Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
EMBO Mol Med ; 16(1): 132-157, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38177536

ABSTRACT

Thoracic aortic aneurysm and dissection (TAAD) is a life-threatening condition associated with Marfan syndrome (MFS), a disease caused by fibrillin-1 gene mutations. While various conditions causing TAAD exhibit aortic accumulation of the proteoglycans versican (Vcan) and aggrecan (Acan), it is unclear whether these ECM proteins are involved in aortic disease. Here, we find that Vcan, but not Acan, accumulated in Fbn1C1041G/+ aortas, a mouse model of MFS. Vcan haploinsufficiency protected MFS mice against aortic dilation, and its silencing reverted aortic disease by reducing Nos2 protein expression. Our results suggest that Acan is not an essential contributor to MFS aortopathy. We further demonstrate that Vcan triggers Akt activation and that pharmacological Akt pathway inhibition rapidly regresses aortic dilation and Nos2 expression in MFS mice. Analysis of aortic tissue from MFS human patients revealed accumulation of VCAN and elevated pAKT-S473 staining. Together, these findings reveal that Vcan plays a causative role in MFS aortic disease in vivo by inducing Nos2 via Akt activation and identify Akt signaling pathway components as candidate therapeutic targets.


Subject(s)
Aortic Aneurysm, Thoracic , Aortic Diseases , Aortic Dissection , Azides , Deoxyglucose , Marfan Syndrome , Animals , Humans , Mice , Aortic Aneurysm, Thoracic/complications , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/metabolism , Aortic Diseases/complications , Deoxyglucose/analogs & derivatives , Marfan Syndrome/complications , Marfan Syndrome/genetics , Marfan Syndrome/metabolism , Nitric Oxide Synthase Type II/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Versicans/metabolism
2.
Int J Mol Sci ; 23(17)2022 Aug 24.
Article in English | MEDLINE | ID: mdl-36076959

ABSTRACT

The sarcomere regulates striated muscle contraction. This structure is composed of several myofibril proteins, isoforms of which are encoded by genes specific to either the heart or skeletal muscle. The chromatin remodeler complex Chd4/NuRD regulates the transcriptional expression of these specific sarcomeric programs by repressing genes of the skeletal muscle sarcomere in the heart. Aberrant expression of skeletal muscle genes induced by the loss of Chd4 in the heart leads to sudden death due to defects in cardiomyocyte contraction that progress to arrhythmia and fibrosis. Identifying the transcription factors (TFs) that recruit Chd4/NuRD to repress skeletal muscle genes in the myocardium will provide important information for understanding numerous cardiac pathologies and, ultimately, pinpointing new therapeutic targets for arrhythmias and cardiomyopathies. Here, we sought to find Chd4 interactors and their function in cardiac homeostasis. We therefore describe a physical interaction between Chd4 and the TF Znf219 in cardiac tissue. Znf219 represses the skeletal-muscle sarcomeric program in cardiomyocytes in vitro and in vivo, similarly to Chd4. Aberrant expression of skeletal-muscle sarcomere proteins in mouse hearts with knocked down Znf219 translates into arrhythmias, accompanied by an increase in PR interval. These data strongly suggest that the physical and genetic interaction of Znf219 and Chd4 in the mammalian heart regulates cardiomyocyte identity and myocardial contraction.


Subject(s)
DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Transcription Factors , Animals , Gene Expression Regulation , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mice , Muscle Proteins/genetics , Muscle Proteins/metabolism , Nucleosomes , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Sci Rep ; 12(1): 14730, 2022 08 30.
Article in English | MEDLINE | ID: mdl-36042288

ABSTRACT

Angiogenesis is a multi-factorial physiological process deregulated in human diseases characterised by excessive or insufficient blood vessel formation. Emerging evidence highlights a novel role for microRNAs as regulators of angiogenesis. Previous studies addressing the effect of miR-133a expression in endothelial cells during blood vessel formation have reported conflicting results. Here, we have assessed the specific effect of mature miR-133a strands in angiogenesis and the expression of endothelial angiogenic genes. Transfection of miR-133a-3p or -5p mimics in primary human endothelial cells significantly inhibited proliferation, migration, and tubular morphogenesis of transfected cells. Screening of gene arrays related to angiogenic processes, and further validation by TaqMan qPCR, revealed that aberrant expression of miR-133a-3p led to a decrease in the expression of genes encoding pro-angiogenic molecules, whilst increasing those with anti-angiogenic functions. Ingenuity Pathway Analysis of a collection of genes differentially expressed in cells harbouring miR-133a-3p, predicted decreased cellular functions related to vasculature branching and cell cycle progression, underlining the inhibitory role of miR-133a-3p in angiogenic cellular processes. Our results suggest that controlled delivery of miR-133a-3p mimics, or antagomirs in diseased endothelial cells, might open new therapeutic interventions to treat patients suffering from cardiovascular pathologies that occur with excessive or insufficient angiogenesis.


Subject(s)
Endothelial Cells , MicroRNAs/genetics , Endothelial Cells/metabolism , Gene Expression , Humans , MicroRNAs/metabolism , Morphogenesis , Transfection
4.
Arterioscler Thromb Vasc Biol ; 42(4): 462-469, 2022 04.
Article in English | MEDLINE | ID: mdl-35196876

ABSTRACT

BACKGROUND: The goal of this study was to determine whether boosting mitochondrial respiration prevents the development of fatal aortic ruptures triggered by atherosclerosis and hypertension. METHODS: Ang-II (angiotensin-II) was infused in ApoE (Apolipoprotein E)-deficient mice fed with a western diet to induce acute aortic aneurysms and lethal ruptures. RESULTS: We found decreased mitochondrial respiration and mitochondrial proteins in vascular smooth muscle cells from murine and human aortic aneurysms. Boosting NAD levels with nicotinamide riboside reduced the development of aortic aneurysms and sudden death by aortic ruptures. CONCLUSIONS: Targetable vascular metabolism is a new clinical strategy to prevent fatal aortic ruptures and sudden death in patients with aortic aneurysms.


Subject(s)
Aortic Rupture , Atherosclerosis , Angiotensin II , Animals , Aortic Rupture/genetics , Aortic Rupture/prevention & control , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Death, Sudden , Humans , Mice , Mitochondrial Proteins
5.
Int J Cardiol ; 352: 125-130, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35092759

ABSTRACT

BACKGROUND: Increased collagen cross-linking (CCL) has been described in hypertensive cardiomyopathy by means of reduced serum ratio of serum carboxyterminal telopeptide of collagen type I (CITP) to matrix metalloproteinase-1 (MMP1). Previous studies have demonstrated the existence of primary impaired diastole in patients with Marfan syndrome (MFS), but little is known about the pathophysiology of this condition. METHODS: 60 MFS patients (without previous cardiovascular surgery or significant valvular regurgitation) and 24 healthy controls (age and sex-matched) were enrolled. All participants underwent a comprehensive transthoracic echocardiographic study, including left atrial and left ventricular speckle-tracking strain analysis. CITP and MMP1 were measured in peripheral blood. RESULTS: All participants had normal diastolic function according to guidelines. Peak left atrial strain in the reservoir phase (LASr) was significantly reduced in the MFS cohort compared to controls (32.2 ± 9.4 vs 43.9 ± 7.0%; p < 0.001). Serum CITP and CITP:MMP1 ratio were lower among MFS patients, showing significant correlations with LASr (R = 0.311; p = 0.020 and R = 0.437; p = 0.001, respectively). The MFS cohort was divided into quartiles of LASr. MFS patients in the lowest quartile of LASr (<26%) had significantly lower values of CITP:MMP1 ratio compared to the other quartiles. CONCLUSIONS: The analysis of serum biomarkers revealed the presence of increased CCL in association with reduced LASr in the MFS cohort. Our results suggest that excessive CCL may play a role in the development of primary myocardial impairment in these patients. Future studies are needed to confirm our findings and evaluate the prognostic role of CCL markers in the MFS population.


Subject(s)
Marfan Syndrome , Biomarkers , Collagen Type I , Diastole , Female , Humans , Male , Marfan Syndrome/complications , Marfan Syndrome/physiopathology , Myocardium
6.
Cardiovasc Res ; 118(16): 3250-3268, 2022 12 29.
Article in English | MEDLINE | ID: mdl-34672341

ABSTRACT

AIMS: Interferon-stimulated gene 15 (ISG15) encodes a ubiquitin-like protein that induces a reversible post-translational modification (ISGylation) and can also be secreted as a free form. ISG15 plays an essential role as host-defence response to microbial infection; however, its contribution to vascular damage associated with hypertension is unknown. METHODS AND RESULTS: Bioinformatics identified ISG15 as a mediator of hypertension-associated vascular damage. ISG15 expression positively correlated with systolic and diastolic blood pressure and carotid intima-media thickness in human peripheral blood mononuclear cells. Consistently, Isg15 expression was enhanced in aorta from hypertension models and in angiotensin II (AngII)-treated vascular cells and macrophages. Proteomics revealed differential expression of proteins implicated in cardiovascular function, extracellular matrix and remodelling, and vascular redox state in aorta from AngII-infused ISG15-/- mice. Moreover, ISG15-/- mice were protected against AngII-induced hypertension, vascular stiffness, elastin remodelling, endothelial dysfunction, and expression of inflammatory and oxidative stress markers. Conversely, mice with excessive ISGylation (USP18C61A) show enhanced AngII-induced hypertension, vascular fibrosis, inflammation and reactive oxygen species (ROS) generation along with elastin breaks, aortic dilation, and rupture. Accordingly, human and murine abdominal aortic aneurysms showed augmented ISG15 expression. Mechanistically, ISG15 induces vascular ROS production, while antioxidant treatment prevented ISG15-induced endothelial dysfunction and vascular remodelling. CONCLUSION: ISG15 is a novel mediator of vascular damage in hypertension through oxidative stress and inflammation.


Subject(s)
Aortic Aneurysm, Abdominal , Hypertension , Mice , Humans , Animals , Elastin/metabolism , Reactive Oxygen Species/metabolism , Angiotensin II/metabolism , Interferons/metabolism , Leukocytes, Mononuclear/metabolism , Carotid Intima-Media Thickness , Oxidative Stress , Hypertension/chemically induced , Hypertension/genetics , Hypertension/metabolism , Oxidation-Reduction , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/prevention & control , Inflammation , Mice, Inbred C57BL
7.
Br J Pharmacol ; 179(7): 1287-1303, 2022 04.
Article in English | MEDLINE | ID: mdl-34599830

ABSTRACT

Recent studies have shown that NO is a central mediator in diseases associated with thoracic aortic aneurysm, such as Marfan syndrome. The progressive dilation of the aorta in thoracic aortic aneurysm ultimately leads to aortic dissection. Unfortunately, current medical treatments have neither halt aortic enlargement nor prevented rupture, leaving surgical repair as the only effective treatment. There is therefore a pressing need for effective therapies to delay or even avoid the need for surgical repair in thoracic aortic aneurysm patients. Here, we summarize the mechanisms through which NO signalling dysregulation causes thoracic aortic aneurysm, particularly in Marfan syndrome. We discuss recent advances based on the identification of new Marfan syndrome mediators related to pathway overactivation that represent potential disease biomarkers. Likewise, we propose iNOS, sGC and PRKG1, whose pharmacological inhibition reverses aortopathy in Marfan syndrome mice, as targets for therapeutic intervention in thoracic aortic aneurysm and are candidates for clinical trials.


Subject(s)
Aortic Aneurysm, Thoracic , Aortic Aneurysm , Aortic Dissection , Marfan Syndrome , Aortic Dissection/complications , Aortic Dissection/surgery , Animals , Aorta , Aortic Aneurysm/complications , Aortic Aneurysm, Thoracic/etiology , Aortic Aneurysm, Thoracic/prevention & control , Aortic Aneurysm, Thoracic/surgery , Cyclic GMP-Dependent Protein Kinase Type I , Humans , Marfan Syndrome/complications , Marfan Syndrome/drug therapy , Marfan Syndrome/surgery , Mice
8.
Stem Cell Reports ; 16(9): 2089-2098, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34450038

ABSTRACT

Regeneration of skeletal muscle requires resident stem cells called satellite cells. Here, we report that the chromatin remodeler CHD4, a member of the nucleosome remodeling and deacetylase (NuRD) repressive complex, is essential for the expansion and regenerative functions of satellite cells. We show that conditional deletion of the Chd4 gene in satellite cells results in failure to regenerate muscle after injury. This defect is principally associated with increased stem cell plasticity and lineage infidelity during the expansion of satellite cells, caused by de-repression of non-muscle-cell lineage genes in the absence of Chd4. Thus, CHD4 ensures that a transcriptional program that safeguards satellite cell identity during muscle regeneration is maintained. Given the therapeutic potential of muscle stem cells in diverse neuromuscular pathologies, CHD4 constitutes an attractive target for satellite cell-based therapies.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , DNA Helicases/genetics , Muscle, Skeletal/physiology , Regeneration , Stem Cells/cytology , Stem Cells/metabolism , Animals , Computational Biology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mice , Models, Biological , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism
9.
Nat Commun ; 12(1): 2628, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976159

ABSTRACT

Thoracic aortic aneurysm, as occurs in Marfan syndrome, is generally asymptomatic until dissection or rupture, requiring surgical intervention as the only available treatment. Here, we show that nitric oxide (NO) signaling dysregulates actin cytoskeleton dynamics in Marfan Syndrome smooth muscle cells and that NO-donors induce Marfan-like aortopathy in wild-type mice, indicating that a marked increase in NO suffices to induce aortopathy. Levels of nitrated proteins are higher in plasma from Marfan patients and mice and in aortic tissue from Marfan mice than in control samples, indicating elevated circulating and tissue NO. Soluble guanylate cyclase and cGMP-dependent protein kinase are both activated in Marfan patients and mice and in wild-type mice treated with NO-donors, as shown by increased plasma cGMP and pVASP-S239 staining in aortic tissue. Marfan aortopathy in mice is reverted by pharmacological inhibition of soluble guanylate cyclase and cGMP-dependent protein kinase and lentiviral-mediated Prkg1 silencing. These findings identify potential biomarkers for monitoring Marfan Syndrome in patients and urge evaluation of cGMP-dependent protein kinase and soluble guanylate cyclase as therapeutic targets.


Subject(s)
Aortic Aneurysm, Thoracic/pathology , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Marfan Syndrome/complications , Soluble Guanylyl Cyclase/metabolism , Animals , Aorta/cytology , Aorta/diagnostic imaging , Aorta/drug effects , Aorta/pathology , Aortic Aneurysm, Thoracic/diagnosis , Aortic Aneurysm, Thoracic/etiology , Aortic Aneurysm, Thoracic/prevention & control , Biomarkers/blood , Biomarkers/metabolism , Carbazoles/administration & dosage , Cyclic GMP/blood , Cyclic GMP/metabolism , Disease Models, Animal , Female , Fibrillin-1/genetics , Gene Knockdown Techniques , Humans , Male , Marfan Syndrome/blood , Marfan Syndrome/genetics , Marfan Syndrome/pathology , Mice , Muscle, Smooth, Vascular/cytology , Mutation , Myocytes, Smooth Muscle , Nitric Oxide/metabolism , Nitric Oxide Donors/administration & dosage , Primary Cell Culture , Soluble Guanylyl Cyclase/antagonists & inhibitors , Ultrasonography
10.
Circulation ; 143(21): 2091-2109, 2021 05 25.
Article in English | MEDLINE | ID: mdl-33709773

ABSTRACT

BACKGROUND: Marfan syndrome (MFS) is an autosomal dominant disorder of the connective tissue caused by mutations in the FBN1 (fibrillin-1) gene encoding a large glycoprotein in the extracellular matrix called fibrillin-1. The major complication of this connective disorder is the risk to develop thoracic aortic aneurysm. To date, no effective pharmacologic therapies have been identified for the management of thoracic aortic disease and the only options capable of preventing aneurysm rupture are endovascular repair or open surgery. Here, we have studied the role of mitochondrial dysfunction in the progression of thoracic aortic aneurysm and mitochondrial boosting strategies as a potential treatment to managing aortic aneurysms. METHODS: Combining transcriptomics and metabolic analysis of aortas from an MFS mouse model (Fbn1c1039g/+) and MFS patients, we have identified mitochondrial dysfunction alongside with mtDNA depletion as a new hallmark of aortic aneurysm disease in MFS. To demonstrate the importance of mitochondrial decline in the development of aneurysms, we generated a conditional mouse model with mitochondrial dysfunction specifically in vascular smooth muscle cells (VSMC) by conditional depleting Tfam (mitochondrial transcription factor A; Myh11-CreERT2Tfamflox/flox mice). We used a mouse model of MFS to test for drugs that can revert aortic disease by enhancing Tfam levels and mitochondrial respiration. RESULTS: The main canonical pathways highlighted in the transcriptomic analysis in aortas from Fbn1c1039g/+ mice were those related to metabolic function, such as mitochondrial dysfunction. Mitochondrial complexes, whose transcription depends on Tfam and mitochondrial DNA content, were reduced in aortas from young Fbn1c1039g/+ mice. In vitro experiments in Fbn1-silenced VSMCs presented increased lactate production and decreased oxygen consumption. Similar results were found in MFS patients. VSMCs seeded in matrices produced by Fbn1-deficient VSMCs undergo mitochondrial dysfunction. Conditional Tfam-deficient VSMC mice lose their contractile capacity, showed aortic aneurysms, and died prematurely. Restoring mitochondrial metabolism with the NAD precursor nicotinamide riboside rapidly reverses aortic aneurysm in Fbn1c1039g/+ mice. CONCLUSIONS: Mitochondrial function of VSMCs is controlled by the extracellular matrix and drives the development of aortic aneurysm in Marfan syndrome. Targeting vascular metabolism is a new available therapeutic strategy for managing aortic aneurysms associated with genetic disorders.


Subject(s)
Aortic Aneurysm/physiopathology , Marfan Syndrome/genetics , Mitochondria/metabolism , Animals , Disease Models, Animal , Humans , Marfan Syndrome/physiopathology , Mice
12.
Arterioscler Thromb Vasc Biol ; 40(10): 2408-2424, 2020 10.
Article in English | MEDLINE | ID: mdl-32847388

ABSTRACT

OBJECTIVE: microRNAs are master regulators of gene expression with essential roles in virtually all biological processes. miR-217 has been associated with aging and cellular senescence, but its role in vascular disease is not understood. Approach and Results: We have used an inducible endothelium-specific knock-in mouse model to address the role of miR-217 in vascular function and atherosclerosis. miR-217 reduced NO production and promoted endothelial dysfunction, increased blood pressure, and exacerbated atherosclerosis in proatherogenic apoE-/- mice. Moreover, increased endothelial miR-217 expression led to the development of coronary artery disease and altered left ventricular heart function, inducing diastolic and systolic dysfunction. Conversely, inhibition of endogenous vascular miR-217 in apoE-/- mice improved vascular contractility and diminished atherosclerosis. Transcriptome analysis revealed that miR-217 regulates an endothelial signaling hub and downregulates a network of eNOS (endothelial NO synthase) activators, including VEGF (vascular endothelial growth factor) and apelin receptor pathways, resulting in diminished eNOS expression. Further analysis revealed that human plasma miR-217 is a biomarker of vascular aging and cardiovascular risk. CONCLUSIONS: Our results highlight the therapeutic potential of miR-217 inhibitors in aging-related cardiovascular disease.


Subject(s)
Aging/metabolism , Atherosclerosis/metabolism , Endothelial Cells/metabolism , MicroRNAs/metabolism , Plaque, Atherosclerotic , Age Factors , Aged, 80 and over , Aging/genetics , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Case-Control Studies , Cells, Cultured , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Disease Models, Animal , Endothelial Cells/pathology , Female , Hemodynamics , Humans , Mice, Inbred C57BL , Mice, Knockout, ApoE , MicroRNAs/blood , MicroRNAs/genetics , Middle Aged , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Signal Transduction , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left
13.
Cell Rep ; 31(7): 107652, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32433961

ABSTRACT

Somatic stem cells expand massively during tissue regeneration, which might require control of cell fitness, allowing elimination of non-competitive, potentially harmful cells. How or if such cells are removed to restore organ function is not fully understood. Here, we show that a substantial fraction of muscle stem cells (MuSCs) undergo necroptosis because of epigenetic rewiring during chronic skeletal muscle regeneration, which is required for efficient regeneration of dystrophic muscles. Inhibition of necroptosis strongly enhances suppression of MuSC expansion in a non-cell-autonomous manner. Prevention of necroptosis in MuSCs of healthy muscles is mediated by the chromatin remodeler CHD4, which directly represses the necroptotic effector Ripk3, while CHD4-dependent Ripk3 repression is dramatically attenuated in dystrophic muscles. Loss of Ripk3 repression by inactivation of Chd4 causes massive necroptosis of MuSCs, abolishing regeneration. Our study demonstrates how programmed cell death in MuSCs is tightly controlled to achieve optimal tissue regeneration.


Subject(s)
Epigenesis, Genetic/genetics , Muscle, Skeletal/metabolism , Necroptosis/genetics , Humans
14.
Front Physiol ; 11: 277, 2020.
Article in English | MEDLINE | ID: mdl-32372967

ABSTRACT

Short-chain fatty acids (SCFAs) are among the main classes of bacterial metabolic products and are mainly synthesized in the colon through bacterial fermentation. Short-chain fatty acids, such as acetate, butyrate, and propionate, reduce endothelial activation induced by proinflammatory mediators, at least in part, by activation of G protein-coupled receptors (GPRs): GPR41 and GPR43. The objective of the study was to analyze the possible protective effects of SCFAs on endothelial dysfunction induced by angiotensin II (AngII). Rat aortic endothelial cells (RAECs) and rat aortas were incubated with AngII (1 µM) for 6 h in the presence or absence of SCFAs (5-10 mM). In RAECs, we found that AngII reduces the production of nitric oxide (NO) stimulated by calcium ionophore A23187; increases the production of reactive oxygen species (ROS), both from the nicotinamide adenine dinucleotide phosphate oxidase system and the mitochondria; diminishes vasodilator-stimulated phosphoprotein (VASP) phosphorylation at Ser239; reduces GPR41 and GPR43 mRNA level; and reduces the endothelium-dependent relaxant response to acetylcholine in aorta. Coincubation with butyrate and acetate, but not with propionate, increases both NO production and pSer239-VASP, reduces the concentration of intracellular ROS, and improves relaxation to acetylcholine. The beneficial effects of butyrate were inhibited by the GPR41 receptor antagonist, ß-hydroxybutyrate, and by the GPR43 receptor antagonist, GLPG0794. Butyrate inhibited the down-regulation of GPR41 and GPR43 induced by AngII, being without effect acetate and propionate. Neither ß-hydroxybutyrate nor GLPG0794 affects the protective effect of acetate in endothelial dysfunction. In conclusion, acetate and butyrate improve endothelial dysfunction induced by AngII by increasing the bioavailability of NO. The effect of butyrate seems to be related to GPR41/43 activation, whereas acetate effects were independent of GPR41/43.

15.
Br J Pharmacol ; 177(9): 2006-2023, 2020 05.
Article in English | MEDLINE | ID: mdl-31883108

ABSTRACT

BACKGROUND AND PURPOSE: Hypertension is associated with gut dysbiosis. Here we have evaluated the effects of the angiotensin receptor antagonist losartan on gut microbiota in spontaneously hypertensive rats (SHR) to assess their contribution to its antihypertensive effects. EXPERIMENTAL APPROACH: Twenty-week-old Wistar Kyoto rats (WKY) and SHR were treated with losartan for 5 weeks (SHR-losartan). Faecal microbiota transplantation (FMT) was performed from donor SHR-losartan group to recipient untreated-SHR. Blood pressure (BP) was measured using tail-cuff plethysmography. Composition of the gut microbiota was assessed by amplification of the V3-V4 region of 16S rRNA gene. T cells were analysed in gut/aorta by flow cytometry. KEY RESULTS: Faeces from SHR showed gut dysbiosis, characterised by higher Firmicutes/Bacteroidetes ratios, lower acetate- and higher lactate-producing bacteria, and lower levels of strict anaerobic bacteria, effects which were restored to normal by losartan. Improvement of gut dysbiosis was linked to higher colonic integrity and lower sympathetic drive in the gut. In contrast, hydralazine reduced BP, but it neither restored gut dysbiosis nor colonic integrity. FMT from SHR-losartan to SHR reduced BP, improved the aortic endothelium-dependent relaxation to ACh, and reduced NADPH oxidase activity. These vascular changes were accompanied by both increased Treg and decreased Th17 cell populations in the vascular wall. CONCLUSION AND IMPLICATIONS: In SHR, losartan treatment reduced gut dysbiosis and sympathetic drive in the gut, thus improving gut integrity. The changes induced by losartan in gut microbiota contributed, in part, to protecting the vasculature and reducing BP, possibly by modulating the immune system in the gut.


Subject(s)
Gastrointestinal Microbiome , Hypertension , Animals , Antihypertensive Agents/pharmacology , Blood Pressure , Hypertension/drug therapy , Losartan/pharmacology , RNA, Ribosomal, 16S , Rats , Rats, Inbred SHR
16.
Sci Rep ; 9(1): 9361, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31249342

ABSTRACT

Calcium/Calcineurin/Nuclear Factor of Activated T cells (Ca/CN/NFAT) signalling pathway is the main calcium (Ca2+) dependent signalling pathway involved in the homeostasis of brain tissue. Here, we study the presence of NFATc members in human glioma by using U251 cells and a collection of primary human glioblastoma (hGB) cell lines. We show that NFATc3 member is the predominant member. Furthermore, by using constitutive active NFATc3 mutant and shRNA lentiviral vectors to achieve specific silencing of this NFATc member, we describe cytokines and molecules regulated by this pathway which are required for the normal biology of cancer cells. Implanting U251 in an orthotopic intracranial assay, we show that specific NFATc3 silencing has a role in tumour growth. In addition NFATc3 knock-down affects both the proliferation and migration capacities of glioma cells in vitro. Our data open the possibility of NFATc3 as a target for the treatment of glioma.


Subject(s)
Astrocytoma/genetics , NFATC Transcription Factors/genetics , Animals , Astrocytoma/metabolism , Astrocytoma/pathology , Biomarkers , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement/genetics , Cell Proliferation/genetics , Disease Models, Animal , Gene Expression , Gene Expression Regulation, Neoplastic , Gene Silencing , Heterografts , Humans , Mice , Muscle Proteins/genetics , Muscle Proteins/metabolism , NFATC Transcription Factors/metabolism
17.
FEBS J ; 286(1): 46-65, 2019 01.
Article in English | MEDLINE | ID: mdl-30548183

ABSTRACT

Previous studies have demonstrated that activation of calcineurin induces pathological cardiac hypertrophy (CH). In these studies, loss-of-function was mostly achieved by systemic administration of the calcineurin inhibitor cyclosporin A. The lack of conditional knockout models for calcineurin function has impeded progress toward defining the role of this protein during the onset and the development of CH in adults. Here, we exploited a mouse model of CH based on the infusion of a hypertensive dose of angiotensin II (AngII) to model the role of calcineurin in CH in adulthood. AngII-induced CH in adult mice was reduced by treatment with cyclosporin A, without affecting the associated increase in blood pressure, and also by induction of calcineurin deletion in adult mouse cardiomyocytes, indicating that cardiomyocyte calcineurin is required for AngII-induced CH. Surprisingly, cardiac-specific deletion of calcineurin, but not treatment of mice with cyclosporin A, significantly reduced AngII-induced cardiac fibrosis and apoptosis. Analysis of profibrotic genes revealed that AngII-induced expression of Tgfß family members and Lox was not inhibited by cyclosporin A but was markedly reduced by cardiac-specific calcineurin deletion. These results show that AngII induces a direct, calcineurin-dependent prohypertrophic effect in cardiomyocytes, as well as a systemic hypertensive effect that is independent of calcineurin activity.


Subject(s)
Calcineurin/physiology , Cardiomegaly/pathology , Fibrosis/pathology , Myocytes, Cardiac/pathology , Angiotensin II/toxicity , Animals , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Disease Progression , Fibrosis/chemically induced , Fibrosis/metabolism , Gene Expression Profiling , Mice , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Signal Transduction , Vasoconstrictor Agents/toxicity
18.
Nat Commun ; 9(1): 4795, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30442942

ABSTRACT

Aortic intramural hematoma (IMH) can evolve toward reabsorption, dissection or aneurysm. Hypertension is the most common predisposing factor in IMH and aneurysm patients, and the hypertensive mediator angiotensin-II induces both in mice. We have previously shown that constitutive deletion of Rcan1 isoforms prevents Angiotensin II-induced aneurysm in mice. Here we generate mice conditionally lacking each isoform or all isoforms in vascular smooth muscle cells, endothelial cells, or ubiquitously, to determine the contribution to aneurysm development of Rcan1 isoforms in vascular cells. Surprisingly, conditional Rcan1 deletion in either vascular cell-type induces a hypercontractile phenotype and aortic medial layer disorganization, predisposing to hypertension-mediated aortic rupture, IMH, and aneurysm. These processes are blocked by ROCK inhibition. We find that Rcan1 associates with GSK-3ß, whose inhibition decreases myosin activation. Our results identify potential therapeutic targets for intervention in IMH and aneurysm and call for caution when interpreting phenotypes of constitutively and inducibly deficient mice.


Subject(s)
Aortic Dissection/genetics , Aortic Rupture/genetics , Glycogen Synthase Kinase 3 beta/genetics , Hematoma/genetics , Hypertension/genetics , Intracellular Signaling Peptides and Proteins/genetics , Muscle Proteins/genetics , rho-Associated Kinases/genetics , Aortic Dissection/metabolism , Aortic Dissection/pathology , Aortic Dissection/prevention & control , Animals , Antihypertensive Agents/pharmacology , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Aortic Rupture/metabolism , Aortic Rupture/pathology , Aortic Rupture/prevention & control , Calcium-Binding Proteins , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Deletion , Gene Expression Regulation , Genetic Predisposition to Disease , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/metabolism , Hematoma/metabolism , Hematoma/pathology , Hematoma/prevention & control , Humans , Hypertension/drug therapy , Hypertension/metabolism , Hypertension/pathology , Intracellular Signaling Peptides and Proteins/deficiency , Male , Mice , Mice, Knockout , Muscle Proteins/deficiency , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Primary Cell Culture , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Kinase Inhibitors/pharmacology , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
19.
J Exp Med ; 215(11): 2901-2918, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30327417

ABSTRACT

MHCII in antigen-presenting cells (APCs) is a key regulator of adaptive immune responses. Expression of MHCII genes is controlled by the transcription coactivator CIITA, itself regulated through cell type-specific promoters. Here we show that the transcription factor NFAT5 is needed for expression of Ciita and MHCII in macrophages, but not in dendritic cells and other APCs. NFAT5-deficient macrophages showed defective activation of MHCII-dependent responses in CD4+ T lymphocytes and attenuated capacity to elicit graft rejection in vivo. Ultrasequencing analysis of NFAT5-immunoprecipitated chromatin uncovered an NFAT5-regulated region distally upstream of Ciita This region was required for CIITA and hence MHCII expression, exhibited NFAT5-dependent characteristics of active enhancers such as H3K27 acetylation marks, and required NFAT5 to interact with Ciita myeloid promoter I. Our results uncover an NFAT5-regulated mechanism that maintains CIITA and MHCII expression in macrophages and thus modulates their T lymphocyte priming capacity.


Subject(s)
Enhancer Elements, Genetic/immunology , Gene Expression Regulation/immunology , Histocompatibility Antigens Class II/immunology , Macrophages/immunology , Nuclear Proteins/immunology , Trans-Activators/immunology , Transcription Factors/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Gene Rearrangement/immunology , Histocompatibility Antigens Class II/genetics , Macrophages/cytology , Mice , Mice, Knockout , Nuclear Proteins/genetics , Trans-Activators/genetics , Transcription Factors/genetics
20.
Trends Mol Med ; 24(10): 825-837, 2018 10.
Article in English | MEDLINE | ID: mdl-30213701

ABSTRACT

Lentiviral vectors (LVs) transduce quiescent cells and provide stable integration to maintain transgene expression. Several approaches have been adopted to optimize LV safety profiles. Similarly, LV targeting has been tailored through strategies including the modification of envelope components, the use of specific regulatory elements, and the selection of appropriate administration routes. Models of aortic disease based on a single injection of pleiotropic LVs have been developed that efficiently transduce the three aorta layers in wild type mice. This approach allows the dissection of pathways involved in aortic aneurysm formation and the identification of targets for gene therapy in aortic diseases. LVs provide a fast, efficient, and affordable alternative to genetically modified mice to study disease mechanisms and develop therapeutic tools.


Subject(s)
ADAMTS1 Protein/genetics , Aortic Aneurysm/therapy , Genetic Therapy/methods , Genetic Vectors/chemistry , Lentivirus/genetics , ADAMTS1 Protein/antagonists & inhibitors , ADAMTS1 Protein/immunology , Animals , Aortic Aneurysm/genetics , Aortic Aneurysm/immunology , Aortic Aneurysm/pathology , Clinical Trials as Topic , Disease Models, Animal , Gene Expression Regulation , Genetic Vectors/immunology , Humans , Immunity, Innate , Lentivirus/immunology , Mice , Patient Safety , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transduction, Genetic/methods , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...