Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
1.
Cells ; 10(12)2021 11 24.
Article in English | MEDLINE | ID: mdl-34943801

ABSTRACT

Chronic hypoxia increases the resistance of pulmonary arteries by stimulating their contraction and augmenting their coverage by smooth muscle cells (SMCs). While these responses require adjustment of the vascular SMC transcriptome, regulatory elements are not well defined in this context. Here, we explored the functional role of the transcription factor nuclear factor of activated T-cells 5 (NFAT5/TonEBP) in the hypoxic lung. Regulatory functions of NFAT5 were investigated in cultured artery SMCs and lungs from control (Nfat5fl/fl) and SMC-specific Nfat5-deficient (Nfat5(SMC)-/-) mice. Exposure to hypoxia promoted the expression of genes associated with metabolism and mitochondrial oxidative phosphorylation (OXPHOS) in Nfat5(SMC)-/- versus Nfat5fl/fl lungs. In vitro, hypoxia-exposed Nfat5-deficient pulmonary artery SMCs elevated the level of OXPHOS-related transcripts, mitochondrial respiration, and production of reactive oxygen species (ROS). Right ventricular functions were impaired while pulmonary right ventricular systolic pressure (RVSP) was amplified in hypoxia-exposed Nfat5(SMC)-/- versus Nfat5fl/fl mice. Scavenging of mitochondrial ROS normalized the raise in RVSP. Our findings suggest a critical role for NFAT5 as a suppressor of OXPHOS-associated gene expression, mitochondrial respiration, and ROS production in pulmonary artery SMCs that is vital to limit ROS-dependent arterial resistance in a hypoxic environment.


Subject(s)
Hypoxia/pathology , Lung/pathology , Mitochondria/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/pathology , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism , Vascular Resistance , Animals , Blood Pressure , Electrocardiography , Gene Expression Regulation , Heart Ventricles/diagnostic imaging , Heart Ventricles/physiopathology , Metabolome , Mice , Myocytes, Smooth Muscle/pathology , Oxidative Phosphorylation , Oxygen Consumption , Protein Transport , Systole , Transcription Factors/deficiency , Vascular Resistance/genetics
2.
Biol Reprod ; 105(1): 192-203, 2021 07 02.
Article in English | MEDLINE | ID: mdl-33739377

ABSTRACT

Preeclampsia is a pregnancy-related hypertensive disorder with unclear mechanisms. While hypersensitivity to angiotensin II via vasoconstrictive angiotensin type-1 receptor (AT1R) is observed in preeclampsia, the importance of vasodilatory angiotensin type-2 receptor (AT2R) in the control of vascular dysfunction is less clear. We assessed whether AT1R, AT2R, and endothelial nitric oxide synthase (eNOS) expression are altered in placental vessels of preeclamptic women and tested if ex vivo incubation with AT2R agonist Compound 21 (C21; 1 µM) could restore AT1R, AT2R, and eNOS balance. Further, using a rat model of gestational hypertension induced by elevated testosterone, we examined whether C21 (1 µg/kg/day, oral) could preserve AT1R and AT2R balance and improve blood pressure, uterine artery blood flow, and vascular function. Western blots revealed that AT1R protein level was higher while AT2R and eNOS protein were reduced in preeclamptic placental vessels, and AT2R agonist C21 decreased AT1R and increased AT2R and eNOS protein levels in preeclamptic vessels. In testosterone dams, blood pressure was higher, and uterine artery blood flow was reduced, and C21 treatment reversed these levels similar to those in controls dams. C21 attenuated the exaggerated Ang II contraction and improved endothelium-dependent vasorelaxation in uterine arteries of testosterone dams. These C21-mediated vascular effects were associated with decreased AT1R and increased AT2R and eNOS protein levels. C21 also increased serum nitrate/nitrite and bradykinin production in testosterone dams and attenuated the fetoplacental growth restriction. Thus, AT1R upregulation and AT2R downregulation are observed in preeclampsia and testosterone model, and increasing AT2R activity could help restore AT1R and AT2R balance and improve gestational vascular function.


Subject(s)
Hypertension/metabolism , Nitric Oxide Synthase Type III/genetics , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 2/genetics , Testosterone/adverse effects , Uterus/physiology , Vascular Resistance/genetics , Adult , Animals , Female , Humans , Nitric Oxide Synthase Type III/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Young Adult
3.
Int J Mol Sci ; 22(3)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33513830

ABSTRACT

In advanced cirrhosis, the TNFα-mediated intestinal inflammation and bacteria dysbiosis are involved in the development of inflammation and vasoconstriction-related renal dysfunction. In colitis and acute kidney injury models, activation of SIRT1 attenuates the TNFα-mediated intestinal and renal abnormalities. This study explores the impacts of intestinal SIRT1 deficiency and TNFα-mediated intestinal abnormalities on the development of cirrhosis-related renal dysfunction. Systemic and renal hemodynamics, intestinal dysbiosis [cirrhosis dysbiosis ratio (CDR) as marker of dysbiosis], and direct renal vasoconstrictive response (renal vascular resistance (RVR) and glomerular filtration rate (GFR)) to cumulative doses of TNFα were measured in bile duct ligated (BDL)-cirrhotic ascitic mice. In SIRT1IEC-KO-BDL-ascitic mice, the worsening of intestinal dysbiosis exacerbates intestinal inflammation/barrier dysfunction, the upregulation of the expressions of intestinal/renal TNFα-related pathogenic signals, higher TNFα-induced increase in RVR, and decrease in GFR in perfused kidney. In intestinal SIRT1 knockout groups, the positive correlations were identified between intestinal SIRT1 activity and CDR. Particularly, the negative correlations were identified between CDR and RVR, with the positive correlation between CDR and GFR. In mice with advanced cirrhosis, the expression of intestinal SIRT1 is involved in the linkage between intestinal dysbiosis and vasoconstriction/hypoperfusion-related renal dysfunction through the crosstalk between intestinal/renal TNFα-related pathogenic inflammatory signals.


Subject(s)
Inflammation/metabolism , Intestinal Mucosa/metabolism , Kidney/abnormalities , Liver Cirrhosis/metabolism , Sirtuin 1/deficiency , Tumor Necrosis Factor-alpha/metabolism , Urogenital Abnormalities/metabolism , Animals , Gastrointestinal Microbiome/genetics , Glomerular Filtration Rate/genetics , Inflammation/genetics , Inflammation/pathology , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiopathology , Intestines/microbiology , Intestines/physiopathology , Kidney/metabolism , Kidney/physiopathology , Liver Cirrhosis/genetics , Liver Cirrhosis/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sirtuin 1/genetics , Sirtuin 1/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Urogenital Abnormalities/genetics , Urogenital Abnormalities/physiopathology , Vascular Resistance/genetics
4.
Hepatology ; 73(2): 726-737, 2021 02.
Article in English | MEDLINE | ID: mdl-32407592

ABSTRACT

BACKGROUND AND AIMS: Portopulmonary hypertension (POPH) was previously associated with a single-nucleotide polymorphism (SNP) rs7175922 in aromatase (cytochrome P450 family 19 subfamily A member 1 [CYP19A1]). We sought to determine whether genetic variants and metabolites in the estrogen signaling pathway are associated with POPH. APPROACH AND RESULTS: We performed a multicenter case-control study. POPH patients had mean pulmonary artery pressure >25 mm Hg, pulmonary vascular resistance >240 dyn-sec/cm-5 , and pulmonary artery wedge pressure ≤15 mm Hg without another cause of pulmonary hypertension. Controls had advanced liver disease, right ventricular (RV) systolic pressure <40 mm Hg, and normal RV function by echocardiography. We genotyped three SNPs in CYP19A1 and CYP1B1 using TaqMan and imputed SNPs in estrogen receptor 1 using genome-wide markers. Estrogen metabolites were measured in blood and urine samples. There were 37 patients with POPH and 290 controls. Mean age was 57 years, and 36% were female. The risk allele A in rs7175922 (CYP19A1) was significantly associated with higher levels of estradiol (P = 0.02) and an increased risk of POPH (odds ratio [OR], 2.36; 95% confidence interval [CI], 1.12-4.91; P = 0.02) whereas other SNPs were not. Lower urinary 2-hydroxyestrogen/16-α-hydroxyestrone (OR per 1-ln decrease = 2.04; 95% CI, 1.16-3.57; P = 0.01), lower plasma levels of dehydroepiandrosterone-sulfate (OR per 1-ln decrease = 2.38; 95% CI, 1.56-3.85; P < 0.001), and higher plasma levels of 16-α-hydroxyestradiol (OR per 1-ln increase = 2.16; 95% CI, 1.61-2.98; P < 0.001) were associated with POPH. CONCLUSIONS: Genetic variation in aromatase and changes in estrogen metabolites were associated with POPH.


Subject(s)
Aromatase/genetics , End Stage Liver Disease/complications , Estrogens/metabolism , Hypertension, Portal/genetics , Hypertension, Pulmonary/genetics , Aged , Aromatase/metabolism , Case-Control Studies , Cytochrome P-450 CYP1B1/genetics , Cytochrome P-450 CYP1B1/metabolism , Echocardiography , End Stage Liver Disease/blood , End Stage Liver Disease/genetics , End Stage Liver Disease/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/blood , Estrogens/urine , Female , Humans , Hypertension, Portal/blood , Hypertension, Portal/metabolism , Hypertension, Portal/urine , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/urine , Liver Function Tests , Male , Middle Aged , Polymorphism, Single Nucleotide , Prospective Studies , Signal Transduction/genetics , Vascular Resistance/genetics
5.
Aging (Albany NY) ; 12(12): 12376-12392, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32575070

ABSTRACT

Telomeres are short segments in chromosome ends, the length of which is reduced during cell lifecycles. We examined the association of mean leukocyte telomere length (LTL) and short telomere proportion (STP) with hemodynamic variables in normotensive and never-treated hypertensive volunteers (n=566, 19-72 years). STP and mean LTL were determined using Southern blotting, and supine hemodynamics recorded using continuous tonometric pulse wave analysis and whole-body impedance cardiography. The analyses were adjusted for age, body mass index (BMI), alcohol use, smoking, plasma chemistry, and estimated glomerular filtration rate (eGFR). In univariate analyses, mean LTL and STP both correlated with age, BMI, eGFR, aortic blood pressure, augmentation index, and pulse wave velocity (p<0.05 for all). Mean LTL also correlated with systemic vascular resistance (p<0.05). In linear regression analyses of all hemodynamic variables, mean LTL was only an independent explanatory factor for augmentation index (Beta -0.006, p=0.032), while STP was not an explanatory factor for any of the hemodynamic variables, in contrast to age, BMI and several cardiovascular risk factors. To conclude, augmentation index was predominantly related with chronological aging, but also with mean LTL, suggesting that this variable of central wave reflection is a modest marker of vascular biological aging.


Subject(s)
Aging/genetics , Blood Pressure/genetics , Hypertension/genetics , Telomere Homeostasis/physiology , Vascular Resistance/genetics , Adult , Aged , Cardiography, Impedance , Female , Humans , Hypertension/blood , Leukocytes/metabolism , Male , Middle Aged , Prospective Studies , Pulse Wave Analysis , Young Adult
6.
Eur Heart J ; 41(35): 3314-3322, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32357239

ABSTRACT

AIMS: Haemodynamic determinants of blood pressure (BP) include cardiac output (CO), systemic vascular resistance (SVR), and arterial stiffness. We investigated the heritability of these phenotypes, their association with BP-related single-nucleotide polymorphisms (SNPs), and the causal association between BP and arterial stiffness. METHODS AND RESULTS: We assessed BP, central BP components, and haemodynamic properties (during a single visit) including CO, SVR, and pulse wave velocity (PWV, measure of arterial stiffness) in 3531 (1934 monozygotic, 1586 dizygotic) female TwinsUK participants. Heritability was estimated using structural equation modelling. Association with 984 BP-associated SNP was examined using least absolute shrinkage and selection operator (LASSO) and generalized estimating equation regression. One and two-sample Mendelian randomization (MR) was used to estimate the causal direction between BP and arterial stiffness including data on 436 419 UK Biobank participants. We found high heritability for systolic and pulsatile components of BP (>50%) and PWV (65%) with overlapping genes accounting for >50% of their observed correlation. Environmental factors explained most of the variability of CO and SVR (>80%). Regression identified SNPs (n = 5) known to be associated with BP to also be associated with PWV. One-sample MR showed evidence of bi-directional causal association between BP and PWV in TwinsUK participants. Two-sample MR, confirmed a bi-directional causal effect of PWV on BP (inverse variance weighted (IVW) beta = 0.11, P < 0.02) and BP on arterial stiffness (IVW beta = 0.004, P < 0.0001). CONCLUSION: The genetic basis of BP is mediated not only by genes regulating BP but also by genes that influence arterial stiffness. Mendelian randomization indicates a bi-directional causal association between BP and arterial stiffness.


Subject(s)
Vascular Stiffness , Blood Pressure/genetics , Female , Mendelian Randomization Analysis , Pulse Wave Analysis , Vascular Resistance/genetics , Vascular Stiffness/genetics
7.
Ann Rheum Dis ; 79(3): 370-378, 2020 03.
Article in English | MEDLINE | ID: mdl-31818805

ABSTRACT

BACKGROUND: In this study, we investigated the impact of the new haemodynamic definition of pulmonary arterial hypertension (PAH) as proposed by the 6th PH World Symposium on phenotypes and survival in patients with systemic sclerosis (SSc). METHODS: In SSc patients who were prospectively and consecutively screened for PAH including right heart catheterisation in Heidelberg or Zurich, haemodynamic and clinical variables have been reassessed according to the new PAH definition. Patients have been followed for 3.7±3.7 (median 3.4) years; Kaplan-Meier survival analysis was performed. Patients with significant lung or left heart disease were excluded from comparative analyses. RESULTS: The final dataset included 284 SSc patients, 146 patients (49.2%) had mean pulmonary arterial pressure (mPAP) ≤20 mm Hg, 19.3% had mPAP 21-24 mm Hg and 29.4% had mPAP ≥25 mm Hg. In the group of mildly elevated mPAP, only four patients (1.4% of the whole SSc cohort) had pulmonary vascular resistance (PVR) values ≥3 Wood Units (WU) and could be reclassified as manifest SSc-APAH. Twenty-eight (9.8%) patients with mPAP of 21-24 mm Hg and PVR ≥2 WU already presented with early pulmonary vascular disease with decreased 6 min walking distance (6MWD) (p<0.001), TAPSE (p=0.004) and pulmonary arterial compliance (p<0.001). A PVR ≥2 WU was associated with reduced long-term survival (p=0.002). PVR and 6MWD were independent prognostic predictors in multivariate analysis. CONCLUSION: The data of this study show that a PVR threshold ≥3 WU is too high to enable an early diagnosis of PAH. A PVR threshold ≥2 WU was already associated with pulmonary vascular disease, significantly reduced survival and would be more appropriate in SSc patients with mild PAH.


Subject(s)
Hemodynamics/genetics , Pulmonary Arterial Hypertension/mortality , Scleroderma, Systemic/mortality , Aged , Arterial Pressure/genetics , Female , Humans , Kaplan-Meier Estimate , Lung/physiopathology , Male , Middle Aged , Multivariate Analysis , Phenotype , Prognosis , Prospective Studies , Pulmonary Arterial Hypertension/genetics , Pulmonary Artery/physiopathology , Scleroderma, Systemic/complications , Scleroderma, Systemic/genetics , Vascular Resistance/genetics
8.
J Am Heart Assoc ; 8(9): e010917, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31030617

ABSTRACT

Background Decreased uterine blood flow is known to contribute to pregnancy complications such as gestational hypertension and preeclampsia. Previously, we showed that the loss of regulator of G protein signaling 2 ( RGS 2), a GTP ase activating protein for Gq/11 and Gi/o class G proteins, decreases uterine blood flow in the nonpregnant state in mice. Here, we examined the effects of the absence of RGS 2 and 5 on uterine blood flow and uterine vascular structure and function at early, mid, and late gestation, as well as peripartum period in mice. Methods and Results Abdominal Doppler ultrasonography was performed on adult female wild-type, Rgs2-/-, and Rgs5-/- mice at pre-pregnancy, gestational days 10, 15, and 18, and postpartum day 3. Uterine artery structure and function were also assessed by vessel myograph studies. At mid-pregnancy, uterine blood flow decreased in both Rgs2-/- and Rgs5-/- mice, whereas resistive index increased only in Rgs2-/- mice. In uterine arteries from wild-type mice, mRNA expression of RGS 2 and 4 increased, whereas RGS 5 expression remained elevated at mid-pregnancy. These changes in gene expression were unique to uterine arteries because they were absent in mesenteric arteries and the aorta of wild-type mice. In Rgs2-/- mice, uterine artery medial cross-sectional area and G protein-coupled receptor-mediated vasoconstriction increased in mid-pregnancy, implicating a role for RGS 2 in structural and functional remodeling of uterine arteries during pregnancy. In contrast, RGS 5 absence increased vasoconstriction only in the peripartum period. Conclusions These data together indicate that RGS 2 plays a critical role in the structural and functional remodeling of uterine arteries to impact uterine blood flow during pregnancy. Targeting the signaling pathway regulated by RGS 2 may therefore be a therapeutic strategy for ameliorating utero-placental perfusion disorders during pregnancy.


Subject(s)
Pregnancy, Animal/genetics , RGS Proteins/genetics , Regional Blood Flow/genetics , Uterine Artery/metabolism , Vascular Remodeling/genetics , Animals , Female , Mice , Mice, Knockout , Pregnancy , Pregnancy, Animal/metabolism , Pregnancy, Animal/physiology , RGS Proteins/metabolism , RNA, Messenger/metabolism , Ultrasonography, Doppler , Uterine Artery/diagnostic imaging , Uterine Artery/physiology , Uterine Artery/physiopathology , Vascular Resistance/genetics
9.
Microcirculation ; 25(4): e12452, 2018 05.
Article in English | MEDLINE | ID: mdl-29577514

ABSTRACT

OBJECTIVE: Receptors and ion channels of smooth muscle cells (SMCs) and endothelial cells (ECs) are integral to the regulation of vessel diameter and tissue blood flow. Physiological roles of ion channels and receptors in skeletal muscle and mesenteric arteries have been identified; however, their gene expression profiles are undefined. We tested the hypothesis that expression profiles for ion channels and receptors governing vascular reactivity vary with cell type, vascular bed, and age. METHODS: Mesenteric and superior epigastric arteries were dissected from Old (24-26 months) and Young (3-6 months) C57BL/6J mice. ECs and SMCs were collected for analysis with custom qRT-PCR arrays to determine expression profiles of 80 ion channel and receptor genes. Bioinformatics analyses were applied to gain insight into functional interactions. RESULTS: We identified 68 differences in gene expression with respect to cell type, vessel type, and age. Heat maps illustrate differential expression, and distance matrices predict patterns of coexpression. Gene networks based upon protein-protein interaction datasets and KEGG pathways illustrate biological processes affected by specific differences in gene expression. CONCLUSIONS: Differences in gene expression profiles are most pronounced between microvascular ECs and SMCs with subtle variations between vascular beds and age groups.


Subject(s)
Ion Channels/genetics , Myocytes, Smooth Muscle/metabolism , Transcriptome , Vascular Resistance , Age Factors , Animals , Computational Biology , Endothelial Cells/metabolism , Mesenteric Arteries , Mice , Microvessels/cytology , Microvessels/metabolism , Muscle, Skeletal/blood supply , Vascular Resistance/genetics
10.
J Hypertens ; 36(4): 815-823, 2018 04.
Article in English | MEDLINE | ID: mdl-29303831

ABSTRACT

AIM: Established essential hypertension is associated with increased arterial stiffness and peripheral resistance, but the extent of vascular changes in persons genetically predisposed for essential hypertension is uncertain. METHODS: Participants from the Danish Hypertension Prevention Project (DHyPP) (both parents hypertensive) (n = 95, 41 ±â€Š1 years, 53% men) were compared with available spouses (n = 45, 41 ±â€Š1 years) using measurements of ambulatory blood pressure (BP), left ventricular mass index (LVMI), pulse wave velocity, central BP and augmentation index (AIx) in addition to forearm resting and minimal resistance [forearm resting vascular resistance (Rrest) and forearm minimal vascular resistance (Rmin)]. RESULTS: DHyPP patients with participating spouses had higher 24-h mean BP (94 ±â€Š1 vs. 88 ±â€Š1 mmHg, P < 0.01), LVMI (94 ±â€Š3 vs. 80 ±â€Š2 g/m, P < 0.01), central SBP (121 ±â€Š2 vs. 111 ±â€Š2 mmHg, P < 0.01) and AIx (16.0 ±â€Š1.2 vs. 10.5 ±â€Š1.7%, P < 0.01), but similar carotid-femoral pulse wave velocity (7.5 ±â€Š0.2 vs. 7.1 ±â€Š0.2 m/s), Rrest (53 ±â€Š3 vs. 51 ±â€Š3 mmHg/ml/min/100 ml) and log Rmin (0.58 ±â€Š0.02 vs. 0.55 ±â€Š0.02 mmHg/ml/min/100 ml) when compared with spouses. Using multiple linear regression analysis (adjusting for sex, age, BMI, creatinine clearance and 24-h BP, heart rate and sodium excretion) AIx and LVMI remained elevated in DHyPP patients [4.2% (0.7; 7.7), P = 0.02 and 6.3 g/m (0.7; 11.9), P = 0.03]. For the entire DHyPP cohort AIx, Rrest and Rmin were higher in women than men (P < 0.01), and the same was true for AIx and Rmin among spouses (P < 0.05). Furthermore, AIx was linearly associated with Rrest and Rmin. CONCLUSION: Young to middle-aged individuals genetically predisposed for essential hypertension display increased AIx and LVMI, although vascular stiffness and peripheral resistance are still normal.


Subject(s)
Essential Hypertension/genetics , Genetic Predisposition to Disease , Heart Ventricles/pathology , Vascular Resistance/genetics , Vascular Stiffness/genetics , Adult , Blood Pressure , Blood Pressure Monitoring, Ambulatory , Echocardiography , Female , Forearm/blood supply , Heart Ventricles/diagnostic imaging , Humans , Male , Organ Size , Parents , Pulse Wave Analysis , Sex Factors
11.
Aging Cell ; 16(5): 1104-1113, 2017 10.
Article in English | MEDLINE | ID: mdl-28795531

ABSTRACT

Hypertension is a well-established risk factor for adverse cardiovascular events, and older age is a risk factor for the development of hypertension. Genomewide association studies have linked ATP2B1, the gene for the plasma membrane calcium ATPase 1 (PMCA1), to blood pressure (BP) and hypertension. Here, we present the effects of reduction in the expression of PMCA1 on BP and small artery structure and function when combined with advancing age. Heterozygous PMCA1 null mice (PMCA1Ht ) were generated and conscious BP was measured at 6 to 18 months of age. Passive and active properties of isolated small mesenteric arteries were examined by pressure myography. PMCA1Ht mice exhibited normal BP at 6 and 9 months of age but developed significantly elevated BP when compared to age-matched wild-type controls at ≥12 months of age. Decreased lumen diameter, increased wall thickness and increased wall:lumen ratio were observed in small mesenteric arteries from animals 9 months of age and older, indicative of eutrophic remodelling. Increases in mesenteric artery intrinsic tone and global intracellular calcium were evident in animals at both 6 and 18 months of age. Thus, decreased expression of PMCA1 is associated with increased BP when combined with advancing age. Changes in arterial structure precede the elevation of BP. Pathways involving PMCA1 may be a novel target for BP regulation in the elderly.


Subject(s)
Aging/genetics , Hypertension/genetics , Mesenteric Arteries/metabolism , Plasma Membrane Calcium-Transporting ATPases/genetics , Vascular Remodeling/genetics , Vascular Resistance/genetics , Aging/metabolism , Animals , Blood Pressure/physiology , Calcium/metabolism , Gene Expression , Heterozygote , Hypertension/metabolism , Hypertension/physiopathology , Male , Mesenteric Arteries/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myography , Plasma Membrane Calcium-Transporting ATPases/deficiency
12.
Med. clín (Ed. impr.) ; 148(7): 310-313, abr. 2017. tab, ilus
Article in Spanish | IBECS | ID: ibc-161456

ABSTRACT

Introducción. La hipertensión portopulmonar (HPP) es una enfermedad rara de baja incidencia y sin una alteración genética claramente identificada. El principal objetivo de este estudio fue analizar los genes y modificadores genéticos relacionados con la hipertensión arterial pulmonar en pacientes con HPP. Pacientes. Se seleccionaron 6 pacientes diagnosticados de HPP y se amplificaron las regiones exónicas y sus límites intrónicos de los genes y la región de interés en los modificadores genéticos. Resultados. Se analizaron 6 pacientes diagnosticados de HPP y se compararon con 55 individuos sanos. Se identificaron mutaciones potencialmente patogénicas en 5 pacientes en alguno de los genes analizados. Ninguna de estas mutaciones, que se encuentran altamente conservadas a lo largo de la evolución, fue detectada en los controles analizados ni en las diferentes bases de datos consultadas (1000 Genomas, ExAC y DECIPHER). Tras el análisis de los modificadores genéticos encontramos diferentes variaciones que podrían favorecer el desarrollo de la enfermedad. Conclusiones. El análisis genético en esta pequeña serie de pacientes con HPP ha mostrado un elevado número de mutaciones, siendo el gen ENG el que muestra una mayor frecuencia mutacional (AU)


Introduction. Portopulmonary hypertension (PPH) is a rare disease with a low incidence and without a clearly-identified genetic component. The aim of this work was to check genes and genetic modifiers related to pulmonary arterial hypertension in patients with PPH in order to clarify the molecular basis of the pathology. Patients. e selected a total of 6 patients with PPH and amplified the exonic regions and intronic flanking regions of the relevant genes and regions of interest of the genetic modifiers. Results. Six patients diagnosed with PPH were analyzed and compared to 55 healthy individuals. Potentially-pathogenic mutations were identified in the analyzed genes of 5 patients. None of these mutations, which are highly conserved throughout evolution, were detected in the control patients nor different databases analyzed (1000 Genomes, ExAC and DECIPHER). After analyzing for genetic modifiers, we found different variations that could favor the onset of the disease. Conclusions. The genetic analysis carried out in this small cohort of patients with PPH revealed a large number of mutations, with the ENG gene showing the greatest mutational frequency (AU)


Subject(s)
Humans , Male , Female , Middle Aged , Hypertension, Pulmonary/genetics , Genome/genetics , Mutagenesis/genetics , DNA Mutational Analysis/methods , Polymorphism, Genetic/genetics , Liver Diseases/complications , Liver Cirrhosis, Alcoholic/complications , Vascular Resistance/genetics
13.
J Pathol ; 241(2): 273-280, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27770446

ABSTRACT

Group 1 pulmonary hypertension or pulmonary arterial hypertension (PAH) is a rare disease characterized by proliferation and occlusion of small pulmonary arterioles, leading to progressive elevation of pulmonary artery pressure and pulmonary vascular resistance, and right ventricular failure. Historically, it has been associated with a high mortality rate, although, over the last decade, treatment has improved survival. PAH includes idiopathic PAH (IPAH), heritable PAH (HPAH), and PAH associated with certain medical conditions. The aetiology of PAH is heterogeneous, and genetics play an important role in some cases. Mutations in BMPR2, encoding bone morphogenetic protein receptor 2, a member of the transforming growth factor-ß superfamily of receptors, have been identified in 70% of cases of HPAH, and in 10-40% of cases of IPAH. Other genetic causes of PAH include mutations in the genes encoding activin receptor-like type 1, endoglin, SMAD9, caveolin 1, and potassium two-pore-domain channel subfamily K member 3. Mutations in the gene encoding T-box 4 have been identified in 10-30% of paediatric PAH patients, but rarely in adults with PAH. PAH in children is much more heterogeneous than in adults, and can be associated with several genetic syndromes, congenital heart disease, pulmonary disease, and vascular disease. In addition to rare mutations as a monogenic cause of HPAH, common variants in the gene encoding cerebellin 2 increase the risk of PAH by approximately two-fold. A PAH panel of genes is available for clinical testing, and should be considered for use in clinical management, especially for patients with a family history of PAH. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Genetic Predisposition to Disease , Heart Defects, Congenital/genetics , Hypertension, Pulmonary/genetics , Vascular Resistance/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Humans , Mutation/genetics
14.
Free Radic Res ; 50(8): 920-8, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27297811

ABSTRACT

A loss of arterial elasticity increases the risk for cardiovascular events. Oxidative injury to the vessel wall may be one of the underlying mechanisms influencing arterial elasticity. We compared markers of oxidative stress, antioxidant capacity, inflammation, windkessel compliance (Cwk), and total peripheral resistance (TPR) in black and white South Africans. Associations of arterial compliance and vascular resistance (as indicated by TPR) with oxidative stress, antioxidant capacity and inflammatory markers were also investigated. We included 146 black and 181 white men and women. Measurements from the Finometer device were used to calculate Cwk and TPR while thiobarbituric acids reactive substances (TBARS), glutathione peroxidase (GPx), C-reactive protein (CRP), and interleukin-6 (IL-6) were analyzed in serum or urine samples. Black participants had higher TPR, TBARS, GPx, CRP, and IL-6 levels (all p ≤ 0.018) and lower Cwk (both p ≤ 0.013) compared to white participants. Multiple regression analyses revealed independent associations of Cwk (ß = -0.27, p = 0.015) and TPR (ß = 0.18, p = 0.018) with TBARS in black participants, while Cwk (ß = -0.10; p = 0.019) and TPR (ß = 0.13, p = 0.047) were independently associated with GPx in white participants. Decreased arterial compliance and increased vascular resistance associated with increased oxidative damage independent of hypertensive status in black participants. These results suggest that oxidative stress plays a role in early vascular changes in a black population prone to the development of cardiovascular disease.


Subject(s)
Oxidative Stress/genetics , Thiobarbituric Acid Reactive Substances/metabolism , Vascular Resistance/genetics , Cohort Studies , Cross-Sectional Studies , Ethnicity , Female , Glutathione Peroxidase , Humans , Male , Middle Aged , Prospective Studies
15.
Int J Cardiol ; 211: 132-6, 2016 May 15.
Article in English | MEDLINE | ID: mdl-27002414

ABSTRACT

BACKGROUND: Pulmonary arterial hypertension (PAH) frequently arises in patients with congenital heart disease (CHD) and can lead to pulmonary vascular disease (PVD). The present study was initiated to distinguish the predisposing effect of bone morphogenetic protein receptor 2 (BMPR2) in CHD by comparing the different mutation features of BMPR2 between CHD patients with or without PVD. METHODS AND RESULTS: 294 CHD-PVD and 161 CHD without PVD patients were enrolled. PAH was diagnosed by heart catheterization at rest after CHD was first recognized by echocardiography. PVD was defined as a pulmonary vascular resistance (PVR) more than 3 Wood units. BMPR2 gene was screened by direct sequencing. A total of 24 mutations were identified, accounting for 22 of the 294 patients with CHD-PVD (7.5%) and 2 of the 161 CHD patients without PVD (1.2%, P=0.004). Female/male CHD-PVD patient ratio was 1.6:1, while in the BMPR2 mutation carriers female patients were more dominant (4.5:1, P=0.042). A significant higher BMPR2 mutation rate (12.6%) was found in repaired CHD-PVD (P=0.010). BMPR2 mutations in CHD-PVD patients were identified in different clinical phenotypes. Missense mutation of BMPR2 is the dominant mutation type. CONCLUSION: Genetic predisposing factor may be an important component in the process of development of PVD in CHD patients. Female, repaired patients are more likely to be detected with genetic mutations.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/genetics , Genetic Predisposition to Disease/genetics , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/genetics , Mutation/genetics , Vascular Resistance/genetics , Adolescent , Adult , Female , Genetic Predisposition to Disease/epidemiology , Heart Defects, Congenital/epidemiology , Humans , Male , Middle Aged , Risk Factors , Young Adult
16.
J Pharmacol Exp Ther ; 357(2): 415-22, 2016 May.
Article in English | MEDLINE | ID: mdl-26941168

ABSTRACT

Elevated blood pressure was an unexpected outcome in some cholesteryl ester transfer protein (CETP) inhibitor trials, possibly due to vascular effects of these drugs. We investigated whether CETP inhibitors (torcetrapib, dalcetrapib, anacetrapib) influence vascular function and explored the putative underlying molecular mechanisms. Resistance arteries and vascular smooth muscle cells (VSMC) from rats, which lack the CETP gene, were studied. CETP inhibitors increased phenylephrine-stimulated vascular contraction (logEC50 (:) 6.6 ± 0.1; 6.4 ± 0.06, and 6.2 ± 0.09 for torcetrapib, dalcetrapib, and anacetrapib, respectively, versus control 5.9 ± 0.05). Only torcetrapib reduced endothelium-dependent vasorelaxation. The CETP inhibitor effects were ameliorated by N-acetylcysteine (NAC), a reactive oxygen species (ROS) scavenger, and by S3I-201 [2-hydroxy-4-[[2-(4-methylphenyl)sulfonyloxyacetyl]amino]benzoic acid], a signal transducer and activator of transcription 3 (STAT3) inhibitor. CETP inhibitors increased the phosphorylation (2- to 3-fold) of vascular myosin light chain (MLC) and myosin phosphatase target subunit 1 (MYPT1) (procontractile proteins) and stimulated ROS production. CETP inhibitors increased the phosphorylation of STAT3 (by 3- to 4-fold), a transcription factor important in cell activation. Activation of MLC was reduced by NAC, GKT137831 [2-(2-chlorophenyl)-4-[3-(dimethylamino)phenyl]-5-methyl-1H-pyrazolo[4,3-c]pyridine-3,6-dione] (Nox1/4 inhibitor), and S3I-201. The phosphorylation of STAT3 was unaffected by NAC and GKT137831. CETP inhibitors did not influence activation of mitogen-activated proteins kinases (MAPK) or c-Src. Our data demonstrate that CETP inhibitors influence vascular function and contraction through redox-sensitive, STAT3-dependent, and MAPK-independent processes. These phenomena do not involve CETP because the CETP gene is absent in rodents. Findings from our study indicate that CETP inhibitors have vasoactive properties, which may contribute to the adverse cardiovascular effects of these drugs such as hypertension.


Subject(s)
Cholesterol Ester Transfer Proteins/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , Acetylcysteine/pharmacology , Animals , Cells, Cultured , Free Radical Scavengers/pharmacology , Male , Mitogen-Activated Protein Kinases/drug effects , Myocytes, Smooth Muscle/drug effects , NADPH Oxidases/metabolism , Oxidation-Reduction , Phosphorylation , Protein Phosphatase 1/drug effects , Pyrazoles/pharmacology , Pyrazolones , Pyridines/pharmacology , Pyridones , Rats , Rats, Inbred WKY , Vascular Resistance/drug effects , Vascular Resistance/genetics , Vasoconstriction/drug effects , Vasoconstriction/genetics , Vasodilator Agents/pharmacology
17.
Clin Exp Hypertens ; 38(2): 238-44, 2016.
Article in English | MEDLINE | ID: mdl-26818475

ABSTRACT

BACKGROUND AND OBJECTIVES: High-altitude essential hypertension (HAEH) is a disease occurring in permanent residents of high-altitude regions. The disease is characterized with SBP ≥140 mmHg and DBP ≥90 mmHg. HAEH is known to run in families, i.e. the disease has genetic component. Rho-associated coiled-coil containing protein kinase 2 (ROCK2) is a stress-activated serine-threonine kinase known to disturb vascular-homeostasis leading to an increase in systemic vascular resistance, hallmark of HAEH. ROCK2 is implicated in sea-level essential hypertension but its role in HAEH is yet to be elucidated. METHODS: The present study deals with genotyping 13 polymorphisms of ROCK2 gene in demographicaly matched human cases (n = 65) and controls (n = 38) by Sequenom MS (TOF)-based MassARRAY platform using iPLEX Gold technology. RESULTS: A significant association was observed for GG genotype (SNP, rs978906), AA genotype (SNP, rs6753921), GG genotype (SNP, rs10495582) and AA genotype (SNP, rs2230774) with HAEH (p < 0.05). The 4 SNPs were tagged to each other and formed a 35 kb LD block (r(2 )> 0.90). Haplotype AGCC, composed of wild-type alleles of the SNPs was over represented in controls. In contrast, haplotype GAGA, composed of variant-alleles was observed to be in higher proportion in cases. Moreover, SBP levels (mmHg) were higher in cases with risk genotype against the ones having protective genotype (p = 0.05). Bioinformatic analysis revealed binding of a critical transcription factor, SRF to variant-allele G of SNP rs10495582. SRF has been reported in previous studies to promote ROCK2 transcriptional expression. INTERPRETATION AND CONCLUSIONS: The data clearly suggests association of ROCK2 polymorphisms and haplotypes with HAEH.


Subject(s)
Altitude , Haplotypes/genetics , Hypertension/genetics , Vascular Resistance/genetics , White People/genetics , rho-Associated Kinases/genetics , Adult , Aged , Alleles , Case-Control Studies , Essential Hypertension , Female , Genotype , Humans , India , Male , Middle Aged , Polymorphism, Genetic , Polymorphism, Single Nucleotide , Young Adult
18.
Adv Exp Med Biol ; 888: 237-52, 2015.
Article in English | MEDLINE | ID: mdl-26663186

ABSTRACT

Pulmonary arterial hypertension (PAH) is a lethal vasculopathy associated with complex etiology that involves remodeling of distal pulmonary arteries leading to elevation of pulmonary vascular resistance. This process results in right ventricular (RV) hypertrophy and ultimately RV failure. In addition, PAH is associated with systemic impairment in the skeletal muscle contributing to exercise intolerance. It has only been a few decades since microRNAs (miRNAs) have been implied in the development and progression of PAH regarding every organ affected by the disease. Indeed, impairment of miRNA's expression has been involved in vascular cell remodeling processes such as adventitial fibroblast (AdvFB) migration; pulmonary arterial smooth muscle cell (PASMC) proliferation and pulmonary arterial endothelial cell (PAEC) dysfunction observed in PAH. At the molecular level miRNAs have been described in the control of ion channels and mitochondrial function as well as the regulation of the BMPR2 signaling pathways contributing to PAH lung impairment. Recently miRNAs have also been specifically implicated in RV dysfunction and systemic angiogenic impairment, observed in PAH. In this chapter, we will summarize the knowledge on miRNA in PAH and highlight their crucial role in the etiology of this disease.


Subject(s)
Hypertension, Pulmonary/genetics , MicroRNAs/genetics , Pulmonary Artery/metabolism , Vascular Resistance/genetics , Humans , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/physiopathology , Models, Cardiovascular , Models, Genetic , Pulmonary Artery/physiopathology , Signal Transduction/genetics , Vascular Remodeling/genetics , Vascular Resistance/physiology
19.
Hum Genet ; 134(6): 627-36, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25813623

ABSTRACT

Proprotein convertase subtilisin/kexin (PCSK) enzymes cleave and convert their immature substrates into biologically active forms. Polymorphisms in the PCSK genes have been reported to associate with human diseases and phenotypes, including hypercholesterolemia and blood pressure (BP), and targeting PCSKs is considered a promising future form of drug therapy. PCSK processing is readily induced upon upregulation of the enzyme, but the genetic factors contributing to PCSK expression have not been thoroughly characterized. To gain a comprehensive understanding of the genetic regulation of PCSK expression, we performed, for the first time, a genome-wide expression quantitative trait loci (eQTL) analysis using mRNA expression in >1400 human peripheral blood samples from the Cardiovascular Risk in Young Finns Study and ca. ten million single-nucleotide polymorphisms (SNPs). The expression data showed clear expression for FURIN, PCSK5, PCSK7 and MBTPS1 (membrane-bound transcription factor peptidase, site 1) mRNAs in virtually all tested samples. A discovery analysis demonstrated a genome-wide significant (p < 5 × 10(-8)) association with the selected PCSK probes for 1024 variants, which were located at ten independent loci. Of these loci, 5/10 could be confirmed to regulate PCSK expression in two additional and independent sample sets. Finally, a phenotypic analysis demonstrated that a novel cis-eQTL SNP rs4702 for FURIN is strongly associated with both diastolic (p = 0.012) and systolic (p = 0.035) BP levels, as well as peripheral vascular resistance (p = 0.003). These findings indicate that the expression of the PCSK enzymes is regulated by genetic factors, which have biological roles in health and disease.


Subject(s)
Blood Pressure , Furin , Gene Expression Regulation, Enzymologic , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Female , Furin/biosynthesis , Furin/genetics , Humans , Male , Proprotein Convertases/biosynthesis , Proprotein Convertases/genetics , Serine Endopeptidases/biosynthesis , Serine Endopeptidases/genetics , Subtilisins/biosynthesis , Subtilisins/genetics , Vascular Resistance/genetics
20.
Am J Physiol Lung Cell Mol Physiol ; 308(4): L314-24, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25398989

ABSTRACT

Sickle cell disease (SCD) is an autosomal recessive disorder in the gene encoding the ß-chain of hemoglobin. Deoxygenation causes the mutant hemoglobin S to polymerize, resulting in rigid, adherent red blood cells that are entrapped in the microcirculation and hemolyze. Cardinal features include severe painful crises and episodic acute lung injury, called acute chest syndrome. This population, with age, develops chronic organ injury, such as chronic kidney disease and pulmonary hypertension. A major risk factor for developing chronic organ injury is hemolytic anemia, which releases red blood cell contents into the circulation. Cell free plasma hemoglobin, heme, and arginase 1 disrupt endothelial function, drive oxidative and inflammatory stress, and have recently been referred to as erythrocyte damage-associated molecular pattern molecules (eDAMPs). Studies suggest that in addition to effects of cell free plasma hemoglobin on scavenging nitric oxide (NO) and generating reactive oxygen species (ROS), heme released from plasma hemoglobin can bind to the toll-like receptor 4 to activate the innate immune system. Persistent intravascular hemolysis over decades leads to chronic vasculopathy, with ∼10% of patients developing pulmonary hypertension. Progressive obstruction of small pulmonary arterioles, increase in pulmonary vascular resistance, decreased cardiac output, and eventual right heart failure causes death in many patients with this complication. This review provides an overview of the pathobiology of hemolysis-mediated endothelial dysfunction and eDAMPs and a summary of our present understanding of diagnosis and management of pulmonary hypertension in sickle cell disease, including a review of recent American Thoracic Society (ATS) consensus guidelines for risk stratification and management.


Subject(s)
Acute Chest Syndrome , Hypertension, Pulmonary , Vascular Diseases , Acute Chest Syndrome/blood , Acute Chest Syndrome/genetics , Acute Chest Syndrome/immunology , Acute Chest Syndrome/physiopathology , Acute Chest Syndrome/therapy , Age Factors , Erythrocytes, Abnormal , Heart Failure/blood , Heart Failure/genetics , Heart Failure/immunology , Heart Failure/physiopathology , Heart Failure/therapy , Heme/genetics , Heme/immunology , Heme/metabolism , Hemoglobin, Sickle/genetics , Hemoglobin, Sickle/immunology , Hemoglobin, Sickle/metabolism , Hemolysis/genetics , Hemolysis/immunology , Humans , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/therapy , Immunity, Innate/genetics , Mutation , Nitric Oxide/blood , Nitric Oxide/genetics , Nitric Oxide/immunology , Oxidative Stress/genetics , Oxidative Stress/immunology , Practice Guidelines as Topic , Reactive Oxygen Species/blood , Reactive Oxygen Species/immunology , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/therapy , Toll-Like Receptor 4/blood , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Vascular Diseases/blood , Vascular Diseases/genetics , Vascular Diseases/immunology , Vascular Diseases/physiopathology , Vascular Diseases/therapy , Vascular Resistance/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...