Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters











Publication year range
1.
J Biochem ; 171(6): 673-693, 2022 May 27.
Article in English | MEDLINE | ID: mdl-35325168

ABSTRACT

Gram-negative intracellular pathogen Vibrio parahaemolyticus manifests its infection through a series of effector proteins released into the host via the type III secretion system. Most of these effector proteins alter signalling pathways of the host to facilitate survival and proliferation of bacteria inside host cells. Here, we report V. parahaemolyticus (serotype O3:K6) infection-induced histone deacetylation in host intestinal epithelial cells, particularly deacetylation of H3K9, H3K56, H3K18 and H4K16 residues. We found a putative NAD+-dependent deacetylase, vp1524 (vpCobB) of V. parahaemolyticus, was overexpressed during infection. Biochemical assays revealed that Vp1524 is a functional NAD+-dependent Sir2 family deacetylase in vitro, which was capable of deacetylating acetylated histones. Furthermore, we observed that vp1524 is expressed and localized to the nuclear periphery of the host cells during infection. Consequently, Vp1524 translocated to nuclear compartments of transfected cells, deacetylated histones, specifically causing deacetylation of those residues (K56, K16, K18) associated with V. parahaemolyticus infection. This infection induced deacetylation resulted in transcriptional repression of several host genes involved in epigenetic regulation, immune response, autophagy etc. Thus, our study shows that a V. parahaemolyticus lysine deacetylase Vp1524 is secreted inside the host cells during infection, modulating host gene expression through histone deacetylation.


Subject(s)
Group III Histone Deacetylases/metabolism , Vibrio parahaemolyticus , Epigenesis, Genetic , Histones/metabolism , Immunity , NAD/genetics , NAD/metabolism , Vibrio parahaemolyticus/genetics , Vibrio parahaemolyticus/metabolism
2.
Eur J Pharmacol ; 914: 174666, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34861210

ABSTRACT

Diabetes mellitus (DM) is associated with accelerated cognitive decline. However, the mechanism of diabetic cognitive impairment remains poorly understood. In this study, we found that the expression of Sirtuin 1 (SIRT1), a nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylase, was downregulated significantly in the hippocampus of streptozotocin (STZ)-induced diabetic cognitive impairment rats. Viral overexpression of hippocampal SIRT1 ameliorated cognitive impairment in diabetic rats, but viral knockdown of hippocampal SIRT1 mimicked the diabetic effect, eliciting the cognitive decline in normal animals. Further study showed that the decreased level of SIRT1 may result in the increase of acetylated tau protein in the hippocampus, which may mediate the development of diabetic cognitive impairment. These results suggest that SIRT1 may be a key epigenetic regulator that guards against the development of diabetic cognitive impairment by deacetylating the tau protein. SIRT1 activator may serve as a new therapeutic approach for the treatment of diabetic cognitive impairment.


Subject(s)
Cognitive Dysfunction , Diabetes Complications/metabolism , Sirtuin 1/metabolism , tau Proteins/metabolism , Acetylation , Animals , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Diabetes Mellitus, Experimental , Down-Regulation , Epigenesis, Genetic , Group III Histone Deacetylases/metabolism , Hippocampus/metabolism , Protein Processing, Post-Translational , Rats
3.
Gene ; 704: 134-141, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-30981839

ABSTRACT

To maintain normal function of cartilage tissue normally, the presence of a sufficient amount of type II collagen and aggrecan is essential, and their synthesis is tightly regulated. Therefore, understanding the mechanisms that control the expression of type II collagen and aggrecan would be useful for understanding gene expression changes in diseases such as osteoarthritis. Recently, we have identified two pairs of enhancer elements, termed E1 and E2 in the type II collagen gene and Ea and Eb in the aggrecan gene. However, their different mechanisms of action remained unclear. Thus, the central aim of this study was to clarify the different transcriptional regulation mediated through each enhancer element. To this end, we established different stable reporter cell lines that express a reporter gene under the control of different enhancer elements using a silent reporter system we previously constructed. Using these cell lines, we found that dexamethasone, forskolin, and trichostatin A affect the gene expression of type II collagen and aggrecan via different enhancer elements. Moreover, we clarified that E1 and E2 enhancer activities are regulated through distinct epigenetic modifications by histone deacetylase 10 and sirtuin 6.


Subject(s)
Aggrecans/genetics , Collagen Type II/genetics , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic/physiology , Aggrecans/metabolism , Animals , Chondrocytes/metabolism , Chondrocytes/pathology , Collagen Type II/metabolism , Gene Expression Regulation , Group III Histone Deacetylases/metabolism , Promoter Regions, Genetic , Rats , Sirtuins/metabolism , Tumor Cells, Cultured
4.
Curr Opin Clin Nutr Metab Care ; 21(4): 252-259, 2018 07.
Article in English | MEDLINE | ID: mdl-29847446

ABSTRACT

PURPOSE OF REVIEW: We present a current perspective of epigenetic alterations that can lead to cardiovascular disease (CVD) and the potential of dietary factors to counteract their actions. In addition, we discuss the challenges and opportunities of dietary treatments as epigenetic modifiers for disease prevention and therapy. RECENT FINDINGS: Recent epigenome-wide association studies along with candidate gene approaches and functional studies in cell culture and animal models have delineated mechanisms through which nutrients, food compounds and dietary patterns may affect the epigenome. Several risk factors for CVD, including adiposity, inflammation and oxidative stress, have been associated with changes in histone acetylation, lower global DNA methylation levels and shorter telomere length. A surplus of macronutrients such as in a high-fat diet or deficiencies of specific nutrients such as folate and other B-vitamins can affect the activity of DNA methyltransferases and histone-modifying enzymes, affecting foetal growth, glucose/lipid metabolism, oxidative stress, inflammation and atherosclerosis. Bioactive compounds such as polyphenols (resveratrol, curcumin) or epigallocatechin may activate deacetylases Sirtuins (SIRTs), histone deacetylases or acetyltransferases and in turn the response of inflammatory mediators. Adherence to cardioprotective dietary patterns, such as the Mediterranean diet (MedDiet), has been associated with altered methylation and expression of genes related to inflammation and immuno-competence. SUMMARY: The mechanisms through which nutrients and dietary patterns may alter the cardiovascular epigenome remain elusive. The research challenge is to determine which of these nutriepigenetic effects are reversible, so that novel findings translate into effective dietary interventions to prevent CVD or its progression.


Subject(s)
Cardiovascular Diseases , DNA Methylation , Diet , Epigenesis, Genetic , Histones/metabolism , Nutritional Status , Acetylation , Animals , Cardiovascular Diseases/diet therapy , Cardiovascular Diseases/etiology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Group III Histone Deacetylases/metabolism , Humans , Inflammation/genetics , Polyphenols , Protein Processing, Post-Translational
5.
PLoS One ; 13(3): e0193602, 2018.
Article in English | MEDLINE | ID: mdl-29543820

ABSTRACT

The de novo crystal structure of the Leishmania infantum Silent Information Regulator 2 related protein 1 (LiSir2rp1) has been solved at 1.99Å in complex with an acetyl-lysine peptide substrate. The structure is broadly commensurate with Hst2/SIRT2 proteins of yeast and human origin, reproducing many of the structural features common to these sirtuin deacetylases, including the characteristic small zinc-binding domain, and the larger Rossmann-fold domain involved in NAD+-binding interactions. The two domains are linked via a cofactor binding loop ordered in open conformation. The peptide substrate binds to the LiSir2rp1 protein via a cleft formed between the small and large domains, with the acetyl-lysine side chain inserting further into the resultant hydrophobic tunnel. Crystals were obtained only with recombinant LiSir2rp1 possessing an extensive internal deletion of a proteolytically-sensitive region unique to the sirtuins of kinetoplastid origin. Deletion of 51 internal amino acids (P253-E303) from LiSir2rp1 did not appear to alter peptide substrate interactions in deacetylation assays, but was indispensable to obtain crystals. Removal of this potentially flexible region, that otherwise extends from the classical structural elements of the Rossmann-fold, specifically the ß8-ß9 connector, appears to result in lower accumulation of the protein when expressed from episomal vectors in L. infantum SIR2rp1 single knockout promastigotes. The biological function of the large serine-rich insertion in kinetoplastid/trypanosomatid sirtuins, highlighted as a disordered region with strong potential for post-translational modification, remains unknown but may confer additional cellular functions that are distinct from their human counterparts. These unique molecular features, along with the resolution of the first kinetoplastid sirtuin deacetylase structure, present novel opportunities for drug design against a protein target previously established as essential to parasite survival and proliferation.


Subject(s)
Group III Histone Deacetylases/chemistry , Group III Histone Deacetylases/metabolism , Leishmania infantum/metabolism , Binding Sites , Crystallography, X-Ray , Drug Design , Humans , Models, Molecular , Peptides/metabolism , Protein Binding , Protein Structure, Secondary , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism
6.
J Biol Chem ; 293(14): 5281-5294, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29440391

ABSTRACT

Heart failure is an aging-associated disease that is the leading cause of death worldwide. Sirtuin family members have been largely studied in the context of aging and aging-associated diseases. Sirtuin 2 (SIRT2) is a cytoplasmic protein in the family of sirtuins that are NAD+-dependent class III histone deacetylases. In this work, we studied the role of SIRT2 in regulating nuclear factor of activated T-cells (NFAT) transcription factor and the development of cardiac hypertrophy. Confocal microscopy analysis indicated that SIRT2 is localized in the cytoplasm of cardiomyocytes and SIRT2 levels are reduced during pathological hypertrophy of the heart. SIRT2-deficient mice develop spontaneous pathological cardiac hypertrophy, remodeling, fibrosis, and dysfunction in an age-dependent manner. Moreover, young SIRT2-deficient mice develop exacerbated agonist-induced hypertrophy. In contrast, SIRT2 overexpression attenuated agonist-induced cardiac hypertrophy in cardiomyocytes in a cell-autonomous manner. Mechanistically, SIRT2 binds to and deacetylates NFATc2 transcription factor. SIRT2 deficiency stabilizes NFATc2 and enhances nuclear localization of NFATc2, resulting in increased transcription activity. Our results suggest that inhibition of NFAT rescues the cardiac dysfunction in SIRT2-deficient mice. Thus, our study establishes SIRT2 as a novel endogenous negative regulator of NFAT transcription factor.


Subject(s)
Cardiomegaly/metabolism , NFATC Transcription Factors/metabolism , Sirtuin 2/metabolism , Acetylation , Animals , Gene Expression Regulation/genetics , Group III Histone Deacetylases/metabolism , Heart Failure/metabolism , Homeostasis , Mice , Mice, Knockout , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Sirtuin 2/physiology
7.
J Neuromuscul Dis ; 5(1): 59-73, 2018.
Article in English | MEDLINE | ID: mdl-29278895

ABSTRACT

BACKGROUND: Mutations in the LAMA2 gene encoding laminin-α2 cause congenital muscular dystrophy Type 1A (MDC1A), a severe recessive disease with no effective treatment. Previous studies have shown that aberrant activation of caspases and cell death through a pathway regulated by BAX and KU70 is a significant contributor to pathogenesis in laminin-α2-deficiency. OBJECTIVES: To identify mechanisms of pathogenesis in MDC1A. METHODS: We used immunocytochemical and molecular studies of human myogenic cells and mouse muscles-comparing laminin-α2-deficient vs. healthy controls-to identify mechanisms that regulate pathological activation of caspase in laminin-α2-deficiency. RESULTS: In cultures of myogenic cells from MDC1A donors, p53 accumulated in a subset of nuclei and aberrant caspase activation was inhibited by the p53 inhibitor pifithrin-alpha. Also, the p53 target BBC3 (PUMA) was upregulated in both MDC1A myogenic cells and Lama2-/- mouse muscles. In addition, studies with sirtuin inhibitors and SIRT1 overexpression showed that caspase activation in MDC1A myotubes was inversely related to sirtuin deacetylase activity. Caspase activation in laminin-α2-deficiency was, however, not associated with increased phosphorylation of p38 MAPK. CONCLUSIONS: Aberrant caspase activation in MDC1A cells was mediated both by sirtuin deacetylase activity and by p53. Interventions that inhibit aberrant caspase activation by targeting sirtuin or p53 function could potentially be useful in ameliorating MDC1A.


Subject(s)
Caspases/metabolism , Laminin/genetics , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophies/metabolism , Sirtuins/metabolism , Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Benzothiazoles/pharmacology , Group III Histone Deacetylases/metabolism , Humans , Laminin/metabolism , Mice , Mice, Knockout , Muscle Development , Muscle Fibers, Skeletal/drug effects , Muscular Dystrophies/genetics , Phosphorylation , Proto-Oncogene Proteins/metabolism , Sirtuin 1/metabolism , Stem Cells/drug effects , Toluene/analogs & derivatives , Toluene/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Mol Microbiol ; 107(4): 577-594, 2018 02.
Article in English | MEDLINE | ID: mdl-29266439

ABSTRACT

Protein acetylation is a rapid mechanism for control of protein function. Acetyl-CoA synthetase (AMP-forming, Acs) is the paradigm for the control of metabolic enzymes by lysine acetylation. In many bacteria, type I or II protein acetyltransferases acetylate Acs, however, in actinomycetes type III protein acetyltransferases control the activity of Acs. We measured changes in the activity of the Streptomyces lividans Acs (SlAcs) enzyme upon acetylation by PatB using in vitro and in vivo analyses. In addition to the acetylation of residue K610, residue S608 within the acetylation motif of SlAcs was also acetylated (PKTRSGK610 ). S608 acetylation rendered SlAcs inactive and non-acetylatable by PatB. It is unclear whether acetylation of S608 is enzymatic, but it was clear that this modification occurred in vivo in Streptomyces. In S. lividans, an NAD+ -dependent sirtuin deacetylase from Streptomyces, SrtA (a homologue of the human SIRT4 protein) was needed to maintain SlAcs function in vivo. We have characterized a sirtuin-dependent reversible lysine acetylation system in Streptomyces lividans that targets and controls the Acs enzyme of this bacterium. These studies raise questions about acetyltransferase specificity, and describe the first Acs enzyme in any organism whose activity is modulated by O-Ser and Nɛ -Lys acetylation.


Subject(s)
Acetate-CoA Ligase/metabolism , Acetyl Coenzyme A/metabolism , Lysine/metabolism , Serine/metabolism , Streptomyces lividans/enzymology , Acetate-CoA Ligase/genetics , Acetylation , Acetyltransferases/genetics , Acetyltransferases/metabolism , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , DNA, Bacterial/genetics , Gene Deletion , Group III Histone Deacetylases/genetics , Group III Histone Deacetylases/metabolism , NAD/metabolism , Streptomyces lividans/genetics
9.
Med Res Rev ; 38(1): 147-200, 2018 01.
Article in English | MEDLINE | ID: mdl-28094444

ABSTRACT

Sirtuins are NAD+ -dependent protein deacylases that cleave off acetyl, as well as other acyl groups, from the ε-amino group of lysines in histones and other substrate proteins. Seven sirtuin isotypes (Sirt1-7) have been identified in mammalian cells. As sirtuins are involved in the regulation of various physiological processes such as cell survival, cell cycle progression, apoptosis, DNA repair, cell metabolism, and caloric restriction, a dysregulation of their enzymatic activity has been associated with the pathogenesis of neoplastic, metabolic, infectious, and neurodegenerative diseases. Thus, sirtuins are promising targets for pharmaceutical intervention. Growing interest in a modulation of sirtuin activity has prompted the discovery of several small molecules, able to inhibit or activate certain sirtuin isotypes. Herein, we give an update to our previous review on the topic in this journal (Schemies, 2010), focusing on recent developments in sirtuin biology, sirtuin modulators, and their potential as novel therapeutic agents.


Subject(s)
Group III Histone Deacetylases/antagonists & inhibitors , Group III Histone Deacetylases/metabolism , Histone Deacetylase Inhibitors/pharmacology , Animals , Humans , Molecular Targeted Therapy
11.
Free Radic Biol Med ; 108: 929-939, 2017 07.
Article in English | MEDLINE | ID: mdl-28506746

ABSTRACT

Silent information regulator 2 (Sir2) enzymes which catalyze NAD+-dependent protein/histone deacetylation. The mammalian sirtuin family SIRT1, SIRT2, SIRT3 and SIRT6 can regulate oxidative stress. The probiotics (Bifidobacterium longum(B.longum) and Lactobacillus acidophilus(L. acidophilus)) have Sir2 gene family and have antioxidant activity in human body. it remains unknown whether probiotics Sir2 has a direct role in regulating oxidative stress. To this end, we knockout BL-sir2(sir2 B. longum) and LA-sir2(sir2 L.acidophilus) in low oxygen level. The antioxidant activities of two sir2 deficient strains was decreased, while when reintroduction of BL-sir2 and LA-sir2, the antioxidant activities were recoveried. In order to understand the regulation mechanism of probiotics Sir2 oxidation response. Then, we screened 65 acetylated protein, and found that SigH (σH) was a substrate of BL-Sir2. In addition, the acetylation level of σH decreased with the increase of BL-Sir2 level in B. longum. Thus, BL-Sir2 deacetylated σH in response to oxidative stress. Next, we transfected BL-Sir2 into H2O2-induced oxidative damage of 293T cells, BL-Sir2 increased the activity of manganese superoxide dismutase (MnSOD/SOD2) and catalase (CAT) and reduced reactive oxygen species(ROS). Then, we analyzed the differential gene by RNA sequencing and Gene ontology (GO) and found that BL-Sir2 regulated forkhead transcription factor (FOXO3a) mediated antioxidant genes in overexpressed BL-Sir2 HEK293T cells. Our study is the first to link probiotics Sir2 with oxidative stress and uncover the antioxidant mechanism of BL-Sir2 in B. longum itself and human body.


Subject(s)
Bacterial Proteins/metabolism , Bifidobacterium longum/physiology , Group III Histone Deacetylases/metabolism , Lactobacillus acidophilus/physiology , Probiotics , Sigma Factor/metabolism , Acetylation , Bacterial Proteins/genetics , HEK293 Cells , Humans , Hydrogen Peroxide/metabolism , Microorganisms, Genetically-Modified , NAD/metabolism , Oxidation-Reduction , Oxidative Stress , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Sigma Factor/genetics , Superoxide Dismutase/metabolism , Transcriptional Activation
12.
G3 (Bethesda) ; 6(10): 3297-3305, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27543294

ABSTRACT

Gene duplication promotes the diversification of protein functions in several ways. Ancestral functions can be partitioned between the paralogs, or a new function can arise in one paralog. These processes are generally viewed as unidirectional. However, paralogous proteins often retain related functions and can substitute for one another. Moreover, in the event of gene loss, the remaining paralog might regain ancestral functions that had been shed. To explore this possibility, we focused on the sirtuin deacetylase SIR2 and its homolog HST1 in the CTG clade of yeasts. HST1 has been consistently retained throughout the clade, whereas SIR2 is only present in a subset of species. These NAD+-dependent deacetylases generate condensed chromatin that represses transcription and stabilizes tandemly repeated sequences. By analyzing phylogenetic trees and gene order, we found that a single duplication of the SIR2/HST1 gene occurred, likely prior to the emergence of the CTG clade. This ancient duplication was followed by at least two independent losses of SIR2 Functional characterization of Sir2 and Hst1 in three species revealed that these proteins have not maintained consistent functions since the duplication. In particular, the rDNA locus is deacetylated by Sir2 in Candida albicans, by Hst1 in C. lusitaniae, and by neither paralog in C. parapsilosis In addition, the subtelomeres in C. albicans are deacetylated by Sir2 rather than by Hst1, which is orthologous to the sirtuin associated with Saccharomyces cerevisiae subtelomeres. These differences in function support the model that sirtuin deacetylases can regain ancestral functions to compensate for gene loss.


Subject(s)
Candida/genetics , Candida/metabolism , Gene Deletion , Gene Duplication , Group III Histone Deacetylases/metabolism , Acetylation , Fungal Proteins/genetics , Fungal Proteins/metabolism , Genetic Loci , Genotype , Histones/metabolism , Phylogeny , Sirtuin 2/genetics , Sirtuin 2/metabolism , Telomere/genetics , Telomere/metabolism
13.
Exp Parasitol ; 169: 28-33, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27423969

ABSTRACT

Sir2 family proteins are highly conserved and catalyze Nicotinamide Adenine Dinucleotide (NAD(+))-dependent protein deacetylation reaction that regulates multiple cellular processes. Little is known about Sir2 family proteins in Giardia. In this research, Sir2 homologs of Giardia were Phylogenetically analyzed. GL50803_10707 (GlSIR2.2) showed strong homology to SIRT1 and was the only parasite SIRT1 homolog being reported to date. Recombinant GlSIR2.2 (rGlSIR2.2) was expressed and purified. The renaturied recombinant protein showed a typical NAD-dependent protein deacetylase activity that could be inhibited by nicotinamide, with IC50 of 4.47 mM rGlSIR2.2 displayed deacetylase activity under varied NAD(+), with Km, kcat and kcat/Km values of 31.71 µM, 1.4 × 10(-3) s(-1), and 4.42 × 10(-5) µM(-1) s(-1). Similarly, the steady-state kinetic parameters with varied ZMAL, yielded Km, kcat and kcat/Km values of 96.89 µM, 4.7 × 10(-3) s(-1), and 4.85 × 10(-5) µM(-1) s(-1). Anti-rGlSIR2.2 serum was used to probe subcellular localization of GlSIR2.2 and strong staining was found predominantly in the nucleus. So we demonstrated that GlSIR2.2 was a SIRT1-like, nuclear-located, NAD(+)-dependent deacetylase. This is the first report of deacetylase activity of Sir2 family protein in Giardia.


Subject(s)
Cell Nucleus/enzymology , Giardia lamblia/enzymology , Group III Histone Deacetylases/metabolism , Sirtuins/metabolism , Amino Acid Sequence , Benzamides/pharmacology , Fluorescent Antibody Technique, Indirect , Giardia lamblia/classification , Giardia lamblia/ultrastructure , Group III Histone Deacetylases/antagonists & inhibitors , Group III Histone Deacetylases/isolation & purification , Humans , Inhibitory Concentration 50 , Naphthalenes/pharmacology , Naphthols/pharmacology , Niacinamide/pharmacology , Phylogeny , Pyrones/pharmacology , Sequence Alignment , Sirtuins/antagonists & inhibitors , Sirtuins/isolation & purification
14.
DNA Repair (Amst) ; 23: 17-26, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25127744

ABSTRACT

In mammals, NAD represents a nodal point for metabolic regulation, and its availability is critical to genome stability. Several NAD-consuming enzymes are induced in various stress conditions and the consequent NAD decline is generally accompanied by the activation of NAD biosynthetic pathways to guarantee NAD homeostasis. In the bacterial world a similar scenario has only recently begun to surface. Here we review the current knowledge on the involvement of NAD homeostasis in bacterial stress response mechanisms. In particular, we focus on the participation of both NAD-consuming enzymes (DNA ligase, mono(ADP-ribosyl) transferase, sirtuins, and RNA 2'-phosphotransferase) and NAD biosynthetic enzymes (both de novo, and recycling enzymes) in the response to DNA/RNA damage. As further supporting evidence for such a link, a genomic context analysis is presented showing several conserved associations between NAD homeostasis and stress responsive genes.


Subject(s)
Bacteria/genetics , Bacteria/metabolism , DNA Damage , NAD/metabolism , ADP Ribose Transferases/metabolism , Adenosine Diphosphate Ribose/metabolism , DNA Ligases/metabolism , DNA, Bacterial/metabolism , Group III Histone Deacetylases/metabolism , Homeostasis/genetics , NAD/biosynthesis , NAD/genetics , Niacinamide/metabolism , RNA, Bacterial/metabolism
15.
PLoS Genet ; 9(10): e1003935, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24204326

ABSTRACT

Deacetylases of the Sir2 or sirtuin family are thought to regulate life cycle progression and life span in response to nutrient availability. This family has undergone successive rounds of duplication and diversification, enabling the enzymes to perform a wide variety of biological functions. Two evolutionarily conserved functions of yeast Sir2 proteins are the generation of repressive chromatin in subtelomeric domains and the suppression of unbalanced recombination within the tandem rDNA array. Here, we describe the function of the Sir2 ortholog ClHst1 in the yeast Clavispora lusitaniae, an occasional opportunistic pathogen. ClHst1 was localized to the non-transcribed spacer regions of the rDNA repeats and deacetylated histones at these loci, indicating that, like other Sir2 proteins, ClHst1 modulates chromatin structure at the rDNA repeats. However, we found no evidence that ClHst1 associates with subtelomeric regions or impacts gene expression directly. This surprising observation highlights the plasticity of sirtuin function. Related yeast species, including Candida albicans, possess an additional Sir2 family member. Thus, it is likely that the ancestral Candida SIR2/HST1 gene was duplicated and subfunctionalized, such that HST1 retained the capacity to regulate rDNA whereas SIR2 had other functions, perhaps including the generation of subtelomeric chromatin. After subsequent species diversification, the SIR2 paralog was apparently lost in the C. lusitaniae lineage. Thus, C. lusitaniae presents an opportunity to discover how subtelomeric chromatin can be reconfigured.


Subject(s)
Evolution, Molecular , Group III Histone Deacetylases/genetics , Heterochromatin/genetics , Saccharomycetales/enzymology , Acetylation , Chromatin/genetics , Conserved Sequence/genetics , Gene Expression Regulation, Fungal , Group III Histone Deacetylases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomycetales/genetics , Telomere/genetics
16.
Neurotherapeutics ; 10(4): 605-20, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24037427

ABSTRACT

Sirtuins are a conserved family of deacetylases whose activities are dependent on nicotinamide adenine dinucleotide (NAD+). Sirtuins act in different cellular compartments, such as the nucleus where they deacetylate histones and transcriptional factors, in the cytoplasm where they modulate cytoskeletal and signaling molecules, and in the mitochondria where they engage components of the metabolic machinery. Collectively, they tune metabolic processes to energy availability, and modulate stress responses, protein aggregation, inflammatory processes, and genome stability. As such, they have garnered much interest and have been widely studied in aging and age-related neurodegeneration. In this chapter, we review the identification of sirtuins and their biological targets. We focus on their biological mechanisms of action and how they might be regulated, including via NAD metabolism, transcriptional and posttranscriptional control, and as targets of pharmacological agents. Lastly, we highlight the numerous studies suggesting that sirtuins are efficacious therapeutic targets in neurodegenerative disease and injury.


Subject(s)
Group III Histone Deacetylases/metabolism , Nervous System Diseases/therapy , Sirtuins/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Gene Expression Regulation , Group III Histone Deacetylases/genetics , Humans , Mitochondria/genetics , Mitochondria/metabolism , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Sirtuins/genetics
17.
PLoS One ; 8(6): e66807, 2013.
Article in English | MEDLINE | ID: mdl-23825566

ABSTRACT

Histone acetylation/deacetylation is an important chromatin modification for epigenetic regulation of gene expression. Silent information regulation2 (Sir2)-related sirtuins are nicotinamide-adenine dinucleotide (NAD(+))-dependent histone deacetylases (HDAC). The mammalian sirtuin family comprises 7 members (SIRT1-7) that act in different cellular compartments to regulate metabolism and aging. The rice genome contains only two Sir2-related genes: OsSRT1 (or SRT701) and OsSRT2 (orSRT702). OsSRT1 is closely related to the mammalian SIRT6, while OsSRT2 is homologous to SIRT4. Previous work has shown that OsSRT1 is required for the safeguard against genome instability and cell damage in rice plant. In this work we investigated the role of OsSRT1 on genome-wide acetylation of histone H3 lysine 9 (H3K9ac) and studied the genome-wide binding targets of OsSRT1. The study reveals that OsSRT1 binds to loci with relatively low levels of H3K9ac and directly regulates H3K9ac and expression of many genes that are related to stress and metabolism, indicating that OsSRT1 is an important site-specific histone deacetylase for gene regulation in rice. In addition, OsSRT1 is found to also target to several families of transposable elements, suggesting that OsSRT1 is directly involved in transposable element repression.


Subject(s)
DNA Transposable Elements/genetics , Genes, Plant/genetics , Group III Histone Deacetylases/metabolism , Oryza/enzymology , Oryza/genetics , Stress, Physiological/genetics , Acetylation , Base Sequence , DNA, Plant/genetics , DNA, Plant/metabolism , Gene Expression Regulation, Plant , Group III Histone Deacetylases/deficiency , Group III Histone Deacetylases/genetics , Histones/metabolism , Oryza/metabolism , Oryza/physiology , RNA Interference , Substrate Specificity
18.
J Biol Chem ; 288(16): 11004-12, 2013 Apr 19.
Article in English | MEDLINE | ID: mdl-23463510

ABSTRACT

In a mouse model of skin repair we found that the class I-IIa histone deacetylase inhibitor trichostatin A accelerated tissue regeneration. Unexpectedly, this effect was suppressed by Sirtinol, a class III histone deacetylase (HDAC) (sirtuin)-selective inhibitor. The role of sirtuins (SIRTs) was then investigated by using resveratrol and a novel SIRT1-2-3 activator, the MC2562 compound we synthesized recently. Both resveratrol and MC2562 were effective in accelerating wound repair. The local administration of natural or synthetic SIRT activators, in fact, significantly accelerated skin regeneration by increasing keratinocyte proliferation. In vitro experiments revealed that the activation of SIRTs stimulated keratinocyte proliferation via endothelial NO synthase phosphorylation and NO production. In this condition, the class I member HDAC2 was found S-nitrosylated on cysteine, a post-transduction modification associated with loss of activity and DNA binding capacity. After deacetylase inhibitor or SIRT activator treatment, ChIP showed, in fact, a significant HDAC2 detachment from the promoter region of insulin growth factor I (IGF-I), fibroblast growth factor 10 (FGF-10), and Epithelial Growth Factor (EGF), which may be the final recipients and effectors of the SIRT-NO-HDAC signaling cascade. Consistently, the effect of SIRT activators was reduced in the presence of NG-nitro-L-arginine methyl ester (L-NAME), a general inhibitor of NO synthesis. In conclusion, the NO-dependent cross-talk among class III and I histone deacetylases suggests an unprecedented signaling pathway important for skin repair.


Subject(s)
Group III Histone Deacetylases/metabolism , Histone Deacetylase 2/metabolism , Nitric Oxide/metabolism , Skin/enzymology , Skin/injuries , Wound Healing/physiology , Animals , Cell Line, Transformed , Enzyme Activators/pharmacology , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 10/metabolism , Group III Histone Deacetylases/antagonists & inhibitors , Humans , Insulin-Like Growth Factor I/metabolism , Male , Mice , NG-Nitroarginine Methyl Ester/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Wound Healing/drug effects
19.
Org Lett ; 14(21): 5456-9, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23083076

ABSTRACT

Cultivation of Chaetomium mollipilium with nicotinamide, a NAD(+)-dependent HDAC inhibitor, stimulated its secondary metabolism, leading to the isolation of structurally diverse new C(13)-polyketides, mollipilin A-E (1-5) as well as two known compounds (6 and 7). Spectroscopic methods, X-ray single crystal diffraction analysis, and VCD elucidated the absolute configurations of structures 1-6, and plausible biosynthetic pathways for 1-7 were proposed based on structural relationships. Mollipilins A (1) and B (2) exhibited moderate growth inhibitory effects on HCT-116 cells.


Subject(s)
Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Chaetomium/chemistry , Group III Histone Deacetylases/metabolism , Histone Deacetylase Inhibitors/pharmacology , Polyketides/isolation & purification , Polyketides/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Crystallography, X-Ray , HCT116 Cells , Histone Deacetylase Inhibitors/chemistry , Humans , Molecular Structure , NAD/metabolism , Niacinamide/metabolism , Nuclear Magnetic Resonance, Biomolecular , Polyketides/chemistry
20.
Neurodegener Dis ; 9(1): 1-10, 2012.
Article in English | MEDLINE | ID: mdl-22041967

ABSTRACT

Sirtuins, commonly known as NAD(+)-dependent class III histone deacetylase enzymes, have been extensively studied to evaluate their potential role in different disease states. Based on the published literature, sirtuins have been implicated in providing a myriad of intrinsic and extrinsic biological effects, which in turn may play an important role in the treatment of various disorders such as type II diabetes, obesity, cancer, aging and different neurodegenerative diseases. In particular, a number of studies have unequivocally supported the idea of sirtuins having therapeutic potential in neurodegenerative diseases such as stroke, ischemic brain injury, Alzheimer's disease, Parkinson's disease, Huntington's disease and amyotrophic lateral sclerosis. To exploit the therapeutic potential of sirtuins, their manipulation in terms of development of small-molecule modulators, inhibitors and analogs has increased dramatically since their inception, in both scientific and industrial worlds. Studies on the structure and catalytic core of sirtuins along with chemical mechanisms and substrate specificity have provided important input into the design and synthesis of sirtuin modulators. To study the role of sirtuins in the biological system, it has become extremely important to understand the molecular and chemical structure of sirtuins. In this review, we have discussed the biological role of sirtuins in various neurodegenerative diseases, and also provided an insight into their chemical structure.


Subject(s)
Neurodegenerative Diseases/drug therapy , Sirtuins , Aging , Alzheimer Disease/drug therapy , Amyotrophic Lateral Sclerosis/drug therapy , Brain Ischemia/drug therapy , Group III Histone Deacetylases/chemistry , Group III Histone Deacetylases/metabolism , Group III Histone Deacetylases/therapeutic use , Humans , Parkinson Disease/drug therapy , Protein Structure, Tertiary , Sirtuins/chemistry , Sirtuins/metabolism , Sirtuins/therapeutic use , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL