Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Pathol ; 243(4): 510-523, 2017 12.
Article in English | MEDLINE | ID: mdl-28862768

ABSTRACT

Asthma is a chronic inflammatory disease of the airways. It is characterized by allergic airway inflammation, airway remodelling, and airway hyperresponsiveness (AHR). Asthma patients, in particular those with chronic or severe asthma, have airway remodelling that is associated with the accumulation of extracellular matrix (ECM) proteins, such as collagens. Fibulin-1 (Fbln1) is an important ECM protein that stabilizes collagen and other ECM proteins. The level of Fbln1c, one of the four Fbln1 variants, which predominates in both humans and mice, is increased in the serum and airways fluids in asthma but its function is unclear. We show that the level of Fbln1c was increased in the lungs of mice with house dust mite (HDM)-induced chronic allergic airway disease (AAD). Genetic deletion of Fbln1c and therapeutic inhibition of Fbln1c in mice with chronic AAD reduced airway collagen deposition, and protected against AHR. Fbln1c-deficient (Fbln1c-/- ) mice had reduced mucin (MUC) 5 AC levels, but not MUC5B levels, in the airways as compared with wild-type (WT) mice. Fbln1c interacted with fibronectin and periostin that was linked to collagen deposition around the small airways. Fbln1c-/- mice with AAD also had reduced numbers of α-smooth muscle actin-positive cells around the airways and reduced airway contractility as compared with WT mice. After HDM challenge, these mice also had fewer airway inflammatory cells, reduced interleukin (IL)-5, IL-13, IL-33, tumour necrosis factor (TNF) and CXCL1 levels in the lungs, and reduced IL-5, IL-33 and TNF levels in lung-draining lymph nodes. Therapeutic targeting of Fbln1c reduced the numbers of GATA3-positive Th2 cells in the lymph nodes and lungs after chronic HDM challenge. Treatment also reduced the secretion of IL-5 and IL-13 from co-cultured dendritic cells and T cells restimulated with HDM extract. Human epithelial cells cultured with Fbln1c peptide produced more CXCL1 mRNA than medium-treated controls. Our data show that Fbln1c may be a therapeutic target in chronic asthma. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Airway Remodeling , Asthma/metabolism , Bronchial Hyperreactivity/metabolism , Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Inflammation/metabolism , Lung/metabolism , Actins/metabolism , Animals , Asthma/immunology , Asthma/physiopathology , Asthma/prevention & control , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/physiopathology , Bronchial Hyperreactivity/prevention & control , Bronchoconstriction , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Cells, Cultured , Coculture Techniques , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Female , Genotype , Humans , Inflammation/immunology , Inflammation/physiopathology , Inflammation/prevention & control , Inflammation Mediators/metabolism , Lung/immunology , Lung/physiopathology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA Interference , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Transfection
2.
J Biol Chem ; 290(35): 21629-41, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26152723

ABSTRACT

Arterial smooth muscle cells (ASMCs) undergo phenotypic changes during development and pathological processes in vivo and during cell culture in vitro. Our previous studies demonstrated that retrovirally mediated expression of the versican V3 splice variant (V3) by ASMCs retards cell proliferation and migration in vitro and reduces neointimal thickening and macrophage and lipid accumulation in animal models of vascular injury and atherosclerosis. However, the molecular pathways induced by V3 expression that are responsible for these changes are not yet clear. In this study, we employed a microarray approach to examine how expression of V3 induced changes in gene expression and the molecular pathways in rat ASMCs. We found that forced expression of V3 by ASMCs affected expression of 521 genes by more than 1.5-fold. Gene ontology analysis showed that components of the extracellular matrix were the most significantly affected by V3 expression. In addition, genes regulating the formation of the cytoskeleton, which also serve as markers of contractile smooth muscle cells (SMCs), were significantly up-regulated. In contrast, components of the complement system, chemokines, chemokine receptors, and transcription factors crucial for regulating inflammatory processes were among the genes most down-regulated. Consistently, we found that the level of myocardin, a key transcription factor promoting contractile SMC phenotype, was greatly increased, and the proinflammatory transcription factors NFκB1 and CCAAT/enhancer-binding protein ß were significantly attenuated in V3-expressing SMCs. Overall, these findings demonstrate that V3 expression reprograms ASMCs promoting differentiated and anti-inflammatory phenotypes.


Subject(s)
Anti-Inflammatory Agents/metabolism , Arteries/cytology , Cell Differentiation , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Versicans/metabolism , Animals , Apoptosis/genetics , Biomarkers/metabolism , Cell Survival/genetics , Cellular Microenvironment , Cluster Analysis , Down-Regulation/genetics , Gene Expression Profiling , Inflammation/genetics , Inflammation/pathology , Molecular Sequence Annotation , Oligonucleotide Array Sequence Analysis , Phenotype , Rats, Inbred F344 , Response Elements/genetics , Software , Up-Regulation/genetics , Versicans/genetics
3.
J Am Soc Nephrol ; 25(5): 1028-36, 2014 May.
Article in English | MEDLINE | ID: mdl-24357674

ABSTRACT

Cubilin is an endocytic receptor highly expressed in renal proximal tubules, where it mediates uptake of albumin and filtered forms of apoA-I/HDL. Cubilin deficiency leads to urinary loss of albumin and apoA-I; however, the consequences of cubilin loss on the homeostasis of blood albumin and apoA-I/HDL have not been studied. Using mice heterozygous for cubilin gene deletion (cubilin HT mice), we show that cubilin haploinsufficiency leads to reduced renal proximal tubular uptake of albumin and apoA-I and significantly increased urinary loss of albumin and apoA-I. Moreover, cubilin HT mice displayed significantly decreased blood levels of albumin, apoA-I, and HDL. The levels of albumin and apoA-I protein or mRNA expressed in the liver, kidney, or intestine of cubilin HT mice did not change significantly. The clearance rate of small HDL3 particles (density>1.13 g/ml) from the blood increased significantly in cubilin HT mice. In contrast, the rate of clearance of larger HDL2 particles from the blood did not change significantly, indicating a decreased half-life for HDL particles capable of filtering through the glomerulus. On the basis of these findings, we conclude that cubilin deficiency reduces renal salvage and delivery back to the blood of albumin and apoA-I, which decreases blood levels of albumin and apoA-I/HDL. These findings raise the possibility that therapeutic increase of renal cubilin expression might reduce proteinuria and increase blood levels of albumin and HDL.


Subject(s)
Albuminuria/etiology , Albuminuria/genetics , Apolipoprotein A-I/urine , Lipoproteins, HDL/blood , Receptors, Cell Surface/physiology , Albumins/antagonists & inhibitors , Albumins/metabolism , Albuminuria/metabolism , Animals , Apolipoprotein A-I/antagonists & inhibitors , Apolipoprotein A-I/blood , Gene Deletion , Genetic Carrier Screening , Humans , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Lipoproteins, HDL/antagonists & inhibitors , Lipoproteins, HDL/biosynthesis , Lipoproteins, HDL3/antagonists & inhibitors , Lipoproteins, HDL3/blood , Lipoproteins, HDL3/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics
4.
Am J Physiol Gastrointest Liver Physiol ; 306(7): G606-13, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24503769

ABSTRACT

Acute Helicobacter pylori infection of gastric epithelial cells induces CagA oncoprotein- and peptidoglycan (SLT)-dependent mobilization of NF-κB p50 homodimers that bind to H-K-ATPase α-subunit (HKα) promoter and repress HKα gene transcription. This process may facilitate gastric H. pylori colonization by induction of transient hypochlorhydria. We hypothesized that H. pylori also regulates HKα expression posttranscriptionally by miRNA interaction with HKα mRNA. In silico analysis of the HKα 3' untranslated region (UTR) identified miR-1289 as a highly conserved putative HKα-regulatory miRNA. H. pylori infection of AGS cells transfected with HKα 3' UTR-Luc reporter construct repressed luciferase activity by 70%, whereas ΔcagA or Δslt H. pylori infections partially abrogated repression. Transfection of AGS cells expressing HKα 3' UTR-Luc construct with an oligoribonucleotide mimetic of miR-1289 induced maximal repression (54%) of UTR activity within 30 min; UTR activity was unchanged by nontargeting siRNA transfection. Gastric biopsies from patients infected with cagA(+) H. pylori showed a significant increase in miR-1289 expression compared with uninfected patients or those infected with cagA(-) H. pylori. Finally, miR-1289 expression was necessary and sufficient to attenuate biopsy HKα protein expression in the absence of infection. Taken together, these data indicate that miR-1289 is upregulated by H. pylori in a CagA- and SLT-dependent manner and targets HKα 3' UTR, affecting HKα mRNA translation. The sensitivity of HKα mRNA 3' UTR to binding of miR-1289 identifies a novel regulatory mechanism of gastric acid secretion and offers new insights into mechanisms underlying transient H. pylori-induced hypochlorhydria.


Subject(s)
Gastric Mucosa/enzymology , H(+)-K(+)-Exchanging ATPase/metabolism , Helicobacter Infections/enzymology , Helicobacter pylori/metabolism , MicroRNAs/metabolism , NF-kappa B p50 Subunit/metabolism , Parietal Cells, Gastric/enzymology , RNA Processing, Post-Transcriptional , 3' Untranslated Regions , Achlorhydria/enzymology , Achlorhydria/microbiology , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cell Line , Gastric Mucosa/microbiology , Gene Expression Regulation, Enzymologic , Genes, Reporter , H(+)-K(+)-Exchanging ATPase/genetics , Helicobacter Infections/complications , Helicobacter Infections/genetics , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Host-Pathogen Interactions , Humans , NF-kappa B p50 Subunit/genetics , Parietal Cells, Gastric/microbiology , Peptidoglycan/metabolism , RNA Interference , RNA, Messenger/metabolism , Time Factors , Transfection , Virulence
5.
Biometals ; 27(1): 135-41, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24337410

ABSTRACT

We sought to examine the relationship between elevated transferrin saturation (TS) and measures of health status (telomere length and patient-reported health-related quality of life) to assess whether elevated TS is associated with negative patient outcomes beyond increased risk for morbidity and mortality, using a cross-sectional analysis of the Hemochromatosis and Iron Overload Screening Study supplemented with assays for leukocyte telomere length in adults ≥25 years old (n = 669). Among individuals with elevated TS (≥45 % for women and ≥50 % for men), who also had a usual source of care, only 5.2 % reported ever being told by a doctor that they had an elevated iron condition. In a fully adjusted general linear regression model controlling for demographic characteristics as well as health conditions associated with iron overload, elevated TS versus non-elevated TS was associated with worse general health status (60.4 vs. 63.8, P < 0.05), mental health status (76.5 vs. 82.2, P < 0.0001) and shorter telomere length (241.4 vs. 261.3, P < 0.05). Increased surveillance of elevated TS may be in order as elevated TS is associated with decreased health status and very few patients with elevated TS are aware of their condition.


Subject(s)
Quality of Life , Telomere/metabolism , Transferrin/analysis , Adult , Female , Humans , Male , Middle Aged , Real-Time Polymerase Chain Reaction , Telomere/genetics , Transferrin/metabolism
6.
J Biol Chem ; 287(53): 44645-53, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23135269

ABSTRACT

Sphingosine 1-phosphate (S1P) is a blood-borne lysosphingolipid that acts to promote endothelial cell (EC) barrier function. In plasma, S1P is associated with both high density lipoproteins (HDL) and albumin, but it is not known whether the carriers impart different effects on S1P signaling. Here we establish that HDL-S1P sustains EC barrier longer than albumin-S1P. We showed that the sustained barrier effects of HDL-S1P are dependent on signaling by the S1P receptor, S1P1, and involve persistent activation of Akt and endothelial NOS (eNOS), as well as activity of the downstream NO target, soluble guanylate cyclase (sGC). Total S1P1 protein levels were found to be higher in response to HDL-S1P treatment as compared with albumin-S1P, and this effect was not associated with increased S1P1 mRNA or dependent on de novo protein synthesis. Several pieces of evidence indicate that long term EC barrier enhancement activity of HDL-S1P is due to specific effects on S1P1 trafficking. First, the rate of S1P1 degradation, which is proteasome-mediated, was slower in HDL-S1P-treated cells as compared with cells treated with albumin-S1P. Second, the long term barrier-promoting effects of HDL-S1P were abrogated by treatment with the recycling blocker, monensin. Finally, cell surface levels of S1P1 and levels of S1P1 in caveolin-enriched microdomains were higher after treatment with HDL-S1P as compared with albumin-S1P. Together, the findings reveal S1P carrier-specific effects on S1P1 and point to HDL as the physiological mediator of sustained S1P1-PI3K-Akt-eNOS-sGC-dependent EC barrier function.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Lipoproteins, HDL/metabolism , Lysophospholipids/metabolism , Receptors, Lysosphingolipid/metabolism , Serum Albumin/metabolism , Sphingosine/analogs & derivatives , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Transport , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Lysosphingolipid/genetics , Signal Transduction , Sphingosine/metabolism
7.
BMC Genomics ; 14: 405, 2013 Jun 18.
Article in English | MEDLINE | ID: mdl-23773363

ABSTRACT

BACKGROUND: Cubilin is an endocytic receptor that is necessary for renal and intestinal absorption of a range of ligands. Endocytosis mediated by cubilin and its co-receptor megalin is the principal mechanism for proximal tubule reabsorption of proteins from the glomerular filtrate. Cubilin is also required for intestinal endocytosis of intrinsic factor-vitamin B12 complex. Despite its importance, little is known about the regulation of cubilin expression. RESULTS: Here we show that cubilin expression is under epigenetic regulation by at least two processes. The first process involves inactivation of expression of one of the cubilin alleles. This monoallelic expression state could not be transformed to biallelic by inhibiting DNA methylation or histone deacetylation. The second process involves transcriptional regulation of cubilin by peroxisome proliferator-activated receptor (PPAR) transcription factors that are themselves regulated by DNA methylation and histone deacetylation. This is supported by findings that inhibitors of DNA methylation and histone deacetylation, 5Aza and TSA, increase cubilin mRNA and protein in renal and intestinal cell lines. Not only was the expression of PPARα and γ inducible by 5Aza and TSA, but the positive effects of TSA and 5Aza on cubilin expression were also dependent on both increased PPAR transcription and activation. Additionally, 5Aza and TSA had similar effects on the expression of the cubilin co-receptor, megalin. CONCLUSIONS: Together, these findings reveal that cubilin and megalin mRNA expression is under epigenetic control and thus point to new avenues for overcoming pathological suppression of these genes through targeting of epigenetic regulatory processes.


Subject(s)
Alleles , Epigenesis, Genetic , Gene Expression Regulation , Peroxisome Proliferator-Activated Receptors/metabolism , Receptors, Cell Surface/genetics , Albumins/metabolism , Animals , Azacitidine/pharmacology , Base Sequence , Caco-2 Cells , Epigenesis, Genetic/drug effects , Exons/genetics , Gene Expression Regulation/drug effects , Humans , Hydroxamic Acids/pharmacology , Intestines/cytology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Male , Membrane Proteins/genetics , Mice , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Protein Transport , Receptors, Cell Surface/deficiency
8.
Cardiovasc Diabetol ; 12: 6, 2013 Jan 07.
Article in English | MEDLINE | ID: mdl-23294625

ABSTRACT

BACKGROUND: Fibulin-1 is one of a few extracellular matrix proteins present in blood in high concentrations. We aimed to define the relationship between plasma fibulin-1 levels and risk markers of cardiovascular disease. METHODS: Plasma fibulin-1 was determined in subjects with chronic kidney disease (n = 32; median age 62.5, inter-quartile range 51 - 73 years) and 60 age-matched control subjects. Among kidney disease patients serological biomarkers related to cardiovascular disease (fibrinogen, interleukin 6, C-reactive protein) were measured. Arterial applanation tonometry was used to determine central hemodynamic and arterial stiffness indices. RESULTS: We observed a positive correlation of fibulin-1 levels with age (r = 0.38; p = 0.033), glycated hemoglobin (r = 0.80; p = 0.003), creatinine (r = 0.35; p = 0.045), and fibrinogen (r = 0.39; p = 0.027). Glomerular filtration rate and fibulin-1 were inversely correlated (r = -0.57; p = 0.022). There was a positive correlation between fibulin-1 and central pulse pressure (r = 0.44; p = 0.011) and central augmentation pressure (r = 0.55; p = 0.001). In a multivariable regression model, diabetes, creatinine, fibrinogen and central augmentation pressure were independent predictors of plasma fibulin-1. CONCLUSION: Increased plasma fibulin-1 levels were associated with diabetes and impaired kidney function. Furthermore, fibulin-1 levels were associated with hemodynamic cardiovascular risk markers. Fibulin-1 is a candidate in the pathogenesis of cardiovascular disease observed in chronic kidney disease and diabetes.


Subject(s)
Calcium-Binding Proteins/blood , Cardiovascular Diseases/blood , Diabetes Mellitus/blood , Extracellular Matrix Proteins/blood , Renal Insufficiency, Chronic/blood , Aged , Biomarkers/blood , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/epidemiology , Diabetes Mellitus/diagnosis , Diabetes Mellitus/epidemiology , Female , Humans , Male , Middle Aged , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/epidemiology , Risk Factors , Up-Regulation/physiology
9.
Cardiovasc Diabetol ; 12: 107, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23866070

ABSTRACT

BACKGROUND: The arterial system in diabetic patients is characterized by generalized non-atherosclerotic alterations in the vascular extracellular matrix causing increased arterial stiffness compared with subjects without diabetes. The underlying pathophysiology remains elusive. The elastin-associated extracellular matrix protein, fibulin-1, was recently found in higher concentrations in the arterial wall and in plasma in patients with long duration type 2 diabetes. Furthermore, plasma fibulin-1 independently predicted total mortality and was associated with pulse pressure, an indirect measure of arterial stiffness. Whether plasma fibulin-1 is associated with arterial stiffness at earlier phases of type 2 diabetes has not been determined. METHODS: In this cross-sectional study, we examined 90 patients with recently diagnosed type 2 diabetes (< 5 years) and 90 gender- and age-matched controls. Plasma fibulin-1 was measured immunochemically. Arterial stiffness was assessed by carotid-femoral Pulse Wave Velocity (PWV). Differences in means were assessed by t-tests. Associations were assessed by multivariate regression analyses. RESULTS: Plasma fibulin-1 levels were lower in the diabetic group compared with the control group, 93 ± 28 vs 106 ± 30 µg/mL, p = 0.005. In unadjusted analysis of the total study sample, plasma fibulin-1 was not associated with PWV, p = 0.46. However, with adjustment for the confounders age, gender, mean blood pressure, heart rate, body mass index, diabetes and glomerular filtration rate, a 10 µg/mL increase in plasma fibulin was associated with 0.09 ± 0.04 m/s increase in PWV, p < 0.05. In subgroup analysis, plasma fibulin-1 was associated with PWV in the diabetes group, (0.16 ± 0.07 m/s increase in PWV per 10 µg/mL increase in plasma fibulin-1, p<0.05), but not controls, ß = 0.021 ± 0.057 m/s per 10 µg/mL, p = 0.70. The association remained significant in the diabetes group after adjustment for covariates, p < 0.05. CONCLUSIONS: Plasma fibulin-1 is independently associated with PWV. Yet, as the plasma level of fibulin-1 was lower in patients with recently diagnosed type 2 diabetes than in healthy controls, plasma fibulin-1 levels are not a simple marker of the degree of arterial stiffening. Further studies are needed to determine the exact role of fibulin-1 in arterial stiffness and cardiovascular risk in patients with type 2 diabetes.


Subject(s)
Calcium-Binding Proteins/blood , Carotid Arteries/physiopathology , Diabetes Mellitus, Type 2/complications , Diabetic Angiopathies/diagnosis , Diabetic Angiopathies/etiology , Femoral Artery/physiopathology , Pulse Wave Analysis , Vascular Stiffness , Aged , Biomarkers/blood , Case-Control Studies , Cross-Sectional Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Diabetic Angiopathies/blood , Diabetic Angiopathies/physiopathology , Humans , Immunoassay , Male , Middle Aged , Multivariate Analysis , Predictive Value of Tests , Risk Factors
10.
Am J Hematol ; 88(6): 492-6, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23512844

ABSTRACT

Elevated body iron stores are associated with morbidity and mortality due to oxidative stress. Hereditary hemochromatosis, a common condition caused by HFE gene mutations, can lead to excess iron storage and disease but clinical penetrance of HFE gene mutations is low and many people with elevated iron stores lack HFE mutations. We analyzed data from the Hemochromatosis and Iron Overload Screening Study to assess the relationship among HFE genotype (individuals with either homozygous or compound heterozygous status for C282Y and/or H63D HFE mutations were defined as genotype positive, or G+), elevated iron phenotype (individuals exceeding gender-specific transferrin saturation and serum ferritin threshold levels were considered phenotype positive, or P+), and leukocyte telomere length, a marker of biological aging and cumulative oxidative stress. In unadjusted analyses in comparison to individuals who were G-P-, G+P- were not significantly different (OR 0.74; 95% CI 0.26-2.04), while the G+P+ (OR 2.03; 95% CI 1.15-3.56), and G-P+ (OR 2.24; 95% CI 1.5-3.29) had increased risk of short telomeres (<=25th percentile) rather than long telomeres (>=75th percentile). In analyses adjusting for age, gender, and race/ethnicity, the effect of individuals with elevated iron phenotypes having short telomeres persisted with G+P+ individuals (OR 1.94; 95% CI 1.02-3.72), and G-P+ individuals (OR 2.17; 95% CI 1.39-3.39) being significantly different from the G-P- group. In conclusion, elevated iron phenotype, but not HFE genotype, was associated with shortened telomeres. Further studies will be needed to determine whether telomere length provides a marker for morbidities specifically associated with iron overload.


Subject(s)
Hemochromatosis/genetics , Histocompatibility Antigens Class I/genetics , Iron/metabolism , Membrane Proteins/genetics , Telomere/ultrastructure , Adult , Female , Genotype , Hemochromatosis/blood , Hemochromatosis Protein , Histocompatibility Antigens Class I/metabolism , Humans , Iron/blood , Iron Overload/genetics , Iron Overload/metabolism , Male , Membrane Proteins/metabolism , Mutation , Phenotype , Telomere/chemistry , Telomere/metabolism
11.
Cardiology ; 126(3): 202-6, 2013.
Article in English | MEDLINE | ID: mdl-24051963

ABSTRACT

OBJECTIVES: Aortic valve stenosis (AS) causes cardiac fibrosis and left ventricular hypertrophy, and over time heart failure can occur. To date, a reliable marker to predict progression of AS or the development of heart failure is still lacking. In this study, we addressed the hypothesis that fibulin-1 levels reflect myocardial fibrosis. METHODS: Patients undergoing heart surgery at the Odense University were investigated. By 2012 data on outcome were obtained. RESULTS: In 293 patients, plasma fibulin-1 levels were measured. Patients with AS or atrial fibrillation (AF) had significantly higher fibulin-1 levels compared to those with coronary artery disease only (p = 0.005). Patients with preoperatively diagnosed chronic AF had significantly higher levels of fibulin-1 compared to those without (p = 0.004). Plasma fibulin-1 levels showed no relationship to echocardiographic size and had no impact on outcome, death or other adverse events. CONCLUSION: This study shows that plasma fibulin-1 levels are increased in patients with AS and AF compared to patients with coronary disease only. Our study results suggest fibulin-1, a vascular extracellular matrix (ECM) protein, as a marker of ECM turnover perhaps due to the increased myocardial stretch that is related to pressure overload.


Subject(s)
Aortic Valve Stenosis/blood , Aortic Valve Stenosis/surgery , Atrial Fibrillation/blood , Calcium-Binding Proteins/blood , Coronary Artery Bypass , Myocardium/pathology , Aged , Aortic Valve Stenosis/complications , Atrial Fibrillation/complications , Atrial Fibrillation/surgery , Cause of Death , Coronary Artery Disease/blood , Female , Fibrosis/blood , Humans , Male , Prognosis
12.
Dev Dyn ; 241(2): 303-14, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22183742

ABSTRACT

BACKGROUND: Trabeculation is an integral component of cardiac ventricular morphogenesis and is dependent on the matrix metalloproteinase, ADAMTS1. A substrate of ADAMTS1 is the proteoglycan versican which is expressed in the developing ventricle and which has been implicated in trabeculation. Fibulin-1 is a versican and ADAMTS1-binding extracellular matrix protein required for ventricular morphogenesis. Here we investigated the involvement of fibulin-1 in ADAMTS1-mediated cleavage of versican in vitro, and the involvement of fibulin-1 in versican cleavage in ventricular morphogenesis. RESULTS: We show that fibulin-1 is a cofactor for ADAMTS1-dependent in vitro cleavage of versican V1, yielding a 70-kDa amino-terminal fragment. Furthermore, fibulin-1-deficiency in mice was found to cause a significant reduction (>90%) in ventricular levels of the 70-kDa versican V1 cleavage product and a 2-fold increase in trabecular cardiomyocyte proliferation. Decreased versican V1 cleavage and augmented trabecular cardiomyocyte proliferation in fibulin-1 null hearts is accompanied by increased ventricular activation of ErbB2 and Erk1/2. By contrast, versican deficiency was found to lead to decreased cardiomyocyte proliferation and reduced ventricular trabeculation. CONCLUSION: We conclude that fibulin-1 regulates versican-dependent events in ventricular morphogenesis by promoting ADAMTS1 cleavage of versican leading to suppression of trabecular cardiomyocyte proliferation mediated by the ErbB2-Map kinase pathway.


Subject(s)
ADAM Proteins/metabolism , Calcium-Binding Proteins/metabolism , Cell Proliferation , Heart Ventricles/embryology , Morphogenesis , Myocytes, Cardiac/physiology , ADAMTS1 Protein , Animals , Calcium-Binding Proteins/genetics , Heart Ventricles/cytology , Heart Ventricles/metabolism , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptor, ErbB-2/metabolism
13.
Pediatr Cardiol ; 33(7): 1154-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22411716

ABSTRACT

Despite the emerging relevance of high-density lipoprotein (HDL) in the inflammatory cascade and vascular barrier integrity, HDL levels in children undergoing cardiac surgery are unexplored. As a measure of HDL levels, the HDL-cholesterol (HDL-C) in single-ventricle patients was quantified before and after the Fontan operation, and it was determined whether relationships existed between the duration and the type of postoperative pleural effusions. The study prospectively enrolled 12 children undergoing the Fontan operation. Plasma HDL-C levels were measured before and after cardiopulmonary bypass. The outcome variables of interest were the duration and type of chest tube drainage (chylous vs. nonchylous). The Kendall rank correlation coefficient and the Wilcoxon rank sum test were used. There were 11 complete observations. The median preoperative HDL-C level for all the subjects was 30 mg/dl (range, 24-53 mg/dl), and the median postcardiopulmonary bypass level was 21 mg/dl (range, 14-46 mg/dl) (p = 0.004). There was a tendency toward a moderate inverse correlation (-0.42) between the postcardiopulmonary bypass HDL-C level and the duration of chest tube drainage, but the result was not statistically significant (p = 0.07). In the chylous effusion group, the median postcardiopulmonary bypass HDL-C tended to be lower (16 vs. 23 mg/dl; p = 0.09). After the Fontan operation, the plasma HDL-C levels in children are significantly reduced. It is reasonable to conclude that the reduction in HDL-C reflects reduced plasma levels of HDL particles, which may have pertinent implications in postoperative pleural effusions given the antiinflammatory and endothelial barrier functions of HDL.


Subject(s)
Cholesterol, HDL/blood , Fontan Procedure , Heart Defects, Congenital/blood , Heart Defects, Congenital/surgery , Cardiopulmonary Bypass , Chest Tubes , Child , Child, Preschool , Female , Humans , Infant , Male , Pleural Effusion/blood , Postoperative Complications/blood , Prospective Studies , Statistics, Nonparametric , Treatment Outcome
14.
Lipids Health Dis ; 10: 70, 2011 May 09.
Article in English | MEDLINE | ID: mdl-21554699

ABSTRACT

BACKGROUND: The lysosphingolipid sphingosine 1-phosphate (S1P) is carried in the blood in association with lipoproteins, predominantly high density lipoproteins (HDL). Emerging evidence suggests that many of the effects of HDL on cardiovascular function may be attributable to its S1P cargo. METHODS: Here we have evaluated how levels of S1P and related sphingolipids in an HDL-containing fraction of human serum correlate with occurrence of ischemic heart disease (IHD). To accomplish this we used liquid chromatography-mass spectrometry to measure S1P levels in the HDL-containing fraction of serum (depleted of LDL and VLDL) from 204 subjects in the Copenhagen City Heart Study (CCHS). The study group consisted of individuals having high serum HDL cholesterol (HDL-C) (females:≥ 73.5 mg/dL; males:≥ 61.9 mg/dL) and verified IHD; subjects with high HDL-C and no IHD; individuals with low HDL-C (females:≤ 38.7 mg/dL; males:≤ 34.1 mg/dL) and IHD, and subjects with low HDL-C and no IHD. RESULTS: The results show a highly significant inverse relationship between the level of S1P in the HDL-containing fraction of serum and the occurrence of IHD. Furthermore, an inverse relationship with IHD was also observed for two other sphingolipids, dihydro-S1P and C24:1-ceramide, in the HDL-containing fraction of serum. Additionally, we demonstrated that the amount of S1P on HDL correlates with the magnitude of HDL-induced endothelial cell barrier signaling. CONCLUSIONS: These findings indicate that compositional differences of sphingolipids in the HDL-containing fraction of human serum are related to the occurrence of IHD, and may contribute to the putative protective role of HDL in IHD.


Subject(s)
Lipoproteins, HDL/blood , Lysophospholipids/blood , Myocardial Ischemia/blood , Myocardial Ischemia/epidemiology , Sphingosine/analogs & derivatives , Cell Movement/drug effects , Ceramides/blood , Chemical Fractionation , Chromatography, Liquid , Denmark/epidemiology , Electric Impedance , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Female , Humans , Lysophospholipids/pharmacology , Male , Mass Spectrometry , Middle Aged , ROC Curve , Sphingolipids/blood , Sphingosine/blood , Sphingosine/pharmacology
15.
Dev Dyn ; 239(2): 398-406, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19918756

ABSTRACT

We evaluated the self-assembly properties of uniluminal vascular spheroids having outer layers of vascular smooth muscle cells and a contiguous inner layer of endothelial cells lining a central lumen. We showed that while pairs of uniluminal vascular spheroids suspended in culture medium fused to form a larger diameter spheroidal structure, spheroids in collagen hydrogels formed elongated structures. These findings highlight the potential use of uniluminal vascular spheroids as modules to engineer blood vessels. We also demonstrate that uniluminal vascular spheroid fusion conforms to models describing the coalescence of liquid drops. Furthermore, the fusion of uniluminal vascular spheroids in vitro closely resembled the in vivo process by which the descending aorta forms from the fusion of the paired dorsal aortae during embryonic development. Together, the findings indicate that tissue liquidity underlies uniluminal vascular spheroid fusion and that in vivo anastomosis of blood vessels may involve a similar mechanism.


Subject(s)
Blood Vessels/embryology , Spheroids, Cellular/physiology , Animals , Aorta/embryology , Cell Fusion , Collagen , Female , Hydrogels , Mice , Models, Cardiovascular , Pregnancy , Rabbits , Rats , Tissue Engineering
16.
Dev Dyn ; 239(7): 2024-33, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20549724

ABSTRACT

Nkx2.5, a transcription factor implicated in human congenital heart disease, is required for regulation of second heart field (SHF) progenitors contributing to outflow tract (OFT). Here, we define a set of genes (Lrrn1, Elovl2, Safb, Slc39a6, Khdrbs1, Hoxb4, Fez1, Ccdc117, Jarid2, Nrcam, and Enpp3) expressed in SHF containing pharyngeal arch tissue whose regulation is dependent on Nkx2.5. Further investigation shows that Jarid2, which has been implicated in OFT morphogenesis, is a direct target of Nkx2.5 regulation. Jarid2 expression was up-regulated in SHF mesoderm of Nkx2.5-deficient embryos. Chromatin immunoprecipitation analysis showed Nkx2.5 interaction with consensus binding sites in the Jarid2 promoter in pharyngeal arch cells. Finally, Jarid2 promoter activity and mRNA expression levels were down-regulated by Nkx2.5 overexpression. Given the role of Jarid2 as a regulator of early cardiac proliferation, these findings highlight Jarid2 as one of several potential mediators of the critical role played by Nkx2.5 during OFT morphogenesis.


Subject(s)
Heart/embryology , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/genetics , Transcription Factors/metabolism , Animals , Binding Sites , Branchial Region/embryology , Branchial Region/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Endoderm/metabolism , Heart Ventricles/embryology , Heart Ventricles/metabolism , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , In Situ Hybridization , Mesoderm/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Polycomb Repressive Complex 2 , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
17.
BMC Bioinformatics ; 11: 166, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20359363

ABSTRACT

BACKGROUND: An important objective of DNA microarray-based gene expression experimentation is determining inter-relationships that exist between differentially expressed genes and biological processes, molecular functions, cellular components, signaling pathways, physiologic processes and diseases. RESULTS: Here we describe GeneMesh, a web-based program that facilitates analysis of DNA microarray gene expression data. GeneMesh relates genes in a query set to categories available in the Medical Subject Headings (MeSH) hierarchical index. The interface enables hypothesis driven relational analysis to a specific MeSH subcategory (e.g., Cardiovascular System, Genetic Processes, Immune System Diseases etc.) or unbiased relational analysis to broader MeSH categories (e.g., Anatomy, Biological Sciences, Disease etc.). Genes found associated with a given MeSH category are dynamically linked to facilitate tabular and graphical depiction of Entrez Gene information, Gene Ontology information, KEGG metabolic pathway diagrams and intermolecular interaction information. Expression intensity values of groups of genes that cluster in relation to a given MeSH category, gene ontology or pathway can be displayed as heat maps of Z score-normalized values. GeneMesh operates on gene expression data derived from a number of commercial microarray platforms including Affymetrix, Agilent and Illumina. CONCLUSIONS: GeneMesh is a versatile web-based tool for testing and developing new hypotheses through relating genes in a query set (e.g., differentially expressed genes from a DNA microarray experiment) to descriptors making up the hierarchical structure of the National Library of Medicine controlled vocabulary thesaurus, MeSH. The system further enhances the discovery process by providing links between sets of genes associated with a given MeSH category to a rich set of html linked tabular and graphic information including Entrez Gene summaries, gene ontologies, intermolecular interactions, overlays of genes onto KEGG pathway diagrams and heatmaps of expression intensity values. GeneMesh is freely available online at http://proteogenomics.musc.edu/genemesh/.


Subject(s)
Gene Expression Profiling/methods , Medical Subject Headings , Oligonucleotide Array Sequence Analysis/methods , Software , Databases, Genetic , Internet , User-Computer Interface
18.
Dev Biol ; 319(2): 336-45, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18538758

ABSTRACT

Here we report that mouse embryos homozygous for a gene trap insertion in the fibulin-1 (Fbln1) gene are deficient in Fbln1 and exhibit cardiac ventricular wall thinning and ventricular septal defects with double outlet right ventricle or overriding aorta. Fbln1 nulls also display anomalies of aortic arch arteries, hypoplasia of the thymus and thyroid, underdeveloped skull bones, malformations of cranial nerves and hemorrhagic blood vessels in the head and neck. The spectrum of malformations is consistent with Fbln1 influencing neural crest cell (NCC)-dependent development of these tissues. This is supported by evidence that Fbln1 expression is associated with streams of cranial NCCs migrating adjacent to rhombomeres 2-7 and that Fbln1-deficient embryos display patterning anomalies of NCCs forming cranial nerves IX and X, which derive from rhombomeres 6 and 7. Additionally, Fbln1-deficient embryos show increased apoptosis in areas populated by NCCs derived from rhombomeres 4, 6 and 7. Based on these findings, it is concluded that Fbln1 is required for the directed migration and survival of cranial NCCs contributing to the development of pharyngeal glands, craniofacial skeleton, cranial nerves, aortic arch arteries, cardiac outflow tract and cephalic blood vessels.


Subject(s)
Calcium-Binding Proteins/genetics , Morphogenesis/physiology , Neural Crest/physiology , Animals , CD4 Antigens/genetics , Calcium-Binding Proteins/deficiency , Cerebrovascular Circulation/genetics , Chromosome Mapping , Crosses, Genetic , Endoplasmic Reticulum/physiology , Fetal Heart/pathology , Fetal Heart/physiology , Genotype , Heart Ventricles/embryology , Heart Ventricles/pathology , Immunohistochemistry , Mice , Mice, Knockout , Polymerase Chain Reaction , beta-Galactosidase/genetics
19.
Histochem Cell Biol ; 132(5): 559-65, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19693531

ABSTRACT

Fibulin-1 is a fibrinogen-binding blood protein and a component of many extracellular matrices (ECM) including those of blood vessels. In this study, the deposition patterns of fibulin-1 and fibrinogen were examined in human coronary artery atherosclerotic lesions excised by atherectomy from 20 patients. Fibulin-1 deposition was found to be closely overlapping with fibrinogen located within the atherosclerotic lesions and in regions containing fresh thrombi. Pronounced intracellular fibulin-1 immunostaining was apparent in lesion areas rich in macrophages and foam cells, although THP-1 macrophages and foam cells were found not to express fibulin-1. Strong ECM deposition of fibulin-1 was observed in acellular atheromatous and myxomatous regions. By contrast, fibulin-1 was present at relatively low levels in the ECM associated with smooth muscle cells within and outside of lesions and was not detected in sclerotic regions. These results reveal the pattern of fibulin-1 within human atherosclerotic lesions and highlight the potential for fibulin-1, perhaps derived from the blood and acting in conjunction with fibrinogen, to play a role in the etiology and cardiovascular disease progression, particularly with respect to thrombotic aspects of atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/pathology , Calcium-Binding Proteins/metabolism , Fibrinogen/metabolism , Adult , Aged , Antibody Specificity , Calcium-Binding Proteins/immunology , Cells, Cultured , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Fibrinogen/immunology , Humans , Immunohistochemistry , Middle Aged
20.
J Appl Physiol (1985) ; 106(2): 582-95, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19074574

ABSTRACT

Spaceflight results in a number of adaptations to skeletal muscle, including atrophy and shifts toward faster muscle fiber types. To identify changes in gene expression that may underlie these adaptations, we used both microarray expression analysis and real-time polymerase chain reaction to quantify shifts in mRNA levels in the gastrocnemius from mice flown on the 11-day, 19-h STS-108 shuttle flight and from normal gravity controls. Spaceflight data also were compared with the ground-based unloading model of hindlimb suspension, with one group of pure suspension and one of suspension followed by 3.5 h of reloading to mimic the time between landing and euthanization of the spaceflight mice. Analysis of microarray data revealed that 272 mRNAs were significantly altered by spaceflight, the majority of which displayed similar responses to hindlimb suspension, whereas reloading tended to counteract these responses. Several mRNAs altered by spaceflight were associated with muscle growth, including the phosphatidylinositol 3-kinase regulatory subunit p85alpha, insulin response substrate-1, the forkhead box O1 transcription factor, and MAFbx/atrogin1. Moreover, myostatin mRNA expression tended to increase, whereas mRNA levels of the myostatin inhibitor FSTL3 tended to decrease, in response to spaceflight. In addition, mRNA levels of the slow oxidative fiber-associated transcriptional coactivator peroxisome proliferator-associated receptor (PPAR)-gamma coactivator-1alpha and the transcription factor PPAR-alpha were significantly decreased in spaceflight gastrocnemius. Finally, spaceflight resulted in a significant decrease in levels of the microRNA miR-206. Together these data demonstrate that spaceflight induces significant changes in mRNA expression of genes associated with muscle growth and fiber type.


Subject(s)
Gene Expression Regulation , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Space Flight , Weightlessness , Adaptation, Physiological/genetics , Animals , Cluster Analysis , Female , Gene Expression Profiling/methods , Hindlimb Suspension , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Atrophy/physiopathology , Myostatin/genetics , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/genetics , Polymerase Chain Reaction , Protein Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/metabolism , Reproducibility of Results , TOR Serine-Threonine Kinases , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL