Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Gerontol A Biol Sci Med Sci ; 75(11): 2037-2041, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32556267

ABSTRACT

SRT1720, a sirtuin1-activator, and metformin (MET), an antidiabetic drug, confer health and life-span benefits when administered individually. It is unclear whether combination of the two compounds could lead to additional benefits. Groups of 56-week-old C57BL/6J male mice were fed a high-fat diet (HFD) alone or supplemented with either SRT1720 (2 g/kg food), a high dose of MET (1% wt/wt food), or a combination of both. Animals were monitored for survival, body weight, food consumption, body composition, and rotarod performance. Mice treated with MET alone did not have improved longevity, and life span was dramatically reduced by combination of MET with SRT1720. Although all groups of animals were consuming similar amounts of food, mice on MET or MET + SRT1720 showed a sharp reduction in body weight. SRT1720 + MET mice also had lower percent body fat combined with better performance on the rotarod compared to controls. These data suggest that co-treatment of SRT1720 with MET is detrimental to survival at the doses used and, therefore, risk-benefits of combining life-span-extending drugs especially in older populations needs to be systematically evaluated.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/pharmacology , Longevity/drug effects , Metformin/pharmacology , Animals , Body Composition , Body Weight , Diet, High-Fat , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Male , Metformin/administration & dosage , Mice , Mice, Inbred C57BL , Models, Animal , Sirtuin 1
2.
Aging Cell ; 13(5): 787-96, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24931715

ABSTRACT

Increased expression of SIRT1 extends the lifespan of lower organisms and delays the onset of age-related diseases in mammals. Here, we show that SRT2104, a synthetic small molecule activator of SIRT1, extends both mean and maximal lifespan of mice fed a standard diet. This is accompanied by improvements in health, including enhanced motor coordination, performance, bone mineral density, and insulin sensitivity associated with higher mitochondrial content and decreased inflammation. Short-term SRT2104 treatment preserves bone and muscle mass in an experimental model of atrophy. These results demonstrate it is possible to design a small molecule that can slow aging and delay multiple age-related diseases in mammals, supporting the therapeutic potential of SIRT1 activators in humans.


Subject(s)
Bone and Bones/drug effects , Heterocyclic Compounds, 2-Ring/pharmacology , Aging , Animals , Body Composition , Body Mass Index , Bone and Bones/metabolism , Diet , Humans , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred C57BL , Survival Analysis
3.
Cell Metab ; 17(3): 448-455, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23473037

ABSTRACT

Type 1 diabetes is caused by autoimmune-mediated ß cell destruction leading to insulin deficiency. The histone deacetylase SIRT1 plays an essential role in modulating several age-related diseases. Here we describe a family carrying a mutation in the SIRT1 gene, in which all five affected members developed an autoimmune disorder: four developed type 1 diabetes, and one developed ulcerative colitis. Initially, a 26-year-old man was diagnosed with the typical features of type 1 diabetes, including lean body mass, autoantibodies, T cell reactivity to ß cell antigens, and a rapid dependence on insulin. Direct and exome sequencing identified the presence of a T-to-C exchange in exon 1 of SIRT1, corresponding to a leucine-to-proline mutation at residue 107. Expression of SIRT1-L107P in insulin-producing cells resulted in overproduction of nitric oxide, cytokines, and chemokines. These observations identify a role for SIRT1 in human autoimmunity and unveil a monogenic form of type 1 diabetes.


Subject(s)
Autoimmunity/genetics , Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease/genetics , Sirtuin 1/genetics , Analysis of Variance , Base Sequence , Chemokines/metabolism , Cytokines/metabolism , Humans , Immunoprecipitation , Male , Molecular Sequence Data , Mutagenesis , Mutation, Missense/genetics , Nitric Oxide/metabolism , Pedigree , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Switzerland
4.
Br J Clin Pharmacol ; 75(1): 186-96, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22616762

ABSTRACT

AIM: SRT2104 is a novel, first-in-class, highly selective small molecule activator of the NAD + dependent deacetylase SIRT1. SRT2104 was dosed to healthy male and female volunteers in a series of phase 1 clinical studies that were designed to elucidate tolerability and pharmacokinetics associated with oral dosing to aid in dose selection for subsequent clinical trials. METHODS: In the first-in-human study, there was both a single dose phase and 7 day repeat dose phase. Doses used ranged from 0.03 to 3.0 g. A radioactive microtracer study was subsequently conducted to determine systemic clearance, bioavailability and preliminary metabolism, and a crossover study was conducted to determine the effect of gender, formulation and feeding state on SRT2104 pharmacokinetics. RESULTS: SRT2104 was well tolerated in all of these studies, with no serious adverse reactions observed. SRT2104 displayed a dose-dependent, but sub-proportional increase in exposure following single dose and repeated dose administration. Accumulation of three-fold or less occurs after 7 days of repeat dosing. The mean bioavailability was circa 14% and the mean clearance was circa 400 ml min(-1). Although there were no substantial effects on exposure resulting from gender or formulation differences, a notable food effect was observed, manifested as up to four-fold increase in exposure parameters. CONCLUSIONS: In the absence of an optimized formulation of SRT2104, the food effect can be used to maximize exposure in future clinical studies. Combined with the good tolerability of all doses demonstrated in these studies, the favourable selectivity profile of SRT2104 allows for the use of this SIRT1 modulator for target validation in the clinic.


Subject(s)
Imidazoles/pharmacokinetics , Sirtuin 1/drug effects , Thiazoles/pharmacokinetics , Biological Availability , Dose-Response Relationship, Drug , Double-Blind Method , Enzyme Activation , Female , Humans , Imidazoles/administration & dosage , Imidazoles/adverse effects , Male , Thiazoles/administration & dosage , Thiazoles/adverse effects
5.
Nature ; 473(7346): 234-8, 2011 05 12.
Article in English | MEDLINE | ID: mdl-21499261

ABSTRACT

Notch signalling is a key intercellular communication mechanism that is essential for cell specification and tissue patterning, and which coordinates critical steps of blood vessel growth. Although subtle alterations in Notch activity suffice to elicit profound differences in endothelial behaviour and blood vessel formation, little is known about the regulation and adaptation of endothelial Notch responses. Here we report that the NAD(+)-dependent deacetylase SIRT1 acts as an intrinsic negative modulator of Notch signalling in endothelial cells. We show that acetylation of the Notch1 intracellular domain (NICD) on conserved lysines controls the amplitude and duration of Notch responses by altering NICD protein turnover. SIRT1 associates with NICD and functions as a NICD deacetylase, which opposes the acetylation-induced NICD stabilization. Consequently, endothelial cells lacking SIRT1 activity are sensitized to Notch signalling, resulting in impaired growth, sprout elongation and enhanced Notch target gene expression in response to DLL4 stimulation, thereby promoting a non-sprouting, stalk-cell-like phenotype. In vivo, inactivation of Sirt1 in zebrafish and mice causes reduced vascular branching and density as a consequence of enhanced Notch signalling. Our findings identify reversible acetylation of the NICD as a molecular mechanism to adapt the dynamics of Notch signalling, and indicate that SIRT1 acts as rheostat to fine-tune endothelial Notch responses.


Subject(s)
Endothelial Cells/enzymology , Gene Expression Regulation , Receptors, Notch/metabolism , Signal Transduction/physiology , Sirtuin 1/genetics , Sirtuin 1/metabolism , Acetylation , Animals , Endothelial Cells/cytology , Gene Knockout Techniques , Gene Silencing , HEK293 Cells , Humans , Mice , Mutation , Receptor, Notch1/metabolism , Zebrafish/embryology , Zebrafish/genetics
6.
Cardiovasc Res ; 89(2): 464-72, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-20978007

ABSTRACT

AIMS: The mammalian silent information regulator-two 1 (Sirt1) blunts the noxious effects of cardiovascular risk factors such as type 2 diabetes mellitus and obesity. Nevertheless, the role of Sirt1 in regulating the expression of tissue factor (TF), the key trigger of coagulation, and arterial thrombus formation remains unknown. METHODS AND RESULTS: Human as well as mouse cell lines were used for in vitro experiments, and C57Bl/6 mice for in vivo procedures. Sirt1 inhibition by splitomicin or sirtinol enhanced cytokine-induced endothelial TF protein expression as well as surface activity, while TF pathway inhibitor protein expression did not change. Sirt1 inhibition further enhanced TF mRNA expression, TF promoter activity, and nuclear translocation as well as DNA binding of the p65 subunit of nuclear factor-kappa B (NFκB/p65). Sirt1 siRNA enhanced TF protein and mRNA expression, and this effect was reduced in NFκB/p65(-/-) mouse embryonic fibroblasts reconstituted with non-acetylatable Lys(310)-mutant NFκB/p65. Activation of the mitogen-activated protein kinases p38, c-Jun NH(2)-terminal kinase, and p44/42 (ERK) remained unaffected. In vivo, mice treated with the Sirt1 inhibitor splitomicin exhibited enhanced TF activity in the arterial vessel wall and accelerated carotid artery thrombus formation in a photochemical injury model. CONCLUSION: We provide pharmacological and genetic evidence that Sirt1 inhibition enhances TF expression and activity by increasing NFκB/p65 activation in human endothelial cells. Furthermore, Sirt1 inhibition induces arterial thrombus formation in vivo. Hence, modulation of Sirt1 may offer novel therapeutic options for targeting thrombosis.


Subject(s)
Endothelial Cells/drug effects , Histone Deacetylase Inhibitors/pharmacology , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/metabolism , Thromboplastin/metabolism , Thrombosis/etiology , Animals , Benzamides/pharmacology , Binding Sites , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/enzymology , Enzyme Activators , Genes, Reporter , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Naphthalenes/pharmacology , Naphthols/pharmacology , Promoter Regions, Genetic , Pyrones/pharmacology , RNA Interference , RNA, Messenger/metabolism , Resveratrol , Sirtuin 1/deficiency , Sirtuin 1/genetics , Stilbenes/pharmacology , Thromboplastin/genetics , Thrombosis/blood , Thrombosis/enzymology , Thrombosis/genetics , Transcription Factor RelA/deficiency , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Transfection
7.
Sci Rep ; 1: 70, 2011.
Article in English | MEDLINE | ID: mdl-22355589

ABSTRACT

Sirt1 is an NAD(+)-dependent deacetylase that extends lifespan in lower organisms and improves metabolism and delays the onset of age-related diseases in mammals. Here we show that SRT1720, a synthetic compound that was identified for its ability to activate Sirt1 in vitro, extends both mean and maximum lifespan of adult mice fed a high-fat diet. This lifespan extension is accompanied by health benefits including reduced liver steatosis, increased insulin sensitivity, enhanced locomotor activity and normalization of gene expression profiles and markers of inflammation and apoptosis, all in the absence of any observable toxicity. Using a conditional SIRT1 knockout mouse and specific gene knockdowns we show SRT1720 affects mitochondrial respiration in a Sirt1- and PGC-1α-dependent manner. These findings indicate that SRT1720 has long-term benefits and demonstrate for the first time the feasibility of designing novel molecules that are safe and effective in promoting longevity and preventing multiple age-related diseases in mammals.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/pharmacology , Longevity/drug effects , Obesity/physiopathology , Animals , Apoptosis/drug effects , Body Composition/drug effects , Dietary Fats/administration & dosage , Gene Expression/drug effects , Glucose/metabolism , Homeostasis/drug effects , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Pancreas/drug effects
8.
Genes Dev ; 24(13): 1403-17, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20595232

ABSTRACT

The sterol regulatory element-binding protein (SREBP) transcription factor family is a critical regulator of lipid and sterol homeostasis in eukaryotes. In mammals, SREBPs are highly active in the fed state to promote the expression of lipogenic and cholesterogenic genes and facilitate fat storage. During fasting, SREBP-dependent lipid/cholesterol synthesis is rapidly diminished in the mouse liver; however, the mechanism has remained incompletely understood. Moreover, the evolutionary conservation of fasting regulation of SREBP-dependent programs of gene expression and control of lipid homeostasis has been unclear. We demonstrate here a conserved role for orthologs of the NAD(+)-dependent deacetylase SIRT1 in metazoans in down-regulation of SREBP orthologs during fasting, resulting in inhibition of lipid synthesis and fat storage. Our data reveal that SIRT1 can directly deacetylate SREBP, and modulation of SIRT1 activity results in changes in SREBP ubiquitination, protein stability, and target gene expression. In addition, chemical activators of SIRT1 inhibit SREBP target gene expression in vitro and in vivo, correlating with decreased hepatic lipid and cholesterol levels and attenuated liver steatosis in diet-induced and genetically obese mice. We conclude that SIRT1 orthologs play a critical role in controlling SREBP-dependent gene regulation governing lipid/cholesterol homeostasis in metazoans in response to fasting cues. These findings may have important biomedical implications for the treatment of metabolic disorders associated with aberrant lipid/cholesterol homeostasis, including metabolic syndrome and atherosclerosis.


Subject(s)
Down-Regulation , Fasting/physiology , Sirtuin 1/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Acetylation , Animals , Benzamides/pharmacology , Caenorhabditis elegans , Cell Line , Cholesterol/biosynthesis , Down-Regulation/drug effects , HeLa Cells , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Lipids/biosynthesis , Mice , Naphthols/pharmacology , Niacinamide/pharmacology , Protein Stability/drug effects , Sirtuins/antagonists & inhibitors
9.
Diabetes ; 59(4): 829-35, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20107110

ABSTRACT

OBJECTIVE: Sirtuin 1 (SIRT1) is implicated in the regulation of mitochondrial function, energy metabolism, and insulin sensitivity in rodents. No studies are available in humans to demonstrate that SIRT1 expression in insulin-sensitive tissues is associated with energy expenditure and insulin sensitivity. RESEARCH DESIGN AND METHODS: Energy expenditure (EE), insulin sensitivity, and SIRT1 mRNA adipose tissue expression (n = 81) were measured by indirect calorimetry, hyperinsulinemic-euglycemic clamp, and quantitative RT-PCR in 247 nondiabetic offspring of type 2 diabetic patients. RESULTS: High EE during the clamp (r = 0.375, P = 2.8 x 10(-9)) and high DeltaEE (EE during the clamp - EE in the fasting state) (r = 0.602, P = 2.5 x 10(-24)) were associated with high insulin sensitivity. Adipose tissue SIRT1 mRNA expression was significantly associated with EE (r = 0.289, P = 0.010) and with insulin sensitivity (r = 0.334, P = 0.002) during hyperinsulinemic-euglycemic clamp. Furthermore, SIRT1 mRNA expression correlated significantly with the expression of several genes regulating mitochondrial function and energy metabolism (e.g., peroxisome proliferator-activated receptor gamma coactivator-1beta, estrogen-related receptor alpha, nuclear respiratory factor-1, and mitochondrial transcription factor A), and with several genes of the respiratory chain (e.g., including NADH dehydrogenase [ubiquinone] 1alpha subcomplex 2, cytochrome c, cytochrome c oxidase subunit IV, and ATP synthase). CONCLUSIONS: Impaired stimulation of EE by insulin and low SIRT1 expression in insulin-sensitive tissues is likely to reflect impaired regulation of mitochondrial function associated with insulin resistance in humans.


Subject(s)
Diabetes Mellitus, Type 2/genetics , RNA, Messenger/genetics , Sirtuin 1/genetics , Adult , Animals , Body Mass Index , Crosses, Genetic , Diabetes Mellitus, Type 2/physiopathology , Energy Metabolism/genetics , Female , Gene Expression Regulation , Glucose Intolerance/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Muscle, Skeletal/physiology , Muscle, Skeletal/physiopathology , Reference Values
10.
Am J Physiol Endocrinol Metab ; 298(3): E419-28, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19996381

ABSTRACT

Chronic inflammation is an important etiology underlying obesity-related disorders such as insulin resistance and type 2 diabetes, and recent findings indicate that the macrophage can be the initiating cell type responsible for this chronic inflammatory state. The mammalian silent information regulator 2 homolog SIRT1 modulates several physiological processes important for life span, and a potential role of SIRT1 in the regulation of insulin sensitivity has been shown. However, with respect to inflammation, the role of SIRT1 in regulating the proinflammatory pathway within macrophages is poorly understood. Here, we show that knockdown of SIRT1 in the mouse macrophage RAW264.7 cell line and in intraperitoneal macrophages broadly activates the JNK and IKK inflammatory pathways and increases LPS-stimulated TNFalpha secretion. Moreover, gene expression profiles reveal that SIRT1 knockdown leads to an increase in inflammatory gene expression. We also demonstrate that SIRT1 activators inhibit LPS-stimulated inflammatory pathways, as well as secretion of TNFalpha, in a SIRT1-dependent manner in RAW264.7 cells and in primary intraperitoneal macrophages. Treatment of Zucker fatty rats with a SIRT1 activator leads to greatly improved glucose tolerance, reduced hyperinsulinemia, and enhanced systemic insulin sensitivity during glucose clamp studies. These in vivo insulin-sensitizing effects were accompanied by a reduction in tissue inflammation markers and a decrease in the adipose tissue macrophage proinflammatory state, fully consistent with the in vitro effects of SIRT1 in macrophages. In conclusion, these results define a novel role for SIRT1 as an important regulator of macrophage inflammatory responses in the context of insulin resistance and raise the possibility that targeting of SIRT1 might be a useful strategy for treating the inflammatory component of metabolic diseases.


Subject(s)
Inflammation/metabolism , Insulin Resistance/genetics , Insulin/metabolism , Macrophage Activation/genetics , Macrophages/metabolism , Sirtuin 1/metabolism , Animals , Cells, Cultured , Gene Expression , Male , Mice , Rats , Rats, Zucker , Signal Transduction
11.
J Biol Chem ; 284(36): 24394-405, 2009 Sep 04.
Article in English | MEDLINE | ID: mdl-19535340

ABSTRACT

SIRT3 is a major mitochondrial NAD(+)-dependent protein deacetylase playing important roles in regulating mitochondrial metabolism and energy production and has been linked to the beneficial effects of exercise and caloric restriction. SIRT3 is emerging as a potential therapeutic target to treat metabolic and neurological diseases. We report the first sets of crystal structures of human SIRT3, an apo-structure with no substrate, a structure with a peptide containing acetyl lysine of its natural substrate acetyl-CoA synthetase 2, a reaction intermediate structure trapped by a thioacetyl peptide, and a structure with the dethioacetylated peptide bound. These structures provide insights into the conformational changes induced by the two substrates required for the reaction, the acetylated substrate peptide and NAD(+). In addition, the binding study by isothermal titration calorimetry suggests that the acetylated peptide is the first substrate to bind to SIRT3, before NAD(+). These structures and biophysical studies provide key insight into the structural and functional relationship of the SIRT3 deacetylation activity.


Subject(s)
Acetate-CoA Ligase/chemistry , Mitochondrial Proteins/chemistry , NAD/chemistry , Peptides/chemistry , Sirtuins/chemistry , Acetate-CoA Ligase/metabolism , Acetylation , Humans , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Peptides/metabolism , Protein Binding/physiology , Protein Structure, Quaternary , Sirtuin 3 , Sirtuins/metabolism , Structure-Activity Relationship
12.
Protein Sci ; 18(3): 514-25, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19241369

ABSTRACT

SIRT3 is a key mitochondrial protein deacetylase proposed to play key roles in regulating mitochondrial metabolism but there has been considerable debate about its actual size, the sequences required for activity, and its subcellular localization. A previously cloned mouse SIRT3 has high sequence similarity with the C-terminus of human SIRT3 but lacks an N-terminal mitochondrial targeting sequence and has no detectable deacetylation activity in vitro. Using 5' rapid amplification of cDNA ends, we cloned the entire sequence of mouse SIRT3, as well as rat and rabbit SIRT3. Importantly, we find that full-length SIRT3 protein localizes exclusively to the mitochondria, in contrast to reports of SIRT3 localization to the nucleus. We demonstrate that SIRT3 has no deacetylation activity in vitro unless the protein is truncated, consistent with human SIRT3. In addition, we determined the inhibition constants and mechanism of action for nicotinamide and a small molecule SIRT3 inhibitor against active mouse SIRT3 and show that the mechanisms are different for the two compounds with respect to peptide substrate and NAD(+). Thus, identification and characterization of the actual SIRT3 sequence should help resolve the debate about the nature of mouse SIRT3 and identify new mechanisms to modulate enzymatic activity.


Subject(s)
Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Protein Sorting Signals , Sirtuins/genetics , Sirtuins/metabolism , Tissue Distribution/genetics , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Cloning, Molecular , Heterocyclic Compounds, 4 or More Rings/metabolism , Mice , Mitochondria/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/chemistry , Molecular Sequence Data , Niacinamide/metabolism , Rabbits , Rats , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sirtuin 3 , Sirtuins/antagonists & inhibitors , Sirtuins/chemistry
13.
Nature ; 450(7170): 712-6, 2007 Nov 29.
Article in English | MEDLINE | ID: mdl-18046409

ABSTRACT

Calorie restriction extends lifespan and produces a metabolic profile desirable for treating diseases of ageing such as type 2 diabetes. SIRT1, an NAD+-dependent deacetylase, is a principal modulator of pathways downstream of calorie restriction that produce beneficial effects on glucose homeostasis and insulin sensitivity. Resveratrol, a polyphenolic SIRT1 activator, mimics the anti-ageing effects of calorie restriction in lower organisms and in mice fed a high-fat diet ameliorates insulin resistance, increases mitochondrial content, and prolongs survival. Here we describe the identification and characterization of small molecule activators of SIRT1 that are structurally unrelated to, and 1,000-fold more potent than, resveratrol. These compounds bind to the SIRT1 enzyme-peptide substrate complex at an allosteric site amino-terminal to the catalytic domain and lower the Michaelis constant for acetylated substrates. In diet-induced obese and genetically obese mice, these compounds improve insulin sensitivity, lower plasma glucose, and increase mitochondrial capacity. In Zucker fa/fa rats, hyperinsulinaemic-euglycaemic clamp studies demonstrate that SIRT1 activators improve whole-body glucose homeostasis and insulin sensitivity in adipose tissue, skeletal muscle and liver. Thus, SIRT1 activation is a promising new therapeutic approach for treating diseases of ageing such as type 2 diabetes.


Subject(s)
Caloric Restriction , Diabetes Mellitus, Type 2/drug therapy , Sirtuins/agonists , Acetylation , Allosteric Site , Animals , Blood Glucose/metabolism , Catalytic Domain , Cell Line , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Disease Models, Animal , Drosophila melanogaster , Heterocyclic Compounds, 4 or More Rings/pharmacology , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Humans , Insulin/metabolism , Insulin/pharmacology , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Rats , Rats, Sprague-Dawley , Rats, Zucker , Resveratrol , Sirtuin 1 , Sirtuins/metabolism , Stilbenes/chemistry , Stilbenes/pharmacology
15.
J Prosthet Dent ; 94(4): 342-9, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16198171

ABSTRACT

STATEMENT OF PROBLEM: Endodontically treated posterior teeth are more likely to fracture compared to posterior teeth with vital pulps. Reinforcement with an extracoronal restoration that covers the cusps is the most commonly recommended method for reducing the risk of fracture. It is not known whether bonded intracoronal restorations without cuspal coverage will reduce the risk of fracture. PURPOSE: The aim of this in vitro study was to investigate whether reinforcement of endodontically treated premolars with MOD preparations could be achieved by insertion of bonded CAD/CAM ceramic inlays. MATERIAL AND METHODS: Forty-five extracted maxillary premolars were equally distributed among 3 groups (END, CER, CTR). In group END (n=15), root canals were enlarged with a rotary NiTi system and obturated with heat-softened gutta-percha around a plastic carrier (Thermafil). After filling of the endodontic access cavities with autopolymerizing composite resin (Luxacore), standardized MOD cavity preparations were made and CAD/CAM ceramic inlays (CEREC) were fabricated and then bonded to the teeth with composite resin (Tetric) and an adhesive system (Syntac Classic). In group CER (n=15), teeth without endodontic treatment were restored with bonded inlays (CEREC). Sound premolars served as controls (group CTR, n=15). Teeth were then thermal cycled (1445 cycles, dwell time: 30 seconds, 5 degrees /55 degrees C). An eccentric load was applied on the buccal incline of the palatal cusp in a universal testing machine until cusp fracture (N). Fracture load was evaluated with the Mann-Whitney test, and type of fracture, with a chi-square analysis (alpha=.05). The type of fracture was determined by visual inspection: type I - supragingival fracture within the palatal cusp; type II - fracture below cemento-enamel junction of palatal cusp; and type III - fracture of palatal cusp and central portion of the tooth exposing the root canal cavity. RESULTS: No significant difference was found among the 3 groups with respect to load required for fracture. Mean fracture load +/- SD was recorded as follows: 291.6 +/- 113.7 N for group END, 363.2 +/- 140.3 N for group CER, and 296.5 +/- 170.5 N for group CTR. Regarding fracture modes, significantly more teeth from group END exhibited fractures of type III and II compared with control specimens. CONCLUSION: Teeth restored with bonded CAD/CAM ceramic inlays (CEREC) fractured with a significantly higher number of severe fractures compared to the control group.


Subject(s)
Bicuspid/physiopathology , Ceramics/chemistry , Computer-Aided Design , Dental Porcelain/chemistry , Inlays , Root Canal Therapy , Tooth Fractures/physiopathology , Tooth, Nonvital/physiopathology , Composite Resins/chemistry , Dental Bonding , Dental Cavity Preparation/methods , Dental Enamel/injuries , Dental Stress Analysis , Dentin-Bonding Agents/chemistry , Gutta-Percha/chemistry , Humans , Resin Cements/chemistry , Root Canal Preparation/instrumentation , Stress, Mechanical , Tooth Cervix/injuries , Tooth Crown/injuries
16.
Biol Reprod ; 70(4): 1206-12, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14681204

ABSTRACT

Ionizing radiation (IR) and consequent induction of DNA double-strand breaks (DSBs) causes activation of the protein ataxia telangiectasia mutated (ATM). Normally, ATM is present as inactive dimers; however, in response to DSBs, the ATM dimer partners cross-phosphorylate each other on serine 1981, and kinase active ATM monomers are subsequently released. We have studied the presence of both nonphosphorylated as well as active serine 1981 phosphorylated ATM (pS1981-ATM) in the mouse testis. In the nonirradiated testis, ATM was present in spermatogonia and spermatocytes until stage VII of the cycle of the seminiferous epithelium, whereas pS1981-ATM was found only to be present in the sex body of pachytene spermatocytes. In response to IR, ATM became activated by pS1981 cross-phosphorylation in spermatogonia and Sertoli cells. Despite the occurrence of endogenous programmed DSBs during the first meiotic prophase and the presence of ATM in both spermatogonia and spermatocytes, pS1981 phosphorylated ATM did not appear in spermatocytes after treatment with IR. These results show that spermatogonial ATM and ATM in the spermatocytes are differentially regulated. In the mitotically dividing spermatogonia, ATM is activated by cross-phosphorylation, whereas during meiosis nonphosphorylated ATM or differently phosphorylated ATM is already active. ATM has been shown to be present at the synapsed axes of the meiotic chromosomes, and in the ATM knock-out mice spermatogenesis stops at pachytene stage IV of the seminiferous epithelium, indicating that indeed nonphosphorylated ATM is functional during meiosis. Additionally, ATM is constitutively phosphorylated in the sex body where its continued presence remains an enigma.


Subject(s)
Protein Serine-Threonine Kinases/physiology , Testis/metabolism , Animals , Apoptosis , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Male , Meiosis , Mice , Mice, Inbred Strains , Mice, Knockout , Phosphorylation , Prophase , Protein Serine-Threonine Kinases/metabolism , Spermatocytes/physiology , Spermatogonia/physiology , Testis/radiation effects , Tissue Distribution , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...