Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Sci Immunol ; 9(96): eadj5465, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38875319

ABSTRACT

Nucleic acids are major structures detected by the innate immune system. Although intracellular single-stranded DNA (ssDNA) accumulates during pathogen infection or disease, it remains unclear whether and how intracellular ssDNA stimulates the innate immune system. Here, we report that intracellular ssDNA triggers cytokine expression and cell death in a CGT motif-dependent manner. We identified Schlafen 11 (SLFN11) as an ssDNA-activated RNase, which is essential for the innate immune responses induced by intracellular ssDNA and adeno-associated virus infection. We found that SLFN11 directly binds ssDNA containing CGT motifs through its carboxyl-terminal domain, translocates to the cytoplasm upon ssDNA recognition, and triggers innate immune responses through its amino-terminal ribonuclease activity that cleaves transfer RNA (tRNA). Mice deficient in Slfn9, a mouse homolog of SLFN11, exhibited resistance to CGT ssDNA-induced inflammation, acute hepatitis, and septic shock. This study identifies CGT ssDNA and SLFN11/9 as a class of immunostimulatory nucleic acids and pattern recognition receptors, respectively, and conceptually couples DNA immune sensing to controlled RNase activation and tRNA cleavage.


Subject(s)
DNA, Single-Stranded , Immunity, Innate , Mice, Inbred C57BL , Animals , Female , Humans , Male , Mice , DNA, Single-Stranded/immunology , HEK293 Cells , Immunity, Innate/immunology , Mice, Knockout , Nuclear Proteins/immunology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ribonucleases/immunology , Ribonucleases/metabolism
2.
Signal Transduct Target Ther ; 8(1): 255, 2023 07 03.
Article in English | MEDLINE | ID: mdl-37394473

ABSTRACT

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.


Subject(s)
Aortic Aneurysm, Thoracic , Sirtuins , Humans , Mice , Animals , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/metabolism , Aortic Aneurysm, Thoracic/pathology , Inflammation/genetics , Angiotensin II/genetics , Angiotensin II/pharmacology , Epigenesis, Genetic/genetics , Sirtuins/genetics
3.
Org Biomol Chem ; 21(17): 3675-3683, 2023 May 03.
Article in English | MEDLINE | ID: mdl-37067868

ABSTRACT

We report a ring-opening trifluoromethylthiolation of cyclopropanols with TsSCF3 by using Cu(OAc)2 as the catalyst. Moreover, by using this strategy, the trifluoromethylselenolation of cyclopropanols with Se-(trifluoromethyl) 4-methoxybenzenesulfonoselenoate to access ß-SeCF3-substituted carbonyl compounds is achieved for the first time. The broad substrate scope, readily accessible reagents and cheap catalyst make this protocol an alternative and efficient method for the synthesis of ß-SCF3-substituted or ß-SeCF3-substituted carbonyl compounds.

4.
Exp Cell Res ; 422(1): 113409, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36356655

ABSTRACT

Oxidative stress caused by excess ROS often leads to cellular macromolecule damage and eventually causes various biological catastrophes. Sirt6, a member of the mammalian homolog family of yeast Sir2 NAD+-dependent histone deacetylases, regulates multiple biological processes. Sirt6 exerts antioxidative functions by enhancing DNA repair and DNA end resection. In our study, we found that Sirt6 expression was induced by H2O2 and paraquat (PQ) in cells. When exposed to PQ, the Sirt6+/- C57BL/6 mice showed more serious liver damage and lower survival rate than the Sirt6+/+ mice. The Nrf2 protein levels and the mRNA levels of its target genes in mouse tissues were decreased by Sirt6 deficiency, and Sirt6 overexpression increased the Nrf2 protein content. Moreover, the endogenous H2O2 levels were increased in the tissues of Sirt6-deficient mice and were decreased in Sirt6 overexpression cells. Then, we found that Nrf2 was degraded faster in the Sirt6-deficient mouse embryonic fibroblasts (MEFs) than in the wild type MEFs and that Sirt6 enhanced the protein accumulation of Nrf2 in the nucleus. Lastly, we found that Sirt6 interacted with Nrf2 in co-IP and GST pull-down assays and that Sirt6 overexpression decreased the binding of Nrf2 to Keap1. Taken together, the results of the present study suggest that Sirt6 exerts antioxidative functions by increasing the Nrf2 protein level via Keap1-mediated regulation.


Subject(s)
NF-E2-Related Factor 2 , Sirtuins , Animals , Mice , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Antioxidants/metabolism , Hydrogen Peroxide/metabolism , Sirtuins/genetics , Sirtuins/metabolism , Mice, Inbred C57BL , Fibroblasts/metabolism , Oxidative Stress , Paraquat , Mammals/metabolism
6.
Nat Cell Biol ; 21(12): 1553-1564, 2019 12.
Article in English | MEDLINE | ID: mdl-31768048

ABSTRACT

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.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Hydrogen Peroxide/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Animals , Female , Liver/metabolism , Liver/physiology , Male , Mammals/metabolism , Mammals/physiology , Mice , Mice, Knockout , Oxidation-Reduction , Period Circadian Proteins/metabolism , Signal Transduction/physiology , Suprachiasmatic Nucleus/metabolism , Suprachiasmatic Nucleus/physiology
7.
iScience ; 17: 155-166, 2019 Jul 26.
Article in English | MEDLINE | ID: mdl-31279933

ABSTRACT

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.

8.
J Genet Genomics ; 45(1): 25-32, 2018 01 20.
Article in English | MEDLINE | ID: mdl-29396144

ABSTRACT

Abdominal aortic aneurysm (AAA) is a vascular degenerative disease. Macrophage polarization and the balance between classically activated macrophages (M1) and alternatively activated macrophages (M2) are crucial for AAA pathogenesis. The present study aims to investigate the roles of macrophage SIRT1 in AAA formation and macrophage polarization. We found that in mouse peritoneal macrophages, SIRT1 expression was decreased after M1 stimulation, but was enhanced after M2 stimulation. Results from SIRT1flox/flox mice and macrophage specific SIRT1 knockout mice with treatment of angiotensin II (Ang II) for 4 weeks showed that macrophage specific deficiency of SIRT1 increased the incidence of AAA and exacerbated the severity, including more severe aneurysm types, enlarged diameter of the aneurysm and increased degradation of elastin. In mouse aortas, SIRT1 deficiency increased the pro-inflammatory M1 molecule inducible nitric oxide synthase (iNOS), and decreased M2 molecules such as arginase 1 (Arg1) and mannose receptor (MR). Furthermore, in peritoneal macrophages, SIRT1 deficiency increased the expression of M1 inflammatory molecules, but decreased the expression of M2 molecules. Overexpression of SIRT1 had the opposite effects. Thus, macrophage specific knockout of SIRT1 influences macrophage polarization and accelerates Ang II-induced AAA formation.


Subject(s)
Aorta/metabolism , Aortic Aneurysm, Abdominal/genetics , Cell Polarity/genetics , Sirtuin 1/genetics , Angiotensin II/administration & dosage , Angiotensin II/adverse effects , Animals , Aorta/physiopathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/pathology , Arginase/genetics , Disease Models, Animal , Humans , Lectins, C-Type/genetics , Macrophages/drug effects , Mannose Receptor , Mannose-Binding Lectins/genetics , Mice , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Receptors, Cell Surface/genetics , Signal Transduction/genetics
9.
Circulation ; 136(21): 2051-2067, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-28947430

ABSTRACT

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.


Subject(s)
Cardiomegaly/prevention & control , Metformin/pharmacology , Myocardium/enzymology , Sirtuin 2/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Acetylation , Age Factors , Aging/metabolism , Angiotensin II , Animals , Cardiomegaly/chemically induced , Cardiomegaly/enzymology , Cardiomegaly/physiopathology , Cells, Cultured , Disease Models, Animal , Fibrosis , Genetic Predisposition to Disease , Lysine , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction/drug effects , Myocardium/pathology , Phenotype , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/pharmacology , Rats , Signal Transduction/drug effects , Sirtuin 2/deficiency , Sirtuin 2/genetics , Stroke Volume/drug effects , Ventricular Remodeling/drug effects
10.
Sci Rep ; 7: 46204, 2017 04 10.
Article in English | MEDLINE | ID: mdl-28393844

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Inflammation/genetics , Inflammation/pathology , Monocytes/metabolism , Monocytes/pathology , NF-kappa B/metabolism , RNA, Long Noncoding/metabolism , Repressor Proteins/metabolism , Aorta/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line , Cell Nucleus/metabolism , Genome-Wide Association Study , Humans , Myocytes, Smooth Muscle/metabolism , Phenotype , Protein Transport , RNA, Long Noncoding/genetics , Receptors, Cell Surface/metabolism , Repressor Proteins/genetics , Signal Transduction/genetics , Up-Regulation
11.
Eur Heart J ; 38(18): 1389-1398, 2017 May 07.
Article in English | MEDLINE | ID: mdl-27099261

ABSTRACT

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.


Subject(s)
Cardiomegaly/enzymology , Mitochondrial Proteins/physiology , Sirtuins/physiology , Superoxide Dismutase/antagonists & inhibitors , Angiotensin II/pharmacology , Animals , Gene Knockdown Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Heart/enzymology , Myocytes, Cardiac/enzymology , Oxidative Stress/physiology , Reactive Oxygen Species/antagonists & inhibitors , Vascular Remodeling/physiology , Vasoconstrictor Agents/pharmacology
12.
J Exp Med ; 213(11): 2473-2488, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27670594

ABSTRACT

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.


Subject(s)
Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/prevention & control , Caloric Restriction , Acetylation , Angiotensin II , Animals , Aorta/metabolism , Aortic Aneurysm, Abdominal/blood , Aortic Aneurysm, Abdominal/metabolism , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Chromatin/metabolism , Epigenesis, Genetic , Glucose/metabolism , Histones/metabolism , Lipid Metabolism/genetics , Lipids/blood , Liver/metabolism , Lysine/metabolism , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Oxidative Phosphorylation , Promoter Regions, Genetic/genetics , Sirtuin 1/metabolism
13.
Sci Rep ; 6: 23912, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27045575

ABSTRACT

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.


Subject(s)
Apolipoproteins E/genetics , Epigenesis, Genetic , NK Cell Lectin-Like Receptor Subfamily K/genetics , Plaque, Atherosclerotic/metabolism , Sirtuins/genetics , Animals , Endarterectomy, Carotid , Female , Fibroblasts/metabolism , Heterozygote , Humans , Ligands , Male , Mice , Mice, Knockout , Promoter Regions, Genetic , Signal Transduction
14.
Nucleic Acids Res ; 44(6): 2613-27, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26615201

ABSTRACT

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.


Subject(s)
Homeodomain Proteins/genetics , Mouse Embryonic Stem Cells/metabolism , Multigene Family , Octamer Transcription Factor-3/genetics , Polycomb Repressive Complex 2/genetics , RNA, Long Noncoding/genetics , Animals , Cell Line , Chromatin/chemistry , Chromatin/metabolism , Gene Expression Regulation , Histones/genetics , Histones/metabolism , Homeodomain Proteins/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Octamer Transcription Factor-3/metabolism , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
15.
Sci China Life Sci ; 58(9): 860-6, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26376810

ABSTRACT

Spatial expression patterns of homeobox (HOX) genes delineate positional identity of primary fibroblasts from different topographic sites. The molecular mechanism underlying the establishing or maintaining of HOX gene expression pattern remains an attractive developmental issue to be addressed. Our previous work suggested a critical role of CTCF/cohesin-mediated higher- order chromatin structure in RA-induced HOXA activation in human teratocarcinoma NT2/D1 cells. This study investigated the recruitment of CTCF and cohesin, and the higher-order chromatin structure of the HOXA locus in fetal lung and adult foreskin fibroblasts, which display complementary HOXA gene expression patterns. Chromatin contacts between the CTCF-binding sites were observed with lower frequency in human foreskin fibroblasts. This observation is consistent with the lower level of cohesin recruitment and 5' HOXA gene expression in the same cells. We also showed that CTCF-binding site A56 (CBSA56) related chromatin structures exhibit the most notable changes in between the two types of cell, and hence may stand for one of the key CTCF-binding sites for cell-type specific chromatin structure organization. Together, these results imply that CTCF/cohesin coordinates HOXA cluster higher-order chromatin structure and expression during development, and provide insight into the relationship between cell-type specific chromatin organization and the spatial collinearity.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Fibroblasts/metabolism , Homeodomain Proteins/genetics , Multigene Family , Repressor Proteins/metabolism , Transcription Factors/genetics , Binding Sites , CCCTC-Binding Factor , Cell Line , Cells, Cultured , Chromatin Immunoprecipitation , Cluster Analysis , Fibroblasts/cytology , Foreskin/cytology , Gene Expression Profiling , Gene Expression Regulation , Genes, Homeobox , HEK293 Cells , Humans , Lung/cytology , Lung/embryology , Male , Protein Binding , Repressor Proteins/genetics , Cohesins
16.
Stem Cells ; 33(7): 2135-47, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25940188

ABSTRACT

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.


Subject(s)
SOXB1 Transcription Factors/metabolism , Sirtuin 1/metabolism , Animals , Cell Differentiation , Cellular Reprogramming , Kruppel-Like Factor 4 , Mice
17.
Mol Cell Biol ; 34(20): 3867-79, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25135475

ABSTRACT

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.


Subject(s)
Chromatin/genetics , Gene Silencing , Homeodomain Proteins/genetics , Polycomb Repressive Complex 2/physiology , Repressor Proteins/physiology , Base Sequence , CCCTC-Binding Factor , Cell Line, Tumor , Chromatin/metabolism , Genetic Loci , Histones/metabolism , Homeodomain Proteins/metabolism , Homeodomain Proteins/ultrastructure , Humans , Molecular Conformation , Protein Stability , Sequence Deletion , Tretinoin/physiology
18.
Aging Cell ; 13(5): 890-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25040736

ABSTRACT

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.


Subject(s)
Cellular Senescence/physiology , Plasminogen Activator Inhibitor 1/genetics , Serpin E2/genetics , Sirtuin 1/genetics , Animals , Down-Regulation , Epigenesis, Genetic , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice , Mice, Transgenic , Plaque, Atherosclerotic/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Serpin E2/metabolism , Sirtuin 1/metabolism
19.
Blood ; 123(2): 261-70, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24255919

ABSTRACT

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.


Subject(s)
Cell Differentiation/physiology , Erythroid Cells/cytology , Erythroid Cells/metabolism , GATA1 Transcription Factor/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Tumor Suppressor Proteins/metabolism , Acetylation , E1A-Associated p300 Protein/metabolism , GATA1 Transcription Factor/chemistry , GATA1 Transcription Factor/metabolism , Gene Expression , Humans , K562 Cells , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Protein Binding , Protein Interaction Domains and Motifs , Protein Isoforms , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription, Genetic , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Zinc Fingers
20.
J Mol Med (Berl) ; 92(4): 347-57, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24352856

ABSTRACT

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.


Subject(s)
Hypertension/enzymology , Sirtuin 1/genetics , Angiotensin II , Animals , Aorta/enzymology , Aorta/pathology , Aorta/physiopathology , Epigenesis, Genetic , Gene Expression , Hypertension/chemically induced , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Reactive Oxygen Species/metabolism , Sirtuin 1/metabolism , Vasculitis/chemically induced , Vasculitis/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...