Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Genome Res ; 30(12): 1695-1704, 2020 12.
Article in English | MEDLINE | ID: mdl-33122304

ABSTRACT

Somatic mosaicism, manifesting as single nucleotide variants (SNVs), mobile element insertions, and structural changes in the DNA, is a common phenomenon in human brain cells, with potential functional consequences. Using a clonal approach, we previously detected 200-400 mosaic SNVs per cell in three human fetal brains (15-21 wk postconception). However, structural variation in the human fetal brain has not yet been investigated. Here, we discover and validate four mosaic structural variants (SVs) in the same brains and resolve their precise breakpoints. The SVs were of kilobase scale and complex, consisting of deletion(s) and rearranged genomic fragments, which sometimes originated from different chromosomes. Sequences at the breakpoints of these rearrangements had microhomologies, suggesting their origin from replication errors. One SV was found in two clones, and we timed its origin to ∼14 wk postconception. No large scale mosaic copy number variants (CNVs) were detectable in normal fetal human brains, suggesting that previously reported megabase-scale CNVs in neurons arise at later stages of development. By reanalysis of public single nuclei data from adult brain neurons, we detected an extrachromosomal circular DNA event. Our study reveals the existence of mosaic SVs in the developing human brain, likely arising from cell proliferation during mid-neurogenesis. Although relatively rare compared to SNVs and present in ∼10% of neurons, SVs in developing human brain affect a comparable number of bases in the genome (∼6200 vs. ∼4000 bp), implying that they may have similar functional consequences.


Subject(s)
Brain/embryology , DNA, Circular/genetics , Genomic Structural Variation , Sequence Analysis, DNA/methods , Clonal Evolution , Female , Genotyping Techniques , Gestational Age , Humans , Mosaicism , Neurogenesis , Pregnancy
2.
J Cell Sci ; 131(10)2018 05 15.
Article in English | MEDLINE | ID: mdl-29678906

ABSTRACT

Myofibroblasts play key roles in wound healing and pathological fibrosis. Here, we used an RNAi screen to characterize myofibroblast regulatory genes, using a high-content imaging approach to quantify α-smooth muscle actin stress fibers in cultured human fibroblasts. Screen hits were validated on physiological compliance hydrogels, and selected hits tested in primary fibroblasts from patients with idiopathic pulmonary fibrosis. Our RNAi screen led to the identification of STAT3 as an essential mediator of myofibroblast activation and function. Strikingly, we found that STAT3 phosphorylation, while responsive to exogenous ligands on both soft and stiff matrices, is innately active on a stiff matrix in a ligand/receptor-independent, but ROCK- and JAK2-dependent fashion. These results demonstrate how a cytokine-inducible signal can become persistently activated by pathological matrix stiffening. Consistent with a pivotal role for this pathway in driving persistent fibrosis, a STAT3 inhibitor attenuated murine pulmonary fibrosis when administered in a therapeutic fashion after bleomycin injury. Our results identify novel genes essential for the myofibroblast phenotype, and point to STAT3 as an important target in pulmonary fibrosis and other fibrotic diseases.


Subject(s)
Janus Kinase 2/metabolism , Myofibroblasts/metabolism , Pulmonary Fibrosis/genetics , RNA Interference , STAT3 Transcription Factor/metabolism , rho-Associated Kinases/metabolism , Animals , Female , Fibroblasts/metabolism , Humans , Janus Kinase 2/genetics , Mice , Mice, Inbred C57BL , Phosphorylation , Pulmonary Fibrosis/metabolism , STAT3 Transcription Factor/genetics , Signal Transduction , rho-Associated Kinases/genetics
3.
FASEB J ; 33(2): 3024-3034, 2019 02.
Article in English | MEDLINE | ID: mdl-30351991

ABSTRACT

Recent studies have demonstrated an effect of neurotrophins, particularly brain-derived neurotrophic factor (BDNF), on airway contractility [ via increased airway smooth muscle (ASM) intracellular calcium [Ca2+]i] and remodeling (ASM proliferation and extracellular matrix formation) in the context of airway disease. In the present study, we examined the role of BDNF in allergen-induced airway inflammation using 2 transgenic models: 1) tropomyosin-related kinase B (TrkB) conditional knockin (TrkBKI) mice allowing for inducible, reversible disruption of BDNF receptor kinase activity by administration of 1NMPP1, a PP1 derivative, and 2) smooth muscle-specific BDNF knockout (BDNFfl/fl/SMMHC11Cre/0) mice. Adult mice were intranasally challenged with PBS or mixed allergen ( Alternaria alternata, Aspergillus fumigatus, house dust mite, and ovalbumin) for 4 wk. Our data show that administration of 1NMPP1 in TrkBKI mice during the 4-wk allergen challenge blunted airway hyperresponsiveness (AHR) and reduced fibronectin mRNA expression in ASM layers but did not reduce inflammation per se. Smooth muscle-specific deletion of BDNF reduced AHR and blunted airway fibrosis but did not significantly alter airway inflammation. Together, our novel data indicate that TrkB signaling is a key modulator of AHR and that smooth muscle-derived BDNF mediates these effects during allergic airway inflammation.-Britt, R. D., Jr., Thompson, M. A., Wicher, S. A., Manlove, L. J., Roesler, A., Fang, Y.-H., Roos, C., Smith, L., Miller, J. D., Pabelick, C. M., Prakash, Y. S. Smooth muscle brain-derived neurotrophic factor contributes to airway hyperreactivity in a mouse model of allergic asthma.


Subject(s)
Asthma/physiopathology , Brain-Derived Neurotrophic Factor/physiology , Bronchial Hyperreactivity/etiology , Disease Models, Animal , Membrane Glycoproteins/physiology , Muscle, Smooth/metabolism , Protein-Tyrosine Kinases/physiology , Respiratory System/physiopathology , Airway Remodeling/drug effects , Allergens/adverse effects , Animals , Asthma/chemically induced , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/pathology , Female , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Male , Membrane Glycoproteins/antagonists & inhibitors , Mice , Mice, Knockout , Mice, Transgenic , Muscle Contraction , Muscle, Smooth/cytology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology
4.
Am J Respir Cell Mol Biol ; 61(1): 51-60, 2019 07.
Article in English | MEDLINE | ID: mdl-30508396

ABSTRACT

Supplemental O2 (hyperoxia; 30-90% O2) is a necessary intervention for premature infants, but it contributes to development of neonatal and pediatric asthma, necessitating better understanding of contributory mechanisms in hyperoxia-induced changes to airway structure and function. In adults, environmental stressors promote formation of senescent cells that secrete factors (senescence-associated secretory phenotype), which can be inflammatory and have paracrine effects that enhance chronic lung diseases. Hyperoxia-induced changes in airway structure and function are mediated in part by effects on airway smooth muscle (ASM). In the present study, using human fetal ASM cells as a model of prematurity, we ascertained the effects of clinically relevant moderate hyperoxia (40% O2) on cellular senescence. Fetal ASM exposed to 40% O2 for 7 days exhibited elevated concentrations of senescence-associated markers, including ß-galactosidase; cell cycle checkpoint proteins p16, p21, and p-p53; and the DNA damage marker p-γH2A.X (phosphorylated γ-histone family member X). The combination of dasatinib and quercetin, compounds known to eliminate senescent cells (senolytics), reduced the number of hyperoxia-exposed ß-galactosidase-, p21-, p16-, and p-γH2A.X-positive ASM cells. The senescence-associated secretory phenotype profile of hyperoxia-exposed cells included both profibrotic and proinflammatory mediators. Naive ASM exposed to media from hyperoxia-exposed senescent cells exhibited increased collagen and fibronectin and higher contractility. Our data show that induction of cellular senescence by hyperoxia leads to secretion of inflammatory factors and has a functional effect on naive ASM. Cellular senescence in the airway may thus contribute to pediatric airway disease in the context of sequelae of preterm birth.


Subject(s)
Cellular Senescence , Fetus/pathology , Hyperoxia/pathology , Lung/embryology , Myocytes, Smooth Muscle/pathology , Biomarkers/metabolism , Cell Cycle/drug effects , Cellular Senescence/drug effects , Cytokines/metabolism , DNA Damage , Dasatinib/pharmacology , Etoposide/pharmacology , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Humans , Inflammation Mediators/metabolism , Models, Biological , Myocytes, Smooth Muscle/drug effects , Phenotype , Quercetin/pharmacology
5.
J Cell Physiol ; 234(8): 14187-14197, 2019 08.
Article in English | MEDLINE | ID: mdl-30624783

ABSTRACT

Airway smooth muscle (ASM) regulation of airway structure and contractility is critical in fetal/neonatal physiology in health and disease. Fetal lungs experience higher Ca2+ environment that may impact extracellular Ca2+ ([Ca2+ ]o ) sensing receptor (CaSR). Well-known in the parathyroid gland, CaSR is also expressed in late embryonic lung mesenchyme. Using cells from 18-22 week human fetal lungs, we tested the hypothesis that CaSR regulates intracellular Ca2+ ([Ca2+ ]i ) in fetal ASM (fASM). Compared with adult ASM, CaSR expression was higher in fASM, while fluorescence Ca2+ imaging showed that [Ca2+ ]i was more sensitive to altered [Ca2+ ]o . The fASM [Ca2+ ]i responses to histamine were also more sensitive to [Ca2+ ]o (0-2 mM) compared with an adult, enhanced by calcimimetic R568 but blunted by calcilytic NPS2143. [Ca2+ ]i was enhanced by endogenous CaSR agonist spermine (again higher sensitivity compared with adult). Inhibition of phospholipase C (U73122; siRNA) or inositol 1,4,5-triphosphate receptor (Xestospongin C) blunted [Ca2+ ]o sensitivity and R568 effects. NPS2143 potentiated U73122 effects. Store-operated Ca2+ entry was potentiated by R568. Traction force microscopy showed responsiveness of fASM cellular contractility to [Ca2+ ]o and NPS2143. Separately, fASM proliferation showed sensitivity to [Ca2+ ]o and NPS2143. These results demonstrate functional CaSR in developing ASM that modulates airway contractility and proliferation.


Subject(s)
Calcium Signaling/genetics , Lung/growth & development , Myoblasts/metabolism , Receptors, Calcium-Sensing/genetics , Calcium/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , Fetus , Humans , Lung/embryology , Lung/metabolism , Macrocyclic Compounds/pharmacology , Muscle, Smooth/metabolism , Naphthalenes/pharmacology , Oxazoles/pharmacology , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/genetics
6.
Am J Physiol Lung Cell Mol Physiol ; 317(5): L525-L536, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31411059

ABSTRACT

Hyperoxia exposure in premature infants increases the risk of subsequent lung diseases, such as asthma and bronchopulmonary dysplasia. Fibroblasts help maintain bronchial and alveolar integrity. Thus, understanding mechanisms by which hyperoxia influences fibroblasts is critical. Cellular senescence is increasingly recognized as important to the pathophysiology of multiple diseases. We hypothesized that clinically relevant moderate hyperoxia (<50% O2) induces senescence in developing fibroblasts. Using primary human fetal lung fibroblasts, we investigated effects of 40% O2 on senescence, endoplasmic reticulum (ER) stress, and autophagy pathways. Fibroblasts were exposed to 21% or 40% O2 for 7 days with etoposide as a positive control to induce senescence, evaluated by morphological changes, ß-galactosidase activity, and DNA damage markers. Senescence-associated secretory phenotype (SASP) profile of inflammatory and profibrotic markers was further assessed. Hyperoxia decreased proliferation but increased cell size. SA-ß-gal activity and DNA damage response, cell cycle arrest in G2/M phase, and marked upregulation of phosphorylated p53 and p21 were noted. Reduced autophagy was noted with hyperoxia. mRNA expression of proinflammatory and profibrotic factors (TNF-α, IL-1, IL-8, MMP3) was elevated by hyperoxia or etoposide. Hyperoxia increased several SASP factors (PAI-1, IL1-α, IL1-ß, IL-6, LAP, TNF-α). The secretome of senescent fibroblasts promoted extracellular matrix formation by naïve fibroblasts. Overall, we demonstrate that moderate hyperoxia enhances senescence in primary human fetal lung fibroblasts with reduced autophagy but not enhanced ER stress. The resulting SASP is profibrotic and may contribute to abnormal repair in the lung following hyperoxia.


Subject(s)
Cellular Senescence/drug effects , Fibroblasts/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation/drug effects , Hyperoxia/genetics , Oxygen/pharmacology , Autophagy/drug effects , Autophagy/genetics , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Proliferation/drug effects , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Endoplasmic Reticulum Stress/drug effects , Etoposide/pharmacology , Extracellular Matrix/chemistry , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fetus , Fibroblasts/cytology , Fibroblasts/metabolism , G2 Phase Cell Cycle Checkpoints/genetics , Humans , Hyperoxia/metabolism , Interleukin-1/genetics , Interleukin-1/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Lung/cytology , Lung/metabolism , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Primary Cell Culture , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
Am J Physiol Lung Cell Mol Physiol ; 317(1): L99-L108, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31042080

ABSTRACT

Reactive airway diseases are significant sources of pulmonary morbidity in neonatal and pediatric patients. Supplemental oxygen exposure in premature infants contributes to airway diseases such as asthma and promotes development of airway remodeling, characterized by increased airway smooth muscle (ASM) mass and extracellular matrix (ECM) deposition. Decreased plasma membrane caveolin-1 (CAV1) expression has been implicated in airway disease and may contribute to airway remodeling and hyperreactivity. Here, we investigated the impact of clinically relevant moderate hyperoxia (50% O2) on airway remodeling and caveolar protein expression in a neonatal mouse model. Within 12 h of birth, litters of B6129SF2J mice were randomized to room air (RA) or 50% hyperoxia exposure for 7 days with or without caveolin-1 scaffolding domain peptide (CSD; caveolin-1 mimic; 10 µl, 0.25 mM daily via intraperitoneal injection) followed by 14 days of recovery in normoxia. Moderate hyperoxia significantly increased airway reactivity and decreased pulmonary compliance at 3 wk. Histologic assessment demonstrated airway wall thickening and increased ASM mass following hyperoxia. RNA from isolated ASM demonstrated significant decreases in CAV1 and cavin-1 in hyperoxia-exposed animals while cavin-3 was increased. Supplementation with intraperitoneal CSD mitigated both the physiologic and histologic changes observed with hyperoxia. Overall, these data show that moderate hyperoxia is detrimental to developing airway and may predispose to airway reactivity and remodeling. Loss of CAV1 is one mechanism through which hyperoxia produces these deleterious effects. Supplementation of CAV1 using CSD or similar analogs may represent a new therapeutic avenue for blunting hyperoxia-induced pulmonary damage in neonates.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bronchial Hyperreactivity/drug therapy , Caveolin 1/pharmacology , Hyperoxia/drug therapy , Lung/drug effects , Peptide Fragments/pharmacology , Airway Remodeling/drug effects , Airway Remodeling/immunology , Animals , Animals, Newborn , Bronchial Hyperreactivity/etiology , Bronchial Hyperreactivity/genetics , Bronchial Hyperreactivity/immunology , Bronchoconstrictor Agents/pharmacology , Caveolin 1/genetics , Caveolin 1/immunology , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Hyperoxia/etiology , Hyperoxia/genetics , Hyperoxia/immunology , Injections, Intraperitoneal , Lung/immunology , Lung/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Methacholine Chloride/pharmacology , Mice , Oxygen/adverse effects , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Signal Transduction
8.
Thorax ; 74(8): 749-760, 2019 08.
Article in English | MEDLINE | ID: mdl-31182654

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a fatal ageing-related disease linked to mitochondrial dysfunction. The present study aimed to determine whether peroxisome proliferator activated receptor gamma co-activator 1-alpha (PPARGC1A, encoding PGC1α), a master regulator of mitochondrial biogenesis, is diminished in IPF and controls pathologic fibroblast activation. Primary human IPF, control lung fibroblasts and fibroblasts sorted from bleomycin-injured mice were used to evaluate the expression and function of PGC1α. In vitro PGC1α manipulation was performed by small interfering RNA knockdown or overexpression. Fibroblast activation was assessed by quantitative PCR, Western blotting, matrix deposition, secreted cytokine array, immunofluorescence and traction force microscopy. Mitochondrial function was assessed by Seahorse analyzer and mitochondria mass and number by flow cytometry, mitochondrial DNA quantification and transmission electron microscopy (TEM). We found that PGC1α levels are stably repressed in IPF fibroblasts. After bleomycin injury in young mice, PGC1α expression drops transiently but then increases prior to fibrosis resolution. In contrast, PGC1α expression fails to recover in aged mice with persistent fibrosis. PGC1α knockdown alone in normal human lung fibroblasts reduces mitochondrial mass and function while enhancing contractile and matrix synthetic fibroblast activation, senescence-related gene expression and soluble profibrotic and prosenescence signalling. Re-expression of PGC1α in IPF fibroblasts ameliorates all of these pathological cellular functions. Pharmacological treatment of IPF fibroblasts with rosiglitazone, but not thyroid hormone, elevated PGC1α expression and attenuated fibroblast activation. The sustained repression of PGC1α and beneficial effects of its rescue in IPF fibroblasts identifies PGC1α as an important regulator of the fibroblast's pathological state in IPF.


Subject(s)
Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Mitochondria/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Actins/genetics , Animals , Bleomycin , Cell Line , Cellular Senescence/genetics , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Cyclin-Dependent Kinase Inhibitor p16/genetics , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression/drug effects , Gene Knockdown Techniques , Humans , Hypoglycemic Agents/pharmacology , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/pathology , Mice , NAD/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Primary Cell Culture , RNA, Small Interfering , Rosiglitazone/pharmacology , Signal Transduction/genetics , Triiodothyronine/pharmacology , beta-Galactosidase/genetics
9.
Am J Physiol Lung Cell Mol Physiol ; 313(2): L360-L370, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28522569

ABSTRACT

Airway remodeling in asthma driven by inflammation involves proliferation of epithelial cells and airway smooth muscle (ASM), as well as enhanced extracellular matrix (ECM) generation and deposition, i.e., fibrosis. Accordingly, understanding profibrotic mechanisms is important for developing novel therapeutic strategies in asthma. Recent studies, including our own, have suggested a role for locally produced growth factors such as brain-derived neurotrophic factor (BDNF) in mediating and modulating inflammation effects. In this study, we explored the profibrotic influence of BDNF in the context of asthma by examining expression, activity, and deposition of ECM proteins in primary ASM cells isolated from asthmatic vs. nonasthmatic patients. Basal BDNF expression and secretion, and levels of the high-affinity BDNF receptor TrkB, were higher in asthmatic ASM. Exogenous BDNF significantly increased ECM production and deposition, especially of collagen-1 and collagen-3 (less so fibronectin) and the activity of matrix metalloproteinases (MMP-2, MMP-9). Exposure to the proinflammatory cytokine TNFα significantly increased BDNF secretion, particularly in asthmatic ASM, whereas no significant changes were observed with IL-13. Chelation of BDNF using TrkB-Fc reversed TNFα-induced increase in ECM deposition. Conditioned media from asthmatic ASM enhanced ECM generation in nonasthmatic ASM, which was blunted by BDNF chelation. Inflammation-induced changes in MMP-2, MMP-9, and tissue inhibitor metalloproteinases (TIMP-1, TIMP-2) were reversed in the presence of TrkB-Fc. These novel data suggest ASM as an inflammation-sensitive source of BDNF within human airways, with autocrine effects on fibrosis relevant to asthma.


Subject(s)
Airway Remodeling/physiology , Asthma/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Fibrosis/metabolism , Respiratory System/metabolism , Collagen Type I/metabolism , Collagen Type III/metabolism , Cytokines/metabolism , Humans , Inflammation/metabolism , Interleukin-13/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
Am J Physiol Lung Cell Mol Physiol ; 308(2): L141-6, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25399436

ABSTRACT

Supplemental oxygen, used to treat hypoxia in preterm and term neonates, increases the risk of neonatal lung diseases, such as bronchopulmonary dysplasia (BPD) and asthma. There is a known sex predilection for BPD, but the underlying mechanisms are not clear. We tested the hypothesis that altered, local estradiol following hyperoxia contributes to pathophysiological changes observed in immature lung. In human fetal airway smooth muscle (fASM) cells exposed to normoxia or hyperoxia, we measured the expression of proteins involved in estrogen metabolism and cell proliferation responses to estradiol. In fASM cells, CYP1a1 expression was increased by hyperoxia, whereas hyperoxia-induced enhancement of cell proliferation was blunted by estradiol. Pharmacological studies indicated that these effects were attributable to upregulation of CYP1a1 and subsequent increased metabolism of estradiol to a downstream intermediate 2-methoxyestradiol. Microarray analysis of mouse lung exposed to 14 days of hyperoxia showed the most significant alteration in CYP1a1 expression, with minimal changes in expression of five other genes related to estrogen receptors, synthesis, and metabolism. Our novel results on estradiol metabolism in fetal and early postnatal lung in the context of hyperoxia indicate CYP1a1 as a potential mechanism for the protective effect of estradiol in hyperoxia-exposed immature lung, which may help explain the sex difference in neonatal lung diseases.


Subject(s)
Cytochrome P-450 CYP1A1/biosynthesis , Estradiol/metabolism , Hyperoxia/physiopathology , Lung/embryology , 2-Methoxyestradiol , Animals , Apoptosis , Aromatase/biosynthesis , Asthma/epidemiology , Bronchopulmonary Dysplasia/epidemiology , Catechol O-Methyltransferase/biosynthesis , Cell Hypoxia/physiology , Cell Proliferation , Cells, Cultured , Cytochrome P-450 CYP1B1/biosynthesis , Estradiol/analogs & derivatives , Estradiol/biosynthesis , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/biosynthesis , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Humans , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred ICR , Muscle, Smooth/metabolism , Oxygen/metabolism , RNA, Messenger/biosynthesis , Reactive Oxygen Species/metabolism , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Sex Factors , Up-Regulation
11.
J Neurointerv Surg ; 11(5): 523-527, 2019 May.
Article in English | MEDLINE | ID: mdl-30415228

ABSTRACT

BACKGROUND: Flow diverters (FDs) are increasingly used in the treatment of intracranial aneurysms, and carry the risk of thromboembolic complications, even in patients treated with dual antiplatelet therapy. The effect of FDs on the downstream vascular is unknown. The aim of the study was to investigate vascular wall pulse wave velocity (PWV) and contractility changes following FD treatment in a rabbit model. METHODS: FDs (Pipeline Embolic Device, Medtronic Inc., Irvine, California, USA) were implanted in the aorta of normal rabbits and sham-operated aorta were used as controls (n=6 per group). Pulse wave imaging with ultra-fast ultrasound at 1600 frames per second (Vantage, Verasonics, Inc., Kirkland, WA) was performed in the vessel wall distal to FD prior to device implantation and at 8- week follow-up to measure the PWV. Force contraction vascular reactivity studies were conducted in the aortic rings using an organ bath. RESULTS: The difference in mean PWV in the follow-up compared with pre-implantation was significantly higher in the distal vessels compared with sham controls (1.18 m/s [SD=0.54] vs. 0.37 m/s [SD=1.09], P=0.03). Conversely, the aortic segments distal to the FD exhibited a 55% increase in vascular contractility compared with proximal segments (P=0.002). We observed a significant positive correlation between mean PWV and mean vascular contractility. CONCLUSION: Implantation of FD was associated with increased PWV and vascular contractility, suggesting that FD implantation causes changes to the vascular wall. Further studies are needed to understand the clinical implication of changes in vascular PWV and contractility.


Subject(s)
Blood Vessel Prosthesis , Cerebrovascular Circulation , Animals , Aorta, Thoracic/anatomy & histology , Aorta, Thoracic/diagnostic imaging , Aorta, Thoracic/surgery , Blood Vessel Prosthesis Implantation , Embolization, Therapeutic , Muscle Contraction , Muscle, Smooth, Vascular , Pulse Wave Analysis , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL