Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Stem Cell Res ; 72: 103213, 2023 10.
Article in English | MEDLINE | ID: mdl-37774637

ABSTRACT

Alagille syndrome (ALGS) is a multisystem disease with high variability in clinical features. ALGS is predominantly caused by pathogenic variants in the Notch ligand JAG1. An iPSC line, NCHi011-A, was generated from a ALGS patient with complex cardiac phenotypes consisting of pulmonic valve and branch pulmonary artery stenosis. NCHi011-A is heterozygous for a single base duplication causing a frameshift in the JAG1 gene. This iPSC line demonstrates normal cellular morphology, expression of pluripotency markers, trilineage differentiation potential, and identity to the source patient. NCHi011-A provides a resource for modeling ALGS and investigating the role of Notch signaling in the disease.


Subject(s)
Alagille Syndrome , Induced Pluripotent Stem Cells , Female , Humans , Young Adult , Adult , Alagille Syndrome/genetics , Alagille Syndrome/metabolism , Induced Pluripotent Stem Cells/metabolism , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Heart , Cell Differentiation
2.
Stem Cell Res ; 71: 103177, 2023 09.
Article in English | MEDLINE | ID: mdl-37549562

ABSTRACT

Alagille syndrome (ALGS) is an autosomal dominant disease affecting the liver, heart and other organs with high variability. About 95% of ALGS cases are associated with pathogenic variants in JAG1, encoding the Jagged1 ligand that binds to Notch receptors. The iPSC line NCHi012-A was derived from an ALGS patient with cholestatic liver disease and mild pulmonary stenosis, who is heterozygous for a 2 bp deletion in the JAG1 coding sequence. We report here an initial characterization of NCHi012-A to evaluate its morphology, pluripotency, differentiation potential, genotype, karyotype and identity to the source patient.


Subject(s)
Alagille Syndrome , Induced Pluripotent Stem Cells , Humans , Alagille Syndrome/genetics , Alagille Syndrome/metabolism , Induced Pluripotent Stem Cells/metabolism , Receptors, Notch/metabolism , Heart , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism
3.
Vascul Pharmacol ; 145: 107087, 2022 08.
Article in English | MEDLINE | ID: mdl-35792302

ABSTRACT

BACKGROUND: Notch signaling is an evolutionarily conserved pathway that functions via direct cell-cell contact. The Notch ligand Jagged1 (Jag1) has been extensively studied in vascular development, particularly for its role in smooth muscle cell maturation. Endothelial cell-expressed Jag1 is essential for blood vessel formation by signaling to nascent vascular smooth muscle cells and promoting their differentiation. Given the established importance of Jag1 in endothelial cell/smooth muscle crosstalk during development, we sought to determine the extent of this communication in the adult vasculature for blood vessel function and homeostasis. METHODS: We conditionally deleted Jag1 in endothelial cells of adult mice and examined the phenotypic consequences on smooth muscle cells of the vasculature. RESULTS: Our results show that genetic loss of Jag1 in endothelial cells has a significant impact on Notch signaling and vascular smooth muscle function in mature blood vessels. Endothelial cell-specific deletion of Jag1 causes a concomitant loss of JAG1 and NOTCH3 expression in vascular smooth muscle cells, resulting in a transition to a less differentiated state. Aortic vascular smooth muscle cells isolated from the endothelial cell-specific Jag1 deficient mice retain an altered phenotype in culture with fixed changes in gene expression and reduced Notch signaling. Utilizing comparative RNA-sequence analysis, we found that Jag1 deficiency preferentially affects extracellular matrix and adhesion protein gene expression. Vasoreactivity studies revealed a reduced contractile response and impaired agonist-induced relaxation in endothelial cell Jag1-deficient aortas compared to controls. CONCLUSIONS: These data are the first to demonstrate that Jag1 in adult endothelial cells is required for the regulation and homeostasis of smooth muscle cell function in arterial vessels partially through the autoregulation of Notch signaling and cell matrix/adhesion components in smooth muscle cells.


Subject(s)
Endothelial Cells , Receptors, Notch , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Endothelial Cells/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Ligands , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Muscle, Smooth, Vascular/metabolism , Phenotype , RNA/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Serrate-Jagged Proteins/genetics , Serrate-Jagged Proteins/metabolism
4.
J Vasc Res ; 59(3): 163-175, 2022.
Article in English | MEDLINE | ID: mdl-35294950

ABSTRACT

Pulmonary arterial hypertension (PAH) is a chronic progressive disease with significant morbidity and mortality. The disease is characterized by vascular remodeling that includes increased muscularization of distal blood vessels and vessel stiffening associated with changes in extracellular matrix deposition. In humans, chronic hypoxia causes PAH, and hypoxia-induced rodent models of PAH have been used for years to study the disease. With the development of single-cell RNA sequencing technology, it is now possible to examine hypoxia-dependent transcriptional changes in vivo at a cell-specific level. In this study, we used single-cell RNA sequencing to compare lungs from wild-type (Wt) mice exposed to hypoxia for 28 days to normoxia-treated control mice. We additionally examined mice deficient for Notch3, a smooth muscle-enriched gene linked to PAH. Data analysis revealed that hypoxia promoted cell number changes in immune and endothelial cell types in the lung, activated the innate immunity pathway, and resulted in specific changes in gene expression in vascular cells. Surprisingly, we found limited differences in lungs from mice deficient for Notch3 compared to Wt controls. These findings provide novel insight into the effects of chronic hypoxia exposure on gene expression and cell phenotypes in vivo and identify unique changes to cells of the vasculature.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Animals , Cell Proliferation , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypoxia/complications , Lung , Mice , Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/metabolism , Sequence Analysis, RNA
5.
Physiol Rep ; 9(17): e15013, 2021 09.
Article in English | MEDLINE | ID: mdl-34523259

ABSTRACT

BACKGROUND: Both downregulation and elevation of microRNA miR-145 has been linked to an array of cardiopulmonary phenotypes, and a host of studies suggest that it is an important contributor in governing the differentiation of cardiac and vascular smooth muscle cell types. METHODS AND RESULTS: To better understand the role of elevated miR-145 in utero within the cardiopulmonary system, we utilized a transgene to overexpress miR-145 embryonically in mice and examined the consequences of this lineage-restricted enhanced expression. Overexpression of miR-145 has detrimental effects that manifest after birth as overexpressor mice are unable to survive beyond postnatal day 18. The miR-145 expressing mice exhibit respiratory distress and fail to thrive. Gross analysis revealed an enlarged right ventricle, and pulmonary dysplasia with vascular hypertrophy. Single cell sequencing of RNA derived from lungs of control and miR-145 transgenic mice demonstrated that miR-145 overexpression had global effects on the lung with an increase in immune cells and evidence of leukocyte extravasation associated with vascular inflammation. CONCLUSIONS: These data provide novel findings that demonstrate a pathological role for miR-145 in the cardiopulmonary system that extends beyond its normal function in governing smooth muscle differentiation.


Subject(s)
Heart Arrest/metabolism , Heart Arrest/mortality , MicroRNAs/biosynthesis , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Animals, Newborn , Cells, Cultured , Female , Heart Arrest/genetics , Humans , Male , Mice , Mice, Transgenic , MicroRNAs/genetics , Mortality, Premature , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology
6.
Front Cardiovasc Med ; 8: 681948, 2021.
Article in English | MEDLINE | ID: mdl-34124207

ABSTRACT

COVID-19 is associated with a large number of cardiovascular sequelae, including dysrhythmias, myocardial injury, myocarditis and thrombosis. The Notch pathway is one likely culprit leading to these complications due to its direct role in viral entry, inflammation and coagulation processes, all shown to be key parts of COVID-19 pathogenesis. This review highlights links between the pathophysiology of SARS-CoV2 and the Notch signaling pathway that serve as primary drivers of the cardiovascular complications seen in COVID-19 patients.

7.
Sci Adv ; 7(6)2021 02.
Article in English | MEDLINE | ID: mdl-33547080

ABSTRACT

Calcific aortic valve disease (CAVD) is an increasingly prevalent condition, and endothelial dysfunction is implicated in its etiology. We previously identified nitric oxide (NO) as a calcification inhibitor by its activation of NOTCH1, which is genetically linked to human CAVD. Here, we show NO rescues calcification by an S-nitrosylation-mediated mechanism in porcine aortic valve interstitial cells and single-cell RNA-seq demonstrated NO regulates the NOTCH pathway. An unbiased proteomic approach to identify S-nitrosylated proteins in valve cells found enrichment of the ubiquitin-proteasome pathway and implicated S-nitrosylation of USP9X (ubiquitin specific peptidase 9, X-linked) in NOTCH regulation during calcification. Furthermore, S-nitrosylated USP9X was shown to deubiquitinate and stabilize MIB1 for NOTCH1 activation. Consistent with this, genetic deletion of Usp9x in mice demonstrated CAVD and human calcified aortic valves displayed reduced S-nitrosylation of USP9X. These results demonstrate a previously unidentified mechanism by which S-nitrosylation-dependent regulation of a ubiquitin-associated pathway prevents CAVD.

8.
Vasc Biol ; 2(1): R115-R128, 2020.
Article in English | MEDLINE | ID: mdl-33283158

ABSTRACT

miRNAs are small regulatory RNAs which govern gene expression post-transcriptionally by primarily binding to the 3'-UTR of mRNA target genes. miR-145 is a well-studied miRNA that has been implicated in controlling a range of biological processes. miR-145 is expressed in a variety of tissues and cell types and acts as a tumor-suppressor by regulating target gene signaling pathways involved in different aspects of tumor growth and progression. There is also strong evidence that highlights the important functions of miR-145 in the cardiovascular system. Here, we review the mechanisms of miR-145 in tumorigenesis and cancer progression and compare and contrast with the roles of miR-145 in cardiovascular development and disease. We discuss the important targets of miR-145 in cancer and their possible link to the cardiovascular system.

9.
Genesis ; 58(9): e23385, 2020 09.
Article in English | MEDLINE | ID: mdl-32648361

ABSTRACT

MicroRNAs are modulators of cellular phenotypes and their functions contribute to development, homeostasis, and disease. miR-145 is a conserved microRNA that has been implicated in regulating an array of phenotypes. These include supporting smooth muscle differentiation, repression of stem cell pluripotency, and inhibition of tumor growth and metastasis. Previously, our lab demonstrated that miR-145 acts to suppress cardiac fibrosis through inhibition of the TGF-ß signaling pathway. The range of effects that miR-145 has on different cell types makes it an attractive microRNA for further study. Here we describe the generation of transgenic mice that conditionally express miR-145 through Cre recombinase-mediated activation. Characterization of individual founder lines indicates that overexpression of miR-145 in the developing cardiovascular system has detrimental effects, with three independent miR-145 transgenic lines exhibiting Cre-dependent lethality. Expression analysis demonstrates that the transgene is robustly expressed and our analysis reveals a novel downstream target of miR-145, Tnnt2. The miR-145 transgenic mice represent a valuable tool to understand the role of miR-145 in diverse cell types and to address its potential as a therapeutic mediator for the treatment of disease.


Subject(s)
Genetic Engineering/methods , MicroRNAs/genetics , Transgenes , Animals , Cell Line , Integrases/genetics , Integrases/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , Rats , Troponin T/genetics , Troponin T/metabolism
10.
Front Physiol ; 11: 29, 2020.
Article in English | MEDLINE | ID: mdl-32116749

ABSTRACT

RATIONALE: Myoendothelial junctions (MEJs) within the fenestrae of the internal elastic lamina (IEL) are critical sites that allow for endothelial cell (EC) - vascular smooth muscle cell (VSMC) contact and communication. Vascular Notch signaling is a critical determinant of normal vasculogenesis and remodeling, and it regulates cell phenotype via contact between ECs and VSMCs. To date, no studies have linked Notch signaling to the MEJ despite it requiring cell-cell contact. Furthermore, very little is known about Notch in the adult coronary circulation or the localization of Notch signaling and activity within the mature intact blood vessel. OBJECTIVE: We tested the hypothesis that vascular Notch signaling between ECs and VSMCs occurs at MEJs. METHODS AND RESULTS: Notch receptor and ligand immunofluorescence was performed in human coronary EC and VSMC co-cultures across transwell inserts (in vitro MEJs) and in the intact mouse coronary circulation. Human coronary VSMC Notch activity induced by human coronary ECs at the in vitro MEJ was assessed using a CBF-luciferase construct. We observed Jagged1, Notch1, Notch2, and Notch3 expression within the in vitro and in vivo MEJs. We also demonstrated a 3-fold induction (p < 0.001) of human coronary VSMC Notch signaling by ECs at the in vitro MEJ, which was completely blocked by the Notch inhibitor, DAPT (p < 0.01). CONCLUSION: We demonstrate for the first time in mature blood vessels that Notch receptors and ligands are expressed within and are active at coronary MEJs, demonstrating a previously unrecognized mode of Notch signaling regulation between the endothelium and smooth muscle.

11.
J Cardiovasc Dev Dis ; 6(2)2019 06 22.
Article in English | MEDLINE | ID: mdl-31234534

ABSTRACT

Mammalian Kiaa1211 and Kiaa1211-like are a homologous pair of uncharacterized, highly conserved genes cloned from fetal and adult brain cDNA libraries. Herein we map the in utero spatiotemporal expression of mKiaa1211 and mKiaa1211L mRNA and their expression patterns in postnatal testis, skin, gastrointestinal, and adipose progenitor tissues. Significantly, mKiaa1211 is present throughout the early stages of mouse heart development, particularly in the second heart field (SHF) lineage as it differentiates from mesenchymal cells into cardiomyocytes. We also show that mKiaa1211 is expressed within several early neuronal tissues destined to give rise to central, peripheral, and sympathetic nervous system structures. Expression profiling revealed that the paralog mKiaa1211L is not expressed during the normal developmental process and that mKiaa1211 expression was noticeably absent from most adult terminally differentiated tissues. Finally, we confirm that a previously uncharacterized CRISPR/CAS-generated mKiaa1211 mouse mutant allele is hypomorphic.

12.
J Vasc Res ; 55(5): 308-318, 2018.
Article in English | MEDLINE | ID: mdl-30347392

ABSTRACT

Loss of vascular pericytes has long been associated with the onset of diabetic retinopathy; however, mechanisms contributing to pericyte dropout are not understood. Notch3 has been implicated in pericyte stability and survival, and linked to vascular integrity. Notch3 mutant mice exhibit progressive loss of retinal pericytes. Given that diabetic retinopathy is associated with pericyte loss, we sought to determine whether perturbation of Notch3 signaling contributes to diabetes-induced pericyte dropout and capillary degeneration. We utilized a pericyte-expressed LacZ transgene (XlacZ4) to examine pericyte loss in retinas of a type I diabetic mouse model (Ins2Akita) and Notch3-deficient mice. Notch3 null animals showed a dramatic loss of the LacZ marker by 8 weeks of age, while Ins2Akita diabetic and Notch3 heterozygous mice exhibited a much slower and subtler loss of LacZ. Although combined Notch3 heterozygosity in Ins2Akita diabetic animals did not show further deficits, the trypsin digest method revealed that Notch3 haploinsufficiency increased the formation of acellular capillaries in diabetic mice. Our data further indicate that Notch signaling is blunted in diabetic retinas and in cells exposed to hyperglycemia. These results are the first to demonstrate an association between Notch3 signaling, pericyte loss, and diabetic retinopathy.


Subject(s)
Diabetic Retinopathy/metabolism , Pericytes/metabolism , Receptor, Notch3/deficiency , Retina/metabolism , Animals , Blood Glucose/metabolism , Cell Death , Cells, Cultured , Diabetic Retinopathy/blood , Diabetic Retinopathy/genetics , Diabetic Retinopathy/pathology , Disease Models, Animal , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Pericytes/pathology , Receptor, Notch3/genetics , Retina/pathology , Signal Transduction
13.
Vascul Pharmacol ; 110: 7-15, 2018 11.
Article in English | MEDLINE | ID: mdl-29969687

ABSTRACT

Cytoglobin is a widely expressed heme protein that binds oxygen, carbon monoxide and nitric oxide. Recent examination of cytoglobin in the vasculature indicates that it contributes to nitric oxide availability, which is central to normal blood vessel function through regulation of smooth muscle cell tone and physiological response. Given the potential implications of cytoglobin in vascular function, we examined how cytoglobin might be uniquely regulated in vascular smooth muscle cells. Our data demonstrate that endothelial cells can increase the expression of cytoglobin in vascular smooth muscle cells, and the induction of cytoglobin is cell contact-dependent. We show that Notch signaling is necessary for endothelial cell-induced cytoglobin expression and Notch2 and Notch3 are sufficient to drive its expression in aortic smooth muscle cells. We further reveal that in cytoglobin-depleted smooth muscle cells there is increased cellular nitric oxide. These data demonstrate that, in addition to being the main producer of vascular nitric oxide, endothelial cells facilitate the ability of smooth muscle cells to metabolize nitric oxide through upregulation of cytoglobin. Our results reveal a novel mechanism by which Notch signaling contributes to vascular function through regulation of a gene that controls nitric oxide levels.


Subject(s)
Cell Communication , Endothelial Cells/metabolism , Globins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nitric Oxide/metabolism , Animals , Cells, Cultured , Coculture Techniques , Cytoglobin , Globins/genetics , Humans , Mice, Inbred C57BL , Receptor, Notch2/metabolism , Receptor, Notch3/metabolism , Signal Transduction , Up-Regulation
14.
JCI Insight ; 2(21)2017 11 02.
Article in English | MEDLINE | ID: mdl-29093270

ABSTRACT

An ascending aortic aneurysm (AscAA) is a life-threatening disease whose molecular basis is poorly understood. Mutations in NOTCH1 have been linked to bicuspid aortic valve (BAV), which is associated with AscAA. Here, we describe a potentially novel role for Notch1 in AscAA. We found that Notch1 haploinsufficiency exacerbated the aneurysmal aortic root dilation seen in the Marfan syndrome mouse model and that heterozygous deletion of Notch1 in the second heart field (SHF) lineage recapitulated this exacerbated phenotype. Additionally, Notch1+/- mice in a predominantly 129S6 background develop aortic root dilation, indicating that loss of Notch1 is sufficient to cause AscAA. RNA sequencing analysis of the Notch1.129S6+/- aortic root demonstrated gene expression changes consistent with AscAA. These findings are the first to our knowledge to demonstrate an SHF lineage-specific role for Notch1 in AscAA and suggest that genes linked to the development of BAV may also contribute to the associated aortopathy.


Subject(s)
Aortic Aneurysm/genetics , Aortic Valve/abnormalities , Genetic Predisposition to Disease , Haploinsufficiency , Receptor, Notch1/genetics , Animals , Aorta , Aortic Aneurysm/pathology , Aortic Valve/diagnostic imaging , Aortic Valve/pathology , Bicuspid Aortic Valve Disease , Disease Models, Animal , Gene Expression , Genetic Association Studies , Heart Valve Diseases , Mice , Mice, Knockout , Mutation , Phenotype
15.
PLoS One ; 12(5): e0178538, 2017.
Article in English | MEDLINE | ID: mdl-28562688

ABSTRACT

AIMS: Infiltration of macrophages and apoptosis of vascular smooth muscle cells (VSMCs) promote the development of abdominal aortic aneurysm (AAA). Previously, we demonstrated that global Notch1 deficiency prevents the formation of AAA in a mouse model. Herein, we sought to explore the cell-specific roles of Notch1 in AAA development. METHODS AND RESULTS: Cell-specific Notch1 haploinsufficient mice, generated on Apoe-/- background using Cre-lox technology, were infused with angiotensin II (1000 ng/min/kg) for 28 days. Notch1 haploinsufficiency in myeloid cells (n = 9) prevented the formation of AAA attributed to decreased inflammation. Haploinsufficiency of Notch1 in SMCs (n = 14) per se did not prevent AAA formation, but histoarchitectural traits of AAA including elastin degradation and aortic remodeling, were minimal in SMC-Notch1+/-;Apoe-/- mice compared to Apoe-/- mice (n = 33). Increased immunostaining of the contractile SMC-phenotype markers and concomitant decreased expression of synthetic SMC-phenotype markers were observed in the aortae of SMC-Notch1+/-;Apoe-/- mice. Expression of connective tissue growth factor (CTGF), a matrix-associated protein that modulates the synthetic VSMC phenotype, increased in the abdominal aorta of Apoe-/- mice and in the adventitial region of the abdominal aorta in human AAA. Notch1 haploinsufficiency decreased the expression of Ctgf in the aorta and in vitro cell culture system. In vitro studies on SMCs using the Notch1 intracellular domain (NICD) plasmid, dominant negative mastermind-like (dnMAML), or specific siRNA suggest that Notch1, not Notch3, directly modulates the expression of CTGF. CONCLUSIONS: Our data suggest that lack of Notch1 in SMCs limits dilation of the abdominal aorta by maintaining contractile SMC-phenotype and preventing matrix-remodeling.


Subject(s)
Aortic Aneurysm, Abdominal/pathology , Connective Tissue Growth Factor/metabolism , Haploinsufficiency , Muscle, Smooth, Vascular/metabolism , Receptor, Notch1/metabolism , Animals , Aortic Aneurysm, Abdominal/metabolism , Cells, Cultured , Coculture Techniques , Matrix Metalloproteinases/biosynthesis , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology
16.
Genesis ; 53(12): 738-48, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26453897

ABSTRACT

The overlapping roles of the predominant Notch receptors in vascular smooth muscle cells, Notch2 and Notch3, have not been clearly defined in vivo. In this study, we use a smooth muscle-specific deletion of Notch2 together with a global Notch3 deletion to produce mice with combinations of mutant and wild-type Notch2/3 alleles in vascular smooth muscle cells. Mice with complete loss of Notch3 and smooth muscle-expressed Notch2 display late embryonic lethality and subcutaneous hemorrhage. Mice without smooth muscle-Notch2 and only one wild-type copy of Notch3 die within one day of birth and present with vascular defects, most notably patent ductus arteriosus (DA) and aortic dilation. These defects were associated with decreased expression of contractile markers in both the DA and aorta. These results demonstrate that Notch2 and Notch3 have overlapping roles in promoting development of vascular smooth muscle cells, and together contribute to functional closure of the DA.


Subject(s)
Ductus Arteriosus, Patent/metabolism , Muscle, Smooth, Vascular/metabolism , Receptor, Notch2/metabolism , Receptors, Notch/metabolism , Animals , Aorta/metabolism , Cell Differentiation/physiology , Ductus Arteriosus, Patent/genetics , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Receptor, Notch2/genetics , Receptor, Notch3 , Receptors, Notch/genetics , Signal Transduction
17.
Ann Thorac Surg ; 100(3): 1013-9; discussion 1019-20, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26163359

ABSTRACT

BACKGROUND: The hybrid palliation for hypoplastic left heart syndrome has emerged as an alternative approach to the Norwood procedure. The development of patent ductus arteriosus (PDA) in-stent stenosis can cause retrograde aortic arch stenosis (RAAS), leading to significant morbidity. This study aimed to identify potential mechanisms of PDA in-stent stenosis contributing to RAAS. METHODS: Tissues from stented PDAs were collected from 17 patients undergoing comprehensive stage II repair between 2009 and 2014. Patients requiring RAAS intervention based on cardiology-surgery consensus were defined as RAAS(+) (n = 10), whereas patients without any RAAS intervention were defined as RAAS(-) (n = 7). Tissues were examined by quantitative polymerase chain reaction analysis for vascular smooth muscle cell (VSMC) differentiation and proliferation markers. RESULTS: Patient characteristics were hypoplastic left heart syndrome with aortic atresia in 6 and with aortic stenosis in 3; unbalanced atrioventricular canal in 3; double-inlet left ventricle/transposition of the great arteries in 3; and double-outlet right ventricle in 2. VSMC differentiation markers (ß-actin, SM22, and calponin) and signaling pathways for VSMC modulation (transforming growth factor-ß1, Notch, and platelet derived growth factor-BB) were significantly higher in the RAAS(+) than in RAAS(-) patients. The proliferation marker Ki67 was increased in RAAS(+) patients. Cell cycle markers were comparable in both groups. CONCLUSIONS: Increased VSMC differentiation and proliferation markers suggest a mechanism for inward neointima formation of the PDA in RAAS. The apparent lack of change in cell cycle markers is contrary to coronary artery in-stent stenosis, suggesting further targets should be examined. Combined primary in vitro PDA cell culture and proteomics can be strong tools to elucidate targets to reduce PDA in-stent stenosis for RAAS in the future.


Subject(s)
Aortic Valve Stenosis/etiology , Ductus Arteriosus, Patent/etiology , Hypoplastic Left Heart Syndrome/surgery , Muscle, Smooth, Vascular/pathology , Postoperative Complications/etiology , Stents , Aortic Valve Stenosis/genetics , Cardiac Surgical Procedures , Cell Differentiation/genetics , Cell Proliferation/genetics , Ductus Arteriosus, Patent/genetics , Humans , Infant, Newborn , Postoperative Complications/genetics
18.
J Biol Chem ; 290(26): 16226-37, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25957400

ABSTRACT

Notch signaling is a key regulator of vascular smooth muscle cell (VSMC) phenotypes, including differentiation, proliferation, and cell survival. However, the exact contribution of the individual Notch receptors has not been thoroughly delineated. In this study, we identify unique roles for NOTCH2 and NOTCH3 in regulating proliferation and cell survival in cultured VSMCs. Our results indicate that NOTCH2 inhibits PDGF-B-dependent proliferation and its expression is decreased by PDGF-B. In contrast, NOTCH3 promotes proliferation and receptor expression is increased by PDGF-B. Additionally, data show that NOTCH3, but not NOTCH2 protects VSMCs from apoptosis and apoptosis mediators degrade NOTCH3 protein. We identified three pro-survival genes specifically regulated by NOTCH3 in cultured VSMCs and in mouse aortas. This regulation is mediated through MAP kinase signaling, which we demonstrate can be activated by NOTCH3, but not NOTCH2. Overall, this study highlights discrete roles for NOTCH2 and NOTCH3 in VSMCs and connects these roles to specific upstream regulators that control their expression.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptor, Notch2/metabolism , Receptors, Notch/metabolism , Animals , Cell Proliferation , Cell Survival , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Receptor, Notch2/genetics , Receptor, Notch3 , Receptors, Notch/genetics , Signal Transduction
19.
J Cardiovasc Dev Dis ; 2(1): 17-30, 2015.
Article in English | MEDLINE | ID: mdl-25914885

ABSTRACT

Thoracic aortic aneurysms (TAA) are a significant cause of morbidity and mortality in humans. While the exact etiology is unknown, genetic factors play an important role. Mutations in NOTCH1 have been linked to bicuspid aortic valve (BAV) and aortopathy in humans. The aim of this study was to determine if haploinsufficiency of Notch1 contributes to aortopathy using Notch1+/-; Nos3-/- mice. Echocardiographic analysis of Notch1+/-; Nos3-/- mice reveals effacement of the sinotubular junction and a trend toward dilation of the aortic sinus. Furthermore, examination of the proximal aorta of Notch1+/-; Nos3-/- mice reveals elastic fiber degradation, a trend toward increased matrix metalloproteinase 2 expression, and increased smooth muscle cell apoptosis, features characteristic of aneurysmal disease. Although at a lower penetrance, we also found features consistent with aortopathic changes in Notch1 heterozygote mice and in Nos3-null mice. Our findings implicate a novel role for Notch1 in aortopathy of the proximal aorta.

20.
Circ Res ; 116(1): 23-34, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25323858

ABSTRACT

RATIONALE: MicroRNA miR145 has been implicated in vascular smooth muscle cell differentiation, but its mechanisms of action and downstream targets have not been fully defined. OBJECTIVE: Here, we sought to explore and define the mechanisms of miR145 function in smooth muscle cells. METHODS AND RESULTS: Using a combination of cell culture assays and in vivo mouse models to modulate miR145, we characterized its downstream actions on smooth muscle phenotypes. Our results show that the miR-143/145 gene cluster is induced in smooth muscle cells by coculture with endothelial cells. Endothelial cell-induced expression of miR-143/145 is augmented by Notch signaling and accordingly expression is reduced in Notch receptor-deficient cells. Screens to identify miR145-regulated genes revealed that the transforming growth factor (TGF)-ß pathway has a significantly high number of putative target genes, and we show that TGFß receptor II is a direct target of miR145. Extracellular matrix genes that are regulated by TGFß receptor II were attenuated by miR145 overexpression, and miR145 mutant mice exhibit an increase in extracellular matrix synthesis. Furthermore, activation of TGFß signaling via angiotensin II infusion revealed a pronounced fibrotic response in the absence of miR145. CONCLUSIONS: These data demonstrate a specific role for miR145 in the regulation of matrix gene expression in smooth muscle cells and suggest that miR145 acts to suppress TGFß-dependent extracellular matrix accumulation and fibrosis, while promoting TGFß-induced smooth muscle cell differentiation. Our findings offer evidence to explain how TGFß signaling exhibits distinct downstream actions via its regulation by a specific microRNA.


Subject(s)
Extracellular Matrix/metabolism , MicroRNAs/physiology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Receptors, Transforming Growth Factor beta/biosynthesis , Animals , Cells, Cultured , Gene Expression Regulation , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Transforming Growth Factor-beta Type II
SELECTION OF CITATIONS
SEARCH DETAIL
...