Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
Dev Dyn ; 253(2): 233-254, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37688792

RESUMEN

BACKGROUND: Latent TGFß binding protein-2 (LTBP2) is a fibrillin 1 binding component of the microfibril. LTBP2 is the only LTBP protein that does not bind any isoforms of TGFß, although it may interfere with the function of other LTBPs or interact with other signaling pathways. RESULTS: Here, we investigate mice lacking Ltbp2 (Ltbp2-/- ) and identify multiple phenotypes that impact bodyweight and fat mass, and affect bone and skin development. The alterations in skin and bone development are particularly noteworthy since the strength of these tissues is differentially affected by loss of Ltbp2. Interestingly, some tissues that express high levels of Ltbp2, such as the aorta and lung, do not have a developmental or homeostatic phenotype. CONCLUSIONS: Analysis of these mice show that LTBP2 has complex effects on development through direct effects on the extracellular matrix (ECM) or on signaling pathways that are known to regulate the ECM.


Asunto(s)
Proteínas Portadoras , Matriz Extracelular , Animales , Ratones , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Matriz Extracelular/metabolismo , Fenotipo , Factor de Crecimiento Transformador beta/metabolismo , Isoformas de Proteínas/metabolismo , Unión Proteica
2.
J Biomech Eng ; 146(5)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-37646627

RESUMEN

Arterial stiffening occurs during natural aging, is associated with an increased risk of adverse cardiovascular events, and can follow different timelines in males and females. One mechanism of arterial stiffening includes remodeling of the extracellular matrix (ECM), which alters the wall material properties. We used elastin haploinsufficient (Eln+/-) and wildtype (Eln+/+) mice to investigate how material properties of two different arteries (ascending aorta and carotid artery) change with age, sex, and ECM composition. We used a constitutive model by Dong and Sun that is based on the Holzapfel-Gasser-Ogden (HGO) type, but does not require a discrete number of fibrous ECM families and allows varied deformation coupling. We find that the amount of deformation coupling for the best fit model depends on the artery type. We also find that remodeling to maintain homeostatic (i.e., young, wildtype) values of biomechanical parameters with age, sex, and ECM composition depends on the artery type, with ascending aorta being more adaptable than carotid artery. Fitted material constants indicate sex-dependent remodeling that may be important for determining the time course of arterial stiffening in males and females. We correlated fitted material constants with ECM composition measured by biochemical (ascending aorta) or histological (carotid artery) methods. We show significant correlations between ECM composition and material parameters for the mean values for each group, with biochemical measurements correlating more strongly than histological measurements. Understanding how arterial stiffening depends on age, sex, ECM composition, and artery type may help design effective, personalized clinical treatment strategies.


Asunto(s)
Aorta , Elastina , Animales , Femenino , Humanos , Masculino , Ratones , Aorta/patología , Arterias Carótidas , Matriz Extracelular , Proteínas de la Matriz Extracelular
3.
Hum Mol Genet ; 29(12): 2035-2050, 2020 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-32412588

RESUMEN

Supravalvular aortic stenosis (SVAS) is a narrowing of the aorta caused by elastin (ELN) haploinsufficiency. SVAS severity varies among patients with Williams-Beuren syndrome (WBS), a rare disorder that removes one copy of ELN and 25-27 other genes. Twenty percent of children with WBS require one or more invasive and often risky procedures to correct the defect while 30% have no appreciable stenosis, despite sharing the same basic genetic lesion. There is no known medical therapy. Consequently, identifying genes that modify SVAS offers the potential for novel modifier-based therapeutics. To improve statistical power in our rare-disease cohort (N = 104 exomes), we utilized extreme-phenotype cohorting, functional variant filtration and pathway-based analysis. Gene set enrichment analysis of exome-wide association data identified increased adaptive immune system variant burden among genes associated with SVAS severity. Additional enrichment, using only potentially pathogenic variants known to differ in frequency between the extreme phenotype subsets, identified significant association of SVAS severity with not only immune pathway genes, but also genes involved with the extracellular matrix, G protein-coupled receptor signaling and lipid metabolism using both SKAT-O and RQTest. Complementary studies in Eln+/-; Rag1-/- mice, which lack a functional adaptive immune system, showed improvement in cardiovascular features of ELN insufficiency. Similarly, studies in mixed background Eln+/- mice confirmed that variations in genes that increase elastic fiber deposition also had positive impact on aortic caliber. By using tools to improve statistical power in combination with orthogonal analyses in mice, we detected four main pathways that contribute to SVAS risk.


Asunto(s)
Estenosis Aórtica Supravalvular/genética , Elastina/genética , Proteínas de Homeodominio/genética , Síndrome de Williams/genética , Adolescente , Animales , Estenosis Aórtica Supravalvular/fisiopatología , Preescolar , Constricción Patológica/genética , Constricción Patológica/fisiopatología , Modelos Animales de Enfermedad , Haploinsuficiencia/genética , Humanos , Masculino , Ratones , Factores de Riesgo , Secuenciación del Exoma , Síndrome de Williams/fisiopatología
4.
Arterioscler Thromb Vasc Biol ; 41(12): 2890-2905, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34587758

RESUMEN

OBJECTIVE: Using a mouse model of Eln (elastin) insufficiency that spontaneously develops neointima in the ascending aorta, we sought to understand the origin and phenotypic heterogeneity of smooth muscle cells (SMCs) contributing to intimal hyperplasia. We were also interested in exploring how vascular cells adapt to the absence of Eln. Approach and Results: We used single-cell sequencing together with lineage-specific cell labeling to identify neointimal cell populations in a noninjury, genetic model of neointimal formation. Inactivating Eln production in vascular SMCs results in rapid intimal hyperplasia around breaks in the ascending aorta's internal elastic lamina. Using lineage-specific Cre drivers to both lineage mark and inactivate Eln expression in the secondary heart field and neural crest aortic SMCs, we found that cells with a secondary heart field lineage are significant contributors to neointima formation. We also identified a small population of secondary heart field-derived SMCs underneath and adjacent to the internal elastic lamina. Within the neointima of SMC-Eln knockout mice, 2 unique SMC populations were identified that are transcriptionally different from other SMCs. While these cells had a distinct gene signature, they expressed several genes identified in other studies of neointimal lesions, suggesting that some mechanisms underlying neointima formation in Eln insufficiency are shared with adult vessel injury models. CONCLUSIONS: These results highlight the unique developmental origin and transcriptional signature of cells contributing to neointima in the ascending aorta. Our findings also show that the absence of Eln, or changes in elastic fiber integrity, influences the SMC biological niche in ways that lead to altered cell phenotypes.


Asunto(s)
Elastina/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neointima/metabolismo , Animales , Aorta/metabolismo , Diferenciación Celular , Matriz Extracelular/metabolismo , Femenino , Masculino , Ratones Endogámicos , Modelos Animales
5.
J Biol Chem ; 295(9): 2687-2697, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31988245

RESUMEN

Microfibril-associated glycoprotein-1 (MAGP-1) is a component of vertebrate extracellular matrix (ECM) microfibrils that, together with the fibrillins, contributes to microfibril function. Many of the phenotypes associated with MAGP-1 gene inactivation are consistent with dysregulation of the transforming growth factor ß (TGFß)/bone morphogenetic protein (BMP) signaling system. We have previously shown that full-length MAGP-1 binds active TGFß-1 and some BMPs. The work presented here further defines the growth factor-binding domain of MAGP-1. Using recombinant domains and synthetic peptides, along with surface plasmon resonance analysis to measure the kinetics of the MAGP-1-TGFß-1 interaction, we localized the TGFß- and BMP-binding site in MAGP-1 to a 19-amino acid-long, highly acidic sequence near the N terminus. This domain was specific for binding active, but not latent, TGFß-1. Growth factor activity experiments revealed that TGFß-1 retains signaling activity when complexed with MAGP-1. Furthermore, when bound to fibrillin, MAGP-1 retained the ability to interact with TGFß-1, and active TGFß-1 did not bind fibrillin in the absence of MAGP-1. The absence of MAGP was sufficient to raise the amount of total TGFß stored in the ECM of cultured cells, suggesting that the MAGPs compete with the TGFß large latent complex for binding to microfibrils. Together, these results indicate that MAGP-1 plays an active role in TGFß signaling in the ECM.


Asunto(s)
Factores de Empalme de ARN/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Fibrilina-1/metabolismo , Humanos , Unión Proteica , Transducción de Señal
6.
Circ Res ; 125(11): 1006-1018, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31590613

RESUMEN

RATIONALE: Elastin is an important ECM (extracellular matrix) protein in large and small arteries. Vascular smooth muscle cells (SMCs) produce the layered elastic laminae found in elastic arteries but synthesize little elastin in muscular arteries. However, muscular arteries have a well-defined internal elastic lamina (IEL) that separates endothelial cells (ECs) from SMCs. The extent to which ECs contribute elastin to the IEL is unknown. OBJECTIVE: To use targeted elastin (Eln) deletion in mice to explore the relative contributions of SMCs and ECs to elastic laminae formation in different arteries. METHODS AND RESULTS: We used SMC- and EC-specific Cre recombinase transgenes with a novel floxed Eln allele to focus gene inactivation in mice. Inactivation of Eln in SMCs using Sm22aCre resulted in depletion of elastic laminae in the arterial wall with the exception of the IEL and SMC clusters in the outer media near the adventitia. Inactivation of elastin in ECs using Tie2Cre or Cdh5Cre resulted in normal medial elastin and a typical IEL in elastic arteries. In contrast, the IEL was absent or severely disrupted in muscular arteries. Interruptions in the IEL resulted in neointimal formation in the ascending aorta but not in muscular arteries. CONCLUSIONS: Combined with lineage-specific fate mapping systems, our knockout results document an unexpected heterogeneity in vascular cells that produce the elastic laminae. SMCs and ECs can independently form an IEL in most elastic arteries, whereas ECs are the major source of elastin for the IEL in muscular and resistance arteries. Neointimal formation at IEL disruptions in the ascending aorta confirms that the IEL is a critical physical barrier between SMCs and ECs in the large elastic arteries. Our studies provide new information about how SMCs and ECs contribute elastin to the arterial wall and how local elastic laminae defects may contribute to cardiovascular disease.


Asunto(s)
Tejido Elástico/metabolismo , Elastina/metabolismo , Células Endoteliales/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Arterias/crecimiento & desarrollo , Arterias/metabolismo , Presión Sanguínea , Linaje de la Célula , Proliferación Celular , Tejido Elástico/crecimiento & desarrollo , Tejido Elástico/ultraestructura , Elastina/deficiencia , Elastina/genética , Células Endoteliales/ultraestructura , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/crecimiento & desarrollo , Músculo Liso Vascular/ultraestructura , Miocitos del Músculo Liso/ultraestructura , Neointima , Transducción de Señal
7.
Am J Physiol Heart Circ Physiol ; 319(6): H1398-H1408, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33035438

RESUMEN

Elastin is a primary structural protein in the arterial wall that contributes to vascular mechanical properties and degrades with aging. Aging is associated with arterial stiffening and an increase in blood pressure. There is evidence that arterial aging follows different timelines with sex. Our objective was to investigate how elastin content affects arterial remodeling in male and female mice with aging. We used male and female wild-type (Eln+/+) and elastin heterozygous (Eln+/-) mice at 6, 12, and 24 mo of age and measured their blood pressure and arterial morphology, wall structure, protein content, circumferential stress, stretch ratio, and stiffness. Two arteries were used with varying contents of elastin: the left common carotid and ascending aorta. We show that Eln+/- arteries start at a different homeostatic set point for circumferential wall stress, stretch, and material stiffness but show similar increases with aging to Eln+/+ mice. With aging, structural stiffness is greatly increased, while material stiffness and circumferential stress are only slightly increased, highlighting the importance of maintaining these homeostatic values. Circumferential stretch shows the smallest change with age and may be important for controlling cellular phenotype. Independent sex differences are mostly associated with males being larger than females; however, many of the measured factors show age × sex and/or genotype × sex interactions, indicating that males and females follow different cardiovascular remodeling timelines with aging and are differentially affected by reduced elastin content.NEW & NOTEWORTHY A comprehensive study on arterial mechanical behavior as a function of elastin content, aging, and sex in mice. Elastin haploinsufficient arteries start at a different homeostatic set point for mechanical parameters such as circumferential stress, stretch, and material stiffness. Structural stiffness of the arterial wall greatly increases with aging, as expected, but there are interactions between sex and aging for most of the mechanical parameters that are important to consider in future work.


Asunto(s)
Aorta/metabolismo , Arteria Carótida Común/metabolismo , Elastina/deficiencia , Haploinsuficiencia , Remodelación Vascular , Factores de Edad , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Aorta/patología , Aorta/fisiopatología , Presión Arterial , Arteria Carótida Común/patología , Arteria Carótida Común/fisiopatología , Elastina/genética , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Factores Sexuales , Rigidez Vascular
8.
Development ; 144(24): 4563-4572, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29122839

RESUMEN

Alveologenesis, the final step of lung development, is characterized by the formation of millions of alveolar septa that constitute the vast gas-exchange surface area. The genetic network driving alveologenesis is poorly understood compared with earlier steps in lung development. FGF signaling through receptors Fgfr3 and Fgfr4 is crucial for alveologenesis, but the mechanisms through which they mediate this process remain unclear. Here we show that in Fgfr3;Fgfr4 (Fgfr3;4) global mutant mice, alveolar simplification is first observed at the onset of alveologenesis at postnatal day 3. This is preceded by disorganization of elastin, indicating defects in the extracellular matrix (ECM). Although Fgfr3 and Fgfr4 are expressed in the mesenchyme and epithelium, inactivation in the mesenchyme, but not the epithelium, recapitulated the defects. Expression analysis of components of the elastogenesis machinery revealed that Mfap5 (also known as Magp2), which encodes an elastin-microfibril bridging factor, is upregulated in Fgfr3;4 mutants. Mfap5 mutation in the Fgfr3;4 mutant background partially attenuated the alveologenesis defects. These data demonstrate that, during normal lung maturation, FGF signaling restricts expression of the elastogenic machinery in the lung mesenchyme to control orderly formation of the elastin ECM, thereby driving alveolar septa formation to increase the gas-exchange surface.


Asunto(s)
Proteínas Contráctiles/biosíntesis , Proteínas de la Matriz Extracelular/biosíntesis , Organogénesis/fisiología , Alveolos Pulmonares/embriología , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Proteínas Contráctiles/genética , Epitelio/metabolismo , Matriz Extracelular/patología , Proteínas de la Matriz Extracelular/genética , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Alveolos Pulmonares/citología , Factores de Empalme de ARN , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal/fisiología
9.
J Physiol ; 597(20): 5093-5108, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31444905

RESUMEN

KEY POINTS: We report that a sodium-activated potassium current, IKNa , has been inadvertently overlooked in both conduit and resistance arterial smooth muscle cells. IKNa is a major K+ resting conductance and is absent in cells of IKNa knockout (KO) mice. The phenotype of the IKNa KO is mild hypertension, although KO mice react more strongly than wild-type with raised blood pressure when challenged with vasoconstrictive agents. IKNa is negatively regulated by angiotensin II acting through Gαq protein-coupled receptors. In current clamp, KO arterial smooth muscle cells have easily evoked Ca2+ -dependent action potentials. ABSTRACT: Although several potassium currents have been reported to play a role in arterial smooth muscle (ASM), we find that one of the largest contributors to membrane conductance in both conduit and resistance ASMs has been inadvertently overlooked. In the present study, we show that IKNa , a sodium-activated potassium current, contributes a major portion of macroscopic outward current in a critical physiological voltage range that determines intrinsic cell excitability; IKNa is the largest contributor to ASM cell resting conductance. A genetic knockout (KO) mouse strain lacking KNa channels (KCNT1 and KCNT2) shows only a modest hypertensive phenotype. However, acute administration of vasoconstrictive agents such as angiotensin II (Ang II) and phenylephrine results in an abnormally large increase in blood pressure in the KO animals. In wild-type animals Ang II acting through Gαq protein-coupled receptors down-regulates IKNa , which increases the excitability of the ASMs. The complete genetic removal of IKNa in KO mice makes the mutant animal more vulnerable to vasoconstrictive agents, thus producing a paroxysmal-hypertensive phenotype. This may result from the lowering of cell resting K+ conductance allowing the cells to depolarize more readily to a variety of excitable stimuli. Thus, the sodium-activated potassium current may serve to moderate blood pressure in instances of heightened stress. IKNa may represent a new therapeutic target for hypertension and stroke.


Asunto(s)
Músculo Liso Vascular/fisiología , Canales de potasio activados por Sodio/metabolismo , Angiotensina II , Animales , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Ratones , Ratones Noqueados , Canales de potasio activados por Sodio/genética , Ratas , Ratas Sprague-Dawley
10.
Exp Eye Res ; 184: 221-226, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31022398

RESUMEN

Retinal capillary basement membrane (BM) thickening is closely associated with the development of vascular lesions in diabetic retinopathy. Thickened capillary BM can compromise blood-retinal-barrier characteristics and contribute to retinal vascular permeability, a significant clinical manifestation of diabetic retinopathy. We have previously shown that high glucose increases the expression and activity of lysyl oxidase (LOX), a crosslinking enzyme, in retinal endothelial cells. Additionally, concomitant with overexpression of LOX, increased vascular permeability was observed in diabetic rat retinas. However, it is unknown whether decreasing LOX overexpression may have protective effects against development of retinal vascular lesions in diabetes. To investigate whether reduced LOX level protects against diabetes-induced development of retinal vascular lesions characteristic of diabetic retinopathy, four groups of mice: wild type (WT) control mice, streptozotocin (STZ)-induced diabetic mice, LOX +/- mice, and STZ-induced diabetic LOX +/- mice were used for this study. Diabetes was maintained for 16 weeks; at the end of the study, retinas were assessed for LOX protein level by Western Blot (WB) analysis, and retinal capillary networks were isolated using retinal trypsin digestion and stained with hematoxylin and periodic acid Schiff to identify the number of acellular capillaries (AC) and pericyte loss (PL). In parallel, TUNEL assay was performed on retinal trypsin digests (RTDs) to detect cells undergoing apoptosis in the retinal capillary networks. Retinal vascular permeability was analyzed following FITC-dextran injection in retinal whole mounts. A significant increase in LOX expression was detected in the diabetic retinas compared to those of the WT control retinas, and as expected, a significant decrease in LOX expression in the diabetic LOX +/- retinas was observed compared to those of the diabetic retinas. RTD images showed significantly increased AC and PL counts in the retinas of diabetic mice compared to those of the WT control mice. Importantly, the number of AC and PL was significantly decreased, as was retinal vascular permeability in the retinas of the diabetic LOX +/- mice compared to those of the diabetic mice. Results suggest that decreasing diabetes-induced LOX overexpression may have protective effects against the development of vascular lesions characteristic of diabetic retinopathy. Therefore, LOX overexpression may be a potential target in preventing retinal vascular cell loss and excess permeability associated with diabetic retinopathy.


Asunto(s)
Permeabilidad Capilar/fisiología , Retinopatía Diabética/metabolismo , Proteína-Lisina 6-Oxidasa/metabolismo , Retina/metabolismo , Vasos Retinianos/patología , Animales , Apoptosis/fisiología , Diabetes Mellitus Experimental/complicaciones , Retinopatía Diabética/patología , Retinopatía Diabética/fisiopatología , Ratones , Ratones Endogámicos C57BL , Vasos Retinianos/fisiopatología
11.
Proc Natl Acad Sci U S A ; 113(31): 8759-64, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27432961

RESUMEN

Thoracic aortic aneurysms and dissections (TAAD) represent a substantial cause of morbidity and mortality worldwide. Many individuals presenting with an inherited form of TAAD do not have causal mutations in the set of genes known to underlie disease. Using whole-genome sequencing in two first cousins with TAAD, we identified a missense mutation in the lysyl oxidase (LOX) gene (c.893T > G encoding p.Met298Arg) that cosegregated with disease in the family. Using clustered regularly interspaced short palindromic repeats (CRISPR)/clustered regularly interspaced short palindromic repeats-associated protein-9 nuclease (Cas9) genome engineering tools, we introduced the human mutation into the homologous position in the mouse genome, creating mice that were heterozygous and homozygous for the human allele. Mutant mice that were heterozygous for the human allele displayed disorganized ultrastructural properties of the aortic wall characterized by fragmented elastic lamellae, whereas mice homozygous for the human allele died shortly after parturition from ascending aortic aneurysm and spontaneous hemorrhage. These data suggest that a missense mutation in LOX is associated with aortic disease in humans, likely through insufficient cross-linking of elastin and collagen in the aortic wall. Mutation carriers may be predisposed to vascular diseases because of weakened vessel walls under stress conditions. LOX sequencing for clinical TAAD may identify additional mutation carriers in the future. Additional studies using our mouse model of LOX-associated TAAD have the potential to clarify the mechanism of disease and identify novel therapeutics specific to this genetic cause.


Asunto(s)
Aneurisma de la Aorta Torácica/genética , Disección Aórtica/genética , Predisposición Genética a la Enfermedad/genética , Mutación con Pérdida de Función , Proteína-Lisina 6-Oxidasa/genética , Adulto , Anciano , Disección Aórtica/enzimología , Animales , Aneurisma de la Aorta Torácica/enzimología , Secuencia de Bases , Análisis Mutacional de ADN/métodos , Salud de la Familia , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Linaje , Proteína-Lisina 6-Oxidasa/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 37(5): 930-941, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28254817

RESUMEN

OBJECTIVE: Williams syndrome is characterized by obstructive aortopathy attributable to heterozygous loss of ELN, the gene encoding elastin. Lesions are thought to result primarily from excessive smooth muscle cell (SMC) proliferation and consequent medial expansion, although an initially smaller caliber and increased stiffness of the aorta may contribute to luminal narrowing. The relative contributions of such abnormalities to the obstructive phenotype had not been defined. APPROACH AND RESULTS: We quantified determinants of luminal stenosis in thoracic aortas of Eln-/- mice incompletely rescued by human ELN. Moderate obstruction was largely because of deficient circumferential growth, most prominently of ascending segments, despite increased axial growth. Medial thickening was evident in these smaller diameter elastin-deficient aortas, with medial area similar to that of larger diameter control aortas. There was no difference in cross-sectional SMC number between mutant and wild-type genotypes at multiple stages of postnatal development. Decreased elastin content was associated with medial fibrosis and reduced aortic distensibility because of increased structural stiffness but preserved material stiffness. Elastin-deficient SMCs exhibited greater contractile-to-proliferative phenotypic modulation in vitro than in vivo. We confirmed increased medial collagen without evidence of increased medial area or SMC number in a small ascending aorta with thickened media of a Williams syndrome subject. CONCLUSIONS: Deficient circumferential growth is the predominant mechanism for moderate obstructive aortic disease resulting from partial elastin deficiency. Our findings suggest that diverse aortic manifestations in Williams syndrome result from graded elastin content, and SMC hyperplasia causing medial expansion requires additional elastin loss superimposed on ELN haploinsufficiency.


Asunto(s)
Aorta Torácica/crecimiento & desarrollo , Enfermedades de la Aorta/fisiopatología , Elastina/metabolismo , Síndrome de Williams/fisiopatología , Adulto , Animales , Aorta Torácica/metabolismo , Aorta Torácica/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Proliferación Celular , Células Cultivadas , Colágeno/metabolismo , Constricción Patológica , Modelos Animales de Enfermedad , Elastina/deficiencia , Elastina/genética , Fibrosis , Predisposición Genética a la Enfermedad , Humanos , Hiperplasia , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Músculo Liso Vascular/crecimiento & desarrollo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fenotipo , Factores de Tiempo , Rigidez Vascular , Vasoconstricción , Síndrome de Williams/genética , Síndrome de Williams/metabolismo , Síndrome de Williams/patología
13.
Am J Physiol Heart Circ Physiol ; 313(2): H446-H456, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28550176

RESUMEN

Mutations in lysyl oxidase (LOX) are associated with thoracic aortic aneurysm and dissection (TAAD). Mice that do not express Lox (Lox-/- ) die soon after birth and have 60% and 40% reductions in elastin- and collagen-specific cross-links, respectively. LOX inactivation could also change the expression of secreted factors, the structural matrix, and matrix-associated proteins that constitute the aortic matrisome. We hypothesized that absence of Lox will change the mechanical behavior of the aortic wall because of reduced elastin and collagen cross-linking and alter the expression levels of matrisome and smooth muscle cell (SMC) genes in a vascular location-specific manner. Using fluorescence microscopy, pressure myography, and gene set enrichment analysis, we visualized the microarchitecture, quantified the mechanical behavior, and examined matrisome and SMC gene expression from ascending aortas (AAs) and descending aortas (DAs) from newborn Lox+/+ and Lox-/- mice. Even though Lox-/- AAs and DAs have fragmented elastic laminae and disorganized SMCs, the unloaded outer diameter and wall thickness were similar to Lox+/+ AAs and DAs. Lox-/- AAs and DAs have altered opening angles, circumferential stresses, and circumferential stretch ratios; however, only Lox-/- AAs have increased pressurized diameters and tangent moduli. Gene set enrichment analysis showed upregulation of the extracellular matrix (ECM) regulator gene set in Lox-/- AAs and DAs as well as differential expression of secreted factors, collagens, ECM-affiliated proteins, ECM glycoproteins, and SMC cell cycle gene sets that depend on the Lox genotype and vascular location. These results provide insights into the local chemomechanical changes induced by Lox inactivation that may be important for TAAD pathogenesis.NEW & NOTEWORTHY Absence of lysyl oxidase (Lox) causes thoracic aortic aneurysms. The aortic mechanical behavior of Lox-/- mice is consistent with reduced elastin and collagen cross-linking but demonstrates vascular location-specific differences. Lox-/- aortas show upregulation of matrix remodeling genes and location-specific differential expression of other matrix and smooth muscle cell gene sets.


Asunto(s)
Aorta Torácica/enzimología , Aneurisma de la Aorta Torácica/enzimología , Proteínas de la Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Proteína-Lisina 6-Oxidasa/metabolismo , Animales , Animales Recién Nacidos , Aorta Torácica/patología , Aorta Torácica/fisiopatología , Aneurisma de la Aorta Torácica/genética , Aneurisma de la Aorta Torácica/patología , Aneurisma de la Aorta Torácica/fisiopatología , Presión Arterial , Fenómenos Biomecánicos , Colágeno/genética , Colágeno/metabolismo , Dilatación Patológica , Modelos Animales de Enfermedad , Elastina/genética , Elastina/metabolismo , Proteínas de la Matriz Extracelular/genética , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad , Mecanotransducción Celular , Ratones Noqueados , Fenotipo , Proteína-Lisina 6-Oxidasa/genética , Estrés Mecánico , Rigidez Vascular
14.
J Biomech Eng ; 139(11)2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28916838

RESUMEN

Elastic fibers are present in low quantities in tendon, where they are located both within fascicles near tenocytes and more broadly in the interfascicular matrix (IFM). While elastic fibers have long been known to be significant in the mechanics of elastin-rich tissue (i.e., vasculature, skin, lungs), recent studies have suggested a mechanical role for elastic fibers in tendons that is dependent on specific tendon function. However, the exact contribution of elastin to properties of different types of tendons (e.g., positional, energy-storing) remains unknown. Therefore, this study purposed to evaluate the role of elastin in the mechanical properties and collagen alignment of functionally distinct supraspinatus tendons (SSTs) and Achilles tendons (ATs) from elastin haploinsufficient (HET) and wild type (WT) mice. Despite the significant decrease in elastin in HET tendons, a slight increase in linear stiffness of both tendons was the only significant mechanical effect of elastin haploinsufficiency. Additionally, there were significant changes in collagen nanostructure and subtle alteration to collagen alignment in the AT but not the SST. Hence, elastin may play only a minor role in tendon mechanical properties. Alternatively, larger changes to tendon mechanics may have been mitigated by developmental compensation of HET tendons and/or the role of elastic fibers may be less prominent in smaller mouse tendons compared to the larger bovine and human tendons evaluated in previous studies. Further research will be necessary to fully elucidate the influence of various elastic fiber components on structure-function relationships in functionally distinct tendons.


Asunto(s)
Elastina/metabolismo , Haploinsuficiencia , Fenómenos Mecánicos , Tendones/citología , Tendones/metabolismo , Animales , Fenómenos Biomecánicos , Masculino , Ensayo de Materiales , Ratones , Estrés Mecánico
15.
J Biol Chem ; 290(35): 21443-59, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26178373

RESUMEN

Fibulin-4 is an extracellular matrix protein essential for elastic fiber formation. Frameshift and missense mutations in the fibulin-4 gene (EFEMP2/FBLN4) cause autosomal recessive cutis laxa (ARCL) 1B, characterized by loose skin, aortic aneurysm, arterial tortuosity, lung emphysema, and skeletal abnormalities. Homozygous missense mutations in FBLN4 are a prevalent cause of ARCL 1B. Here we generated a knock-in mouse strain bearing a recurrent fibulin-4 E57K homozygous missense mutation. The mutant mice survived into adulthood and displayed abnormalities in multiple organ systems, including loose skin, bent forelimb, aortic aneurysm, tortuous artery, and pulmonary emphysema. Biochemical studies of dermal fibroblasts showed that fibulin-4 E57K mutant protein was produced but was prone to dimer formation and inefficiently secreted, thereby triggering an endoplasmic reticulum stress response. Immunohistochemistry detected a low level of fibulin-4 E57K protein in the knock-in skin along with altered expression of selected elastic fiber components. Processing of a precursor to mature lysyl oxidase, an enzyme involved in cross-linking of elastin and collagen, was compromised. The knock-in skin had a reduced level of desmosine, an elastin-specific cross-link compound, and ultrastructurally abnormal elastic fibers. Surprisingly, structurally aberrant collagen fibrils and altered organization into fibers were characteristics of the knock-in dermis and forelimb tendons. Type I collagen extracted from the knock-in skin had decreased amounts of covalent intermolecular cross-links, which could contribute to the collagen fibril abnormalities. Our studies provide the first evidence that fibulin-4 plays a role in regulating collagen fibril assembly and offer a preclinical platform for developing treatments for ARCL 1B.


Asunto(s)
Vasos Sanguíneos/anomalías , Huesos/anomalías , Colágeno Tipo I/metabolismo , Cutis Laxo/patología , Tejido Elástico/anomalías , Proteínas de la Matriz Extracelular/genética , Técnicas de Sustitución del Gen , Piel/patología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Vasos Sanguíneos/patología , Huesos/patología , Colágeno Tipo I/ultraestructura , Reactivos de Enlaces Cruzados/metabolismo , Cutis Laxo/metabolismo , Modelos Animales de Enfermedad , Tejido Elástico/patología , Tejido Elástico/ultraestructura , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/metabolismo , Fibroblastos/enzimología , Fibroblastos/patología , Miembro Anterior/anomalías , Miembro Anterior/diagnóstico por imagen , Miembro Anterior/patología , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Modelos Biológicos , Datos de Secuencia Molecular , Mutación , Biosíntesis de Proteínas , Multimerización de Proteína , Proteína-Lisina 6-Oxidasa/metabolismo , Radiografía , Tendones/anomalías , Tendones/patología , Tendones/ultraestructura
16.
Am J Physiol Renal Physiol ; 311(1): F120-30, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27147675

RESUMEN

Alport syndrome is a familial kidney disease caused by defects in the collagen type IV network of the glomerular basement membrane. Lack of collagen-α3α4α5(IV) changes the glomerular basement membrane morphologically and functionally, rendering it leaky to albumin and other plasma proteins. Filtered albumin has been suggested to be a cause of the glomerular and tubular injuries observed at advanced stages of Alport syndrome. To directly investigate the role that albumin plays in the progression of disease in Alport syndrome, we generated albumin knockout (Alb(-/-)) mice to use as a tool for removing albuminuria as a component of kidney disease. Mice lacking albumin were healthy and indistinguishable from control littermates, although they developed hypertriglyceridemia. Dyslipidemia was observed in Alb(+/-) mice, which displayed half the normal plasma albumin concentration. Alb mutant mice were bred to collagen-α3(IV) knockout (Col4a3(-/-)) mice, which are a model for human Alport syndrome. Lack of circulating and filtered albumin in Col4a3(-/-);Alb(-/-) mice resulted in dramatically improved kidney disease outcomes, as these mice lived 64% longer than did Col4a3(-/-);Alb(+/+) and Col4a3(-/-);Alb(+/-) mice, despite similar blood pressures and serum triglyceride levels. Further investigations showed that the absence of albumin correlated with reduced transforming growth factor-ß1 signaling as well as reduced tubulointerstitial, glomerular, and podocyte pathology. We conclude that filtered albumin is injurious to kidney cells in Alport syndrome and perhaps in other proteinuric kidney diseases, including diabetic nephropathy.


Asunto(s)
Albúminas/metabolismo , Enfermedades Renales/metabolismo , Nefritis Hereditaria/metabolismo , Albúminas/deficiencia , Albúminas/genética , Animales , Autoantígenos/genética , Autoantígenos/metabolismo , Presión Sanguínea , Colágeno Tipo IV/biosíntesis , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Progresión de la Enfermedad , Riñón/patología , Enfermedades Renales/etiología , Enfermedades Renales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nefritis Hereditaria/complicaciones , Nefritis Hereditaria/patología , Análisis de Supervivencia , Factor de Crecimiento Transformador beta1/biosíntesis , Triglicéridos/sangre
17.
Am J Respir Cell Mol Biol ; 52(1): 116-28, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24988442

RESUMEN

The pathogenesis of pulmonary fibrosis involves lung epithelial injury and aberrant proliferation of fibroblasts, and results in progressive pulmonary scarring and declining lung function. In vitro, fibroblast growth factor (FGF) 2 promotes myofibroblast differentiation and proliferation in cooperation with the profibrotic growth factor, transforming growth factor-ß1, but the in vivo requirement for FGF2 in the development of pulmonary fibrosis is not known. The bleomycin model of lung injury and pulmonary fibrosis was applied to Fgf2 knockout (Fgf2(-/-)) and littermate control mice. Weight loss, mortality, pulmonary fibrosis, and histology were analyzed after a single intranasal dose of bleomycin. Inflammation was evaluated in bronchoalveolar lavage (BAL) fluid, and epithelial barrier integrity was assessed by measuring BAL protein and Evans Blue dye permeability. Fgf2 is expressed in mouse and human lung epithelial and inflammatory cells, and, in response to bleomycin, Fgf2(-/-) mice have significantly increased mortality and weight loss. Analysis of BAL fluid and histology show that pulmonary fibrosis is unaltered, but Fgf2(-/-) mice fail to efficiently resolve inflammation, have increased BAL cellularity, and, importantly, deficient recovery of epithelial integrity. Fgf2(-/-) mice similarly have deficient recovery of club cell secretory protein(+) bronchial epithelium in response to naphthalene. We conclude that FGF2 is not required for bleomycin-induced pulmonary fibrosis, but rather is essential for epithelial repair and maintaining epithelial integrity after bleomycin-induced lung injury in mice. These data identify that FGF2 acts as a protective growth factor after lung epithelial injury, and call into question the role of FGF2 as a profibrotic growth factor in vivo.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Bleomicina/efectos adversos , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Lesión Pulmonar , Mucosa Respiratoria/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Bleomicina/farmacología , Lavado Broncoalveolar , Femenino , Factor 2 de Crecimiento de Fibroblastos/genética , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/genética , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Masculino , Ratones , Ratones Noqueados , Naftalenos/farmacología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Mucosa Respiratoria/lesiones , Mucosa Respiratoria/patología
18.
J Physiol ; 593(8): 1931-43, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25627876

RESUMEN

KEY POINTS: Increased large artery stiffness is a hallmark of arterial dysfunction with advancing age and is also present in other disease conditions such as diabetes. Increased large artery stiffness is correlated with resistance artery dysfunction in humans. Using a mouse model of altered arterial elastin content, this is the first study to examine the cause-and-effect relationship between large artery stiffness and peripheral resistance artery function. Our results indicate that mice with genetically greater large artery stiffness have impaired cerebral artery endothelial function, but generally preserved skeletal muscle feed artery endothelial function. The mechanisms for impaired cerebral artery endothelial function are reduced nitric oxide bioavailability and increased oxidative stress. These findings suggest that interventions that target large artery stiffness may be important to reduce disease risk associated with cerebral artery dysfunction in conditions such as advancing age. ABSTRACT: Advancing age as well as diseases such as diabetes are characterized by both increased large artery stiffness and impaired peripheral artery function. It has been hypothesized that greater large artery stiffness causes peripheral artery dysfunction; however, a cause-and-effect relationship has not previously been established. We used elastin heterozygote mice (Eln(+/-) ) as a model of increased large artery stiffness without co-morbidities unrelated to the large artery properties. Aortic stiffness, measured by pulse wave velocity, was ∼35% greater in Eln(+/-) mice than in wild-type (Eln(+/+) ) mice (P = 0.04). Endothelium-dependent dilatation (EDD), assessed by the maximal dilatation to acetylcholine, was ∼40% lower in Eln(+/-) than Eln(+/+) mice in the middle cerebral artery (MCA, P < 0.001), but was similar between groups in the gastrocnemius feed arteries (GFA, P = 0.79). In the MCA, EDD did not differ between groups after incubation with the nitric oxide (NO) synthase inhibitor N(ω) -nitro-l-arginine methyl ester (P > 0.05), indicating that lower NO bioavailability contributed to the impaired EDD in Eln(+/-) mice. Superoxide production and content of the oxidative stress marker nitrotyrosine was higher in MCAs from Eln(+/-) compared with Eln(+/+) mice (P < 0.05). In the MCA, after incubation with the superoxide scavenger TEMPOL, maximal EDD improved by ∼65% in Eln(+/-) (P = 0.002), but was unchanged in Eln(+/+) mice (P = 0.17). These results indicate that greater large artery stiffness has a more profound effect on endothelial function in cerebral arteries compared with skeletal muscle feed arteries. Greater large artery stiffness can cause cerebral artery endothelial dysfunction by reducing NO bioavailability and increasing oxidative stress.


Asunto(s)
Arterias Cerebrales/fisiopatología , Endotelio Vascular/fisiopatología , Músculo Esquelético/irrigación sanguínea , Rigidez Vascular/fisiología , Animales , Arterias Cerebrales/efectos de los fármacos , Modelos Animales de Enfermedad , Elastina/genética , Elastina/metabolismo , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Indometacina/farmacología , Ratones , Ratones Noqueados , NG-Nitroarginina Metil Éster/farmacología , Resistencia Vascular/efectos de los fármacos , Resistencia Vascular/fisiología , Rigidez Vascular/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología
19.
Am J Physiol Heart Circ Physiol ; 309(5): H1008-16, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26232234

RESUMEN

Increased arterial stiffness is a common characteristic of humans with Williams-Beuren syndrome and mouse models of elastin insufficiency. Arterial stiffness is associated with multiple negative cardiovascular outcomes, including myocardial infarction, stroke, and sudden death. Therefore, identifying therapeutic interventions that improve arterial stiffness in response to changes in elastin levels is of vital importance. The goal of this study was to determine the effect of chronic pharmacologic therapy with different classes of antihypertensive medications on arterial stiffness in elastin insufficiency. Elastin-insufficient mice 4-6 wk of age and wild-type littermates were subcutaneously implanted with osmotic micropumps delivering a continuous dose of one of the following: vehicle, losartan, nicardipine, or propranolol for 8 wk. At the end of treatment period, arterial blood pressure and large artery compliance and remodeling were assessed. Our results show that losartan and nicardipine treatment lowered blood pressure and pulse pressure in elastin-insufficient mice. Elastin and collagen content of abdominal aortas as well as ascending aorta and carotid artery biomechanics were not affected by any of the drug treatments in either genotype. By reducing pulse pressure and shifting the working pressure range of an artery to a more compliant region of the pressure-diameter curve, antihypertensive medications may mitigate the consequences of arterial stiffness, an effect that is drug class independent. These data emphasize the importance of early recognition and long-term management of hypertension in Williams-Beuren syndrome and elastin insufficiency.


Asunto(s)
Antihipertensivos/uso terapéutico , Arterias/efectos de los fármacos , Presión Sanguínea , Rigidez Vascular/efectos de los fármacos , Síndrome de Williams/tratamiento farmacológico , Animales , Arterias/fisiopatología , Elastina/deficiencia , Elastina/genética , Losartán/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Nicardipino/uso terapéutico , Propranolol/uso terapéutico , Rigidez Vascular/genética , Síndrome de Williams/fisiopatología
20.
J Biol Chem ; 288(8): 5268-77, 2013 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-23300084

RESUMEN

Type 1 phosphotidylinosotol-4 phosphate 5 kinase γ (PIP5KIγ) is central to generation of phosphotidylinosotol (4,5)P(2) (PI(4,5)P(2)). PIP5KIγ also participates in cytoskeletal organization by delivering talin to integrins, thereby enhancing their ligand binding capacity. As the cytoskeleton is pivotal to osteoclast function, we hypothesized that absence of PIP5KIγ would compromise their resorptive capacity. Absence of the kinase diminishes PI(4,5) abundance and desensitizes precursors to RANK ligand-stimulated differentiation. Thus, PIP5KIγ(-/-) osteoclasts are reduced in number in vitro and confirm physiological relevance in vivo. Despite reduced numbers, PIP5KIγ(-/-) osteoclasts surprisingly have normal cytoskeletons and effectively resorb bone. PIP5KIγ overexpression, which increases PI(4,5)P(2), also delays osteoclast differentiation and reduces cell number but in contrast to cells lacking the kinase, its excess disrupts the cytoskeleton. The cytoskeleton-disruptive effects of excess PIP5KIγ reflect its kinase activity and are independent of talin recognition. The combined arrested differentiation and disorganized cytoskeleton of PIP5KIγ-transduced osteoclasts compromises bone resorption. Thus, optimal PIP5KIγ and PI(4,5)P(2) expression, by osteoclasts, are essential for skeletal homeostasis.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Osteoclastos/citología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Animales , Transporte Biológico , Resorción Ósea , Calcio/metabolismo , Diferenciación Celular , Ligandos , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteoclastos/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositoles/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Plásmidos/metabolismo , Ligando RANK/metabolismo , Espectrometría de Masa por Ionización de Electrospray/métodos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda