768713

RÉSUMÉ

Pulmonary arterial hypertension (PAH) is a progressive and inevitably fatal disease characterized by the progressive increase of pulmonary vascular resistance and obliterative pulmonary vascular remodeling, which lead to right-sided heart failure and premature death. Many of the genetically modified mouse models do not develop severe PH and occlusive vascular remodeling. Egln1Tie2Cre mice with Tie2Cre-mediated deletion of Egln1, which encodes hypoxia-inducible factor (HIF) prolyl hydroxylase 2 (PHD2), is the only mouse model with severe PAH, progressive occlusive pulmonary vascular remodeling, and right-sided heart failure leading to 50-80% mortality from the age of 3-6 months, indicating that the Egln1Tie2Cre mice model is a long-sought-after murine PAH model. However, it is unknown if Egln1Tie2Cre mice respond to FDA-approved PAH drugs in a way similar to PAH patients. Here, we tested the therapeutic effects of the three vasodilators: sildenafil (targeting nitric oxide signaling), ambrisentan (endothelin receptor antagonist), and treprostinil (prostacyclin analog) on Egln1Tie2Cre mice. All of them attenuated right ventricular systolic pressure (RVSP) in Egln1Tie2Cre mice consistent with their role as vasodilators. However, these drugs have no beneficial effects on pulmonary arterial function. Cardiac output was also markedly improved in Egln1Tie2Cre mice by any of the drug treatments. They only partially improved RV function and reduced RV hypertrophy and pulmonary vascular remodeling as well as improving short-term survival in a drug-dependent manner. These data demonstrate that Egln1Tie2Cre mice exhibit similar responses to these drugs as PAH patients seen in clinical trials. Thus, our study provides further evidence that the Egln1Tie2Cre mouse model of severe PAH is an ideal model of PAH and is potentially useful for enabling identification of drug targets and preclinical testing of novel PAH drug candidates.


Sujet(s)
Défaillance cardiaque , Hypertension pulmonaire , Hypertension artérielle pulmonaire , Souris , Animaux , Citrate de sildénafil/pharmacologie , Citrate de sildénafil/usage thérapeutique , Hypertension artérielle pulmonaire/traitement médicamenteux , Hypertension artérielle pulmonaire/génétique , Remodelage vasculaire , Hypertension pulmonaire/traitement médicamenteux , Hypertension artérielle pulmonaire primitive familiale , Vasodilatateurs/pharmacologie , Hypoxia-inducible factor-proline dioxygenases , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/génétique , Artère pulmonaire
2.
Eur Respir J ; 60(6)2022 12.
Article de Anglais | MEDLINE | ID: mdl-35798360

RÉSUMÉ

BACKGROUND: Nitrative stress is a characteristic feature of the pathology of human pulmonary arterial hypertension. However, the role of nitrative stress in the pathogenesis of obliterative vascular remodelling and severe pulmonary arterial hypertension remains largely unclear. METHOD: Our recently identified novel mouse model (Egln1Tie2Cre, Egln1 encoding prolyl hydroxylase 2 (PHD2)) has obliterative vascular remodelling and right heart failure, making it an excellent model to use in this study to examine the role of nitrative stress in obliterative vascular remodelling. RESULTS: Nitrative stress was markedly elevated whereas endothelial caveolin-1 (Cav1) expression was suppressed in the lungs of Egln1Tie2Cre mice. Treatment with a superoxide dismutase mimetic, manganese (III) tetrakis (1-methyl-4-pyridyl) porphyrin pentachloride or endothelial Nos3 knockdown using endothelial cell-targeted nanoparticle delivery of CRISPR-Cas9/guide RNA plasmid DNA inhibited obliterative pulmonary vascular remodelling and attenuated severe pulmonary hypertension in Egln1Tie2Cre mice. Genetic restoration of Cav1 expression in Egln1Tie2Cre mice normalised nitrative stress, reduced pulmonary hypertension and improved right heart function. CONCLUSION: These data suggest that suppression of Cav1 expression secondary to PHD2 deficiency augments nitrative stress through endothelial nitric oxide synthase activation, which contributes to obliterative vascular remodelling and severe pulmonary hypertension. Thus, a reactive oxygen/nitrogen species scavenger might have therapeutic potential for the inhibition of obliterative vascular remodelling and severe pulmonary arterial hypertension.


Sujet(s)
Cavéoline-1 , Hypoxia-inducible factor-proline dioxygenases , Stress nitrosatif , Hypertension artérielle pulmonaire , Remodelage vasculaire , Animaux , Humains , Souris , Cavéoline-1/génétique , Cavéoline-1/métabolisme , Poumon/métabolisme , Hypertension artérielle pulmonaire/génétique , Hypertension artérielle pulmonaire/métabolisme , Espèces réactives de l'azote/métabolisme , Espèces réactives de l'oxygène/métabolisme , Remodelage vasculaire/génétique , Stress nitrosatif/génétique , Hypoxia-inducible factor-proline dioxygenases/génétique , Hypoxia-inducible factor-proline dioxygenases/métabolisme , Modèles animaux de maladie humaine
3.
Pulm Circ ; 12(1): e12056, 2022 Jan.
Article de Anglais | MEDLINE | ID: mdl-35506101

RÉSUMÉ

BMP signaling deficiency is evident in the lungs of patients with pulmonary arterial hypertension. We demonstrated that PHD2 deficiency suppresses BMP signaling in the lung endothelial cells, suggesting the novel mechanisms of dysregulated BMP signaling in the development of pulmonary arterial hypertension.

4.
J Biochem Mol Toxicol ; 36(7): e23069, 2022 Jul.
Article de Anglais | MEDLINE | ID: mdl-35411647

RÉSUMÉ

Paternal nicotine exposure can cause a phenotypic change in offspring. To study whether paternal nicotine exposure influences acute liver injury and repair of the offspring, we established a paternal nicotine exposure model in mice and treated the offspring mice with carbon tetrachloride (CCl4 ) to induce acute liver injury. After the treatment of CCl4 , the levels of alanine aminotransferase and aspartate aminotransferase in offspring serum of paternal nicotine exposed mice are about 37.44%, and 30.21% lower than the control mice, respectively. Transcription profiling screen and bioinformatics analysis of differently expressed genes in F1 mice liver revealed that the Wnt pathway was altered. The results demonstrate that nicotine exposure in male mice could enhance the activation of the Wnt pathway in F1 mice liver. The Wnt pathway facilitates cell proliferation and tissue repair. In conclusion, our findings showed that nicotine exposure of male mice protects hepatic against CCl4 -induced acute injury in offspring by activating the Wnt pathway in the F1 liver.


Sujet(s)
Tétrachloro-méthane , Lésions hépatiques dues aux substances , Alanine transaminase , Animaux , Tétrachloro-méthane/toxicité , Lésions hépatiques dues aux substances/prévention et contrôle , Foie/métabolisme , Mâle , Souris , Nicotine/toxicité
5.
Toxicol Lett ; 343: 44-55, 2021 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-33640489

RÉSUMÉ

Paternal nicotine exposure can alter phenotypes in future generations. The aim of this study is to explore whether paternal nicotine exposure affects the hepatic repair to chronic injury which leads to hepatic fibrosis in offspring. Our results demonstrate that nicotine down regulates mmu-miR-15b expression via the hyper-methylation on its CpG island shore region in the spermatozoa. This epigenetic modification imprinted in the liver of the offspring. The decreased mmu-miR-15b promotes the expression of Wnt4 and activates the Wnt pathway in the offspring mice liver. The activation of the Wnt pathway improves the activation and proliferation of hepatic stellate cells (HSCs) leading to liver fibrosis. Moreover, the Wnt pathway promotes the activation of the TGF-ß pathway and the two pathways cooperate to promote the transcription of extracellular matrix (ECM) genes. In conclusion, this study found that nicotine promotes hepatic fibrosis in the offspring via the activation of Wnt pathway by imprinting the hyper-methylation of mmu-miR-15b.


Sujet(s)
Cirrhose du foie/induit chimiquement , Nicotine/toxicité , Agonistes nicotiniques/toxicité , Exposition paternelle , Animaux , Régulation négative , Épigenèse génétique , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Mâle , Souris , microARN/génétique , microARN/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Protéine Wnt4/génétique , Protéine Wnt4/métabolisme
6.
Toxicology ; 431: 152367, 2020 02 15.
Article de Anglais | MEDLINE | ID: mdl-31945395

RÉSUMÉ

Many substances in cigarette smoke can induce changes in DNA methylation. Our previous studies have confirmed paternal nicotine exposure causes hyperactivity in the offspring via mmu-miR-15b. The main aim of the present study is to explore the molecular mechanism underlying the cross-generation effects of paternal nicotine exposure more comprehensively. The male C57BL/6 mice were exposed to 2 mg/kg/d nicotine for 5 weeks, and then mated with wild-type females. The offspring male mice were subjected to behavioral tests at 8 weeks after birth. The results suggested that, paternal nicotine exposure led to hyperactivity in the offspring. An analysis of the changes in DNA methylation revealed that nicotine exposure induced a rise in the total DNA methylation level of Dat in murine spermatozoa, and the hyper-methylation could imprint in the brains of the offspring mice. Then these epigenetic modifications reduced the expression of DAT in the brain of the offspring, resulting in a rise in the level of extracellular dopamine. The activation of D2 receptors caused the dephosphorylation of AKT, which led to increased activation of GSK3α/ß, and ultimately caused hyperactivity in the offspring mice. Further, in wild-type mice, injection of DAT inhibitors simulated this hyperactive phenotype, while the injection of D2s inhibitors reversed the hyperactivity of the offspring caused by paternal nicotine exposure. In conclusion, all results indicated that paternal nicotine exposure could induce hyperactivity in the offspring via the hyper-methylation of Dat. Consequently, Dat may be one of the genes that mediate the cross-generation effects of nicotine besides mmu-mmiR-15b.


Sujet(s)
Trouble déficitaire de l'attention avec hyperactivité/induit chimiquement , Transporteurs de la dopamine/antagonistes et inhibiteurs , Nicotine/toxicité , Agonistes nicotiniques/toxicité , Exposition paternelle/effets indésirables , Animaux , Comportement animal/effets des médicaments et des substances chimiques , Chimie du cerveau/effets des médicaments et des substances chimiques , Méthylation de l'ADN , Dopamine/métabolisme , Régulation négative/effets des médicaments et des substances chimiques , Mâle , Souris , Souris de lignée C57BL , Récepteur D2 de la dopamine/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Spermatozoïdes/effets des médicaments et des substances chimiques , Spermatozoïdes/métabolisme
7.
Data Brief ; 24: 103825, 2019 Jun.
Article de Anglais | MEDLINE | ID: mdl-31008156

RÉSUMÉ

Data provided in this article is RNA profile represented in RPKM and RPKM based TPM value for the research article titled Nicotine inhibits Murine Leydig cell differentiation and maturation via regulating Hedgehog signal pathway Jiajie et al., 2019. Nicotine treatment changes the RNA profile of Murine Leydig cells. RNA of 12 control group Leydig cells and 12 nicotine treated Leydig cells are sequenced and the data of 29943 genes are achieved. The information of the gene symbol, gene description, gene type, position and transcript length are provided.

8.
Biochem Biophys Res Commun ; 510(1): 1-7, 2019 02 26.
Article de Anglais | MEDLINE | ID: mdl-30683315

RÉSUMÉ

Nicotine, the main toxic substance in cigarette smoke, significantly reduced the differentiation and maturation ratio of Leydig cell in murine testes. To investigate the underlying mechanism, C57BL/6J mice were divided into control (CT) and nicotine treated (NT) groups. Next generation RNA sequencing and bio-informatics analysis were carried out to analysis the effects of nicotine on the RNA profile of Leydig cells. Expression level of 7 pathways remarkably changed after nicotine treatment. As the positive regulating pathway of Leydig cell differentiation, Hedgehog signaling pathway was found among these pathways. PTCH1 and ß-TrCP were down-regulated in nicotine treated mice Leydig cells, while GSK3ß, Gli2 and Gli2 fragments increased significantly. Nicotine stimulated the destabilization of Gli2 via ß-TrCP induced ubiquitination and degradation. Gli2 was phosphorylated by up-expressed GSK3ß during this process. Destabilization of Gli2 reduced the activation rate of target genes of Hedgehog signaling pathway such as Ptch1. The differentiation of Leydig cell positively regulated by Hh pathway was thus inhibited by nicotine exposure. Consequently, the male reproduction process powered by Leydic cell-mediated androgen secretion was thus influenced. In conclusion, we find that nicotine inhibits murine Leydig cell differentiation and maturation via regulating Hedgehog signal pathway.


Sujet(s)
Différenciation cellulaire/effets des médicaments et des substances chimiques , Protéines Hedgehog/métabolisme , Cellules de Leydig/anatomopathologie , Nicotine/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Mâle , Souris , Reproduction/effets des médicaments et des substances chimiques
9.
Nat Commun ; 9(1): 3850, 2018 09 21.
Article de Anglais | MEDLINE | ID: mdl-30242159

RÉSUMÉ

The molecular mechanisms underlying the metabolic shift toward increased glycolysis observed in pulmonary artery smooth muscle cells (PASMC) during the pathogenesis of pulmonary arterial hypertension (PAH) are not fully understood. Here we show that the glycolytic enzyme α-enolase (ENO1) regulates the metabolic reprogramming and malignant phenotype of PASMC. We show that ENO1 levels are elevated in patients with associated PAH and in animal models of hypoxic pulmonary hypertension (HPH). The silencing or inhibition of ENO1 decreases PASMC proliferation and de-differentiation, and induces PASMC apoptosis, whereas the overexpression of ENO1 promotes a synthetic, de- differentiated, and apoptotic-resistant phenotype via the AMPK-Akt pathway. The suppression of ENO1 prevents the hypoxia-induced metabolic shift from mitochondrial respiration to glycolysis in PASMC. Finally, we find that pharmacological inhibition of ENO1 reverses HPH in mice and rats, suggesting ENO1 as a regulator of pathogenic metabolic reprogramming in HPH.


Sujet(s)
Hypertension pulmonaire/étiologie , Myocytes du muscle lisse/enzymologie , Enolase/métabolisme , Adenylate kinase/métabolisme , Animaux , Apoptose , Différenciation cellulaire , Respiration cellulaire , Modèles animaux de maladie humaine , Glycolyse , Humains , Hypertension pulmonaire/enzymologie , Souris , Phénotype , Enolase/antagonistes et inhibiteurs , Culture de cellules primaires , Protéines proto-oncogènes c-akt/métabolisme , Artère pulmonaire/enzymologie , Rats
10.
Cell Signal ; 51: 119-129, 2018 11.
Article de Anglais | MEDLINE | ID: mdl-30092353

RÉSUMÉ

RATIONALE: Forkhead box M1 (FoxM1) is a transcription factor that promotes cell proliferation by regulating a broad spectrum of genes that participate in cell cycle regulation, such as Cyclin B, CDC25B, and Aurora B Kinase. We have shown that hypoxia, a well-known stimulus for pulmonary hypertension (PH), induces FoxM1 in pulmonary artery smooth muscle cells (PASMC) in a HIF-dependent pathway, resulting in PASMC proliferation, while the suppression of FoxM1 prevents hypoxia-induced PASMC proliferation. However, the implications of FoxM1 in the development of PH remain less known. METHODS: We determined FoxM1 levels in the lung samples of idiopathic PAH (pulmonary arterial hypertension) (IPAH) patients and hypoxia-induced PH mice. We generated constitutive and inducible smooth muscle cell (SMC)-specific FoxM1 knockdown or knockout mice as well as FoxM1 transgenic mice which overexpress FoxM1, and exposed them to hypoxia (10% O2, 90% N2) or normoxia (Room air, 21% oxygen) for four weeks, and measured PH indices. We also isolated mouse PASMC (mPASMC) and mouse embryonic fibroblasts (MEF) from these mice to examine the cell proliferation and expression levels of SMC contractile proteins. RESULTS: We showed that in hypertensive human lungs or mouse lungs, FoxM1 levels were elevated. Constitutive knockout of FoxM1 in mouse SMC caused early lethality, whereas constitutive knockdown of FoxM1 in mouse SMC prevented hypoxia-induced PH and PASMC proliferation. Inducible knockout of FoxM1 in SMC reversed hypoxia-induced pulmonary artery wall remodeling in existing PH. Overexpression of FoxM1 enhanced hypoxia-induced pulmonary artery wall remodeling and right ventricular hypertrophy in mice. Alteration of FoxM1 status did not affect hypoxia-induced hypoxia-inducible factor (HIF) activity in mice. Knockout of FoxM1 decreased PASMC proliferation and induced expression of SMC contractile proteins and TGF-ß/Smad3 signaling. CONCLUSIONS: Our studies provide clear evidence that altered FoxM1 expression in PASMC contributes to PH and uncover a correlation between Smad3-dependent signaling in FoxM1-mediated proliferation and de-differentiation of PASMC.


Sujet(s)
Protéine M1 à motif en tête de fourche/physiologie , Hypertension pulmonaire/métabolisme , Muscles lisses vasculaires/métabolisme , Myocytes du muscle lisse/métabolisme , Artère pulmonaire/métabolisme , Animaux , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Prolifération cellulaire , Cellules cultivées , Protéines contractiles/métabolisme , Modèles animaux de maladie humaine , Femelle , Protéine M1 à motif en tête de fourche/génétique , Régulation de l'expression des gènes , Humains , Hypertrophie ventriculaire droite/métabolisme , Hypoxie/métabolisme , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Muscles lisses vasculaires/cytologie , Artère pulmonaire/cytologie , Transduction du signal , Protéine Smad-3/métabolisme , Facteur de croissance transformant bêta/métabolisme , Remodelage vasculaire
11.
Behav Brain Res ; 348: 263-266, 2018 08 01.
Article de Anglais | MEDLINE | ID: mdl-29698694

RÉSUMÉ

Our previous studies showed that paternal nicotine exposure can lead to hyperactivity in the offspring. Nevertheless, the cross-generational effects of maternal and biparental nicotine exposure remain unclear. In this study, female and male mice were exposed respectively by nicotine before pregnancy. The maternal pre-pregnancy nicotine exposure led to depression-like behaviors in the F1 offspring. However, after biparental pre-pregnancy nicotine exposure, seventy percentage of the offspring exhibited a depressive phenotype while 20% were hyperactive, and the remaining exhibited no obvious abnormal behavior. The cross-generational effects appeared to be mediated via disruption of the balance between GSK3 and p-GKS3 by nicotine. These results suggested that pre-pregnancy nicotine exposure can induce alterations in the behavior of the offspring, and the cross-generational effects of maternal nicotine exposure were particularly serious.


Sujet(s)
Comportement maternel/effets des médicaments et des substances chimiques , Hérédité maternelle/effets des médicaments et des substances chimiques , Nicotine/effets indésirables , Animaux , Comportement animal/effets des médicaments et des substances chimiques , Dépression/étiologie , Trouble dépressif/physiopathologie , Femelle , Glycogen Synthase Kinase 3/effets des médicaments et des substances chimiques , Glycogen Synthase Kinase 3/métabolisme , Mâle , Souris , Souris de lignée C57BL , Activité motrice/effets des médicaments et des substances chimiques , Nicotine/métabolisme , Nicotine/pharmacologie , Grossesse , Effets différés de l'exposition prénatale à des facteurs de risque
12.
Am J Physiol Lung Cell Mol Physiol ; 315(2): L149-L161, 2018 08 01.
Article de Anglais | MEDLINE | ID: mdl-29644896

RÉSUMÉ

We have previously reported that miR-17~92 is critically involved in the pathogenesis of pulmonary hypertension (PH). We also identified two novel mR-17/20a direct targets, PDZ and LIM domain protein 5 (PDLIM5) and prolyl hydroxylase 2 (PHD2), and elucidated the signaling pathways by which PDLIM5 and PHD2 regulate functions of pulmonary artery smooth muscle cells (PASMCs). In addition, we have shown that plasminogen activator inhibitor-1 (PAI-1) is also downregulated in PASMCs that overexpress miR-17~92. However, it is unclear whether PAI-1 is a direct target of miR-17~92 and whether it plays a role in regulating the PASMC phenotype. In this study, we have identified PAI-1 as a novel target of miR-19a/b, two members of the miR-17~92 cluster. We found that the 3'-untranslated region (UTR) of PAI-1 contains a miR-19a/b binding site and that miR-19a/b can target this site to suppress PAI-1 protein expression. MiR-17/20a, two other members of miR-17~92, may also indirectly suppress PAI-1 expression through PDLIM5. PAI-1 is a negative regulator of miR-17~92-mediated PASMC proliferation. Silencing of PAI-1 induces Smad2/calponin signaling in PASMCs, suggesting that PAI-1 is a negative regulator of the PASMC contractile phenotype. We also found that PAI-1 is essential for the metabolic gene expression in PASMCs. Furthermore, although there is no significant change in PAI-1 levels in PASMCs isolated from idiopathic pulmonary arterial hypertension and associated pulmonary arterial hypertension patients, PAI-1 is downregulated in hypoxia/Sugen-induced hypertensive rat lungs. These results suggest that miR-17~92 regulates the PASMC contractile phenotype and proliferation coordinately and synergistically by direct and indirect targeting of PAI-1.


Sujet(s)
microARN/métabolisme , Muscles lisses vasculaires/métabolisme , Myocytes du muscle lisse/métabolisme , Inhibiteur-1 d'activateur du plasminogène/biosynthèse , Artère pulmonaire/métabolisme , Transduction du signal , Régions 3' non traduites , Animaux , Prolifération cellulaire , Régulation de l'expression des gènes , Humains , Hypertension pulmonaire/génétique , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/anatomopathologie , Mâle , microARN/génétique , Contraction musculaire/génétique , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/anatomopathologie , Inhibiteur-1 d'activateur du plasminogène/génétique , Artère pulmonaire/anatomopathologie , Rats , Rat Sprague-Dawley
13.
Sci Rep ; 7(1): 7286, 2017 08 04.
Article de Anglais | MEDLINE | ID: mdl-28779169

RÉSUMÉ

The neurobehavioral effects of paternal smoking and nicotine use have not been widely reported. In the present study, nicotine exposure induced depression in the paternal generation, but reduced depression and promoted hyperactivity in F1 offspring. While this intergenerational effect was not passed down to the F2 generation. Further studies revealed that nicotine induced the down-regulation of mmu-miR-15b expression due to hyper-methylation in the CpG island shore region of mmu-miR-15b in both the spermatozoa of F0 mice and the brains of F1 mice. As the target gene of mmu-miR-15b, Wnt4 expression was elevated in the thalamus of F1 mice due to the inheritance of DNA methylation patterns from the paternal generation. Furthermore, the increased expression of Wnt4 elevated the phosphorylation level of its downstream protein GSK-3 through the canonical WNT4 pathway which involved in the behavioral alterations observed in F1 mice. Moreover, in vivo stereotaxic brain injections were used to induce the overexpression of mmu-miR-15b and WNT4 and confirm the neurobehavioral effects in vitro. The behavioral phenotype of the F1 mice resulting from paternal nicotine exposure could be attenuated by viral manipulation of mmu-miR-15b in the thalamus.


Sujet(s)
Comportement animal , Méthylation de l'ADN , microARN/génétique , Nicotine/effets indésirables , Exposition paternelle/effets indésirables , Effets différés de l'exposition prénatale à des facteurs de risque , Animaux , Lignée cellulaire , Ilots CpG , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Dépression , Femelle , Analyse de profil d'expression de gènes , Ordre des gènes , Vecteurs génétiques/génétique , Séquençage nucléotidique à haut débit , Humains , Mâle , Souris , Phénotype , Grossesse , ARN messager/génétique , Transduction du signal , Fumer , Spermatozoïdes/métabolisme , Transcriptome , Protéine Wnt4/génétique , Protéine Wnt4/métabolisme
14.
Cell Signal ; 38: 49-59, 2017 10.
Article de Anglais | MEDLINE | ID: mdl-28652146

RÉSUMÉ

Atypical protein kinase C ζ (PKCζ) forms an apico-basal polarity complex with Partitioning Defective (Pard) 3 and Pard6 to regulate normal epithelial cell apico-basolateral polarization. The dissociation of the PKCζ/Pard3/Pard6 complex is essential for the disassembly of the tight/adherens junction and epithelial-mesenchymal transition (EMT) that is critical for tumor spreading. Loss of cell polarity and epithelial organization is strongly correlated with malignancy and tumor progression in some other cancer types. However, it is unclear whether the PKCζ/Pard3/Pard6 complex plays a role in the progression of non-small-cell lung cancer (NSCLC). We found that hypoxia downregulated the PKCζ/Pard3/Pard6 complex, correlating with induction of lung cancer cell migration and invasion. Silencing of the PKCζ/Pard3/Pard6 polarity complex components induced lung cancer cell EMT, invasion, and colonization in vivo. Suppression of Pard3 was associated with altered expression of genes regulating wound healing, cell apoptosis/death and cell motility, and particularly upregulation of MAP3K1 and fibronectin which are known to contribute to lung cancer progression. Human lung adenocarcinoma tissues expressed less Pard6b and PKCζ than the adjacent normal tissues and in experimental mouse lung adenocarcinoma, the levels of Pard3 and PKCζ were also decreased. In addition, we showed that a methylation locus in the gene body of Pard3 is positively associated with the expression of Pard3 and that methylation of the Pard3 gene increased cellular sensitivity to carboplatin, a common chemotherapy drug. Suppression of Pard3 increased chemoresistance in lung cancer cells. Together, these results suggest that reduced expression of PKCζ/Pard3/Pard6 contributes to NSCLC EMT, invasion, and chemoresistance.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Adénocarcinome/métabolisme , Adénocarcinome/anatomopathologie , Protéines du cycle cellulaire/métabolisme , Transition épithélio-mésenchymateuse , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Protéines membranaires/métabolisme , Protéine kinase C/métabolisme , Cellules A549 , Adénocarcinome pulmonaire , Animaux , Carboplatine/pharmacologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Hypoxie cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Polarité de la cellule/effets des médicaments et des substances chimiques , Cisplatine/pharmacologie , Régulation négative/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Fibronectines/métabolisme , Extinction de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Poumon/anatomopathologie , MAP Kinase Kinase Kinase 1/métabolisme , Souris nude , Invasion tumorale
15.
Biochem Biophys Res Commun ; 472(4): 573-9, 2016 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-26972251

RÉSUMÉ

In mouse testes, germ cell apoptosis can be caused by cigarette smoke and lead to declining quality of semen, but the exact molecular mechanisms remain unclear. To evaluate the effects of nicotine exposure on apoptosis during spermatogenesis, we first constructed a nicotine-treated mouse model and detected germ cell apoptosis activity in the testes using the TUNEL method. Then we analyzed the variation of telomere length and telomerase activity by real-time PCR and TRAP-real-time PCR, respectively. Further, we investigated a highly expressed gene, Nme2, in mouse testes after nicotine treatment from our previous results, which has close correlation with the apoptosis activity predicted by bioinformatics. We performed NME2 overexpression in Hela cells to confirm whether telomere length and telomerase activity were regulated by the Nme2 gene. Finally, we examined methylation of CpG islands in the Nme2 promoter with the Bisulfite Sequencing (BSP) method. The results showed that apoptosis had increased significantly, and then telomerase activity became weak. Further, telomere length was shortened in the germ cells among the nicotine-treated group. In Hela cells, both overexpression of the Nme2 gene and nicotine exposure can suppress the activity of telomerase activity and shorten telomere length. BSP results revealed that the Nme2 promoter appeared with low methylation in mouse testes after nicotine treatment. We assume that nicotine-induced apoptosis may be caused by telomerase activity decline, which is inhibited by the up expression of Nme2 because of its hypomethylation in mouse germ cells.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , NM23 Nucleoside Diphosphate kinases/métabolisme , Nicotine/toxicité , Agonistes nicotiniques/pharmacologie , Homéostasie des télomères/effets des médicaments et des substances chimiques , Testicule/cytologie , Testicule/effets des médicaments et des substances chimiques , Animaux , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Cellules HeLa , Humains , Mâle , Souris , Souris de lignée C57BL , NM23 Nucleoside Diphosphate kinases/génétique , Spermatogenèse/effets des médicaments et des substances chimiques , Telomerase/métabolisme , Testicule/métabolisme
16.
Biol Reprod ; 94(2): 31, 2016 Feb.
Article de Anglais | MEDLINE | ID: mdl-26607717

RÉSUMÉ

Nicotine significantly promoted apoptosis in stages I, VII, VIII, and XI spermatogonia, stages I, VII, VIII, X, and XI spermatocytes, and stages I-V, VII, and VIII elongating spermatids. To explore the underlying molecular mechanisms, sperm mRNA next-generation sequencing of nicotine-treated mice was conducted. Out of the 86 genes related to apoptosis, Tnf (tumor necrosis factor alpha) was screened to be the most significant varied transcript, and the Onto-pathway analysis indicated that the TNF apoptotic pathway was especially activated by nicotine exposure. The TNF pathway was further studied at the gene and protein levels. The results showed that RIP1, the key component in the TNF apoptotic pathway, was up-expressed in its deubiquitinated form in nicotine-treated mice testis. TRIM27, an E3 ubiquitin ligase that activated TNF apoptotic pathway through up-regulating deubiquitinated RIP1, was also overexpressed in nicotine-treated spermatocytes; moreover, four consecutive CpG sites near the Trim27 transcription start site were less frequently methylated. Finally, in vitro experiments of Trim27 overexpression and RNA interference in GC-1 spermatogonial cells confirmed that the RIP1 deubiquitination and TRIM27 hyopmethylation were both positively correlated with spermatocyte apoptosis. In summary, our study suggests that nicotine may induce murine spermatozoal apoptosis via the TNF apoptotic pathway through up-regulation of deubiquitinated RIP1 by Trim27 promoter hypomethylation.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Protéines de liaison à l'ADN/métabolisme , Protéines d'activation de la GTPase/métabolisme , Nicotine/pharmacologie , Protéines nucléaires/métabolisme , Spermatozoïdes/effets des médicaments et des substances chimiques , Régulation positive/effets des médicaments et des substances chimiques , Animaux , Protéines de liaison à l'ADN/génétique , Protéines d'activation de la GTPase/génétique , Mâle , Souris , Protéines nucléaires/génétique , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Spermatozoïdes/métabolisme , Testicule/effets des médicaments et des substances chimiques , Testicule/métabolisme , Ubiquitin-protein ligases
17.
Reproduction ; 151(3): 227-37, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26647419

RÉSUMÉ

Many studies have revealed the hazardous effects of cigarette smoking and nicotine exposure on male fertility, but the actual, underlying molecular mechanism remains relatively unclear. To evaluate the detrimental effects of nicotine exposure on the sperm maturation process, two-dimensional gel electrophoresis and mass spectrometry analyses were performed to screen and identify differentially expressed proteins from the epididymal tissue of mice exposed to nicotine. Data mining analysis indicated that 15 identified proteins were mainly involved in the molecular transportation process and the polyol pathway, indicating impaired epididymal secretory functions. Experiments in vitro confirmed that nicotine inhibited tyrosine phosphorylation levels in capacitated spermatozoa via the downregulated seminal fructose concentration. Sord, a key gene encoding sorbitol dehydrogenase, was further investigated to reveal that nicotine induced hyper-methylation of the promoter region of this gene. Nicotine-induced reduced expression of Sord could be involved in impaired secretory functions of the epididymis and thus prevent the sperm from undergoing proper maturation and capacitation, although further experiments are needed to confirm this hypothesis.


Sujet(s)
Épididyme/effets des médicaments et des substances chimiques , Infertilité masculine/induit chimiquement , L-iditol 2-dehydrogenase/métabolisme , Nicotine/effets indésirables , Animaux , Méthylation de l'ADN , Épididyme/métabolisme , Fructose/métabolisme , Analyse de profil d'expression de gènes , Gene Ontology , L-iditol 2-dehydrogenase/génétique , Mâle , Spectrométrie de masse , Souris de lignée C57BL , Répartition aléatoire , Spermatozoïdes/métabolisme , Tyrosine/métabolisme
18.
Acta Biochim Biophys Sin (Shanghai) ; 47(7): 504-15, 2015 Jul.
Article de Anglais | MEDLINE | ID: mdl-26063603

RÉSUMÉ

Cigarette smoking is associated with lower semen quality, but how cigarette smoking changes the semen quality remains unclear. The aim of this study was to screen the differentially expressed proteins in the sperm of mice with daily exposure to cigarette smoke. The 2D gel electrophoresis (2DE) and mass spectrometry (MS) analyses results showed that the mouse sperm protein profile was altered by cigarette smoking. And 22 of the most abundant proteins that correspond to differentially expressed spots in 2DE gels of the sperm samples were identified. These proteins were classified into different groups based on their functions, such as energy metabolism, reproduction, and structural molecules. Furthermore, the 2DE and MS results of five proteins (Aldoa, ATP5a1, Gpx4, Cs, and Spatc1) were validated by western blot analysis and reverse transcriptase-polymerase chain reaction. Results showed that except Spatc1 the other four proteins showed statistically significant different protein levels between the smoking group and the control group (P < 0.05). The expressions of three genes (Aldoa, Gpx4, and Spatc1) were significantly different (P < 0.05) at transcription level between the smoking group and the control group. In addition, five proteins (Aldoa, ATP5a1, Spatc1, Cs, and Gpx4) in human sperm samples from 30 male smokers and 30 non-smokers were detected by western blot analysis. Two proteins (Aldoa and Cs) that are associated with energy production were found to be significantly altered, suggesting that these proteins may be potential diagnostic markers for evaluation of smoking risk in sperm. Further study of these proteins may provide insight into the pathogenic mechanisms underlying infertility in smoking persons.


Sujet(s)
Nicotiana , Protéines/métabolisme , Fumer/métabolisme , Spermatozoïdes/métabolisme , Animaux , Électrophorèse bidimensionnelle sur gel , Mâle , Souris , Souris de lignée C57BL , Oxygène/métabolisme , Spectrométrie de masse MALDI
19.
Asian J Androl ; 17(6): 954-60, 2015.
Article de Anglais | MEDLINE | ID: mdl-25851659

RÉSUMÉ

The substantial harmful effects of tobacco smoking on fertility and reproduction have become apparent but are not generally appreciated. Tobacco smoke contains more than 4000 kinds of constituents, including nicotine, tar, carbonic monoxide, polycyclic aromatic hydrocarbons, and heavy metals. Because of the complexity of tobacco smoke components, the toxicological mechanism is notably complicated. Most studies have reported reduced semen quality, reproductive hormone system dysfunction and impaired spermatogenesis, sperm maturation, and spermatozoa function in smokers compared with nonsmokers. Underlying these effects, elevated oxidative stress, DNA damage, and cell apoptosis may play important roles collaboratively in the overall effect of tobacco smoking on male fertility. In this review, we strive to focus on both the phenotype of and the molecular mechanism underlying these harmful effects, although current studies regarding the mechanism remain insufficient.


Sujet(s)
Altération de l'ADN , Infertilité masculine/étiologie , Nicotiana , Stress oxydatif , Fumée/effets indésirables , Fumer/effets indésirables , Spermatogenèse , Spermatozoïdes , Monoxyde de carbone/effets indésirables , Fécondité , Gazotransmetteurs/effets indésirables , Humains , Mâle , Métaux lourds/effets indésirables , Nicotine/effets indésirables , Agonistes nicotiniques/effets indésirables , Hydrocarbures aromatiques polycycliques/effets indésirables , Analyse du sperme
20.
J Craniomaxillofac Surg ; 42(8): 1903-8, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25187377

RÉSUMÉ

PURPOSE: A novel, modified presurgical nasoalveolar molding (MPNAM) device with retraction screw was designed and used in patients with bilateral complete cleft lip and palate (BCCLP) to rapidly retract and centralize the protuberant and malpositioned premaxilla and correct the nasolabial and palatal deformities. The orthopedic effects and possible complications were evaluated. PATIENTS AND METHODS: Nine patients with BCCLP who met the inclusion criteria were selected. After the maxillary model was obtained, the new MPNAM device with retraction screw was designed and worn until cheilorrhaphy. Changes in local deformities and complications were observed continuously, and the orthopedic effect was evaluated. RESULTS: All patients quickly adapted to the MPNAM appliance, and the treatment was finished after 5-8 return visits. The columella was significantly prolonged, the nasal tip was elevated, and the collapsed nasal dome was obviously improved. Simultaneously, the premaxilla was rapidly retracted and rotated, and gradually centralized; the clefts were gradually reduced and closed, and a nearly normal dental arch was formed. Although there were some complications, the orthopedic treatment was continued until cheiloplasty. CONCLUSIONS: The MPNAM device with retraction screw can simultaneously correct nasolabial and palatal deformities and also rapidly retract and centralize the premaxilla.


Sujet(s)
Bec-de-lièvre/thérapie , Fente palatine/thérapie , Maxillaire/anatomopathologie , Orthèses , Résines acryliques/composition chimique , Matériaux biocompatibles/composition chimique , Bec-de-lièvre/chirurgie , Fente palatine/chirurgie , Arcade dentaire/anatomopathologie , Conception d'appareillage , Femelle , Humains , Nouveau-né , Mâle , Nez/anatomopathologie , Palais/anatomopathologie , Rotation , Acier inoxydable/composition chimique , Endoprothèses
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...