Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Elife ; 122024 Jun 10.
Article in English | MEDLINE | ID: mdl-38856718

ABSTRACT

Abnormal lung development can cause congenital pulmonary cysts, the mechanisms of which remain largely unknown. Although the cystic lesions are believed to result directly from disrupted airway epithelial cell growth, the extent to which developmental defects in lung mesenchymal cells contribute to abnormal airway epithelial cell growth and subsequent cystic lesions has not been thoroughly examined. In the present study using genetic mouse models, we dissected the roles of bone morphogenetic protein (BMP) receptor 1a (Bmpr1a)-mediated BMP signaling in lung mesenchyme during prenatal lung development and discovered that abrogation of mesenchymal Bmpr1a disrupted normal lung branching morphogenesis, leading to the formation of prenatal pulmonary cystic lesions. Severe deficiency of airway smooth muscle cells and subepithelial elastin fibers were found in the cystic airways of the mesenchymal Bmpr1a knockout lungs. In addition, ectopic mesenchymal expression of BMP ligands and airway epithelial perturbation of the Sox2-Sox9 proximal-distal axis were detected in the mesenchymal Bmpr1a knockout lungs. However, deletion of Smad1/5, two major BMP signaling downstream effectors, from the lung mesenchyme did not phenocopy the cystic abnormalities observed in the mesenchymal Bmpr1a knockout lungs, suggesting that a Smad-independent mechanism contributes to prenatal pulmonary cystic lesions. These findings reveal for the first time the role of mesenchymal BMP signaling in lung development and a potential pathogenic mechanism underlying congenital pulmonary cysts.


Congenital disorders are medical conditions that are present from birth. Although many congenital disorders are rare, they can have a severe impact on the quality of life of those affected. For example, congenital pulmonary airway malformation (CPAM) is a rare congenital disorder that occurs in around 1 out of every 25,000 pregnancies. In CPAM, abnormal, fluid-filled sac-like pockets of tissue, known as cysts, form within the lungs of unborn babies. After birth, these cysts become air-filled and do not behave like normal lung tissue and stop a baby's lungs from working properly. In severe cases, babies with CPAM need surgery immediately after birth. We still do not understand exactly what the underlying causes of CPAM might be. CPAM is not considered to be hereditary ­ that is, it does not appear to be passed down in families ­ nor is it obviously linked to any environmental factors. CPAM is also very difficult to study, because researchers cannot access tissue samples during the critical early stages of the disease. To overcome these difficulties, Luo et al. wanted to find a way to study CPAM in the laboratory. First, they developed a non-human animal 'model' that naturally forms CPAM-like lung cysts, using genetically modified mice where the gene for the signaling molecule Bmpr1a had been deleted in lung cells. Normally, Bmpr1a is part of a set of the molecular instructions, collectively termed BMP signaling, which guide healthy lung development early in life. However, mouse embryos lacking Bmpr1a developed abnormal lung cysts that were similar to those found in CPAM patients, suggesting that problems with BMP signalling might also trigger CPAM in humans. Luo et al. also identified several other genes in the Bmpr1a-deficient mouse lungs that had abnormal patterns of activity. All these genes were known to be controlled by BMP signaling, and to play a role in the development and organisation of lung tissue. This suggests that when these genes are not controlled properly, they could drive formation of CPAM cysts when BMP signaling is compromised. This work is a significant advance in the tools available to study CPAM. Luo et al.'s results also shed new light on the molecular mechanisms underpinning this rare disorder. In the future, Luo et al. hope this knowledge will help us develop better treatments for CPAM, or even help to prevent it altogether.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I , Lung , Mesoderm , Mice, Knockout , Signal Transduction , Animals , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type I/deficiency , Mice , Lung/embryology , Lung/metabolism , Lung/pathology , Mesoderm/embryology , Mesoderm/metabolism , Cysts/metabolism , Cysts/pathology , Cysts/genetics , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Lung Diseases/metabolism , Lung Diseases/pathology , Lung Diseases/genetics , Disease Models, Animal
2.
bioRxiv ; 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-37808788

ABSTRACT

Abnormal lung development can cause congenital pulmonary cysts, the mechanisms of which remain largely unknown. Although the cystic lesions are believed to result directly from disrupted airway epithelial cell growth, the extent to which developmental defects in lung mesenchymal cells contribute to abnormal airway epithelial cell growth and subsequent cystic lesions has not been thoroughly examined. In the present study, we dissected the roles of BMP receptor 1a (Bmpr1a)-mediated BMP signaling in lung mesenchyme during prenatal lung development and discovered that abrogation of mesenchymal Bmpr1a disrupted normal lung branching morphogenesis, leading to the formation of prenatal pulmonary cystic lesions. Severe deficiency of airway smooth muscle cells and subepithelial elastin fibers were found in the cystic airways of the mesenchymal Bmpr1a knockout lungs. In addition, ectopic mesenchymal expression of BMP ligands and airway epithelial perturbation of the Sox2-Sox9 proximal-distal axis were detected in the mesenchymal Bmpr1a knockout lungs. However, deletion of Smad1/5, two major BMP signaling downstream effectors, from the lung mesenchyme did not phenocopy the cystic abnormalities observed in the mesenchymal Bmpr1a knockout lungs, suggesting that a Smad-independent mechanism contributes to prenatal pulmonary cystic lesions. These findings reveal for the first time the role of mesenchymal BMP signaling in lung development and a potential pathogenic mechanism underlying congenital pulmonary cysts.

3.
J Pathol ; 259(3): 331-341, 2023 03.
Article in English | MEDLINE | ID: mdl-36484734

ABSTRACT

Abnormal growth of airway smooth muscle cells is one of the key features in asthmatic airway remodeling, which is associated with asthma severity. The mechanisms underlying inappropriate airway smooth muscle cell growth in asthma remain largely unknown. Myocd has been reported to act as a key transcriptional coactivator in promoting airway-specific smooth muscle development in fetal lungs. Whether Myocd controls airway smooth muscle remodeling in asthma has not been investigated. Mice with lung mesenchyme-specific deletion of Myocd after lung development were generated, and a chronic asthma model was established by sensitizing and challenging the mice with ovalbumin for a prolonged period. Comparison of the asthmatic pathology between the Myocd knockout mice and the wild-type controls revealed that abrogation of Myocd mitigated airway smooth muscle cell hypertrophy and hyperplasia, accompanied by reduced peri-airway inflammation, decreased fibrillar collagen deposition on airway walls, and attenuation of abnormal mucin production in airway epithelial cells. Our study indicates that Myocd is a key transcriptional coactivator involved in asthma airway remodeling. Inhibition of Myocd in asthmatic airways may be an effective approach to breaking the vicious cycle of asthmatic progression, providing a novel strategy in treating severe and persistent asthma. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Airway Remodeling , Asthma , Nuclear Proteins , Animals , Mice , Asthma/genetics , Asthma/metabolism , Disease Models, Animal , Inflammation/pathology , Lung/pathology , Mice, Inbred BALB C , Mice, Knockout , Myocytes, Smooth Muscle/pathology , Nuclear Proteins/metabolism
6.
Value Health ; 23(2): 209-216, 2020 02.
Article in English | MEDLINE | ID: mdl-32113626

ABSTRACT

OBJECTIVES: Proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9is)-innovative yet costly cholesterol-lowering agents-have been subject to substantial prior authorization (PA) requirements and low approval rates. We aimed to investigate trends in insurer approval and reasons for rejection for PCSK9i prescriptions as well as associations between patients' demographic, clinical, pharmacy, payer, and PCSK9i-specific plan/coverage factors and approval. METHODS: We examined trends in PCSK9i approval rates and reasons for rejection using medical and prescription claims from 2015 to 2017 for individuals who received a PCSK9i prescription. We used multinomial logistic regression to estimate quarterly risk-adjusted approval rates for initial PCSK9i prescriptions and approval for any PCSK9i prescription within 30, 90, and 180 days of the initial PCSK9i prescription. For a 2016 subsample for whom we had PCSK9i-specific plan policy data, we examined factors associated with approval including PCSK9i-specific plan formulary coverage, step therapy requirements, and number of PA criteria. RESULTS: The main sample included 12 309 patients (mean age 64.8 years [SD = 10.8], 52.1% female, 51.5% receiving Medicare) and was similar in characteristics to the 2016 subsample (n = 6091). Approval rates varied across quarters but remained low (initial prescription, 13%-23%; within 90 days, 28%-44%). Over time, rejections owing to a lack of formulary coverage decreased and rejections owing to PA requirements increased. Lack of formulary coverage and having ≥11 PA criteria in the plan policy were associated with lower odds of PCSK9i prescription approval. CONCLUSIONS: These findings confirm ongoing PCSK9i access issues and offer a baseline for comparison in future studies examining the impact of recent efforts to improve PCSK9i access.


Subject(s)
Anticholesteremic Agents/therapeutic use , Eligibility Determination/trends , Health Care Rationing/trends , Insurance Coverage/trends , Insurance, Pharmaceutical Services/trends , PCSK9 Inhibitors , Prior Authorization/trends , Serine Proteinase Inhibitors/therapeutic use , Aged , Anticholesteremic Agents/adverse effects , Anticholesteremic Agents/economics , Cross-Sectional Studies , Databases, Factual , Drug Costs , Drug Prescriptions , Eligibility Determination/economics , Female , Formularies as Topic , Health Care Rationing/economics , Health Services Accessibility/economics , Health Services Accessibility/trends , Humans , Insurance Coverage/economics , Insurance, Pharmaceutical Services/economics , Male , Medicare/economics , Medicare/trends , Middle Aged , Prior Authorization/economics , Serine Proteinase Inhibitors/adverse effects , Serine Proteinase Inhibitors/economics , Time Factors , United States
8.
Proc Natl Acad Sci U S A ; 116(47): 23625-23635, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31690663

ABSTRACT

Myocardin-related transcription factor B (MRTFB) is a candidate tumor-suppressor gene identified in transposon mutagenesis screens of the intestine, liver, and pancreas. Using a combination of cell-based assays, in vivo tumor xenograft assays, and Mrtfb knockout mice, we demonstrate here that MRTFB is a human and mouse colorectal cancer (CRC) tumor suppressor that functions in part by inhibiting cell invasion and migration. To identify possible MRTFB transcriptional targets, we performed whole transcriptome RNA sequencing in MRTFB siRNA knockdown primary human colon cells and identified 15 differentially expressed genes. Among the top candidate tumor-suppressor targets were melanoma cell adhesion molecule (MCAM), a known tumor suppressor, and spindle apparatus coiled-coil protein 1 (SPDL1), which has no confirmed role in cancer. To determine whether these genes play a role in CRC, we knocked down the expression of MCAM and SPDL1 in human CRC cells and showed significantly increased invasion and migration of tumor cells. We also showed that Spdl1 expression is significantly down-regulated in Mrtfb knockout mouse intestine, while lower SPDL1 expression levels are significantly associated with reduced survival in CRC patients. Finally, we show that depletion of MCAM and SPDL1 in human CRC cells significantly increases tumor development in xenograft assays, further confirming their tumor-suppressive roles in CRC. Collectively, our findings demonstrate the tumor-suppressive role of MRTFB in CRC and identify several genes, including 2 tumor suppressors, that act downstream of MRTFB to regulate tumor growth and survival in CRC patients.


Subject(s)
Adenocarcinoma/genetics , Cell Cycle Proteins/metabolism , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/physiology , Transcription Factors/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , CD146 Antigen/metabolism , Cell Movement , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Gene Knockdown Techniques , Genes, Tumor Suppressor , HCT116 Cells , HT29 Cells , Heterografts , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasm Transplantation , RNA Interference , RNA, Small Interfering/genetics , Transcription Factors/deficiency , Transcription Factors/genetics
9.
Cell Death Differ ; 25(10): 1732-1748, 2018 11.
Article in English | MEDLINE | ID: mdl-29511336

ABSTRACT

Myocardin is a transcriptional co-activator required for cardiovascular development, but also promotes cardiomyocyte survival through an unclear molecular mechanism. Mitochondrial permeability transition is implicated in necrosis, while pore closure is required for mitochondrial maturation during cardiac development. We show that loss of myocardin function leads to subendocardial necrosis at E9.5, concurrent with elevated expression of the death gene Nix. Mechanistically, we demonstrate that myocardin knockdown reduces microRNA-133a levels to allow Nix accumulation, leading to mitochondrial permeability transition, reduced mitochondrial respiration, and necrosis. Myocardin knockdown elicits calcium release from the endo/sarcoplasmic reticulum with mitochondrial calcium accumulation, while restoration of microRNA-133a function, or knockdown of Nix rescues calcium perturbations. We observed reduced myocardin and elevated Nix expression within the infarct border-zone following coronary ligation. These findings identify a myocardin-regulated pathway that maintains calcium homeostasis and mitochondrial function during development, and is attenuated during ischemic heart disease. Given the diverse role of Nix and microRNA-133a, these findings may have broader implications to metabolic disease and cancer.


Subject(s)
Calcium/metabolism , Mitochondria/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Animals , Cells, Cultured , Doxorubicin/pharmacology , Gene Expression/drug effects , Heart/drug effects , Isoproterenol/pharmacology , Membrane Potential, Mitochondrial/drug effects , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Permeability/drug effects , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , Sarcoplasmic Reticulum/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics
10.
Circ Cardiovasc Qual Outcomes ; 11(1): e003939, 2018 01.
Article in English | MEDLINE | ID: mdl-29326146

ABSTRACT

BACKGROUND: Proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9is) are an innovative treatment option for patients with familial hypercholesterolemia or clinical atherosclerotic cardiovascular disease who require further lowering of low-density lipoprotein cholesterol. However, the high costs of these agents have spurred payers to implement utilization management policies to ensure appropriate use. We examined prior authorization (PA) requirements for PCSK9is across private and public US payers. METHODS AND RESULTS: We conducted an analysis of 2016 formulary coverage and PA data from a large, proprietary database with information on policies governing >95% of Americans with prescription drug coverage (275.3 million lives) within 3872 plans across the 4 major insurance segments (commercial, health insurance exchange, Medicare, and Medicaid). The key measures included administrative PA criteria (prescriber specialty, number of criteria in PA policy or number of fields on PA form, requirements for medical record submission, reauthorization requirements) and clinical/diagnostic PA criteria (approved conditions, required laboratories or other tests, required concomitant therapy, step therapy requirements, continuation criteria) for each of 2 Food and Drug Administration-approved PCSK9is. Select measures (eg, number of PA criteria/fields, medical record submission requirements) were obtained for 2 comparator cardiometabolic drugs (ezetimibe and liraglutide). Between 82% and 97% of individuals were enrolled in plans implementing PA for PCSK9is (depending on insurance segment), and one third to two thirds of these enrollees faced PAs restricting PCSK9i prescribing to a specialist. For patients with familial hypercholesterolemia, diagnostic confirmation via genetic testing or meeting minimum clinical scores/criteria was also required. PA requirements were more extensive for PCSK9is as compared with the other cardiometabolic drugs (ie, contained 3×-11× the number of PA criteria or fields on PA forms and more frequently involved the submission of medical records as supporting documentation). CONCLUSIONS: PA requirements for PCSK9is are greater than for selected other drugs within the cardiometabolic disease area, raising concerns about whether payer policies to discourage inappropriate use may also be restricting access to these drugs in patients who need them.


Subject(s)
Anticholesteremic Agents/therapeutic use , Dyslipidemias/drug therapy , Eligibility Determination , Medical Assistance , PCSK9 Inhibitors , Prior Authorization , Private Sector , Serine Proteinase Inhibitors/therapeutic use , Anticholesteremic Agents/economics , Biomarkers/blood , Cholesterol, LDL/blood , Databases, Factual , Drug Costs , Dyslipidemias/blood , Dyslipidemias/diagnosis , Dyslipidemias/economics , Eligibility Determination/economics , Eligibility Determination/legislation & jurisprudence , Humans , Medical Assistance/economics , Medical Assistance/legislation & jurisprudence , Policy Making , Prior Authorization/economics , Prior Authorization/legislation & jurisprudence , Private Sector/economics , Private Sector/legislation & jurisprudence , Proprotein Convertase 9/metabolism , Serine Proteinase Inhibitors/economics , United States
11.
JAMA Cardiol ; 2(8): 835-836, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28614578
14.
Proc Natl Acad Sci U S A ; 112(14): 4447-52, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25805819

ABSTRACT

Myocardin is a muscle-restricted transcriptional coactivator that activates a serum response factor (SRF)-dependent gene program required for cardiogenesis and embryonic survival. To identify myocardin-dependent functions in smooth muscle cells (SMCs) during postnatal development, mice harboring a SMC-restricted conditional, inducible Myocd null mutation were generated and characterized. Tamoxifen-treated SMMHC-Cre(ERT2)/Myocd(F/F) conditional mutant mice die within 6 mo of Myocd gene deletion, exhibiting profound derangements in the structure of great arteries as well as the gastrointestinal and genitourinary tracts. Conditional mutant mice develop arterial aneurysms, dissection, and rupture, recapitulating pathology observed in heritable forms of thoracic aortic aneurysm and dissection (TAAD). SMCs populating arteries of Myocd conditional mutant mice modulate their phenotype by down-regulation of SMC contractile genes and up-regulation of extracellular matrix proteins. Surprisingly, this is accompanied by SMC autonomous activation of endoplasmic reticulum (ER) stress and autophagy, which over time progress to programmed cell death. Consistent with these observations, Myocd conditional mutant mice develop remarkable dilation of the stomach, small intestine, bladder, and ureters attributable to the loss of visceral SMCs disrupting the muscularis mucosa. Taken together, these data demonstrate that during postnatal development, myocardin plays a unique, and important, role required for maintenance and homeostasis of the vasculature, gastrointestinal, and genitourinary tracts. The loss of myocardin in SMCs triggers ER stress and autophagy, which transitions to apoptosis, revealing evolutionary conservation of myocardin function in SMCs and cardiomyocytes.


Subject(s)
Aortic Aneurysm/metabolism , Gene Expression Regulation, Developmental , Muscle, Smooth, Vascular/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Trans-Activators/genetics , Trans-Activators/physiology , Animals , Aorta/metabolism , Apoptosis , Autophagy , Gastrointestinal Tract/metabolism , Homeostasis , Mice , Mice, Transgenic , Muscle Contraction , Mutation , Myocardium/metabolism , Myocytes, Smooth Muscle/cytology , Phenotype , Tamoxifen/chemistry , Urogenital System/metabolism
15.
Circulation ; 131(13): 1202-1213, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25712206

ABSTRACT

BACKGROUND: Genome-wide association studies have established ADAMTS7 as a locus for coronary artery disease in humans. However, these studies fail to provide directionality for the association between ADAMTS7 and coronary artery disease. Previous reports have implicated ADAMTS7 in the regulation of vascular smooth muscle cell migration, but a role for and the direction of impact of this gene in atherogenesis have not been shown in relevant model systems. METHODS AND RESULTS: We bred an Adamts7 whole-body knockout mouse onto both the Ldlr and Apoe knockout hyperlipidemic mouse models. Adamts7(-/-)/Ldlr(-/-) and Adamts7(-/-)/Apoe(-/-) mice displayed significant reductions in lesion formation in aortas and aortic roots compared with controls. Adamts7 knockout mice also showed reduced neointimal formation after femoral wire injury. Adamts7 expression was induced in response to injury and hyperlipidemia but was absent at later time points, and primary Adamts7 knockout vascular smooth muscle cells showed reduced migration in the setting of tumor necrosis factor-α stimulation. ADAMTS7 localized to cells positive for smooth muscle cell markers in human coronary artery disease lesions, and subcellular localization studies in cultured vascular smooth muscle cells placed ADAMTS7 at the cytoplasm and cell membrane, where it colocalized with markers of podosomes. CONCLUSIONS: These data represent the first in vivo experimental validation of the association of Adamts7 with atherogenesis, likely through modulation of vascular cell migration and matrix in atherosclerotic lesions. These results demonstrate that Adamts7 is proatherogenic, lending directionality to the original genetic association and supporting the concept that pharmacological inhibition of ADAMTS7 should be atheroprotective in humans, making it an attractive target for novel therapeutic interventions.


Subject(s)
ADAM Proteins/analysis , ADAM Proteins/physiology , Atherosclerosis/prevention & control , Coronary Disease/enzymology , Neointima/enzymology , Vascular Remodeling/physiology , ADAM Proteins/deficiency , ADAM Proteins/genetics , ADAMTS7 Protein , Amino Acid Sequence , Animals , Aorta/enzymology , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cell Division , Cell Movement , Cells, Cultured , Coronary Disease/pathology , Diet, Western/adverse effects , Endothelial Cells/metabolism , Female , Femoral Artery/injuries , Femoral Artery/pathology , Human Umbilical Vein Endothelial Cells , Humans , Hyperlipidemias/complications , Hyperlipidemias/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/ultrastructure , Neointima/pathology , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Receptors, LDL/deficiency , Receptors, LDL/genetics , Tumor Necrosis Factor-alpha/pharmacology
16.
Hum Genet ; 133(6): 743-53, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24362460

ABSTRACT

Trisomy 21 (Down syndrome, DS) is the most common human genetic anomaly associated with heart defects. Based on evolutionary conservation, DS-associated heart defects have been modeled in mice. By generating and analyzing mouse mutants carrying different genomic rearrangements in human chromosome 21 (Hsa21) syntenic regions, we found the triplication of the Tiam1-Kcnj6 region on mouse chromosome 16 (Mmu16) resulted in DS-related cardiovascular abnormalities. In this study, we developed two tandem duplications spanning the Tiam1-Kcnj6 genomic region on Mmu16 using recombinase-mediated genome engineering, Dp(16)3Yey and Dp(16)4Yey, spanning the 2.1 Mb Tiam1-Il10rb and 3.7 Mb Ifnar1-Kcnj6 regions, respectively. We found that Dp(16)4Yey/+, but not Dp(16)3Yey/+, led to heart defects, suggesting the triplication of the Ifnar1-Kcnj6 region is sufficient to cause DS-associated heart defects. Our transcriptional analysis of Dp(16)4Yey/+ embryos showed that the Hsa21 gene orthologs located within the duplicated interval were expressed at the elevated levels, reflecting the consequences of the gene dosage alterations. Therefore, we have identified a 3.7 Mb genomic region, the smallest critical genomic region, for DS-associated heart defects, and our results should set the stage for the final step to establish the identities of the causal gene(s), whose elevated expression(s) directly underlie this major DS phenotype.


Subject(s)
Chromosomes, Mammalian , Down Syndrome/genetics , Genome , Heart Defects, Congenital/genetics , Heart/embryology , Animals , Chromosome Mapping , Chromosomes, Human, Pair 21 , Disease Models, Animal , Down Syndrome/embryology , Down Syndrome/pathology , Embryo, Mammalian , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Gene Dosage , Genetic Engineering , Genetic Loci , Guanine Nucleotide Exchange Factors/genetics , Heart Defects, Congenital/embryology , Heart Defects, Congenital/pathology , Humans , Male , Mice , Phenotype , Recombination, Genetic , Synteny , T-Lymphoma Invasion and Metastasis-inducing Protein 1
17.
Cardiovasc Res ; 100(2): 316-24, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23985903

ABSTRACT

AIMS: Dissection and rupture of the ascending aorta are life-threatening conditions resulting in 80% mortality. Ascending aortic replacement in patients presenting with thoracic aortic aneurysm (TAA) is determined by metric measurement. However, a significant number of dissections occur outside of the parameters suggested by the current guidelines. We investigate the correlation among altered haemodynamic condition, oxidative stress, and vascular smooth muscle cell (VSMC) phenotype in controlling tissue homoeostasis. METHODS AND RESULTS: We demonstrate using finite element analysis (FEA) based on computed tomography geometries that TAA patients have higher wall stress in the ascending aorta than non-dilated patients. We also show that altered haemodynamic conditions are associated with increased levels of reactive oxygen species (ROS), direct regulators of the VSMC phenotype in the microregional area of the ascending aorta. Using in vitro and ex vivo studies on human tissues, we show that ROS accumulation correlates with media layer degeneration and increased connective tissue growth factor (CTGF) expression, which modulate the synthetic VSMC phenotype. Results were validated by a murine model of TAA (C57BL/6J) based on Angiotensin II infusion showing that medial thickening and luminal expansion of the proximal aorta is associated with the VSMC synthetic phenotype as seen in human specimens. CONCLUSIONS: Increased peak wall stress correlates with change in VSMC towards a synthetic phenotype mediated by ROS accumulation via CTGF. Understanding the molecular mechanisms that regulate VSMC towards a synthetic phenotype could unveil new regulatory pathways of aortic homoeostasis and impact the risk-stratification tool for patients at risk of aortic dissection and rupture.


Subject(s)
Aortic Aneurysm, Thoracic/metabolism , Connective Tissue Growth Factor/physiology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Oxidative Stress , Angiotensin II/pharmacology , Animals , Finite Element Analysis , Humans , Mice , Mice, Inbred C57BL , Phenotype , Reactive Oxygen Species/metabolism , Serum Response Factor/analysis , Vimentin/metabolism , ets-Domain Protein Elk-1/analysis
18.
Circ Res ; 112(6): 881-3, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23493303

ABSTRACT

Wamstad et al have provided a robust global analysis of histone markers and gene expression at 4 stages of murine embryonic stem (ES) cell differentiation into cardiac myocytes. This detailed data set will provide a rich opportunity for generating and testing hypotheses related to combinatorial transcriptional regulation of gene expression and epigenetic regulation of cell fate decisions in cardiac lineages.

20.
J Clin Invest ; 122(10): 3678-91, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22996691

ABSTRACT

Myocardin is a muscle lineage-restricted transcriptional coactivator that has been shown to transduce extracellular signals to the nucleus required for SMC differentiation. We now report the discovery of a myocardin/BMP10 (where BMP10 indicates bone morphogenetic protein 10) signaling pathway required for cardiac growth, chamber maturation, and embryonic survival. Myocardin-null (Myocd) embryos and embryos harboring a cardiomyocyte-restricted mutation in the Myocd gene exhibited myocardial hypoplasia, defective atrial and ventricular chamber maturation, heart failure, and embryonic lethality. Cardiac hypoplasia was caused by decreased cardiomyocyte proliferation accompanied by a dramatic increase in programmed cell death. Defective chamber maturation and the block in cardiomyocyte proliferation were caused in part by a block in BMP10 signaling. Myocardin transactivated the Bmp10 gene via binding of a serum response factor-myocardin protein complex to a nonconsensus CArG element in the Bmp10 promoter. Expression of p57kip2, a BMP10-regulated cyclin-dependent kinase inhibitor, was induced in Myocd-/- hearts, while BMP10-activated cardiogenic transcription factors, including NKX2.5 and MEF2c, were repressed. Remarkably, when embryonic Myocd-/- hearts were cultured ex vivo in BMP10-conditioned medium, the defects in cardiomyocyte proliferation and p57kip2 expression were rescued. Taken together, these data identify a heretofore undescribed myocardin/BMP10 signaling pathway that regulates cardiomyocyte proliferation and apoptosis in the embryonic heart.


Subject(s)
Bone Morphogenetic Proteins/biosynthesis , Fetal Heart/growth & development , Nuclear Proteins/physiology , Trans-Activators/physiology , Animals , Apoptosis , Bone Morphogenetic Proteins/genetics , Cell Differentiation , Culture Media, Conditioned/pharmacology , Fetal Heart/diagnostic imaging , Fetal Heart/pathology , Gene Expression Regulation, Developmental , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Organ Culture Techniques , Organogenesis/genetics , Signal Transduction , Trans-Activators/deficiency , Trans-Activators/genetics , Transcription Factors/physiology , Transcriptional Activation , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL