Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 36
Filtrer
1.
Nat Commun ; 15(1): 6843, 2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-39122671

RÉSUMÉ

Despite the potential of small molecules and recombinant proteins to enhance the efficiency of homology-directed repair (HDR), single-stranded DNA (ssDNA) donors, as currently designed and chemically modified, remain suboptimal for precise gene editing. Here, we screen the biased ssDNA binding sequences of DNA repair-related proteins and engineer RAD51-preferred sequences into HDR-boosting modules for ssDNA donors. Donors with these modules exhibit an augmented affinity for RAD51, thereby enhancing HDR efficiency across various genomic loci and cell types when cooperated with Cas9, nCas9, and Cas12a. By combining with an inhibitor of non-homologous end joining (NHEJ) or the HDRobust strategy, these modular ssDNA donors achieve up to 90.03% (median 74.81%) HDR efficiency. The HDR-boosting modules targeting an endogenous protein enable a chemical modification-free strategy to improve the efficacy of ssDNA donors for precise gene editing.


Sujet(s)
ADN simple brin , Édition de gène , Rad51 Recombinase , Réparation de l'ADN par recombinaison , ADN simple brin/métabolisme , ADN simple brin/génétique , Humains , Édition de gène/méthodes , Rad51 Recombinase/métabolisme , Rad51 Recombinase/génétique , Systèmes CRISPR-Cas , Cellules HEK293 , Protéines associées aux CRISPR/métabolisme , Protéines associées aux CRISPR/génétique , Protéine-9 associée à CRISPR/métabolisme , Protéine-9 associée à CRISPR/génétique , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Endodeoxyribonucleases/métabolisme , Endodeoxyribonucleases/génétique , Réparation de l'ADN par jonction d'extrémités
2.
Signal Transduct Target Ther ; 8(1): 255, 2023 07 03.
Article de Anglais | MEDLINE | ID: mdl-37394473

RÉSUMÉ

Thoracic aortic aneurysms (TAAs) develop asymptomatically and are characterized by dilatation of the aorta. This is considered a life-threating vascular disease due to the risk of aortic rupture and without effective treatments. The current understanding of the pathogenesis of TAA is still limited, especially for sporadic TAAs without known genetic mutation. Sirtuin 6 (SIRT6) expression was significantly decreased in the tunica media of sporadic human TAA tissues. Genetic knockout of Sirt6 in mouse vascular smooth muscle cells accelerated TAA formation and rupture, reduced survival, and increased vascular inflammation and senescence after angiotensin II infusion. Transcriptome analysis identified interleukin (IL)-1ß as a pivotal target of SIRT6, and increased IL-1ß levels correlated with vascular inflammation and senescence in human and mouse TAA samples. Chromatin immunoprecipitation revealed that SIRT6 bound to the Il1b promoter to repress expression partly by reducing the H3K9 and H3K56 acetylation. Genetic knockout of Il1b or pharmacological inhibition of IL-1ß signaling with the receptor antagonist anakinra rescued Sirt6 deficiency mediated aggravation of vascular inflammation, senescence, TAA formation and survival in mice. The findings reveal that SIRT6 protects against TAA by epigenetically inhibiting vascular inflammation and senescence, providing insight into potential epigenetic strategies for TAA treatment.


Sujet(s)
Anévrysme de l'aorte thoracique , Sirtuines , Humains , Souris , Animaux , Anévrysme de l'aorte thoracique/génétique , Anévrysme de l'aorte thoracique/métabolisme , Anévrysme de l'aorte thoracique/anatomopathologie , Inflammation/génétique , Angiotensine-II/génétique , Angiotensine-II/pharmacologie , Épigenèse génétique/génétique , Sirtuines/génétique
4.
Chin Med Sci J ; 35(1): 43-53, 2020 Mar 31.
Article de Anglais | MEDLINE | ID: mdl-32299537

RÉSUMÉ

Objective Angiotensin Ⅱ (Ang Ⅱ)-induced vascular damage is a major risk of hypertension. However, the underlying molecular mechanism of AngⅡ-induced vascular damage is still unclear. In this study, we explored the novel mechanism associated with Ang II-induced hypertension. Methods We treated 8- to 12-week-old C57BL/6J male mice with saline and Ang Ⅱ(0.72 mg/kg·d) for 28 days, respectively. Then the RNA of the media from the collected mice aortas was extracted for transcriptome sequencing. Principal component analysis was applied to show a clear separation of different samples and the distribution of differentially expressed genes was manifested by Volcano plot. Functional annotations including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway were performed to reveal the molecular mechanism of Ang Ⅱ-induced hypertension. Finally, the differentially expressed genes were validated by using quantitative real-time PCR. Results The result revealed that a total of 773 genes, including 599 up-regulated genes and 174 down-regulated genes, were differentially expressed in the aorta of Ang Ⅱ-induced hypertension mice model. Functional analysis of differentially expressed genes manifested that various cellular processes may be involved in the Ang Ⅱ-induced hypertension, including some pathways associated with hypertension such as extracellular matrix, inflammation and immune response. Interestingly, we also found that the differentially expressed genes were enriched in vascular aging pathway, and further validated that the expression levels of insulin-like growth factor 1 and adiponectin were significantly increased (P<0.05). Conclusion We identify that vascular aging is involved in Ang Ⅱ-induced hypertension, and insulin-like growth factor 1 and adiponectin may be important candidate genes leading to vascular aging.


Sujet(s)
Vieillissement , Aorte/métabolisme , Analyse de profil d'expression de gènes/méthodes , Hypertension artérielle/génétique , Angiotensine-II , Animaux , Aorte/physiopathologie , Pression sanguine/génétique , Gene Ontology , Hypertension artérielle/induit chimiquement , Mâle , Souris de lignée C57BL , RT-PCR
5.
Nat Cell Biol ; 21(12): 1553-1564, 2019 12.
Article de Anglais | MEDLINE | ID: mdl-31768048

RÉSUMÉ

Redox balance, an essential feature of healthy physiological steady states, is regulated by circadian clocks, but whether or how endogenous redox signalling conversely regulates clockworks in mammals remains unknown. Here, we report circadian rhythms in the levels of endogenous H2O2 in mammalian cells and mouse livers. Using an unbiased method to screen for H2O2-sensitive transcription factors, we discovered that rhythmic redox control of CLOCK directly by endogenous H2O2 oscillations is required for proper intracellular clock function. Importantly, perturbations in the rhythm of H2O2 levels induced by the loss of p66Shc, which oscillates rhythmically in the liver and suprachiasmatic nucleus (SCN) of mice, disturb the rhythmic redox control of CLOCK function, reprogram hepatic transcriptome oscillations, lengthen the circadian period in mice and modulate light-induced clock resetting. Our findings suggest that redox signalling rhythms are intrinsically coupled to the circadian system through reversible oxidative modification of CLOCK and constitute essential mechanistic timekeeping components in mammals.


Sujet(s)
Horloges circadiennes/physiologie , Rythme circadien/physiologie , Peroxyde d'hydrogène/métabolisme , Protéine transformante-1 contenant un domaine d'homologie-2 de Src/métabolisme , Animaux , Femelle , Foie/métabolisme , Foie/physiologie , Mâle , Mammifères/métabolisme , Mammifères/physiologie , Souris , Souris knockout , Oxydoréduction , Protéines circadiennes Period/métabolisme , Transduction du signal/physiologie , Noyau suprachiasmatique/métabolisme , Noyau suprachiasmatique/physiologie
6.
iScience ; 17: 155-166, 2019 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-31279933

RÉSUMÉ

Both caloric restriction (CR) and mitochondrial proteostasis are linked to longevity, but how CR maintains mitochondrial proteostasis in mammals remains elusive. MicroRNAs (miRNAs) are well known for gene silencing in cytoplasm and have recently been identified in mitochondria, but knowledge regarding their influence on mitochondrial function is limited. Here, we report that CR increases miRNAs, which are required for the CR-induced activation of mitochondrial translation, in mouse liver. The ablation of miR-122, the most abundant miRNA induced by CR, or the retardation of miRNA biogenesis via Drosha knockdown significantly reduces the CR-induced activation of mitochondrial translation. Importantly, CR-induced miRNAs cause the overproduction of mtDNA-encoded proteins, which induces the mitochondrial unfolded protein response (UPRmt), and consequently improves mitochondrial proteostasis and function. These findings establish a physiological role of miRNA-enhanced mitochondrial function during CR and reveal miRNAs as critical mediators of CR in inducing UPRmt to improve mitochondrial proteostasis.

7.
Circulation ; 136(21): 2051-2067, 2017 Nov 21.
Article de Anglais | MEDLINE | ID: mdl-28947430

RÉSUMÉ

BACKGROUND: Pathological cardiac hypertrophy induced by stresses such as aging and neurohumoral activation is an independent risk factor for heart failure and is considered a target for the treatment of heart failure. However, the mechanisms underlying pathological cardiac hypertrophy remain largely unknown. We aimed to investigate the roles of SIRT2 in aging-related and angiotensin II (Ang II)-induced pathological cardiac hypertrophy. METHODS: Male C57BL/6J wild-type and Sirt2 knockout mice were subjected to the investigation of aging-related cardiac hypertrophy. Cardiac hypertrophy was also induced by Ang II (1.3 mg/kg/d for 4 weeks) in male C57BL/6J Sirt2 knockout mice, cardiac-specific SIRT2 transgenic (SIRT2-Tg) mice, and their respective littermates (8 to ≈12 weeks old). Metformin (200 mg/kg/d) was used to treat wild-type and Sirt2 knockout mice infused with Ang II. Cardiac hypertrophy, fibrosis, and cardiac function were examined in these mice. RESULTS: SIRT2 protein expression levels were downregulated in hypertrophic hearts from mice. Sirt2 knockout markedly exaggerated cardiac hypertrophy and fibrosis and decreased cardiac ejection fraction and fractional shortening in aged (24-month-old) mice and Ang II-infused mice. Conversely, cardiac-specific SIRT2 overexpression protected the hearts against Ang II-induced cardiac hypertrophy and fibrosis and rescued cardiac function. Mechanistically, SIRT2 maintained the activity of AMP-activated protein kinase (AMPK) in aged and Ang II-induced hypertrophic hearts in vivo as well as in cardiomyocytes in vitro. We identified the liver kinase B1 (LKB1), the major upstream kinase of AMPK, as the direct target of SIRT2. SIRT2 bound to LKB1 and deacetylated it at lysine 48, which promoted the phosphorylation of LKB1 and the subsequent activation of LKB1-AMPK signaling. Remarkably, the loss of SIRT2 blunted the response of AMPK to metformin treatment in mice infused with Ang II and repressed the metformin-mediated reduction of cardiac hypertrophy and protection of cardiac function. CONCLUSIONS: SIRT2 promotes AMPK activation by deacetylating the kinase LKB1. Loss of SIRT2 reduces AMPK activation, promotes aging-related and Ang II-induced cardiac hypertrophy, and blunts metformin-mediated cardioprotective effects. These findings indicate that SIRT2 will be a potential target for therapeutic interventions in aging- and stress-induced cardiac hypertrophy.


Sujet(s)
Cardiomégalie/prévention et contrôle , Metformine/pharmacologie , Myocarde/enzymologie , Sirtuine-2/métabolisme , AMP-activated protein kinase kinases , AMP-Activated Protein Kinases/métabolisme , Acétylation , Facteurs âges , Vieillissement/métabolisme , Angiotensine-II , Animaux , Cardiomégalie/induit chimiquement , Cardiomégalie/enzymologie , Cardiomégalie/physiopathologie , Cellules cultivées , Modèles animaux de maladie humaine , Fibrose , Prédisposition génétique à une maladie , Lysine , Mâle , Souris de lignée C57BL , Souris knockout , Contraction myocardique/effets des médicaments et des substances chimiques , Myocarde/anatomopathologie , Phénotype , Liaison aux protéines , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/pharmacologie , Rats , Transduction du signal/effets des médicaments et des substances chimiques , Sirtuine-2/déficit , Sirtuine-2/génétique , Débit systolique/effets des médicaments et des substances chimiques , Remodelage ventriculaire/effets des médicaments et des substances chimiques
8.
Sci Rep ; 7: 46204, 2017 04 10.
Article de Anglais | MEDLINE | ID: mdl-28393844

RÉSUMÉ

Accumulating data from genome-wide association studies (GWAS) have provided a collection of novel candidate genes associated with complex diseases, such as atherosclerosis. We identified an atherosclerosis-associated single-nucleotide polymorphism (SNP) located in the intron of the long noncoding RNA (lncRNA) LINC00305 by searching the GWAS database. Although the function of LINC00305 is unknown, we found that LINC00305 expression is enriched in atherosclerotic plaques and monocytes. Overexpression of LINC00305 promoted the expression of inflammation-associated genes in THP-1 cells and reduced the expression of contractile markers in co-cultured human aortic smooth muscle cells (HASMCs). We showed that overexpression of LINC00305 activated nuclear factor-kappa beta (NF-κB) and that inhibition of NF-κB abolished LINC00305-mediated activation of cytokine expression. Mechanistically, LINC00305 interacted with lipocalin-1 interacting membrane receptor (LIMR), enhanced the interaction of LIMR and aryl-hydrocarbon receptor repressor (AHRR), and promoted protein expression as well as nuclear localization of AHRR. Moreover, LINC00305 activated NF-κB exclusively in the presence of LIMR and AHRR. In light of these findings, we propose that LINC00305 promotes monocyte inflammation by facilitating LIMR and AHRR cooperation and the AHRR activation, which eventually activates NF-κB, thereby inducing HASMC phenotype switching.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Inflammation/génétique , Inflammation/anatomopathologie , Monocytes/métabolisme , Monocytes/anatomopathologie , Facteur de transcription NF-kappa B/métabolisme , ARN long non codant/métabolisme , Protéines de répression/métabolisme , Aorte/anatomopathologie , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Lignée cellulaire , Noyau de la cellule/métabolisme , Étude d'association pangénomique , Humains , Myocytes du muscle lisse/métabolisme , Phénotype , Transport des protéines , ARN long non codant/génétique , Récepteurs de surface cellulaire/métabolisme , Protéines de répression/génétique , Transduction du signal/génétique , Régulation positive
9.
Eur Heart J ; 38(18): 1389-1398, 2017 May 07.
Article de Anglais | MEDLINE | ID: mdl-27099261

RÉSUMÉ

AIMS: Oxidative stress contributes to the development of cardiac hypertrophy and heart failure. One of the mitochondrial sirtuins, Sirt4, is highly expressed in the heart, but its function remains unknown. The aim of the present study was to investigate the role of Sirt4 in the pathogenesis of pathological cardiac hypertrophy and the molecular mechanism by which Sirt4 regulates mitochondrial oxidative stress. METHODS AND RESULTS: Male C57BL/6 Sirt4 knockout mice, transgenic (Tg) mice exhibiting cardiac-specific overexpression of Sirt4 (Sirt4-Tg) and their respective controls were treated with angiotensin II (Ang II, 1.1 mg/kg/day). At 4 weeks, hypertrophic growth of cardiomyocytes, fibrosis and cardiac function were analysed. Sirt4 deficiency conferred resistance to Ang II infusion by significantly suppressing hypertrophic growth, and the deposition of fibrosis. In Sirt4-Tg mice, aggravated hypertrophy and reduced cardiac function were observed compared with non-Tg mice following Ang II treatment. Mechanistically, Sirt4 inhibited the binding of manganese superoxide dismutase (MnSOD) to Sirt3, another member of the mitochondrial sirtuins, and increased MnSOD acetylation levels to reduce its activity, resulting in elevated reactive oxygen species (ROS) accumulation upon Ang II stimulation. Furthermore, inhibition of ROS with manganese 5, 10, 15, 20-tetrakis-(4-benzoic acid) porphyrin, a mimetic of SOD, blocked the Sirt4-mediated aggravation of the hypertrophic response in Ang II-treated Sirt4-Tg mice. CONCLUSIONS: Sirt4 promotes hypertrophic growth, the generation of fibrosis and cardiac dysfunction by increasing ROS levels upon pathological stimulation. These findings reveal a role of Sirt4 in pathological cardiac hypertrophy, providing a new potential therapeutic strategy for this disease.


Sujet(s)
Cardiomégalie/enzymologie , Protéines mitochondriales/physiologie , Sirtuines/physiologie , Superoxide dismutase/antagonistes et inhibiteurs , Angiotensine-II/pharmacologie , Animaux , Techniques de knock-down de gènes , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Mitochondries du myocarde/enzymologie , Myocytes cardiaques/enzymologie , Stress oxydatif/physiologie , Espèces réactives de l'oxygène/antagonistes et inhibiteurs , Remodelage vasculaire/physiologie , Vasoconstricteurs/pharmacologie
10.
J Exp Med ; 213(11): 2473-2488, 2016 10 17.
Article de Anglais | MEDLINE | ID: mdl-27670594

RÉSUMÉ

Abdominal aortic aneurysm (AAA), characterized by a localized dilation of the abdominal aorta, is a life-threatening vascular pathology. Because of the current lack of effective treatment for AAA rupture, prevention is of prime importance for AAA management. Calorie restriction (CR) is a nonpharmacological intervention that delays the aging process and provides various health benefits. However, whether CR prevents AAA formation remains untested. In this study, we subjected Apoe-/- mice to 12 wk of CR and then examined the incidence of angiotensin II (AngII)-induced AAA formation. We found that CR markedly reduced the incidence of AAA formation and attenuated aortic elastin degradation in Apoe-/- mice. The expression and activity of Sirtuin 1 (SIRT1), a key metabolism/energy sensor, were up-regulated in vascular smooth muscle cells (VSMCs) upon CR. Importantly, the specific ablation of SIRT1 in smooth muscle cells abolished the preventive effect of CR on AAA formation in Apoe-/- mice. Mechanistically, VSMC-SIRT1-dependent deacetylation of histone H3 lysine 9 on the matrix metallopeptidase 2 (Mmp2) promoter was required for CR-mediated suppression of AngII-induced MMP2 expression. Together, our findings suggest that CR may be an effective intervention that protects against AAA formation.


Sujet(s)
Anévrysme de l'aorte abdominale/induit chimiquement , Anévrysme de l'aorte abdominale/prévention et contrôle , Restriction calorique , Acétylation , Angiotensine-II , Animaux , Aorte/métabolisme , Anévrysme de l'aorte abdominale/sang , Anévrysme de l'aorte abdominale/métabolisme , Apolipoprotéines E/déficit , Apolipoprotéines E/métabolisme , Chromatine/métabolisme , Épigenèse génétique , Glucose/métabolisme , Histone/métabolisme , Métabolisme lipidique/génétique , Lipides/sang , Foie/métabolisme , Lysine/métabolisme , Mâle , Matrix metalloproteinase 2/génétique , Matrix metalloproteinase 2/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/métabolisme , Phosphorylation oxydative , Régions promotrices (génétique)/génétique , Sirtuine-1/métabolisme
11.
Oncotarget ; 7(17): 24719-33, 2016 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-27034160

RÉSUMÉ

The axon guidance factor netrin-1 promotes tumorigenesis in multiple types of cancers, particularly at their advanced stages. Here, we investigate whether netrin-1 is involved in the in vivo growth of pancreatic adenocarcinoma. We show that netrin-1 is significantly under-expressed in stage-I/II pancreatic ductal adenocarcinoma (PDAC). Netrin-1 over-expression effectively arrests the growth of xenografted PDAC cells without decreasing cell proliferation or increasing apoptosis in two-dimensional cultures in vitro. Integrin-beta4 (ITGB4) expression is significantly reduced, and ITGB4-knockdown mimics the tumor-suppressive effect of netrin-1, implying that ITGB4 is a main target of netrin-1 in constraining PDAC. We further show that netrin-1 signals to UNC5B/FAK to stimulate nitric oxide production, which promotes PP2A-mediated inhibition of the MEK/ERK pathway and decreases phosphorylated-c-Jun recruitment to the ITGB4 promoter. Our findings suggest that netrin-1 can suppress the growth of PDAC and provide a mechanistic insight into this suppression.


Sujet(s)
Carcinome du canal pancréatique/métabolisme , Extracellular Signal-Regulated MAP Kinases/génétique , Intégrine bêta4/métabolisme , Système de signalisation des MAP kinases , Nétrine-1/métabolisme , Tumeurs du pancréas/métabolisme , Animaux , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/physiologie , Embryon de poulet , Extracellular Signal-Regulated MAP Kinases/métabolisme , Cellules HEK293 , Hétérogreffes , Humains , Souris , Nétrine-1/génétique , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie
12.
Sci Rep ; 6: 23912, 2016 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-27045575

RÉSUMÉ

Sirt6 is a member of the class III histone deacetylase family which is associated with aging and longevity. Sirt6 deficient mice show an aging-like phenotype, while male transgenic mice of Sirt6 show increased longevity. Sirt6 acts as a tumor suppressor and deficiency of Sirt6 leads to cardiac hypertrophy and heart failure. Whether Sirt6 is involved in atherosclerosis development, the major cause of cardiovascular diseases, is unknown. We found that the expression of Sirt6 is lower in human atherosclerotic plaques than that in controls. When Sirt6(+/-)ApoE(-/-) and ApoE(-/-) mice are fed with high fat diet for 16 weeks, Sirt6(+/-)ApoE(-/-) mice show increased plaque fromation and exhibit feature of plaque instability. Furthermore, Sirt6 downregulation increases expression of NKG2D ligands, which leads to increased cytokine expression. Blocking NKG2D ligand almost completely blocks this effect. Mechanistically, Sirt6 binds to promoters of NKG2D ligand genes and regulates the H3K9 and H3K56 acetylation levels.


Sujet(s)
Apolipoprotéines E/génétique , Épigenèse génétique , Sous-famille K des récepteurs de cellules NK de type lectine/génétique , Plaque d'athérosclérose/métabolisme , Sirtuines/génétique , Animaux , Endartériectomie carotidienne , Femelle , Fibroblastes/métabolisme , Hétérozygote , Humains , Ligands , Mâle , Souris , Souris knockout , Régions promotrices (génétique) , Transduction du signal
13.
Nucleic Acids Res ; 44(6): 2613-27, 2016 Apr 07.
Article de Anglais | MEDLINE | ID: mdl-26615201

RÉSUMÉ

The Hox genes encode transcription factors that determine embryonic pattern formation. In embryonic stem cells, the Hox genes are silenced by PRC2. Recent studies have reported a role for long noncoding RNAs in PRC2 recruitment in vertebrates. However, little is known about how PRC2 is recruited to the Hox genes in ESCs. Here, we used stable knockdown and knockout strategies to characterize the function of the long noncoding RNAGm15055 in the regulation of Hoxa genes in mouse ESCs. We found that Gm15055 is highly expressed in mESCs and its expression is maintained by OCT4.Gm15055 represses Hoxa gene expression by recruiting PRC2 to the cluster and maintaining the H3K27me3 modification on Hoxa promoters. A chromosome conformation capture assay revealed the close physical association of the Gm15055 locus to multiple sites at the Hoxa gene cluster in mESCs, which may facilitate the in cis targeting of Gm15055RNA to the Hoxa genes. Furthermore, an OCT4-responsive positive cis-regulatory element is found in the Gm15055 gene locus, which potentially regulates both Gm15055 itself and the Hoxa gene activation. This study suggests how PRC2 is recruited to the Hoxa locus in mESCs, and implies an elaborate mechanism for Hoxa gene regulation in mESCs.


Sujet(s)
Protéines à homéodomaine/génétique , Cellules souches embryonnaires de souris/métabolisme , Famille multigénique , Facteur de transcription Oct-3/génétique , Complexe répresseur Polycomb-2/génétique , ARN long non codant/génétique , Animaux , Lignée cellulaire , Chromatine/composition chimique , Chromatine/métabolisme , Régulation de l'expression des gènes , Histone/génétique , Histone/métabolisme , Protéines à homéodomaine/métabolisme , Souris , Cellules souches embryonnaires de souris/cytologie , Facteur de transcription Oct-3/métabolisme , Complexe répresseur Polycomb-2/métabolisme , Régions promotrices (génétique) , ARN long non codant/antagonistes et inhibiteurs , ARN long non codant/métabolisme , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Transduction du signal
14.
Stem Cells ; 33(7): 2135-47, 2015 Jul.
Article de Anglais | MEDLINE | ID: mdl-25940188

RÉSUMÉ

Mouse somatic cells can be reprogrammed into induced pluripotent stem cells by defined factors known to regulate pluripotency, including Oct4, Sox2, Klf4, and c-Myc. Together with Oct4, Sox2 plays a major role as a master endogenous pluripotent genes trigger in reprogramming. It has been reported that Sirtuin 1 (Sirt1), a member of the Sirtuin family of NAD(+) -dependent protein deacetylases, is involved in embryonic stem cell antioxidation, differentiation, and individual development. However, as a deacetylation enzyme, whether Sirt1 influences reprogramming through its post-translational modification function remains unknown. In this study, we provide evidence that deacetylation of Sox2 by Sirt1 is required for reprogramming. We found that a low level of Sox2 acetylation could significantly increase reprogramming efficiency. Furthermore, we found that Sox2 can be deacetylated by Sirt1 in an Oct4-mediated manner. Compared with wild-type cells, Sirt1-null mouse embryonic fibroblasts exhibit decreased reprogramming efficiency, and overexpression of Sirt1 rescues this defect. In addition, Sirt1 functions in the regulation of reprogramming through deacetylating Sox2. Taken together, we have identified a new regulatory role of Sirt1 in reprogramming and provided a link between deacetylation events and somatic cell reprogramming. Stem Cells 2015;33:2135-2147.


Sujet(s)
Facteurs de transcription SOX-B1/métabolisme , Sirtuine-1/métabolisme , Animaux , Différenciation cellulaire , Reprogrammation cellulaire , Facteur-4 de type Kruppel , Souris
15.
Mol Cell Biol ; 34(20): 3867-79, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-25135475

RÉSUMÉ

HOX cluster genes are activated sequentially in their positional order along the chromosome during vertebrate development. This phenomenon, known as temporal colinearity, depends on transcriptional silencing of 5' HOX genes. Chromatin looping was recently identified as a conserved feature of silent HOX clusters, with CCCTC-binding factor (CTCF) binding sites located at the loop bases. However, the potential contribution of CTCF to HOX cluster silencing and the underlying mechanism have not been established. Here, we demonstrate that the HOXA locus is organized by CTCF into chromatin loops and that CTCF depletion causes significantly enhanced activation of HOXA3 to -A7, -A9 to -A11, and -A13 in response to retinoic acid, with the highest effect observed for HOXA9. Our subsequent analyses revealed that CTCF facilitates the stabilization of Polycomb repressive complex 2 (PRC2) and trimethylated lysine 27 of histone H3 (H3K27me3) at the human HOXA locus. Our results reveal that CTCF functions as a controller of HOXA cluster silencing and mediates PRC2-repressive higher-order chromatin structure.


Sujet(s)
Chromatine/génétique , Extinction de l'expression des gènes , Protéines à homéodomaine/génétique , Complexe répresseur Polycomb-2/physiologie , Protéines de répression/physiologie , Séquence nucléotidique , Facteur de liaison à la séquence CCCTC , Lignée cellulaire tumorale , Chromatine/métabolisme , Locus génétiques , Histone/métabolisme , Protéines à homéodomaine/métabolisme , Protéines à homéodomaine/ultrastructure , Humains , Conformation moléculaire , Stabilité protéique , Délétion de séquence , Trétinoïne/physiologie
16.
Aging Cell ; 13(5): 890-9, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-25040736

RÉSUMÉ

The inactivation of plasminogen activator inhibitor-1 (PAI-1) has been shown to exert beneficial effects in age-related vascular diseases. Limited information is available on the molecular mechanisms regarding the negatively regulated expression of PAI-1 in the vascular system. In this study, we observed an inverse correlation between SIRT1, a class III histone deacetylase, and PAI-1 expression in human atherosclerotic plaques and the aortas of old mice, suggesting that internal negative regulation exists between SIRT1 and PAI-1. SIRT1 overexpression reversed the increased PAI-1 expression in senescent human umbilical vein endothelial cells (HUVECs) and aortas of old mice, accompanied by decreased SA-ß-gal activity in vitro and improved endothelial function and reduced arterial stiffness in vivo. Moreover, the SIRT1-mediated inhibition of PAI-1 expression exerted an antisenescence effect in HUVECs. Furthermore, we demonstrated that SIRT1 is able to bind to the PAI-1 promoter, resulting in a decrease in the acetylation of histone H4 lysine 16 (H4K16) on the PAI-1 promoter region. Thus, our findings suggest that the SIRT1-mediated epigenetic inhibition of PAI-1 expression exerts a protective effect in vascular endothelial senescence.


Sujet(s)
Vieillissement de la cellule/physiologie , Inhibiteur-1 d'activateur du plasminogène/génétique , Serpine E2/génétique , Sirtuine-1/génétique , Animaux , Régulation négative , Épigenèse génétique , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Humains , Mâle , Souris , Souris transgéniques , Plaque d'athérosclérose/métabolisme , Inhibiteur-1 d'activateur du plasminogène/métabolisme , Serpine E2/métabolisme , Sirtuine-1/métabolisme
17.
J Mol Med (Berl) ; 92(4): 347-57, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24352856

RÉSUMÉ

UNLABELLED: Angiotensin II (AngII) induces the development of vascular hypertrophy and hypertension. We have shown previously that overexpression of class III deacetylase SIRT1 inhibits AngII-induced hypertrophy in vascular smooth muscle cells (VSMCs). However, the direct role of SIRT1 in VSMCs in response to AngII infusion in vivo remains unclear. Here, we found that the expression and activity of SIRT1 in mouse aortas was decreased significantly by AngII infusion. VSMC-specific SIRT1 transgene (SV-Tg) prevented the increase in systolic blood pressure (SBP) caused by AngII infusion without affecting heart function in mice. SIRT1 overexpression alleviated vascular remodeling in mouse thoracic and renal aortas induced by AngII infusion, and significantly inhibited reactive oxygen species (ROS) generation, vascular inflammation, and collagen synthesis in arterial walls. Reduced expression of transforming growth factor-ß 1 (TGF-ß1) was also observed in the aortas of AngII-infused SV-Tg mice. Moreover, SIRT1 overexpression decreased AngII-increased binding of nuclear factor-κB on its specific binding sites on TGF-ß1 promoter. Taken together, these data demonstrate that SIRT1 overexpression in VSMCs reduces SBP and inhibits AngII-induced vascular remodeling in mice. The inhibition of vascular remodeling contributes, at least in part, to the antihypertensive effect of SIRT1. KEY MESSAGE: SIRT1 is reduced in aortas of AngII-infused hypertensive mice. SIRT1 VSMC transgene alleviates AngII-increased systolic blood pressure. SIRT1 VSMC transgene attenuates AngII-induced vascular remodeling. VSMC SIRT1 overexpression inhibits remodeling-related pathological changes. VSMC SIRT1 overexpression reduces AngII-induced TGF-ß1 expression.


Sujet(s)
Hypertension artérielle/enzymologie , Sirtuine-1/génétique , Angiotensine-II , Animaux , Aorte/enzymologie , Aorte/anatomopathologie , Aorte/physiopathologie , Épigenèse génétique , Expression des gènes , Hypertension artérielle/induit chimiquement , Souris de lignée C57BL , Souris transgéniques , Muscles lisses vasculaires/enzymologie , Myocytes du muscle lisse/enzymologie , Espèces réactives de l'oxygène/métabolisme , Sirtuine-1/métabolisme , Vascularite/induit chimiquement , Vascularite/enzymologie
18.
Blood ; 123(2): 261-70, 2014 Jan 09.
Article de Anglais | MEDLINE | ID: mdl-24255919

RÉSUMÉ

Promyelocytic leukemia protein (PML) has been implicated as a participant in multiple cellular processes including senescence, apoptosis, proliferation, and differentiation. Studies of PML function in hematopoietic differentiation previously focused principally on its myeloid activities and also indicated that PML is involved in erythroid colony formation. However, the exact role that PML plays in erythropoiesis is essentially unknown. In this report, we found that PML4, a specific PML isoform expressed in erythroid cells, promotes endogenous erythroid genes expression in K562 and primary human erythroid cells. We show that the PML4 effect is GATA binding protein 1 (GATA-1) dependent using GATA-1 knockout/rescued G1E/G1E-ER4 cells. PML4, but not other detected PML isoforms, directly interacts with GATA-1 and can recruit it into PML nuclear bodies. Furthermore, PML4 facilitates GATA-1 trans-activation activity in an interaction-dependent manner. Finally, we present evidence that PML4 enhances GATA-1 occupancy within the globin gene cluster and stimulates cooperation between GATA-1 and its coactivator p300. These results demonstrate that PML4 is an important regulator of GATA-1 and participates in erythroid differention by enhancing GATA-1 trans-activation activity.


Sujet(s)
Différenciation cellulaire/physiologie , Cellules érythroïdes/cytologie , Cellules érythroïdes/métabolisme , Facteur de transcription GATA-1/génétique , Protéines nucléaires/métabolisme , Facteurs de transcription/métabolisme , Activation de la transcription , Protéines suppresseurs de tumeurs/métabolisme , Acétylation , Protéine p300-E1A/métabolisme , Facteur de transcription GATA-1/composition chimique , Facteur de transcription GATA-1/métabolisme , Expression des gènes , Humains , Cellules K562 , Protéines nucléaires/composition chimique , Protéines nucléaires/génétique , Protéine de la leucémie promyélocytaire , Liaison aux protéines , Motifs et domaines d'intéraction protéique , Isoformes de protéines , Transactivateurs/génétique , Transactivateurs/métabolisme , Facteurs de transcription/composition chimique , Facteurs de transcription/génétique , Transcription génétique , Protéines suppresseurs de tumeurs/composition chimique , Protéines suppresseurs de tumeurs/génétique , Doigts de zinc
19.
Basic Res Cardiol ; 108(4): 364, 2013 Jul.
Article de Anglais | MEDLINE | ID: mdl-23744058

RÉSUMÉ

Nkx2.5 plays protective roles in cardiac homeostasis and survival in the postnatal hearts. However, the underlying molecular mechanisms that mediate the protective functions of Nkx2.5 remain unknown. Here, we showed that Nkx2.5 was downregulated in response to various stresses and was required for protection against the stress-induced apoptosis of cardiomyocytes. SIRT1, a member of the sirtuin family of proteins, was found to be a direct transcriptional target of Nkx2.5 and was required for the Nkx2.5-mediated protection of cardiomyocytes from doxorubicin (DOX)-induced apoptosis. Moreover, using chromatin immunoprecipitation assays, we found that Nkx2.5 was able to bind to the SIRT1 promoter and that this binding was significantly decreased in DOX-treated mouse hearts. Furthermore, the cardiac-specific overexpression of SIRT1 decreased the DOX-induced apoptosis of cardiomyocytes in SIRT1 transgenic mouse hearts compared with the hearts of their wild-type littermates. These findings demonstrate that SIRT1 acts as a direct transcriptional target of Nkx2.5 that maintains cardiomyocyte homeostasis and survival.


Sujet(s)
Protéines à homéodomaine/physiologie , Myocytes cardiaques/physiologie , Sirtuine-1/physiologie , Stress physiologique/physiologie , Facteurs de transcription/physiologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Apoptose/physiologie , Survie cellulaire/physiologie , Cellules cultivées , Doxorubicine/pharmacologie , Protéine homéotique Nkx-2.5 , Homéostasie/physiologie , Souris , Souris transgéniques , Modèles animaux , Myocytes cardiaques/cytologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Rats , Rat Sprague-Dawley , Transduction du signal/physiologie , Sirtuine-1/génétique , Régulation positive/physiologie
20.
Nucleic Acids Res ; 40(11): 4804-15, 2012 Jun.
Article de Anglais | MEDLINE | ID: mdl-22328728

RÉSUMÉ

The higher order chromatin structure has recently been revealed as a critical new layer of gene transcriptional control. Changes in higher order chromatin structures were shown to correlate with the availability of transcriptional factors and/or MAR (matrix attachment region) binding proteins, which tether genomic DNA to the nuclear matrix. How posttranslational modification to these protein organizers may affect higher order chromatin structure still pending experimental investigation. The type III histone deacetylase silent mating type information regulator 2, S. cerevisiae, homolog 1 (SIRT1) participates in many physiological processes through targeting both histone and transcriptional factors. We show that MAR binding protein SATB1, which mediates chromatin looping in cytokine, MHC-I and ß-globin gene loci, as a new type of SIRT1 substrate. SIRT1 expression increased accompanying erythroid differentiation and the strengthening of ß-globin cluster higher order chromatin structure, while knockdown of SIRT1 in erythroid k562 cells weakened the long-range interaction between two SATB1 binding sites in the ß-globin locus, MAR(HS2) and MAR(ε). We also show that SIRT1 activity significantly affects ε-globin gene expression in a SATB1-dependent manner and that knockdown of SIRT1 largely blocks ε-globin gene activation during erythroid differentiation. Our work proposes that SIRT1 orchestrates changes in higher order chromatin structure during erythropoiesis, and reveals the dynamic higher order chromatin structure regulation at posttranslational modification level.


Sujet(s)
Régulation de l'expression des gènes , Protéines de liaison aux séquences d'ADN MAR/métabolisme , Régions d'ancrage à la matrice nucléaire , Sirtuine-1/métabolisme , Globines epsilon/génétique , Cellules cultivées , Cellules érythroïdes/effets des médicaments et des substances chimiques , Cellules érythroïdes/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Hémine/pharmacologie , Humains , Cellules K562 , Région de contrôle de locus , Globines bêta/génétique , Globines epsilon/biosynthèse
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE