Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Oxid Med Cell Longev ; 2018: 3967213, 2018.
Article in English | MEDLINE | ID: mdl-29765495

ABSTRACT

Marfan syndrome (MFS) cardiovascular manifestations such as aortic aneurysms and cardiomyopathy carry substantial morbidity/mortality. We investigated the effects of lipoic acid, an antioxidant, on ROS production and aortic remodeling in a MFS mgΔloxPneo mouse model. MFS and WT (wild-type) 1-month-old mice were allocated to 3 groups: untreated, treated with losartan, and treated with lipoic acid. At 6 months old, echocardiography, ROS production, and morphological analysis of aortas were performed. Aortic ROS generation in 6-month-old MFS animals was higher at advanced stages of disease in MFS. An unprecedented finding in MFS mice analyzed by OCT was the occurrence of focal inhomogeneous regions in the aortic arch, either collagen-rich extremely thickened or collagen-poor hypotrophic regions. MFS animals treated with lipoic acid showed markedly reduced ROS production and lower ERK1/2 phosphorylation; meanwhile, aortic dilation and elastic fiber breakdown were unaltered. Of note, lipoic acid treatment associated with the absence of focal inhomogeneous regions in MFS animals. Losartan reduced aortic dilation and elastic fiber breakdown despite no change in ROS generation. In conclusion, oxidant generation by itself seems neutral with respect to aneurysm progression in MFS; however, lipoic acid-mediated reduction of inhomogeneous regions may potentially associate with less anisotropy and reduced chance of dissection/rupture.


Subject(s)
Aortic Diseases/drug therapy , Marfan Syndrome/drug therapy , Reactive Oxygen Species/metabolism , Animals , Disease Models, Animal , Female , Humans , Mice , Phenotype
2.
Free Radic Biol Med ; 118: 44-58, 2018 04.
Article in English | MEDLINE | ID: mdl-29471108

ABSTRACT

Marfan syndrome (MFS) is characterized by the formation of ascending aortic aneurysms resulting from altered assembly of extracellular matrix fibrillin-containing microfibrils and dysfunction of TGF-ß signaling. Here we identify the molecular targets of redox stress in aortic aneurysms from MFS patients, and investigate the role of NOX4, whose expression is strongly induced by TGF-ß, in aneurysm formation and progression in a murine model of MFS. Working models included aortae and cultured vascular smooth muscle cells (VSMC) from MFS patients, and a NOX4-deficient Marfan mouse model (Fbn1C1039G/+-Nox4-/-). Increased tyrosine nitration and reactive oxygen species levels were found in the tunica media of human aortic aneurysms and in cultured VSMC. Proteomic analysis identified nitrated and carbonylated proteins, which included smooth muscle α-actin (αSMA) and annexin A2. NOX4 immunostaining increased in the tunica media of human Marfan aorta and was transcriptionally overexpressed in VSMC. Fbn1C1039G/+-Nox4-/- mice aortas showed a reduction of fragmented elastic fibers, which was accompanied by an amelioration in the Marfan-associated enlargement of the aortic root. Increase in the contractile phenotype marker calponin in the tunica media of MFS mice aortas was abrogated in Fbn1C1039G/+-Nox4-/- mice. Endothelial dysfunction evaluated by myography in the Marfan ascending aorta was prevented by the absence of Nox4 or catalase-induced H2O2 decomposition. We conclude that redox stress occurs in MFS, whose targets are actin-based cytoskeleton members and regulators of extracellular matrix homeostasis. Likewise, NOX4 have an impact in the progression of the aortic dilation in MFS and in the structural organization of the aortic tunica media, the VSMC phenotypic modulation, and endothelial function.


Subject(s)
Aortic Aneurysm/metabolism , Marfan Syndrome/metabolism , Marfan Syndrome/pathology , NADPH Oxidase 4/metabolism , Oxidative Stress/physiology , Adult , Animals , Aortic Aneurysm/etiology , Female , Humans , Male , Marfan Syndrome/complications , Mice , Mice, Knockout , Middle Aged , Muscle, Smooth, Vascular/metabolism , Oxidation-Reduction , Young Adult
3.
Front Physiol ; 8: 933, 2017.
Article in English | MEDLINE | ID: mdl-29187826

ABSTRACT

Marfan syndrome (MFS) is a hereditary disorder of the connective tissue that causes life-threatening aortic aneurysm, which initiates at the aortic root and can progress into the ascending portion. However, analysis of ascending aorta reactivity in animal models of MFS has remained elusive. Epidemiologic evidence suggests that although MFS is equally prevalent in men and women, men are at a higher risk of aortic complications than non-pregnant women. Nevertheless, there is no experimental evidence to support this hypothesis. The aim of this study was to explore whether there are regional and sex differences in the thoracic aorta function of mice heterozygous for the fibrillin 1 (Fbn1) allele encoding a missense mutation (Fbn1C1039G/+), the most common class of mutation in MFS. Ascending and descending thoracic aorta reactivity was evaluated by wire myography. Ascending aorta mRNA and protein levels, and elastic fiber integrity were assessed by qRT-PCR, Western blotting, and Verhoeff-Van Gieson histological staining, respectively. MFS differently altered reactivity in the ascending and descending thoracic aorta by either increasing or decreasing phenylephrine contractions, respectively. When mice were separated by sex, contractions to phenylephrine increased progressively from 3 to 6 months of age in MFS ascending aortas of males, whereas contractions in females were unchanged. Endothelium-dependent relaxation was unaltered in the MFS ascending aorta of either sex; an effect related to augmented endothelium-dependent hyperpolarization-type dilations. In MFS males, the non-selective cyclooxygenase (COX) inhibitor indomethacin prevented the MFS-induced enhancement of phenylephrine contractions linked to increased COX-2 expression. In MFS mice of both sexes, the non-selective nitric oxide synthase inhibitor L-NAME revealed negative feedback of nitric oxide on phenylephrine contractions, which was associated with upregulation of eNOS in females. Finally, MFS ascending aortas showed a greater number of elastic fiber breaks than the wild-types, and males exhibited more breaks than females. These results show regional and sex differences in Fbn1C1039G/+ mice thoracic aorta contractility and aortic media injuries. The presence of more pronounced aortic alterations in male mice provides experimental evidence to support that male MFS patients are at increased risk of suffering aortic complications.

4.
J Am Heart Assoc ; 6(9)2017 Sep 25.
Article in English | MEDLINE | ID: mdl-28947563

ABSTRACT

BACKGROUND: Marfan syndrome (MF) leads to aortic root dilatation and a predisposition to aortic dissection, mitral valve prolapse, and primary and secondary cardiomyopathy. Overall, regular physical exercise is recommended for a healthy lifestyle, but dynamic sports are strongly discouraged in MF patients. Nonetheless, evidence supporting this recommendation is lacking. Therefore, we studied the role of long-term dynamic exercise of moderate intensity on the MF cardiovascular phenotype. METHODS AND RESULTS: In a transgenic mouse model of MF (Fbn1C1039G/+), 4-month-old wild-type and MF mice were subjected to training on a treadmill for 5 months; sedentary littermates served as controls for each group. Aortic and cardiac remodeling was assessed by echocardiography and histology. The 4-month-old MF mice showed aortic root dilatation, elastic lamina rupture, and tunica media fibrosis, as well as cardiac hypertrophy, left ventricular fibrosis, and intramyocardial vessel remodeling. Over the 5-month experimental period, aortic root dilation rate was significantly greater in the sedentary MF group, compared with the wild-type group (∆mm, 0.27±0.07 versus 0.13±0.02, respectively). Exercise significantly blunted the aortic root dilation rate in MF mice compared with sedentary MF littermates (∆mm, 0.10±0.04 versus 0.27±0.07, respectively). However, these 2 groups were indistinguishable by aortic root stiffness, tunica media fibrosis, and elastic lamina ruptures. In MF mice, exercise also produced cardiac hypertrophy regression without changes in left ventricular fibrosis. CONCLUSIONS: Our results in a transgenic mouse model of MF indicate that moderate dynamic exercise mitigates the progression of the MF cardiovascular phenotype.


Subject(s)
Aortic Aneurysm/prevention & control , Aortic Dissection/prevention & control , Cardiomyopathies/prevention & control , Exercise Therapy , Marfan Syndrome/therapy , Physical Conditioning, Animal/methods , Aortic Dissection/genetics , Aortic Dissection/pathology , Aortic Dissection/physiopathology , Animals , Aorta/pathology , Aorta/physiopathology , Aortic Aneurysm/genetics , Aortic Aneurysm/pathology , Aortic Aneurysm/physiopathology , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Dilatation, Pathologic , Disease Models, Animal , Disease Progression , Female , Fibrillin-1/genetics , Fibrosis , Genetic Predisposition to Disease , Male , Marfan Syndrome/genetics , Marfan Syndrome/pathology , Marfan Syndrome/physiopathology , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Sex Factors , Time Factors , Vascular Remodeling , Ventricular Function, Left , Ventricular Remodeling
5.
Am J Physiol Heart Circ Physiol ; 310(9): H1081-90, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26945079

ABSTRACT

Marfan syndrome (MFS) is a connective tissue disorder that is often associated with the fibrillin-1 (Fbn1) gene mutation and characterized by cardiovascular alterations, predominantly ascending aortic aneurysms. Although neurovascular complications are uncommon in MFS, the improvement in Marfan patients' life expectancy is revealing other secondary alterations, potentially including neurovascular disorders. However, little is known about small-vessel pathophysiology in MFS. MFS is associated with hyperactivated transforming growth factor (TGF)-ß signaling, which among numerous other downstream effectors, induces the NADPH oxidase 4 (Nox4) isoform of NADPH oxidase, a strong enzymatic source of H2O2 We hypothesized that MFS induces middle cerebral artery (MCA) alterations and that Nox4 contributes to them. MCA properties from 3-, 6-, or 9-mo-old Marfan (Fbn1(C1039G/+)) mice were compared with those from age/sex-matched wild-type littermates. At 6 mo, Marfan compared with wild-type mice developed higher MCA wall/lumen (wild-type: 0.081 ± 0.004; Marfan: 0.093 ± 0.002; 60 mmHg; P < 0.05), coupled with increased reactive oxygen species production, TGF-ß, and Nox4 expression. However, wall stiffness and myogenic autoregulation did not change. To investigate the influence of Nox4 on cerebrovascular properties, we generated Marfan mice with Nox4 deficiency (Nox4(-/-)). Strikingly, Nox4 deletion in Marfan mice aggravated MCA wall thickening (cross-sectional area; Marfan: 6,660 ± 363 µm(2); Marfan Nox4(-/-): 8,795 ± 824 µm(2); 60 mmHg; P < 0.05), accompanied by decreased TGF-ß expression and increased collagen deposition and Nox1 expression. These findings provide the first evidence that Nox4 mitigates cerebral artery structural changes in a murine model of MFS.


Subject(s)
Cerebrovascular Disorders/prevention & control , Marfan Syndrome/complications , Middle Cerebral Artery/enzymology , NADPH Oxidases/metabolism , Vascular Remodeling , Animals , Arterial Pressure , Cerebrovascular Disorders/enzymology , Cerebrovascular Disorders/genetics , Cerebrovascular Disorders/pathology , Collagen/metabolism , Disease Models, Animal , Disease Progression , Female , Fibrillin-1/genetics , Genetic Predisposition to Disease , Male , Marfan Syndrome/enzymology , Marfan Syndrome/genetics , Mechanotransduction, Cellular , Mice, Knockout , Middle Cerebral Artery/pathology , Middle Cerebral Artery/physiopathology , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1 , NADPH Oxidase 4 , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , Phenotype , Reactive Oxygen Species/metabolism , Stress, Mechanical , Time Factors , Transforming Growth Factor beta/metabolism , Vascular Stiffness
6.
PLoS One ; 11(3): e0152124, 2016.
Article in English | MEDLINE | ID: mdl-27003297

ABSTRACT

Early morbidity and mortality in patients with Marfan syndrome (MFS) -a connective tissue disease caused by mutations in fibrillin-1 gene- are mainly caused by aorta aneurysm and rupture. However, the increase in the life expectancy of MFS patients recently achieved by reparatory surgery promotes clinical manifestations in other organs. Although some studies have reported respiratory alterations in MFS, our knowledge of how this connective tissue disease modifies lung mechanics is scarce. Hence, we assessed whether the stiffness of the whole lung and of its extracellular matrix (ECM) is affected in a well-characterized MFS mouse model (FBN1C1039G/+). The stiffness of the whole lung and of its ECM were measured by conventional mechanical ventilation and atomic force microscopy, respectively. We studied 5-week and 9-month old mice, whose ages are representative of early and late stages of the disease. At both ages, the lungs of MFS mice were significantly more compliant than in wild type (WT) mice. By contrast, no significant differences were found in local lung ECM stiffness. Moreover, histopathological lung evaluation showed a clear emphysematous-like pattern in MFS mice since alveolar space enlargement was significantly increased compared with WT mice. These data suggest that the mechanism explaining the increased lung compliance in MFS is not a direct consequence of reduced ECM stiffness, but an emphysema-like alteration in the 3D structural organization of the lung. Since lung alterations in MFS are almost fully manifested at an early age, it is suggested that respiratory monitoring could provide early biomarkers for diagnosis and/or follow-up of patients with the Marfan syndrome.


Subject(s)
Lung/pathology , Marfan Syndrome/pathology , Animals , Disease Models, Animal , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Fibrillin-1 , Fibrillins , Marfan Syndrome/genetics , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Mutation/genetics
7.
Int J Biochem Cell Biol ; 71: 81-91, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26718974

ABSTRACT

Fibrillin-1 mutations promote Marfan syndrome (MFS) via complex yet unclear pathways. The roles of endoplasmic reticulum (ER) and the major ER redox chaperone protein disulfide isomerase-A1 in the processing of normal and mutated fibrillin-1 and ensuing protein secretion and/or intracellular retention are unclear. Our results in mouse embryonic fibroblasts bearing the exon-skipping mgΔ(lox-P-neo) (mgΔ(lpn)) mutation, which associates in vivo with MFS and in vitro with disrupted microfibrils, indicate a preserved ER-dependent proteostasis or redox homeostasis. Rather, mutated fibrillin-1 is secreted normally through Golgi-dependent pathways and is not intracellularly retained. Similar results occurred for the C1039G point mutation. In parallel, we provide evidence that PDIA1 physically interacts with fibrillin-1 in the ER. Moreover, siRNA against PDIA1 augmented fibrillin-1 secretion rates in wild-type cells. However, fibrillin-1 with the mgΔ(lpn) mutation bypassed PDI checkpoint delay, while the C1039G mutation did not. This heretofore undisclosed PDIA1-mediated mechanism may be important to control the extracellular availability of function-competent fibrillin-1, an important determinant of disease phenotype. Moreover, our results may reveal a novel, holdase-like, PDI function associated with ER protein quality control.


Subject(s)
Homeostasis/genetics , Marfan Syndrome/genetics , Microfilament Proteins/genetics , Mutation , Protein Disulfide-Isomerases/metabolism , Animals , Cell Line , Endoplasmic Reticulum/metabolism , Fibrillin-1 , Fibrillins , Gene Silencing , Mice , Microfibrils/metabolism , Phenotype , Protein Disulfide-Isomerases/deficiency , Protein Disulfide-Isomerases/genetics
8.
Arterioscler Thromb Vasc Biol ; 35(4): 960-72, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25593132

ABSTRACT

OBJECTIVE: Marfan's syndrome is characterized by the formation of ascending aortic aneurysms resulting from altered assembly of extracellular matrix microfibrils and chronic tissue growth factor (TGF)-ß signaling. TGF-ß is a potent regulator of the vascular smooth muscle cell (VSMC) phenotype. We hypothesized that as a result of the chronic TGF-ß signaling, VSMC would alter their basal differentiation phenotype, which could facilitate the formation of aneurysms. This study explores whether Marfan's syndrome entails phenotypic alterations of VSMC and possible mechanisms at the subcellular level. APPROACH AND RESULTS: Immunohistochemical and Western blotting analyses of dilated aortas from Marfan patients showed overexpression of contractile protein markers (α-smooth muscle actin, smoothelin, smooth muscle protein 22 alpha, and calponin-1) and collagen I in comparison with healthy aortas. VSMC explanted from Marfan aortic aneurysms showed increased in vitro expression of these phenotypic markers and also of myocardin, a transcription factor essential for VSMC-specific differentiation. These alterations were generally reduced after pharmacological inhibition of the TGF-ß pathway. Marfan VSMC in culture showed more robust actin stress fibers and enhanced RhoA-GTP levels, which was accompanied by increased focal adhesion components and higher nuclear localization of myosin-related transcription factor A. Marfan VSMC and extracellular matrix measured by atomic force microscopy were both stiffer than their respective controls. CONCLUSIONS: In Marfan VSMC, both in tissue and in culture, there are variable TGF-ß-dependent phenotypic changes affecting contractile proteins and collagen I, leading to greater cellular and extracellular matrix stiffness. Altogether, these alterations may contribute to the known aortic rigidity that precedes or accompanies Marfan's syndrome aneurysm formation.


Subject(s)
Aortic Aneurysm/etiology , Cell Differentiation , Marfan Syndrome/complications , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Actins/metabolism , Aorta/metabolism , Aorta/pathology , Aortic Aneurysm/metabolism , Aortic Aneurysm/pathology , Biomarkers/metabolism , Calcium-Binding Proteins/metabolism , Case-Control Studies , Cell Line, Tumor , Collagen Type I/metabolism , Cytoskeletal Proteins/metabolism , Dilatation, Pathologic , Focal Adhesions/metabolism , Humans , Marfan Syndrome/metabolism , Marfan Syndrome/pathology , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Nuclear Proteins/metabolism , Phenotype , Signal Transduction , Stress Fibers/metabolism , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Vascular Remodeling , rhoA GTP-Binding Protein/metabolism , Calponins
9.
BMC Cell Biol ; 15: 21, 2014 Jun 11.
Article in English | MEDLINE | ID: mdl-24915973

ABSTRACT

BACKGROUND: Upon activation neutrophil releases microparticles - small plasma membrane vesicles that contain cell surface proteins and cytoplasmic matter, with biological activities. In this study we investigated the potential role of myeloperoxidase in the endothelial cell injury caused by neutrophil-derived microparticles. RESULTS: Microparticles were produced by activating human neutrophils with a calcium ionophore and characterized by flow cytometry and transmission and scanning electron microscopy. Myeloperoxidase activity was measured by luminol-dependent chemiluminescence. Neutrophil microparticles-induced injuries and morphological alterations in human umbilical vein endothelial cells (HUVECs) were evaluated by microscopy and flow cytometry. Neutrophil microparticles were characterized as structures bounded by lipid bilayers and were less than 1 µm in diameter. The microparticles also expressed CD66b, CD62L and myeloperoxidase, which are all commonly expressed on the surface of neutrophils, as well as exposition of phosphatidylserine. The activity of the myeloperoxidase present on the microparticles was confirmed by hypochlorous acid detection. This compound is only catalyzed by myeloperoxidase in the presence of hydrogen peroxide and chloride ion. The addition of sodium azide or taurine inhibited and reduced enzymatic activity, respectively. Exposure of HUVEC to neutrophil microparticles induced a loss of cell membrane integrity and morphological changes. The addition of sodium azide or myeloperoxidase-specific inhibitor-I consistently reduced the injury to the endothelial cells. Taurine addition reduced HUVEC morphological changes. CONCLUSIONS: We have demonstrated the presence of active myeloperoxidase in neutrophil microparticles and that the microparticle-associated myeloperoxidase cause injury to endothelial cells. Hence, the microparticle-associated myeloperoxidase-hydrogen peroxide-chloride system may contribute to widespread endothelial cell damage in conditions of neutrophil activation as observed in vasculitis and sepsis.


Subject(s)
Cell-Derived Microparticles/enzymology , Endothelial Cells/pathology , Neutrophils/metabolism , Peroxidase/metabolism , Calcium/metabolism , Cell Membrane/metabolism , Cell Membrane/pathology , Cell Membrane/ultrastructure , Cell-Derived Microparticles/metabolism , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Human Umbilical Vein Endothelial Cells , Humans , Hydrogen Peroxide/metabolism , Oxidative Stress
10.
Respir Res ; 11: 51, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20433763

ABSTRACT

BACKGROUND: The dust mite Blomia tropicalis is an important source of aeroallergens in tropical areas. Although a mouse model for B. tropicalis extract (BtE)-induced asthma has been described, no study comparing different mouse strains in this asthma model has been reported. The relevance and reproducibility of experimental animal models of allergy depends on the genetic background of the animal, the molecular composition of the allergen and the experimental protocol. OBJECTIVES: This work had two objectives. The first was to study the anti-B. tropicalis allergic responses in different mouse strains using a short-term model of respiratory allergy to BtE. This study included the comparison of the allergic responses elicited by BtE with those elicited by ovalbumin in mice of the strain that responded better to BtE sensitization. The second objective was to investigate whether the best responder mouse strain could be used in an experimental model of allergy employing relatively low BtE doses. METHODS: Groups of mice of four different syngeneic strains were sensitized subcutaneously with 100 microg of BtE on days 0 and 7 and challenged four times intranasally, at days 8, 10, 12, and 14, with 10 microg of BtE. A/J mice, that were the best responders to BtE sensitization, were used to compare the B. tropicalis-specific asthma experimental model with the conventional experimental model of ovalbumin (OVA)-specific asthma. A/J mice were also sensitized with a lower dose of BtE. RESULTS: Mice of all strains had lung inflammatory-cell infiltration and increased levels of anti-BtE IgE antibodies, but these responses were significantly more intense in A/J mice than in CBA/J, BALB/c or C57BL/6J mice. Immunization of A/J mice with BtE induced a more intense airway eosinophil influx, higher levels of total IgE, similar airway hyperreactivity to methacholine but less intense mucous production, and lower levels of specific IgE, IgG1 and IgG2 antibodies than sensitization with OVA. Finally, immunization with a relatively low BtE dose (10 microg per subcutaneous injection per mouse) was able to sensitize A/J mice, which were the best responders to high-dose BtE immunization, for the development of allergy-associated immune and lung inflammatory responses. CONCLUSIONS: The described short-term model of BtE-induced allergic lung disease is reproducible in different syngeneic mouse strains, and mice of the A/J strain was the most responsive to it. In addition, it was shown that OVA and BtE induce quantitatively different immune responses in A/J mice and that the experimental model can be set up with low amounts of BtE.


Subject(s)
Allergens/administration & dosage , Asthma/immunology , Pyroglyphidae/immunology , Administration, Intranasal , Animals , Antigens, Plant , Asthma/genetics , Asthma/physiopathology , Bronchial Hyperreactivity/immunology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Immunologic , Immunoglobulin E/blood , Injections, Subcutaneous , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Ovalbumin , Pulmonary Eosinophilia/immunology , Rats , Rats, Wistar , Species Specificity , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...