Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732160

ABSTRACT

Despite the end of the pandemic, coronavirus disease 2019 (COVID-19) remains a major public health concern. The first waves of the virus led to a better understanding of its pathogenesis, highlighting the fact that there is a specific pulmonary vascular disorder. Indeed, COVID-19 may predispose patients to thrombotic disease in both venous and arterial circulation, and many cases of severe acute pulmonary embolism have been reported. The demonstrated presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) within the endothelial cells suggests that direct viral effects, in addition to indirect effects of perivascular inflammation and coagulopathy, may contribute to pulmonary vasculopathy in COVID-19. In this review, we discuss the pathological mechanisms leading to pulmonary vascular damage during acute infection, which appear to be mainly related to thromboembolic events, an impaired coagulation cascade, micro- and macrovascular thrombosis, endotheliitis and hypoxic pulmonary vasoconstriction. As many patients develop post-COVID symptoms, including dyspnea, we also discuss the hypothesis of pulmonary vascular damage and pulmonary hypertension as a sequela of the infection, which may be involved in the pathophysiology of long COVID.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , COVID-19/complications , COVID-19/virology , COVID-19/pathology , SARS-CoV-2/pathogenicity , Lung/blood supply , Lung/pathology , Lung/virology , Pulmonary Embolism/virology , Pulmonary Embolism/etiology , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/virology , Hypertension, Pulmonary/pathology , Post-Acute COVID-19 Syndrome , Thrombosis/virology , Thrombosis/etiology , Thrombosis/pathology
2.
J Biol Chem ; 296: 100671, 2021.
Article in English | MEDLINE | ID: mdl-33864814

ABSTRACT

The SAGA-like complex SLIK is a modified version of the Spt-Ada-Gcn5-Acetyltransferase (SAGA) complex. SLIK is formed through C-terminal truncation of the Spt7 SAGA subunit, causing loss of Spt8, one of the subunits that interacts with the TATA-binding protein (TBP). SLIK and SAGA are both coactivators of RNA polymerase II transcription in yeast, and both SAGA and SLIK perform chromatin modifications. The two complexes have been speculated to uniquely contribute to transcriptional regulation, but their respective contributions are not clear. To investigate, we assayed the chromatin modifying functions of SAGA and SLIK, revealing identical kinetics on minimal substrates in vitro. We also examined the binding of SAGA and SLIK to TBP and concluded that interestingly, both protein complexes have similar affinity for TBP. Additionally, despite the loss of Spt8 and C-terminus of Spt7 in SLIK, TBP prebound to SLIK is not released in the presence of TATA-box DNA, just like TBP prebound to SAGA. Furthermore, we determined a low-resolution cryo-EM structure of SLIK, revealing a modular architecture identical to SAGA. Finally, we performed a comprehensive study of DNA-binding properties of both coactivators. Purified SAGA and SLIK both associate with ssDNA and dsDNA with high affinity (KD = 10-17 nM), and the binding is sequence-independent. In conclusion, our study shows that the cleavage of Spt7 and the absence of the Spt8 subunit in SLIK neither drive any major conformational differences in its structure compared with SAGA, nor significantly affect HAT, DUB, or DNA-binding activities in vitro.


Subject(s)
Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Protein Binding , Protein Conformation , Protein Subunits , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Trans-Activators/genetics
3.
Cardiovasc Drugs Ther ; 36(2): 245-256, 2022 04.
Article in English | MEDLINE | ID: mdl-33661433

ABSTRACT

PURPOSE: To evaluate the effectiveness of vitamin D3 supplementation, in secondary prevention, on cardiac remodeling and function, as well as lipid profile, in a mouse model of diet-induced type 2 diabetes. METHODS: Mice were fed a high fat and sucrose diet for 10 weeks. Afterward, diet was maintained for 15 more weeks and two groups were formed, with and without cholecalciferol supplementation. A control group was fed with normal chow. Glucose homeostasis and cardiac function were assessed at baseline and at the 10th and 24th weeks. Animals were killed at the 10th and 25th weeks for plasma and cardiac sample analysis. Cardiac lipid profile was characterized by LC-MS/MS. RESULTS: After 10 weeks of diet, mice exhibited pre-diabetes, mild left ventricle hypertrophy, and impaired longitudinal strain, but preserved myocardial circumferential as well as global diastolic and systolic cardiac function. After 15 more weeks of diet, animals presented with well-established type 2 diabetes, pathological cardiac hypertrophy, and impaired regional myocardial function. Cholecalciferol supplementation had no effect on glucose homeostasis but improved cardiac remodeling and regional myocardial function. After 25 weeks, non-supplemented mice exhibited increased myocardial levels of ceramides and diacylglycerol, both of which were normalized by vitamin D3 supplementation. CONCLUSION: This work brought to light the beneficial effects of cholecalciferol supplementation, in secondary prevention, on cardiac remodeling and function in a mouse model of diet-induced type 2 diabetes. Those cardioprotective effects may be, at least in part, attributed to the modulation of myocardial levels of lipotoxic species by vitamin D.


Subject(s)
Diabetes Mellitus, Type 2 , Ventricular Dysfunction, Left , Animals , Cholecalciferol/pharmacology , Chromatography, Liquid , Diabetes Mellitus, Type 2/drug therapy , Diet , Dietary Supplements , Disease Models, Animal , Glucose , Mice , Tandem Mass Spectrometry , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/prevention & control , Ventricular Remodeling
4.
Nano Lett ; 21(2): 1175-1183, 2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33416334

ABSTRACT

Active sites and catalytic activity of heterogeneous catalysts is determined by their surface atomic structures. However, probing the surface structure at an atomic resolution is difficult, especially for solution ensembles of catalytic nanocrystals, which consist of heterogeneous particles with irregular shapes and surfaces. Here, we constructed 3D maps of the coordination number (CN) and generalized CN (CN_) for individual surface atoms of sub-3 nm Pt nanocrystals. Our results reveal that the synthesized Pt nanocrystals are enclosed by islands of atoms with nonuniform shapes that lead to complex surface structures, including a high ratio of low-coordination surface atoms, reduced domain size of low-index facets, and various types of exposed high-index facets. 3D maps of CN_ are directly correlated to catalytic activities assigned to individual surface atoms with distinct local coordination structures, which explains the origin of high catalytic performance of small Pt nanocrystals in important reactions such as oxygen reduction reactions and CO electro-oxidation.

5.
Bioinformatics ; 36(7): 2237-2243, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31790146

ABSTRACT

MOTIVATION: No rigorous statistical tests for detecting point-group symmetry in three-dimensional (3D) charge density maps obtained by electron microscopy (EM) and related techniques have been developed. RESULTS: We propose a method for determining the point-group symmetry of 3D charge density maps obtained by EM and related techniques. Our ab initio algorithm does not depend on atomic coordinates but utilizes the density map directly. We validate the approach for a range of publicly available single-particle cryo-EM datasets. In straightforward cases, our method enables fully automated single-particle 3D reconstruction without having to input an arbitrarily selected point-group symmetry. When pseudo-symmetry is present, our method provides statistics quantifying the degree to which the 3D density agrees with the different point-groups tested. AVAILABILITY AND IMPLEMENTATION: The software is freely available at https://github.com/hael/SIMPLE3.0.


Subject(s)
Algorithms , Software , Cryoelectron Microscopy , Imaging, Three-Dimensional
6.
Basic Res Cardiol ; 116(1): 66, 2021 12 23.
Article in English | MEDLINE | ID: mdl-34940922

ABSTRACT

Endothelial nitric oxide synthase (eNOS) activation in the heart plays a key role in exercise-induced cardioprotection during ischemia-reperfusion, but the underlying mechanisms remain unknown. We hypothesized that the cardioprotective effect of exercise training could be explained by the re-localization of eNOS-dependent nitric oxide (NO)/S-nitrosylation signaling to mitochondria. By comparing exercised (5 days/week for 5 weeks) and sedentary Wistar rats, we found that exercise training increased eNOS level and activation by phosphorylation (at serine 1177) in mitochondria, but not in the cytosolic subfraction of cardiomyocytes. Using confocal microscopy, we confirmed that NO production in mitochondria was increased in response to H2O2 exposure in cardiomyocytes from exercised but not sedentary rats. Moreover, by S-nitrosoproteomic analysis, we identified several key S-nitrosylated proteins involved in mitochondrial function and cardioprotection. In agreement, we also observed that the increase in Ca2+ retention capacity by mitochondria isolated from the heart of exercised rats was abolished by exposure to the NOS inhibitor L-NAME or to the reducing agent ascorbate, known to denitrosylate proteins. Pre-incubation with ascorbate or L-NAME also increased mitochondrial reactive oxygen species production in cardiomyocytes from exercised but not from sedentary animals. We confirmed these results using isolated hearts perfused with L-NAME before ischemia-reperfusion. Altogether, these results strongly support the hypothesis that exercise training increases eNOS/NO/S-nitrosylation signaling in mitochondria, which might represent a key mechanism of exercise-induced cardioprotection.


Subject(s)
Hydrogen Peroxide , Protein S , Animals , Mitochondria , Myocytes, Cardiac , Nitric Oxide , Nitric Oxide Synthase Type III , Rats , Rats, Wistar
8.
Nitric Oxide ; 86: 63-67, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30836135

ABSTRACT

OBJECTIVE: This study evaluated in obese rats the effect of exercise training on eNOS expressed in perivascular adipose tissue (PVAT) and its consequences on vascular function. METHODS: Wistar rats were divided in 3 groups: control (standard diet), obese (high fat/high sucrose diet, HFS for 15 weeks), and exercised obese (HFS diet and exercise from week 6 to week 15, HFS-Ex) rats. The eNOS-adiponectin pathway and reactive oxygen species (ROS) were evaluated. Vascular reactivity was assessed on isolated aortic rings with or without PVAT and/or endothelium and exposed or not to the conditioned media of PVAT. RESULTS: Obesity reduced eNOS level and phosphorylation on its activation site in the PVAT and had no impact on the vascular wall. Exercise training was able to increase eNOS and P-eNOS both in the vascular wall and in the PVAT. Interestingly, this was associated with increased level of adiponectin in the PVAT and to lower ROS in the vascular wall. Finally, PVAT of HFS-Ex aorta has eNOS-dependent anticontractile effects on endothelium denuded aortic rings and has beneficial effects on the endothelium-dependent vasorelaxation to ACh. CONCLUSION: Exercise training in obese rats is able to impact PVAT eNOS with subsequent beneficial impact on vascular function.


Subject(s)
Adipose Tissue/metabolism , Endothelium, Vascular/metabolism , Nitric Oxide Synthase Type III/metabolism , Obesity/metabolism , Adiponectin/metabolism , Animals , Aorta/metabolism , Diet, Carbohydrate Loading/adverse effects , Diet, High-Fat/adverse effects , Enzyme Activation/physiology , Male , Nitric Oxide Synthase Type III/chemistry , Obesity/prevention & control , Phosphorylation/physiology , Physical Conditioning, Animal , Rats, Wistar , Reactive Oxygen Species/metabolism
9.
J Struct Biol ; 204(2): 172-181, 2018 11.
Article in English | MEDLINE | ID: mdl-30092280

ABSTRACT

Cryogenic electron microscopy (cryo-EM) and single-particle analysis enables determination of near-atomic resolution structures of biological molecules. However, large computational requirements limit throughput and rapid testing of new image processing tools. We developed PRIME, an algorithm part of the SIMPLE software suite, for determination of the relative 3D orientations of single-particle projection images. PRIME has primarily found use for generation of an initial ab initio 3D reconstruction. Here we show that the strategy behind PRIME, iterative estimation of per-particle orientation distributions with stochastic hill climbing, provides a competitive approach to near-atomic resolution single-particle 3D reconstruction. A number of mathematical techniques for accelerating the convergence rate are introduced, leading to a speedup of nearly two orders of magnitude. We benchmarked our developments on numerous publicly available data sets and conclude that near-atomic resolution ab initio 3D reconstructions can be obtained with SIMPLE in a matter of hours, using standard over-the-counter CPU workstations.


Subject(s)
Image Processing, Computer-Assisted/methods , Software , Algorithms , Cryoelectron Microscopy
10.
Am J Physiol Heart Circ Physiol ; 314(3): H497-H507, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29127233

ABSTRACT

Sympathetic hyperactivation, a common feature of obesity and metabolic syndrome, is a key trigger of hypertension. However, some obese subjects with autonomic imbalance present a dissociation between sympathetic activity-mediated vasoconstriction and increased blood pressure. Here, we aimed to determine in a rat model of metabolic syndrome whether the endothelium endothelial nitric oxide (NO) synthase (eNOS)-NO pathway contributes to counteract the vasopressor effect of the sympathetic system. Rats were fed a high-fat and high-sucrose (HFS) diet for 15 wk. Sympathovagal balance was evaluated by spectral analysis of heart rate variability and plasmatic catecholamine measurements. Blood pressure was measured in the presence or absence of N-nitro-l-arginine methyl ester (l-NAME) to inhibit the contribution of eNOS. Vascular reactivity was assessed on isolated aortic rings in response to α1-adrenergic agonist. The HFS diet increased sympathetic tone, which is characterized by a higher low on the high-frequency spectral power ratio and a higher plasmatic concentration of epinephrine. Despite this, no change in blood pressure was observed. Interestingly, HFS rats exhibited vascular hyporeactivity (-23.6%) to α1-adrenergic receptor stimulation that was abolished by endothelial removal or eNOS inhibition (l-NAME). In addition, eNOS phosphorylation (Ser1177) was increased in response to phenylephrine in HFS rats only. Accordingly, eNOS inhibition in vivo revealed higher blood pressure in HFS rats compared with control rats (147 vs. 126 mmHg for mean blood pressure, respectively). Restrain of adrenergic vasopressor action by endothelium eNOS is increased in HFS rats and contributes to maintained blood pressure in the physiological range. NEW & NOTEWORTHY Despite the fact that prohypertensive sympathetic nervous system activity is markedly increased in rats with early metabolic syndrome, they present with normal blood pressure. These observations appear to be explained by increased endothelial nitric oxide synthase response to adrenergic stimulation, which results in vascular hyporeactivity to α-adrenergic stimulation, and therefore blood pressure is preserved in the physiological range. Listen to this article's corresponding podcast at http://www.physiology.org/doi/10.1152/ajpheart.00217.2017 .


Subject(s)
Aorta/innervation , Arterial Pressure , Endothelium, Vascular/innervation , Metabolic Syndrome/physiopathology , Sympathetic Nervous System/physiopathology , Vasoconstriction , Animals , Aorta/metabolism , Diet, High-Fat , Dietary Sucrose , Disease Models, Animal , Endothelium, Vascular/metabolism , Epinephrine/blood , Heart Rate , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Norepinephrine/blood , Rats, Wistar , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction , Sympathetic Nervous System/metabolism
11.
PLoS Biol ; 13(2): e1002049, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25654333

ABSTRACT

Membrane attack complex/perforin-like (MACPF) proteins comprise the largest superfamily of pore-forming proteins, playing crucial roles in immunity and pathogenesis. Soluble monomers assemble into large transmembrane pores via conformational transitions that remain to be structurally and mechanistically characterised. Here we present an 11 Å resolution cryo-electron microscopy (cryo-EM) structure of the two-part, fungal toxin Pleurotolysin (Ply), together with crystal structures of both components (the lipid binding PlyA protein and the pore-forming MACPF component PlyB). These data reveal a 13-fold pore 80 Å in diameter and 100 Å in height, with each subunit comprised of a PlyB molecule atop a membrane bound dimer of PlyA. The resolution of the EM map, together with biophysical and computational experiments, allowed confident assignment of subdomains in a MACPF pore assembly. The major conformational changes in PlyB are a ∼70° opening of the bent and distorted central ß-sheet of the MACPF domain, accompanied by extrusion and refolding of two α-helical regions into transmembrane ß-hairpins (TMH1 and TMH2). We determined the structures of three different disulphide bond-trapped prepore intermediates. Analysis of these data by molecular modelling and flexible fitting allows us to generate a potential trajectory of ß-sheet unbending. The results suggest that MACPF conformational change is triggered through disruption of the interface between a conserved helix-turn-helix motif and the top of TMH2. Following their release we propose that the transmembrane regions assemble into ß-hairpins via top down zippering of backbone hydrogen bonds to form the membrane-inserted ß-barrel. The intermediate structures of the MACPF domain during refolding into the ß-barrel pore establish a structural paradigm for the transition from soluble monomer to pore, which may be conserved across the whole superfamily. The TMH2 region is critical for the release of both TMH clusters, suggesting why this region is targeted by endogenous inhibitors of MACPF function.


Subject(s)
Cell Membrane/chemistry , Complement Membrane Attack Complex/chemistry , Fungal Proteins/chemistry , Hemolysin Proteins/chemistry , Pleurotus/chemistry , Recombinant Fusion Proteins/chemistry , Animals , Complement Membrane Attack Complex/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Erythrocytes/chemistry , Erythrocytes/cytology , Escherichia coli/genetics , Escherichia coli/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Models, Molecular , Protein Binding , Protein Folding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sheep
12.
Arterioscler Thromb Vasc Biol ; 37(6): 1250-1260, 2017 06.
Article in English | MEDLINE | ID: mdl-28408372

ABSTRACT

OBJECTIVE: To assess vascular function during acute hyperglycemia induced by commercial sugar-sweetened beverage (SSB) consumption and its effect on underlying mechanisms of the nitric oxide pathway. APPROACH AND RESULTS: In a randomized, single-blind, crossover trial, 12 healthy male participants consumed 600 mL (20 oz.) of water or a commercial SSB across 2 visits. Endothelial and vascular smooth muscle functions were assessed in the microcirculation using laser speckle contrast imaging coupled with iontophoresis and in the macrocirculation using brachial artery ultrasound with flow- and nitrate-mediated dilation. Compared with water, SSB consumption impaired microvascular and macrovascular endothelial function as indicated by a decrease in the vascular response to acetylcholine iontophoresis (208.3±24.3 versus 144.2±15.7%, P<0.01) and reduced flow-mediated dilation (0.019±0.002 versus 0.014±0.002%/s, P<0.01), respectively. Systemic vascular smooth muscle remained preserved. Similar decreases in endothelial function were observed during acute hyperglycemia in an in vivo rat model. However, function was fully restored by treatment with the antioxidants, N-acetylcysteine and apocynin. In addition, ex vivo experiments revealed that although the production of reactive oxygen species was increased during acute hyperglycemia, the bioavailability of nitric oxide in the endothelium was decreased, despite no change in the activation state of endothelial nitric oxide synthase. CONCLUSIONS: To our knowledge, this is the first study to assess the vascular effects of acute hyperglycemia induced by commercial SSB consumption alone. These findings suggest that SSB-mediated endothelial dysfunction is partly due to increased oxidative stress that decreases nitric oxide bioavailability. CLINICAL TRIAL REGISTRATION: URL: https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=366442&isReview=true. Australian New Zealand Clinical Trials Registry Number: ACTRN12614000614695.


Subject(s)
Beverages/adverse effects , Brachial Artery/drug effects , Dietary Sucrose/adverse effects , Hyperglycemia/chemically induced , Microvessels/drug effects , Skin/blood supply , Vasodilation/drug effects , Adult , Animals , Antioxidants/pharmacology , Blood Flow Velocity , Blood Glucose/drug effects , Blood Glucose/metabolism , Brachial Artery/metabolism , Brachial Artery/physiopathology , Cross-Over Studies , Dietary Sucrose/administration & dosage , Disease Models, Animal , Healthy Volunteers , Humans , Hyperglycemia/diagnosis , Hyperglycemia/physiopathology , Iontophoresis , Laser-Doppler Flowmetry , Male , Microcirculation , Microvessels/metabolism , Microvessels/physiopathology , Nitric Oxide/metabolism , Rats, Wistar , Single-Blind Method , Time Factors , Ultrasonography, Doppler , Vasodilator Agents/administration & dosage , Victoria
13.
Proc Natl Acad Sci U S A ; 112(50): 15360-5, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26627714

ABSTRACT

The lethal factor in stonefish venom is stonustoxin (SNTX), a heterodimeric cytolytic protein that induces cardiovascular collapse in humans and native predators. Here, using X-ray crystallography, we make the unexpected finding that SNTX is a pore-forming member of an ancient branch of the Membrane Attack Complex-Perforin/Cholesterol-Dependent Cytolysin (MACPF/CDC) superfamily. SNTX comprises two homologous subunits (α and ß), each of which comprises an N-terminal pore-forming MACPF/CDC domain, a central focal adhesion-targeting domain, a thioredoxin domain, and a C-terminal tripartite motif family-like PRY SPla and the RYanodine Receptor immune recognition domain. Crucially, the structure reveals that the two MACPF domains are in complex with one another and arranged into a stable early prepore-like assembly. These data provide long sought after near-atomic resolution insights into how MACPF/CDC proteins assemble into prepores on the surface of membranes. Furthermore, our analyses reveal that SNTX-like MACPF/CDCs are distributed throughout eukaryotic life and play a broader, possibly immune-related function outside venom.


Subject(s)
Fish Venoms/chemistry , Perforin/chemistry , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Cholesterol/chemistry , Complement Membrane Attack Complex/chemistry , Crystallography, X-Ray , Microscopy, Electron, Transmission , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Binding , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/chemistry , Solubility , Structural Homology, Protein
15.
Biochim Biophys Acta ; 1858(3): 475-86, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26607011

ABSTRACT

Pore Forming Toxins (PFTs) represent a key mechanism for permitting the passage of proteins and small molecules across the lipid membrane. These proteins are typically produced as soluble monomers that self-assemble into ring-like oligomeric structures on the membrane surface. Following such assembly PFTs undergo a remarkable conformational change to insert into the lipid membrane. While many different protein families have independently evolved such ability, members of the Membrane Attack Complex PerForin/Cholesterol Dependent Cytolysin (MACPF/CDC) superfamily form distinctive giant ß-barrel pores comprised of up to 50 monomers and up to 300Å in diameter. In this review we focus on recent advances in understanding the structure of these giant MACPF/CDC pores as well as the underlying molecular mechanisms leading to their formation. Commonalities and evolved variations of the pore forming mechanism across the superfamily are discussed. This article is part of a Special Issue entitled: Pore-Forming Toxins edited by Mauro Dalla Serra and Franco Gambale.


Subject(s)
Cell Membrane/metabolism , Evolution, Molecular , Perforin/classification , Perforin/metabolism , Animals , Cell Membrane/chemistry , Humans , Perforin/chemistry , Protein Structure, Secondary , Structure-Activity Relationship
16.
Biochim Biophys Acta Mol Basis Dis ; 1863(6): 1568-1574, 2017 06.
Article in English | MEDLINE | ID: mdl-28254494

ABSTRACT

In the view of the relationships between excessive sodium intake, immunity and target organ damage, we hypothesized that reduction in dietary sodium would be beneficial in the prevention of cardiac alterations through a restrained local immunity response in a rat model of metabolic syndrome. Sprague-Dawley rats were fed a 60% fructose diet with either a normal sodium (0.64% NaCl) or a low sodium content (<0.01% NaCl) for 8weeks. After 4weeks, rats were infused or not with angiotensin II (200ng·kg-1·min-1, sc) for 4weeks. Tail-cuff blood pressure was determined in conscious rats. Heart and left ventricle weight, cardiomyocyte size, and cardiac fibrosis were evaluated. We performed a transcriptomic analysis in order to identify differentially regulated cardiac mRNAs between normal and low sodium diets. We validated those results using qPCR and immunohistochemistry. Angiotensin II-induced blood pressure rise was blunted (~50%) in the low-sodium fed rats while cardiac hypertrophy and fibrosis were prevented. Transcriptomic analysis revealed 66 differentially regulated genes including 13 downregulated genes under the low sodium diet and implicated in the innate immune response. This was confirmed by reduced cardiac macrophages infiltration under the low sodium diet. Dietary sodium restriction prevents structural alterations of the heart of rats with fructose-induced insulin resistance and angiotensin II-hypertension. The reduction of cardiac inflammation and macrophage infiltration suggests that innate immunity has an important role in the beneficial effect of sodium restriction on cardiac remodeling.


Subject(s)
Cardiomegaly , Diet, Sodium-Restricted , Dietary Carbohydrates/adverse effects , Fructose/adverse effects , Immunity, Innate , Metabolic Syndrome , Animals , Cardiomegaly/diet therapy , Cardiomegaly/immunology , Dietary Carbohydrates/pharmacology , Disease Models, Animal , Fibrosis , Fructose/pharmacology , Metabolic Syndrome/chemically induced , Metabolic Syndrome/diet therapy , Metabolic Syndrome/immunology , Rats , Rats, Sprague-Dawley , Sodium Chloride, Dietary/adverse effects , Sodium Chloride, Dietary/pharmacology
17.
Proc Natl Acad Sci U S A ; 111(25): E2524-9, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24927554

ABSTRACT

The human neuroendocrine enzyme glutamate decarboxylase (GAD) catalyses the synthesis of the inhibitory neurotransmitter gamma-aminobutyric acid (GABA) using pyridoxal 5'-phosphate as a cofactor. GAD exists as two isoforms named according to their respective molecular weights: GAD65 and GAD67. Although cytosolic GAD67 is typically saturated with the cofactor (holoGAD67) and constitutively active to produce basal levels of GABA, the membrane-associated GAD65 exists mainly as the inactive apo form. GAD65, but not GAD67, is a prevalent autoantigen, with autoantibodies to GAD65 being detected at high frequency in patients with autoimmune (type 1) diabetes and certain other autoimmune disorders. The significance of GAD65 autoinactivation into the apo form for regulation of neurotransmitter levels and autoantibody reactivity is not understood. We have used computational and experimental approaches to decipher the nature of the holo → apo conversion in GAD65 and thus, its mechanism of autoinactivation. Molecular dynamics simulations of GAD65 reveal coupling between the C-terminal domain, catalytic loop, and pyridoxal 5'-phosphate-binding domain that drives structural rearrangement, dimer opening, and autoinactivation, consistent with limited proteolysis fragmentation patterns. Together with small-angle X-ray scattering and fluorescence spectroscopy data, our findings are consistent with apoGAD65 existing as an ensemble of conformations. Antibody-binding kinetics suggest a mechanism of mutually induced conformational changes, implicating the flexibility of apoGAD65 in its autoantigenicity. Although conformational diversity may provide a mechanism for cofactor-controlled regulation of neurotransmitter biosynthesis, it may also come at a cost of insufficient development of immune self-tolerance that favors the production of GAD65 autoantibodies.


Subject(s)
Autoimmunity , Glutamate Decarboxylase , Homeostasis/immunology , Molecular Dynamics Simulation , Neurotransmitter Agents , gamma-Aminobutyric Acid , Autoantibodies/immunology , Diabetes Mellitus, Type 1/immunology , Glutamate Decarboxylase/chemistry , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/immunology , Humans , Neurotransmitter Agents/chemistry , Neurotransmitter Agents/genetics , Neurotransmitter Agents/immunology , Protein Multimerization , Structure-Activity Relationship , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/genetics , gamma-Aminobutyric Acid/immunology
18.
Basic Res Cardiol ; 111(4): 40, 2016 07.
Article in English | MEDLINE | ID: mdl-27164904

ABSTRACT

Obesity and diabetes are associated with higher cardiac vulnerability to ischemia-reperfusion (IR). The cardioprotective effect of regular exercise has been attributed to ß3-adrenergic receptor (ß3AR) stimulation and increased endothelial nitric oxide synthase (eNOS) activation. Here, we evaluated the role of the ß3AR-eNOS pathway and NOS isoforms in exercise-induced cardioprotection of C57Bl6 mice fed with high fat and sucrose diet (HFS) for 12 weeks and subjected or not to exercise training during the last 4 weeks (HFS-Ex). HFS animals were more sensitive to in vivo and ex vivo IR injuries than control (normal diet) and HFS-Ex mice. Cardioprotection in HFS-Ex mice was not associated with increased myocardial eNOS activation and NO metabolites storage, possibly due to the ß3AR-eNOS pathway functional loss in their heart. Indeed, a selective ß3AR agonist (BRL37344) increased eNOS activation and had a protective effect against IR in control, but not in HFS hearts. Moreover, iNOS expression, nitro-oxidative stress (protein s-nitrosylation and nitrotyrosination) and ROS production during early reperfusion were increased in HFS, but not in control mice. Exercise normalized iNOS level and reduced protein s-nitrosylation, nitrotyrosination and ROS production in HFS-Ex hearts during early reperfusion. The iNOS inhibitor 1400 W reduced in vivo infarct size in HFS mice to control levels, supporting the potential role of iNOS normalization in the cardioprotective effects of exercise training in HFS-Ex mice. Although the ß3AR-eNOS pathway is defective in the heart of HFS mice, regular exercise can protect their heart against IR by reducing iNOS expression and nitro-oxidative stress.


Subject(s)
Myocardial Reperfusion Injury/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Obesity/complications , Physical Conditioning, Animal/physiology , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Myocardial Ischemia/metabolism , Nitric Oxide Synthase Type III/metabolism , Receptors, Adrenergic, beta-3/metabolism
19.
Arterioscler Thromb Vasc Biol ; 35(9): 2060-72, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26112007

ABSTRACT

OBJECTIVES: Controversy exists over the effect of acute hyperglycemia on vascular function. In this systematic review, we compared the effect of acute hyperglycemia on endothelial and vascular smooth muscle functions across healthy and cardiometabolic diseased subjects. APPROACH AND RESULTS: A systematic search of MEDLINE, EMBASE, and Web of Science from inception until July 2014 identified articles evaluating endothelial or vascular smooth muscle function during acute hyperglycemia and normoglycemia. Meta-analyses compared the standardized mean difference (SMD) in endothelial and vascular smooth muscle functions between acute hyperglycemia and normoglycemia. Subgroup analyses and metaregression identified sources of heterogeneity. Thirty-nine articles (525 healthy and 540 cardiometabolic subjects) were analyzed. Endothelial function was decreased (39 studies; n=1065; SMD, -1.25; 95% confidence interval, -1.52 to -0.98; P<0.01), whereas vascular smooth muscle function was preserved (6 studies; n=144; SMD, -0.07; 95% confidence interval, -0.30 to 0.16; P=0.55) during acute hyperglycemia compared with normoglycemia. Significant heterogeneity was detected among endothelial function studies (P<0.01). A subgroup analysis revealed that endothelial function was decreased in the macrocirculation (30 studies; n=884; SMD, -1.40; 95% confidence interval, -1.68 to -1.12; P<0.01) but not in the microcirculation (9 studies; n=181; SMD, -0.63; 95% confidence interval, -1.36 to 0.11; P=0.09). Similar results were observed according to health status. Macrovascular endothelial function was inversely associated with age, blood pressure, and low-density lipoprotein cholesterol and was positively associated with the postocclusion interval of vascular assessment. CONCLUSIONS: To our knowledge, this is the first systematic review and meta-analysis of its kind. In healthy and diseased subjects, we found evidence for macrovascular but not microvascular endothelial dysfunction during acute hyperglycemia.


Subject(s)
Cardiovascular Diseases , Hyperglycemia , Metabolic Syndrome , Vasodilation/physiology , Acute Disease , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/physiopathology , Global Health , Healthy Volunteers , Humans , Hyperglycemia/blood , Hyperglycemia/complications , Hyperglycemia/physiopathology , Metabolic Syndrome/epidemiology , Metabolic Syndrome/etiology , Metabolic Syndrome/physiopathology , Morbidity/trends
SELECTION OF CITATIONS
SEARCH DETAIL