Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 89
1.
JCI Insight ; 8(10)2023 05 22.
Article En | MEDLINE | ID: mdl-37022786

To improve our limited understanding of the pathogenesis of thoracic aortic aneurysm (TAA) that leads to acute aortic dissection, single-cell RNA sequencing (scRNA-seq) was employed to profile disease-relevant transcriptomic changes of aortic cell populations in a well-characterized mouse model of the most commonly diagnosed form of Marfan syndrome (MFS). As result, 2 discrete subpopulations of aortic cells (SMC3 and EC4) were identified only in the aorta of Fbn1mgR/mgR mice. SMC3 cells highly express genes related to extracellular matrix formation and nitric oxide signaling, whereas the EC4 transcriptional profile is enriched in smooth muscle cell (SMC), fibroblast, and immune cell-related genes. Trajectory analysis predicted close phenotypic modulation between SMC3 and EC4, which were therefore analyzed together as a discrete MFS-modulated (MFSmod) subpopulation. In situ hybridization of diagnostic transcripts located MFSmod cells at the intima of Fbn1mgR/mgR aortas. Reference-based data set integration revealed transcriptomic similarity between MFSmod- and SMC-derived cell clusters modulated in human TAA. Consistent with the angiotensin II type I receptor (At1r) contribution to TAA development, MFSmod cells were absent in the aorta of Fbn1mgR/mgR mice treated with the At1r antagonist losartan. Altogether, our findings indicate that a discrete dynamic alteration of aortic cell identity is associated with dissecting TAA in MFS mice and increased risk of aortic dissection in MFS patients.


Aortic Aneurysm, Thoracic , Aortic Aneurysm , Aortic Dissection , Marfan Syndrome , Humans , Mice , Animals , Transcriptome , Losartan/pharmacology , Marfan Syndrome/complications , Marfan Syndrome/genetics , Marfan Syndrome/pathology , Aortic Aneurysm/genetics , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/pathology , Aorta/pathology , Aortic Dissection/genetics
2.
Hum Mol Genet ; 31(19): 3281-3289, 2022 09 29.
Article En | MEDLINE | ID: mdl-35567544

A disproportionate tall stature is the most evident manifestation in Marfan syndrome (MFS), a multisystem condition caused by mutations in the extracellular protein and TGFß modulator, fibrillin-1. Unlike cardiovascular manifestations, there has been little effort devoted to unravel the molecular mechanism responsible for long bone overgrowth in MFS. By combining the Cre-LoxP recombination system with metatarsal bone cultures, here we identify the outer layer of the perichondrium as the tissue responsible for long bone overgrowth in MFS mice. Analyses of differentially expressed genes in the fibrillin-1-deficient perichondrium predicted that loss of TGFß signaling may influence chondrogenesis in the neighboring epiphyseal growth plate (GP). Immunohistochemistry revealed that fibrillin-1 deficiency in the outer perichondrium is associated with decreased accumulation of latent TGFß-binding proteins (LTBPs)-3 and -4, and reduced levels of phosphorylated (activated) Smad2. Consistent with these findings, mutant metatarsal bones grown in vitro were longer and released less TGFß than the wild-type counterparts. Moreover, addition of recombinant TGFß1 normalized linear growth of mutant metatarsal bones. We conclude that longitudinal bone overgrowth in MFS is accounted for by diminished sequestration of LTBP-3 and LTBP-4 into the fibrillin-1-deficient matrix of the outer perichondrium, which results in less TGFß signaling locally and improper GP differentiation distally.


Marfan Syndrome , Animals , Fibrillin-1/genetics , Fibrillin-2 , Fibrillins , Latent TGF-beta Binding Proteins/genetics , Latent TGF-beta Binding Proteins/metabolism , Marfan Syndrome/genetics , Mice , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
3.
Int J Mol Sci ; 23(3)2022 Feb 08.
Article En | MEDLINE | ID: mdl-35163812

Fibrillin-1 is the major structural component of the 10 nm-diameter microfibrils that confer key physical and mechanical properties to virtually every tissue, alone and together with elastin in the elastic fibers. Mutations in fibrillin-1 cause pleiotropic manifestations in Marfan syndrome (MFS), including dissecting thoracic aortic aneurysms, myocardial dysfunction, progressive bone loss, disproportionate skeletal growth, and the dislocation of the crystalline lens. The characterization of these MFS manifestations in mice, that replicate the human phenotype, have revealed that the underlying mechanisms are distinct and organ-specific. This brief review summarizes relevant findings supporting this conclusion.


Fibrillin-1/genetics , Marfan Syndrome/pathology , Animals , Disease Models, Animal , Humans , Marfan Syndrome/genetics , Mice , Mutation , Organ Specificity
4.
Biomolecules ; 12(1)2022 01 14.
Article En | MEDLINE | ID: mdl-35053276

About 20% of individuals afflicted with thoracic aortic disease have single-gene mutations that predispose the vessel to aneurysm formation and/or acute aortic dissection often without associated syndromic features. One widely studied exception is Marfan syndrome (MFS) in which mutations in the extracellular protein fibrillin-1 cause additional abnormalities in the heart, eyes, and skeleton. Mouse models of MFS have been instrumental in delineating major cellular and molecular determinants of thoracic aortic disease. In spite of research efforts, translating experimental findings from MFS mice into effective drug therapies for MFS patients remains an unfulfilled promise. Here, we describe a series of studies that have implicated endothelial dysfunction and improper angiotensin II and TGFß signaling in driving thoracic aortic disease in MFS mice. We also discuss how these investigations have influenced the way we conceptualized possible new therapies to slow down or even halt aneurysm progression in this relatively common connective tissue disorder.


Aortic Aneurysm, Thoracic , Aortic Dissection , Marfan Syndrome , Aortic Dissection/genetics , Angiotensin II , Animals , Aorta/metabolism , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/metabolism , Humans , Marfan Syndrome/complications , Marfan Syndrome/drug therapy , Marfan Syndrome/genetics , Mice
5.
Dev Dyn ; 251(1): 95-104, 2022 01.
Article En | MEDLINE | ID: mdl-33742701

The purpose of this review is to discuss the transforming growth factor beta (TGFB) binding proteins (LTBP) with respect to their participation in the activity of TGFB. We first describe pertinent aspects of the biology and cell function of the LTBPs. We then summarize the physiological consequences of LTBP loss in humans and mice. Finally, we consider a number of outstanding questions relating to LTBP function.


Latent TGF-beta Binding Proteins , Animals , Humans , Latent TGF-beta Binding Proteins/genetics , Latent TGF-beta Binding Proteins/metabolism , Mice , Protein Binding , Signal Transduction , Transforming Growth Factor beta/metabolism
6.
Arterioscler Thromb Vasc Biol ; 41(9): 2483-2493, 2021 09.
Article En | MEDLINE | ID: mdl-34320838

Objective: Despite considerable research, the goal of finding nonsurgical remedies against thoracic aortic aneurysm and acute aortic dissection remains elusive. We sought to identify a novel aortic PK (protein kinase) that can be pharmacologically targeted to mitigate aneurysmal disease in a well-established mouse model of early-onset progressively severe Marfan syndrome (MFS). Approach and Results: Computational analyses of transcriptomic data derived from the ascending aorta of MFS mice predicted a probable association between thoracic aortic aneurysm and acute aortic dissection development and the multifunctional, stress-activated HIPK2 (homeodomain-interacting protein kinase 2). Consistent with this prediction, Hipk2 gene inactivation significantly extended the survival of MFS mice by slowing aneurysm growth and delaying transmural rupture. HIPK2 also ranked among the top predicted PKs in computational analyses of DEGs (differentially expressed genes) in the dilated aorta of 3 MFS patients, which strengthened the clinical relevance of the experimental finding. Additional in silico analyses of the human and mouse data sets identified the TGF (transforming growth factor)-ß/Smad3 signaling pathway as a potential target of HIPK2 in the MFS aorta. Chronic treatment of MFS mice with an allosteric inhibitor of HIPK2-mediated stimulation of Smad3 signaling validated this prediction by mitigating thoracic aortic aneurysm and acute aortic dissection pathology and partially improving aortic material stiffness. Conclusions: HIPK2 is a previously unrecognized determinant of aneurysmal disease and an attractive new target for antithoracic aortic aneurysm and acute aortic dissection multidrug therapy.


Aorta, Thoracic/drug effects , Aortic Aneurysm, Thoracic/prevention & control , Aortic Dissection/prevention & control , Fibrillin-1/genetics , Marfan Syndrome/genetics , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Vascular Remodeling/drug effects , Adult , Aortic Dissection/enzymology , Aortic Dissection/genetics , Aortic Dissection/pathology , Animals , Aorta, Thoracic/enzymology , Aorta, Thoracic/pathology , Aortic Aneurysm, Thoracic/enzymology , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Dilatation, Pathologic , Disease Models, Animal , Disease Progression , Humans , Male , Marfan Syndrome/complications , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Severity of Illness Index , Signal Transduction , Smad3 Protein/metabolism
7.
Physiol Rep ; 7(21): e14267, 2019 11.
Article En | MEDLINE | ID: mdl-31724332

Fibrillin-1 mutations cause pathological changes in connective tissue that constitute the complex phenotype of Marfan syndrome. In this study, we used fibrillin-1 hypomorphic and haploinsufficient mice (Fbn1mgr/mgR and Fbn1+/- mice, respectively) to investigate the impact of fibrillin-1 deficiency alone or in combination with regular physical activity on tendon tissue morphology and mechanical properties. Morphological and biomechanical analyses revealed that Fbn1mgr/mgR but not Fbn1+/- mice displayed smaller tendons with physical properties that were unremarkable when normalized to tendon size. Fbn1mgR/mgR mice (n = 43) Fbn1+/- mice (n = 27) and wild-type mice (WT, n = 25) were randomly assigned to either control cage conditions (n = 54) or to a running on a running wheel for 4 weeks (n = 41). Both fibrillin-1-deficient mice ran voluntarily on the running wheel in a manner similar to WT mice (3-4 km/24 h). Regular exercise did not mitigate aneurysm progression in Fbn1mgR/mgR mice (P < 0.05) as evidenced by unmodified median survival. In spite of the smaller size, tendons of fibrillin-1-deficient mice subjected to regular exercise showed no evidence of overt histopathological changes or tissue overload. We therefore concluded that lack of optimal fibrillin-1 synthesis leads to a down regulation of integrated tendon formation, rather than to a loss of tendon quality, which also implies that fibrillin-1 deficiency in combination with exercise is not a suitable animal model for studying the development of tendon overuse (tendinopathy).


Fibrillin-1/physiology , Physical Conditioning, Animal/physiology , Tendons/pathology , Tendons/physiopathology , Animals , Biomechanical Phenomena , Fibrillin-1/genetics , Male , Mice, Inbred C57BL , Mice, Transgenic
8.
JCI Insight ; 4(11)2019 06 06.
Article En | MEDLINE | ID: mdl-31167969

Marfan syndrome (MFS) is associated with mutations in fibrillin-1 that predispose afflicted individuals to progressive thoracic aortic aneurysm (TAA) leading to dissection and rupture of the vessel wall. Here we combined computational and experimental approaches to identify and test FDA-approved drugs that may slow or even halt aneurysm progression. Computational analyses of transcriptomic data derived from the aortas of MFS patients and MFS mice (Fbn1mgR/mgR mice) predicted that subcellular pathways associated with reduced muscle contractility are key TAA determinants that could be targeted with the GABAB receptor agonist baclofen. Systemic administration of baclofen to Fbn1mgR/mgR mice validated our computational prediction by mitigating arterial disease progression at the cellular and physiological levels. Interestingly, baclofen improved muscle contraction-related subcellular pathways by upregulating a different set of genes than those downregulated in the aorta of vehicle-treated Fbn1mgR/mgR mice. Distinct transcriptomic profiles were also associated with drug-treated MFS and wild-type mice. Thus, systems pharmacology approaches that compare patient- and mouse-derived transcriptomic data for subcellular pathway-based drug repurposing represent an effective strategy to identify potential new treatments of human diseases.


Aortic Aneurysm, Thoracic , Drug Repositioning/methods , Transcriptome/drug effects , Animals , Aortic Aneurysm, Thoracic/drug therapy , Aortic Aneurysm, Thoracic/etiology , Aortic Aneurysm, Thoracic/prevention & control , Cardiovascular Agents/pharmacology , Cardiovascular Agents/therapeutic use , Disease Models, Animal , Gene Expression Profiling , Humans , Marfan Syndrome/complications , Mice , Mice, Transgenic
9.
Arterioscler Thromb Vasc Biol ; 39(2): 126-136, 2019 02.
Article En | MEDLINE | ID: mdl-30651002

Thoracic aortic aneurysms that progress to acute aortic dissections are often fatal. Thoracic aneurysms have been managed with treatment with ß-adrenergic blocking agents (ß-blockers) and routine surveillance imaging, followed by surgical repair of the aneurysm when the risk of dissection exceeds the risk for repair. Thus, there is a window to initiate therapies to slow aortic enlargement and delay or ideally negate the need for surgical repair of the aneurysm to prevent a dissection. Mouse models of Marfan syndrome-a monogenic disorder predisposing to thoracic aortic disease-have been used extensively to identify such therapies. The initial finding that TGFß (transformation growth factor-ß) signaling was increased in the aortic media of a Marfan syndrome mouse model and that its inhibition via TGFß neutralization or At1r (Ang II [angiotensin II] type I receptor) antagonism prevented aneurysm development was generally viewed as a groundbreaking discovery that could be translated into the first cure of thoracic aortic disease. However, several large randomized trials of pediatric and adult patients with Marfan syndrome have subsequently yielded no evidence that At1r antagonism by losartan slows aortic enlargement more effectively than conventional treatment with ß-blockers. Subsequent studies in mouse models have begun to resolve the complex molecular pathophysiology underlying onset and progression of aortic disease and have emphasized the need to preserve TGFß signaling to prevent aneurysm formation. This review describes critical experiments that have influenced the evolution of our understanding of thoracic aortic disease, in addition to discussing old controversies and identifying new therapeutic opportunities.


Aortic Aneurysm, Thoracic/therapy , Aortic Dissection/therapy , Acute Disease , Angiotensin II/physiology , Animals , Aortic Aneurysm, Thoracic/etiology , Humans , Losartan/therapeutic use , Mice , Signal Transduction/physiology , Transforming Growth Factor beta/physiology
10.
Curr Top Dev Biol ; 130: 193-216, 2018.
Article En | MEDLINE | ID: mdl-29853177

The extracellular matrix (ECM) is a highly heterogeneous mixture of macromolecules capable of self-assembling into tissue-specific suprastructures that constitute the architectural elements supporting organ function. Contrary to the traditional view of being a static scaffold that supports tissue integrity along with cell adhesion and migration, the ECM is an inherently dynamic system that specifies cellular function and defines the limits and patterns of tissue organization. Throughout evolution, the composition and organization of the ECM have changed to accommodate basic and new tissue functions, both in terms of providing structural support and integrating multivalent signals to cells. In this review, we will highlight some of these bidirectional cell-matrix interactions that guide the development of a mechanically compliant vascular system. Specifically, we will focus on studies that have investigated how ECM composition and physical properties influence cell fate decisions associated with vascular tissue development and homeostasis and implicitly, vascular disease.


Arteries/growth & development , Arteries/physiology , Extracellular Matrix/physiology , Homeostasis/physiology , Morphogenesis/physiology , Animals , Cardiovascular System/growth & development , Cell Differentiation , Humans , Invertebrates/growth & development , Invertebrates/physiology , Vertebrates/growth & development , Vertebrates/physiology
11.
JCI Insight ; 3(5)2018 03 08.
Article En | MEDLINE | ID: mdl-29515038

Proteoglycan accumulation is a hallmark of medial degeneration in thoracic aortic aneurysm and dissection (TAAD). Here, we defined the aortic proteoglycanome using mass spectrometry, and based on the findings, investigated the large aggregating proteoglycans aggrecan and versican in human ascending TAAD and a mouse model of severe Marfan syndrome. The aortic proteoglycanome comprises 20 proteoglycans including aggrecan and versican. Antibodies against these proteoglycans intensely stained medial degeneration lesions in TAAD, contrasting with modest intralamellar staining in controls. Aggrecan, but not versican, was increased in longitudinal analysis of Fbn1mgR/mgR aortas. TAAD and Fbn1mgR/mgR aortas had increased aggrecan and versican mRNAs, and reduced expression of a key proteoglycanase gene, ADAMTS5, was seen in TAAD. Fbn1mgR/mgR mice with ascending aortic dissection and/or rupture had dramatically increased aggrecan staining compared with mice without these complications. Thus, aggrecan and versican accumulation in ascending TAAD occurs via increased synthesis and/or reduced proteolytic turnover, and correlates with aortic dissection/rupture in Fbn1mgR/mgR mice. Tissue swelling imposed by aggrecan and versican is proposed to be profoundly deleterious to aortic wall mechanics and smooth muscle cell homeostasis, predisposing to type-A dissections. These proteoglycans provide potential biomarkers for refined risk stratification and timing of elective aortic aneurysm repair.


Aggrecans/metabolism , Aortic Aneurysm, Thoracic/pathology , Aortic Dissection/pathology , Versicans/metabolism , ADAMTS5 Protein/metabolism , Adult , Aged , Aged, 80 and over , Aortic Dissection/diagnosis , Aortic Dissection/etiology , Aortic Dissection/prevention & control , Animals , Aorta, Thoracic/pathology , Aortic Aneurysm, Thoracic/diagnosis , Aortic Aneurysm, Thoracic/etiology , Aortic Aneurysm, Thoracic/surgery , Biomarkers/metabolism , Disease Models, Animal , Female , Fibrillin-1/genetics , Gene Expression Profiling , Humans , Male , Marfan Syndrome/complications , Marfan Syndrome/genetics , Marfan Syndrome/pathology , Mice, Knockout , Middle Aged , RNA, Messenger/metabolism , Risk Assessment/methods , Tunica Media/pathology
12.
Arterioscler Thromb Vasc Biol ; 38(3): 588-591, 2018 03.
Article En | MEDLINE | ID: mdl-29371244

OBJECTIVE: Two were the aims of this study: first, to translate whole-genome expression profiles into computational predictions of functional associations between signaling pathways that regulate aorta homeostasis and the activity of angiotensin II type 1a receptor (At1ar) in either vascular endothelial or smooth muscle cells; and second, to characterize the impact of endothelial cell- or smooth muscle cell-specific At1ar disruption on the development of thoracic aortic aneurysm in fibrillin-1 hypomorphic (Fbn1mgR/mgR ) mice, a validated animal model of early onset progressively severe Marfan syndrome. APPROACH AND RESULTS: Cdh5-Cre and Sm22-Cre transgenic mice were used to inactivate the At1ar-coding gene (Agt1ar) in either intimal or medial cells of both wild type and Marfan syndrome mice, respectively. Computational analyses of differentially expressed genes predicted dysregulated signaling pathways of cell survival and matrix remodeling in Agt1arCdh5-/- aortas and of cell adhesion and contractility in Agt1arSm22-/- aortas. Characterization of Fbn1mgR/mgR;Agt1arCdh5-/- mice revealed increased median survival associated with mitigated aneurysm growth and media degeneration, as well as reduced levels of phosphorylated (p-) Erk1/2 but not p-Smad2. By contrast, levels of both p-Erk1/2 and p-Smad2 proteins were normalized in Fbn1mgR/mgR;Agt1arSm22-/- aortas in spite of them showing no appreciable changes in thoracic aortic aneurysm pathology. CONCLUSIONS: Physiological At1ar signaling in the intimal and medial layers is associated with distinct regulatory processes of aorta homeostasis and function; improper At1ar activity in the vascular endothelium is a significant determinant of thoracic aortic aneurysm development in Marfan syndrome mice.


Aorta, Thoracic/metabolism , Aortic Aneurysm, Thoracic/metabolism , Endothelial Cells/metabolism , Myocytes, Smooth Muscle/metabolism , Receptor, Angiotensin, Type 1/metabolism , Animals , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/pathology , Aortic Aneurysm, Thoracic/physiopathology , Computational Biology , Dilatation, Pathologic , Disease Models, Animal , Endothelial Cells/pathology , Fibrillin-1/genetics , Fibrillin-1/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation , Homeostasis , Male , Marfan Syndrome/genetics , Marfan Syndrome/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/pathology , Receptor, Angiotensin, Type 1/deficiency , Receptor, Angiotensin, Type 1/genetics , Signal Transduction
13.
Matrix Biol ; 71-72: 82-89, 2018 10.
Article En | MEDLINE | ID: mdl-28782645

Mutations in fibrillin-1 cause Marfan syndrome (MFS), the most common heritable disorder of connective tissue. Fibrillin-1 assemblies (microfibrils and elastic fibers) represent a unique dual-function component of the architectural matrix. The first role is structural for they endow tissues with tensile strength and elasticity, transmit forces across them and demarcate functionally discrete areas within them. The second role is instructive in that these macroaggregates modulate a large variety of sub-cellular processes by interacting with mechanosensors, and integrin and syndecan receptors, and by modulating the bioavailability of local TGFß signals. The multifunctional, tissue-specific nature of fibrillin-1 assemblies is reflected in the variety of clinical manifestations and disease mechanisms associated with the MFS phenotype. Characterization of mice with ubiquitous or cell type-restricted fibrillin-1 deficiency has unraveled some pathophysiological mechanisms associated with the MFS phenotype, such as altered mechanotransduction in the heart, dysregulated TGFß signaling in the ascending aorta and perturbed stem cell fate in the bone marrow. In each case, potential druggable targets have also been identified. However, the finding that distinct disease mechanisms underlie different organ abnormalities strongly argues for developing multi-drug strategies to mitigate or even prevent both life-threatening and morbid manifestations in pediatric and adult MFS patients.


Fibrillin-1/genetics , Marfan Syndrome/metabolism , Mechanotransduction, Cellular , Stem Cells/metabolism , Transforming Growth Factor beta/metabolism , Animals , Disease Models, Animal , Humans , Marfan Syndrome/genetics , Mutation , Myocardium/metabolism , Signal Transduction
14.
Annu Rev Med ; 68: 51-67, 2017 01 14.
Article En | MEDLINE | ID: mdl-28099082

Thoracic aortic diseases, including aneurysms and dissections of the thoracic aorta, are a major cause of morbidity and mortality. Risk factors for thoracic aortic disease include increased hemodynamic forces on the ascending aorta, typically due to poorly controlled hypertension, and heritable genetic variants. The altered genes predisposing to thoracic aortic disease either disrupt smooth muscle cell (SMC) contraction or adherence to an impaired extracellular matrix, or decrease canonical transforming growth factor beta (TGF-ß) signaling. Paradoxically, TGF-ß hyperactivity has been postulated to be the primary driver for the disease. More recently, it has been proposed that the response of aortic SMCs to the hemodynamic load on a structurally defective aorta is the primary driver of thoracic aortic disease, and that TGF-ß overactivity in diseased aortas is a secondary, unproductive response to restore tissue function. The engineering of mouse models of inherited aortopathies has identified potential therapeutic agents to prevent thoracic aortic disease.


Angiotensin II/metabolism , Aortic Aneurysm, Thoracic/genetics , Aortic Dissection/genetics , Marfan Syndrome/therapy , Transforming Growth Factor beta/metabolism , Aortic Dissection/metabolism , Aortic Dissection/prevention & control , Animals , Antihypertensive Agents/therapeutic use , Aortic Aneurysm, Thoracic/metabolism , Aortic Aneurysm, Thoracic/prevention & control , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Losartan/therapeutic use , Marfan Syndrome/genetics , Mechanoreceptors , Mice , Muscle, Smooth, Vascular/physiopathology , Signal Transduction
15.
Ann Biomed Eng ; 44(10): 2994-3006, 2016 10.
Article En | MEDLINE | ID: mdl-27090893

Marfan syndrome (MFS) is an autosomal dominant disease of the connective tissue due to mutations in the fibrillin-1 gene (FBN1). This study aimed at characterizing microelastic properties of the ascending aortic wall and lung parenchyma tissues from wild type (WT) and age-matched Fbn1 hypomorphic mice (Fbn1(mgR/mgR) mice) to identify tissue-specific biomechanical effects of aging and disease in MFS. Atomic force microscopy was used to indent lung parenchyma and aortic wall tissues, using Hybrid Eshelby Decomposition analysis to extract layer-specific properties of the intima and media. The intima stiffened with age and was not different between WT and Fbn1(mgR/mgR) tissues, whereas the media layer of MFS aortas showed progressive structural and mechanical degradation with a modulus that was 50% softer than WT by 3.5 months of age. Similarly, MFS mice displayed progressive structural and mechanical deterioration of lung tissue, which was over 85% softer than WT by 3.5 months of age. Chronic treatment with the angiotensin type I receptor antagonist, losartan, attenuated the aorta and lung tissue degradation, resulting in structural and mechanical properties not significantly different from age-matched WT controls. By revealing micromechanical softening of elastin-rich aorta and lung tissues with disease progression in fibrillin-1 deficient mice, our findings support the use of losartan as a prophylactic treatment that may abrogate the life-threatening symptoms of MFS.


Aorta , Losartan/pharmacology , Lung , Marfan Syndrome , Animals , Aorta/metabolism , Aorta/pathology , Aorta/physiopathology , Disease Models, Animal , Fibrillin-1/genetics , Fibrillin-1/metabolism , Humans , Lung/metabolism , Lung/pathology , Lung/physiopathology , Marfan Syndrome/drug therapy , Marfan Syndrome/genetics , Marfan Syndrome/metabolism , Marfan Syndrome/physiopathology , Mice , Mice, Transgenic
16.
Matrix Biol ; 52-54: 88-94, 2016.
Article En | MEDLINE | ID: mdl-26610678

We have recently demonstrated that fibrillin-1 assemblies regulate the fate of skeletal stem cells (aka, mesenchymal stem cells [MSCs]) by modulating TGFß activity within the microenvironment of adult bone marrow niches. Since MSCs can also influence hematopoietic stem cell (HSC) activities, here we investigated adult hematopoiesis in mice with Cre-mediated inactivation of the fibrillin-1 (Fbn1) gene in the mesenchyme of the forming limbs (Fbn1(Prx1-/-) mice). Analyses of 3-month-old Fbn1(Prx1-/-) mice revealed a statistically significant increase of circulating red blood cells, which a differentiation assay correlated with augmented erythropoiesis. This finding, together with evidence of fibrillin-1 deposition in erythroblastic niches, supported the notion that this extracellular matrix protein normally restricts differentiation of erythroid progenitors. Whereas flow cytometry measurements identified a decreased HSC frequency in mutant relative to wild type mice, no appreciable differences were noted with regard to the relative abundance and differentiation potential of myeloid progenitor cells. Together these findings implied that fibrillin-1 normally promotes HSC expansion but does not influence cell lineage commitment. Since local TGFß hyperactivity has been associated with abnormal osteogenesis in Fbn1(Prx1-/-) mice, 1-month-old mutant and wild type animals were systemically treated for 8weeks with either a pan-TGF-ß-neutralizing antibody or an antibody of the same IgG1 isotype. The distinct outcomes of these pharmacological interventions strongly suggest that fibrillin-1 differentially modulates TGFß activity in HSC vs. erythroid niches.


Adult Stem Cells/metabolism , Bone Marrow Cells/cytology , Fibrillin-1/genetics , Hematopoiesis , Animals , Cell Differentiation , Fibrillin-1/metabolism , Flow Cytometry , Gene Knockout Techniques , Humans , Mice , Signal Transduction , Stem Cell Niche , Transforming Growth Factor beta/metabolism
17.
Matrix Biol ; 52-54: 191-197, 2016.
Article En | MEDLINE | ID: mdl-26408953

The severe skeletal abnormalities associated with Marfan syndrome (MFS) and congenital contractural arachnodactyly (CCA) underscore the notion that fibrillin assemblies (microfibrils and elastic fibers) play a critical role in bone formation and function in spite of representing a low abundance component of skeletal matrices. Studies of MFS and CCA mice have correlated the skeletal phenotypes of these mutant animals with distinct pathophysiological mechanisms that reflect the contextual contribution of fibrillin-1 and -2 scaffolds to TGFß and BMP signaling during bone patterning, growth and metabolism. Illustrative examples include the unique role of fibrillin-2 in regulating BMP-dependent limb patterning and the distinct impact of the two fibrillin proteins on the commitment and differentiation of marrow mesenchymal stem cells. Collectively, these findings have important implication for our understanding of the pathophysiological mechanisms that drive age- and injury-related processes of bone degeneration.


Arachnodactyly/pathology , Contracture/pathology , Fibrillin-1/genetics , Fibrillin-2/genetics , Marfan Syndrome/pathology , Animals , Arachnodactyly/genetics , Arachnodactyly/metabolism , Body Patterning , Cell Differentiation , Contracture/genetics , Contracture/metabolism , Disease Models, Animal , Fibrillin-1/metabolism , Fibrillin-2/metabolism , Humans , Marfan Syndrome/genetics , Marfan Syndrome/metabolism , Mesenchymal Stem Cells/cytology , Mice , Mutation , Osteogenesis , Signal Transduction , Transforming Growth Factor beta/metabolism
18.
J Bone Miner Res ; 31(1): 86-97, 2016 Jan.
Article En | MEDLINE | ID: mdl-26189658

A full understanding of the microenvironmental factors that control the activities of skeletal stem cells (also known as mesenchymal stem cells [MSCs]) in the adult bone marrow holds great promise for developing new therapeutic strategies to mitigate age-related diseases of bone and cartilage degeneration. Bone loss is an understudied manifestation of Marfan syndrome, a multisystem disease associated with mutations in the extracellular matrix protein and TGFß modulator fibrillin-1. Here we demonstrate that progressive loss of cancellous bone in mice with limbs deficient for fibrillin-1 (Fbn1(Prx1-/-) mice) is accounted for by premature depletion of MSCs and osteoprogenitor cells combined with constitutively enhanced bone resorption. Longitudinal analyses of Fbn1(Prx1-/-) mice showed incremental bone loss and trabecular microarchitecture degeneration accompanied by a progressive decrease in the number and clonogenic potential of MSCs. Significant paucity of marrow fat cells in the long bones of Fbn1(Prx1-/-) mice, together with reduced adipogenic potential of marrow stromal cell cultures, indicated an additional defect in MSC differentiation. This postulate was corroborated by showing that an Fbn1-silenced osteoprogenitor cell line cultured in the presence of insulin yielded fewer than normal adipocytes and exhibited relatively lower PPARγ levels. Consonant with fibrillin-1 modulation of TGFß bioavailability, cultures of marrow stromal cells from Fbn1(Prx1-/-) limb bones showed improper overactivation of latent TGFß. In line with this finding, systemic TGFß neutralization improved bone mass and trabecular microarchitecture along with normalizing the number of MSCs, osteoprogenitor cells, and marrow adipocytes. Collectively, our findings show that fibrillin-1 regulates MSC activity by modulating TGFß bioavailability within the microenvironment of marrow niches.


Bone Marrow/metabolism , Cell Differentiation/physiology , Mesenchymal Stem Cells/metabolism , Microfilament Proteins/metabolism , Stem Cell Niche/physiology , Transforming Growth Factor beta/metabolism , Animals , Fibrillin-1 , Fibrillins , Mice , Mice, Knockout , Microfilament Proteins/genetics , Transforming Growth Factor beta/genetics
19.
Proc Natl Acad Sci U S A ; 112(45): 14012-7, 2015 Nov 10.
Article En | MEDLINE | ID: mdl-26494287

Marfan syndrome (MFS) is an autosomal dominant disorder of connective tissue, caused by mutations of the microfibrillar protein fibrillin-1, that predisposes affected individuals to aortic aneurysm and rupture and is associated with increased TGFß signaling. TGFß is secreted from cells as a latent complex consisting of TGFß, the TGFß propeptide, and a molecule of latent TGFß binding protein (LTBP). Improper extracellular localization of the latent complex can alter active TGFß levels, and has been hypothesized as an explanation for enhanced TGFß signaling observed in MFS. We previously reported the absence of LTBP-3 in matrices lacking fibrillin-1, suggesting that perturbed TGFß signaling in MFS might be due to defective interaction of latent TGFß complexes containing LTBP-3 with mutant fibrillin-1 microfibrils. To test this hypothesis, we genetically suppressed Ltbp3 expression in a mouse model of progressively severe MFS. Here, we present evidence that MFS mice lacking LTBP-3 have improved survival, essentially no aneurysms, reduced disruption and fragmentation of medial elastic fibers, and decreased Smad2/3 and Erk1/2 activation in their aortas. These data suggest that, in MFS, improper localization of latent TGFß complexes composed of LTBP-3 and TGFß contributes to aortic disease progression.


Aortic Aneurysm, Thoracic/metabolism , Latent TGF-beta Binding Proteins/metabolism , Marfan Syndrome/complications , Marfan Syndrome/genetics , Multiprotein Complexes/metabolism , Transforming Growth Factor beta/metabolism , Analysis of Variance , Animals , Aortic Aneurysm, Thoracic/etiology , DNA, Complementary/biosynthesis , Fibrillin-1 , Fibrillins , Immunohistochemistry , Latent TGF-beta Binding Proteins/deficiency , Mice , Microfilament Proteins/genetics , Muscle, Smooth, Vascular/cytology , Real-Time Polymerase Chain Reaction
20.
PLoS Genet ; 11(6): e1005340, 2015 Jun.
Article En | MEDLINE | ID: mdl-26114882

Fibrillins are large extracellular macromolecules that polymerize to form the backbone structure of connective tissue microfibrils. Mutations in the gene for fibrillin-1 cause the Marfan syndrome, while mutations in the gene for fibrillin-2 cause Congenital Contractural Arachnodactyly. Both are autosomal dominant disorders, and both disorders affect musculoskeletal tissues. Here we show that Fbn2 null mice (on a 129/Sv background) are born with reduced muscle mass, abnormal muscle histology, and signs of activated BMP signaling in skeletal muscle. A delay in Myosin Heavy Chain 8, a perinatal myosin, was found in Fbn2 null forelimb muscle tissue, consistent with the notion that muscle defects underlie forelimb contractures in these mice. In addition, white fat accumulated in the forelimbs during the early postnatal period. Adult Fbn2 null mice are already known to demonstrate persistent muscle weakness. Here we measured elevated creatine kinase levels in adult Fbn2 null mice, indicating ongoing cycles of muscle injury. On a C57Bl/6 background, Fbn2 null mice showed severe defects in musculature, leading to neonatal death from respiratory failure. These new findings demonstrate that loss of fibrillin-2 results in phenotypes similar to those found in congenital muscular dystrophies and that FBN2 should be considered as a candidate gene for recessive congenital muscular dystrophy. Both in vivo and in vitro evidence associated muscle abnormalities and accumulation of white fat in Fbn2 null mice with abnormally activated BMP signaling. Genetic rescue of reduced muscle mass and accumulation of white fat in Fbn2 null mice was accomplished by deleting a single allele of Bmp7. In contrast to other reports that activated BMP signaling leads to muscle hypertrophy, our findings demonstrate the exquisite sensitivity of BMP signaling to the fibrillin-2 extracellular environment during early postnatal muscle development. New evidence presented here suggests that fibrillin-2 can sequester BMP complexes in a latent state.


Bone Morphogenetic Proteins/metabolism , Microfilament Proteins/genetics , Muscular Diseases/genetics , Animals , Animals, Newborn , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Proteins/genetics , Creatine Kinase/blood , Female , Fibrillin-1 , Fibrillin-2 , Fibrillins , Gene Expression Regulation , Limb Deformities, Congenital/genetics , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Microfilament Proteins/metabolism , Muscle, Skeletal/abnormalities , Muscle, Skeletal/pathology , Muscular Diseases/physiopathology , Muscular Dystrophies/genetics , Organ Culture Techniques , Signal Transduction/genetics
...