Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 151
Filter
Add more filters

Publication year range
1.
PLoS Pathog ; 19(1): e1011063, 2023 01.
Article in English | MEDLINE | ID: mdl-36634048

ABSTRACT

The Coronavirus Disease 2019 (COVID-19) caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and its sublineages pose a new challenge to healthcare systems worldwide due to its ability to efficiently spread in immunized populations and its resistance to currently available therapies. COVID-19, although targeting primarily the respiratory system, is also now well established that later affects every organ in the body. Most importantly, despite the available therapy and vaccine-elicited protection, the long-term consequences of viral infection in breakthrough and asymptomatic individuals are areas of concern. In the past two years, investigators accumulated evidence on how the virus triggers our immune system and the molecular signals involved in the cross-talk between immune cells and structural cells in the pulmonary vasculature to drive pathological lung complications such as endothelial dysfunction and thrombosis. In the review, we emphasize recent updates on the pathophysiological inflammatory and immune responses associated with SARS-CoV-2 infection and their potential long-term consequences that may consequently lead to the development of pulmonary vascular diseases.


Subject(s)
COVID-19 , Coinfection , Humans , SARS-CoV-2 , Lung , Cross Reactions
2.
Am J Respir Cell Mol Biol ; 71(1): 95-109, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38546978

ABSTRACT

Pulmonary arterial (PA) hypertension (PAH) is a severe cardiopulmonary disease that may be triggered by exposure to drugs such as dasatinib or facilitated by genetic predispositions. The incidence of dasatinib-associated PAH is estimated at 0.45%, suggesting individual predispositions. The mechanisms of dasatinib-associated PAH are still incomplete. We discovered a KCNK3 gene (Potassium channel subfamily K member 3; coding for outward K+ channel) variant in a patient with dasatinib-associated PAH and investigated the impact of this variant on KCNK3 function. Additionally, we assessed the effects of dasatinib exposure on KCNK3 expression. In control human PA smooth muscle cells (hPASMCs) and human pulmonary endothelial cells (hPECs), we evaluated the consequences of KCNK3 knockdown on cell migration, mitochondrial membrane potential, ATP production, and in vitro tube formation. Using mass spectrometry, we determined the KCNK3 interactome. Patch-clamp experiments revealed that the KCNK3 variant represents a loss-of-function variant. Dasatinib contributed to PA constriction by decreasing KCNK3 function and expression. In control hPASMCs, KCNK3 knockdown promotes mitochondrial membrane depolarization and glycolytic shift. Dasatinib exposure or KCNK3 knockdown reduced the number of caveolae in hPECs. Moreover, KCNK3 knockdown in control hPECs reduced migration, proliferation, and in vitro tubulogenesis. Using proximity labeling and mass spectrometry, we identified the KCNK3 interactome, revealing that KCNK3 interacts with various proteins across different cellular compartments. We identified a novel pathogenic variant in KCNK3 and showed that dasatinib downregulates KCNK3, emphasizing the relationship between dasatinib-associated PAH and KCNK3 dysfunction. We demonstrated that a loss of KCNK3-dependent signaling contributes to endothelial dysfunction in PAH and glycolytic switch of hPASMCs.


Subject(s)
Dasatinib , Endothelial Cells , Potassium Channels, Tandem Pore Domain , Dasatinib/pharmacology , Dasatinib/adverse effects , Humans , Potassium Channels, Tandem Pore Domain/metabolism , Potassium Channels, Tandem Pore Domain/genetics , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Endothelial Cells/pathology , Cell Movement/drug effects , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/pathology , Membrane Potential, Mitochondrial/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Male , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Pulmonary Artery/drug effects , Nerve Tissue Proteins
3.
Am J Respir Cell Mol Biol ; 69(2): 147-158, 2023 08.
Article in English | MEDLINE | ID: mdl-36917789

ABSTRACT

Reduced expression and/or activity of Kv1.5 channels (encoded by KCNA5) is a common hallmark in human or experimental pulmonary arterial hypertension (PAH). Likewise, genetic variants in KCNA5 have been found in patients with PAH, but their functional consequences and potential impact on the disease are largely unknown. Herein, this study aimed to characterize the functional consequences of seven KCNA5 variants found in a cohort of patients with PAH. Potassium currents were recorded by patch-clamp technique in HEK293 cells transfected with wild-type or mutant Kv1.5 cDNA. Flow cytometry, Western blot, and confocal microscopy techniques were used for measuring protein expression and cell apoptosis in HEK293 and human pulmonary artery smooth muscle cells. KCNA5 variants (namely, Arg184Pro and Gly384Arg) found in patients with PAH resulted in a clear loss of potassium channel function as assessed by electrophysiological and molecular modeling analyses. The Arg184Pro variant also resulted in a pronounced reduction of Kv1.5 expression. Transfection with Arg184Pro or Gly384Arg variants decreased apoptosis of human pulmonary artery smooth muscle cells compared with the wild-type cells, demonstrating that KCNA5 dysfunction in both variants affects cell viability. Thus, in addition to affecting channel activity, both variants were associated with impaired apoptosis, a crucial process linked to the disease. The estimated prevalence of dysfunctional KCNA5 variants in the PAH population analyzed was around 1%. The data indicate that some KCNA5 variants found in patients with PAH have critical consequences for channel function, supporting the idea that KCNA5 pathogenic variants may be a causative or contributing factor for PAH.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Humans , Pulmonary Arterial Hypertension/metabolism , HEK293 Cells , Hypertension, Pulmonary/metabolism , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Familial Primary Pulmonary Hypertension/metabolism , Pulmonary Artery/pathology
4.
J Physiol ; 601(17): 3717-3737, 2023 09.
Article in English | MEDLINE | ID: mdl-37477289

ABSTRACT

Potassium channel subfamily K member 3 (KCNK3), encoded by the KCNK3 gene, is part of the two-pore domain potassium channel family, constitutively active at resting membrane potentials in excitable cells, including smooth muscle and cardiac cells. Several physiological and pharmacological mediators, such as intracellular signalling pathways, extracellular pH, hypoxia and anaesthetics, regulate KCNK3 channel function. Recent studies show that modulation of KCNK3 channel expression and function strongly influences pulmonary vascular cell and cardiomyocyte function. The altered activity of KCNK3 in pathological situations such as atrial fibrillation, pulmonary arterial hypertension and right ventricular dysfunction demonstrates the crucial role of KCNK3 in cardiovascular homeostasis. Furthermore, loss of function variants of KCNK3 have been identified in patients suffering from pulmonary arterial hypertension and atrial fibrillation. This review focuses on current knowledge of the role of the KCNK3 channel in pulmonary circulation and the heart, in healthy and pathological conditions.


Subject(s)
Atrial Fibrillation , Potassium Channels, Tandem Pore Domain , Pulmonary Arterial Hypertension , Humans , Pulmonary Circulation , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Membrane Potentials , Lung/metabolism , Potassium Channels, Tandem Pore Domain/metabolism
5.
Pharmacol Res ; 197: 106985, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37949331

ABSTRACT

IL-11 is linked to fibrotic diseases, but its role in pulmonary hypertension is unclear. We examined IL-11's involvement in idiopathic pulmonary arterial hypertension (iPAH). Using samples from control (n = 20) and iPAH (n = 6) subjects, we assessed IL-11 and IL-11Rα expression and localization through RT-qPCR, ELISA, immunohistochemistry, and immunofluorescence. A monocrotaline-induced PAH model helped evaluate the impact of siRNA-IL-11 on pulmonary artery remodeling and PH. The effects of recombinant human IL-11 and IL-11Rα on human pulmonary artery smooth muscle cell (HPASMC) proliferation, pulmonary artery endothelial cell (HPAEC) mesenchymal transition, monocyte interactions, endothelial tube formation, and precision cut lung slice (PCLS) pulmonary artery remodeling and contraction were evaluated. IL-11 and IL-11Rα were over-expressed in pulmonary arteries (3.2-fold and 75-fold respectively) and serum (1.5-fold and 2-fold respectively) of patients with iPAH. Therapeutic transient transfection with siRNA targeting IL-11 resulted in a significant reduction in pulmonary artery remodeling (by 98%), right heart hypertrophy (by 66%), and pulmonary hypertension (by 58%) in rats exposed to monocrotaline treatment. rhIL-11 and soluble rhIL-11Rα induce HPASMC proliferation and HPAEC to monocyte interactions, mesenchymal transition, and tube formation. Neutralizing monoclonal IL-11 and IL-11Rα antibodies inhibited TGFß1 and EDN-1 induced HPAEC to mesenchymal transition and HPASMC proliferation. In 3D PCLS, rhIL-11 and soluble rhIL-11Rα do not promote pulmonary artery contraction but sensitize PCLS pulmonary artery contraction induced by EDN-1. In summary, IL-11 and IL-11Rα are more highly expressed in the pulmonary arteries of iPAH patients and contribute to pulmonary artery remodeling and the development of PH.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Humans , Animals , Rats , Familial Primary Pulmonary Hypertension , Interleukin-11 , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Monocrotaline , Pulmonary Artery , RNA, Small Interfering/genetics
6.
Pharmacol Res ; 194: 106834, 2023 08.
Article in English | MEDLINE | ID: mdl-37343646

ABSTRACT

The corpus cavernosum (CC) is a highly vascularized tissue and represents an excellent example of microcirculation. Indeed, erectile dysfunction is considered an early index of cardiovascular disease. Hydrogen sulfide (H2S) at the vascular level is endogenously produced from L-cysteine mainly by the action of cystathionine-γ-lyase (CSE) and plays a role in CC vascular homeostasis. Here we have evaluated the involvement of the endogenous H2S in the regulation of the soluble guanylate cyclase (sCG) redox state. The lack of CSE-derived endogenous H2S, in CSE-/- mice, disrupted the eNOS/NO/sGC/PDE pathway. Indeed, the absence of CSE-derived endogenous H2S caused a significant reduction of the relaxant response to riociguat, an sGC redox-dependent stimulator. Conversely, the response to cinaciguat, an sGC redox-independent activator, was not modified. The relevance of the role played at the redox level of the endogenous H2S was confirmed by the findings that in CC harvested from CSE-/- mice there was a significant reduction of GCß1 expression coupled with a decrease in CYP5R3, a reductase involved in the regulation of the redox state of sGC. These molecular changes driven by the lack of endogenous H2S translate into a significant reduction in cGMP levels. The replenishment of the lack of H2S with an H2S donor rescued the relaxant response to riociguat in CC of CSE-/- mice. In conclusion, the endogenous CSE-derived H2S plays a physiological key role in the regulation of the redox state of sGC in CC microcirculation.


Subject(s)
Hydrogen Sulfide , Microcirculation , Soluble Guanylyl Cyclase , Animals , Male , Mice , Cystathionine gamma-Lyase/metabolism , Hydrogen Sulfide/metabolism , Oxidation-Reduction , Penis/blood supply , Soluble Guanylyl Cyclase/metabolism
7.
Pharmacol Res ; 189: 106684, 2023 03.
Article in English | MEDLINE | ID: mdl-36740150

ABSTRACT

KV1.5 channels are key players in the regulation of vascular tone and atrial excitability and their impairment is associated with cardiovascular diseases including pulmonary arterial hypertension (PAH) and atrial fibrillation (AF). Unfortunately, pharmacological strategies to improve KV1.5 channel function are missing. Herein, we aimed to study whether the chaperone sigma-1 receptor (S1R) is able to regulate these channels and represent a new strategy to enhance their function. By using different electrophysiological and molecular techniques in X. laevis oocytes and HEK293 cells, we demonstrate that S1R physically interacts with KV1.5 channels and regulate their expression and function. S1R induced a bimodal regulation of KV1.5 channel expression/activity, increasing it at low concentrations and decreasing it at high concentrations. Of note, S1R agonists (PRE084 and SKF10047) increased, whereas the S1R antagonist BD1047 decreased, KV1.5 expression and activity. Moreover, PRE084 markedly increased KV1.5 currents in pulmonary artery smooth muscle cells and attenuated vasoconstriction and proliferation in pulmonary arteries. We also show that both KV1.5 channels and S1R, at mRNA and protein levels, are clearly downregulated in samples from PAH and AF patients. Moreover, the expression of both genes showed a positive correlation. Finally, the ability of PRE084 to increase KV1.5 function was preserved under sustained hypoxic conditions, as an in vitro PAH model. Our study provides insight into the key role of S1R in modulating the expression and activity of KV1.5 channels and highlights the potential role of this chaperone as a novel pharmacological target for pathological conditions associated with KV1.5 channel dysfunction.


Subject(s)
Atrial Fibrillation , Receptors, sigma , Humans , HEK293 Cells , Lung/pathology , Pulmonary Artery , Receptors, sigma/metabolism , Sigma-1 Receptor
8.
Int J Mol Sci ; 24(15)2023 Aug 02.
Article in English | MEDLINE | ID: mdl-37569725

ABSTRACT

Recent evidence suggests that vitamin D is involved in the development of pulmonary arterial hypertension (PAH). The aim of this study was to analyze the electrophysiological and contractile properties of pulmonary arteries (PAs) in vitamin D receptor knockout mice (Vdr-/-). PAs were dissected and mounted in a wire myograph. Potassium membrane currents were recorded in freshly isolated PA smooth muscle cells (PASMCs) using the conventional whole-cell configuration of the patch-clamp technique. Potential vitamin D response elements (VDREs) in Kv7 channels coding genes were studied, and their protein expression was analyzed. Vdr-/- mice did not show a pulmonary hypertensive phenotype, as neither right ventricular hypertrophy nor endothelial dysfunction was apparent. However, resistance PA from these mice exhibited increased response to retigabine, a Kv7 activator, compared to controls and heterozygous mice. Furthermore, the current sensitive to XE991, a Kv7 inhibitor, was also higher in PASMCs from knockout mice. A possible VDRE was found in the gene coding for KCNE4, the regulatory subunit of Kv7.4. Accordingly, Vdr-/- mice showed an increased expression of KCNE4 in the lungs, with no changes in Kv7.1 and Kv7.4. These results indicate that the absence of Vdr in mice, as occurred with vitamin D deficient rats, is not sufficient to induce PAH. However, the contribution of Kv7 channel currents to the regulation of PA tone is increased in Vdr-/- mice, resembling animals and humans suffering from PAH.


Subject(s)
Potassium Channels, Voltage-Gated , Pulmonary Artery , Animals , Humans , Mice , Rats , KCNQ Potassium Channels/metabolism , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Potassium Channels, Voltage-Gated/metabolism , Pulmonary Artery/metabolism , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Vitamin D/pharmacology , Vitamin D/metabolism
9.
Clin Exp Hypertens ; 44(4): 355-365, 2022 May 19.
Article in English | MEDLINE | ID: mdl-35311436

ABSTRACT

Chronic treatment with sildenafil (SILD) is an effective protector on the development of cardiovascular complications of pulmonary hypertension (PH) and diabetes. However, to date, no studies have evaluated the effect of SILD on cardiopulmonary pathophysiology during PH secondary to type 1 diabetes. AIM: The present study aimed to evaluate the beneficial effects of chronic SILD treatment on pulmonary arterial pressure, right ventricular hypertrophy (RVH) and cardiac autonomic dysfunction in rats with PH secondary to diabetes. METODOLOGY: Male Sprague Dawley rats were randomly distributed into the control group (saline), diabetic group (60 mg/kg with streptozotocin), SILD-treated control group (20 mg/kg) and SILD-treated diabetic group. RESULTS: After 8 weeks the type 1 diabetic animals presented PH, endothelial dysfunction of the pulmonary arteries, electrocardiographic alterations, RVH and overexpression of phosphodiesterase type 5 in the heart. In type 1 diabetic animals, SILD treatment prevented the development of PH, endothelial dysfunction and RVH. SILD treatment also prevented alterations in the corrected QT period and heart rate variability and prevented overexpression of phosphodiesterase type 5. CONCLUSION: Our results indicate for the first time that SILD treatment prevents pulmonary arterial endothelial dysfunction, pulmonary hypertension, right ventricular hypertrophy and improves heart rate variability in type 1 diabetic rats.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Hypertension, Pulmonary , Rats , Male , Animals , Sildenafil Citrate/pharmacology , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/prevention & control , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/prevention & control , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Heart Rate , Cyclic Nucleotide Phosphodiesterases, Type 5 , Diabetes Mellitus, Type 1/complications , Rats, Sprague-Dawley , Disease Models, Animal
10.
FASEB J ; 34(10): 13626-13640, 2020 10.
Article in English | MEDLINE | ID: mdl-32780919

ABSTRACT

Many probiotics that affect gut microbial ecology have been shown to produce beneficial effects on renin-angiotensin-dependent rodent models and human hypertension. We hypothesized that Bifidobacterium breve CECT7263 (BFM) would attenuate hypertension in deoxycorticosterone acetate (DOCA)-salt rats, a renin-independent model of hypertension. Rats were randomly divided into five groups: control, DOCA-salt, treated DOCA-salt-BFM, treated DOCA-salt-butyrate, and treated DOCA-salt-acetate, for 5 weeks. BFM prevented the increase in systolic blood pressure, cardiac weight, and renal damage induced by DOCA-salt. BFM increased acetate-producing bacterial population and gut acetate levels, improved colonic integrity, normalized endotoxemia, plasma trimethylamine (TMA) levels, and restored the Th17 and Treg content in mesenteric lymph nodes and aorta. Furthermore, BFM improved nitric oxide-dependent vasorelaxation induced by acetylcholine in aortic rings and reduced NADPH oxidase activity in DOCA-salt animals. These protective effects were mimicked by acetate, but not by butyrate supplementation. These data demonstrate that BFM induces changes in gut microbiota linked with attenuation of endothelial dysfunction and increase in blood pressure in this low-renin form of hypertension. These beneficial effects seem to be mediated by increased acetate and reduced TMA production by gut microbiota, thus, improving gut integrity and restoring Th17/Tregs polarization and endotoxemia.


Subject(s)
Bifidobacterium breve , Blood Pressure , Gastrointestinal Microbiome , Hypertension/therapy , Probiotics/therapeutic use , Vasodilation , Animals , Desoxycorticosterone Acetate , Hypertension/chemically induced , Male , Rats , Rats, Wistar
11.
Clin Exp Hypertens ; 43(3): 242-253, 2021 Apr 03.
Article in English | MEDLINE | ID: mdl-33349077

ABSTRACT

Clinical studies suggest that diabetes is a risk factor in the development of pulmonary arterial hypertension. The increase in blood pressure in the pulmonary area is characterized by the increase in the afterload and hypertrophy of the right ventricle. The objective of this study was to conduct a longitudinal follow-up of the morphological and functional changes in the right ventricle in a rat model with pulmonary arterial hypertension secondary to diabetes. Male Sprague Dawley rats were randomly divided into a control group (saline solution) and a diabetic group (60 mg/kg with streptozotocin). For 12 weeks, an echocardiography for longitudinal (in vivo) image analysis of the pulmonary pressure was performed at the same time as the evaluation of myocardial remodeling and right ventricular. After this period, the pulmonary pressure was measured by means of a pulmonary artery catheterization, and the presence of hypertrophy was determined by means of the Fulton index. The plasma concentration of brain natriuretic peptide was measured by means of the ELISA technique. It was found that the diabetic rats showed an increase in pressure in the pulmonary arteries, an increase in the Fulton index, and an increase in brain natriuretic peptide. The echocardiographic follow-up showed that the diabetic rats presented an increase in the pulmonary artery from the fourth week, while hypertrophy and right ventricular systolic dysfunction occurred until the twelfth week. In conclusion, pulmonary arterial hypertension induced by experimental diabetes generated hypertrophy and systolic dysfunction of the right ventricle.


Subject(s)
Echocardiography , Heart Ventricles/diagnostic imaging , Hypertension, Pulmonary/diagnostic imaging , Animals , Blood Glucose/metabolism , Blood Pressure/physiology , Body Weight , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diastole/physiology , Follow-Up Studies , Heart Failure/physiopathology , Heart Rate/physiology , Heart Ventricles/physiopathology , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/physiopathology , Male , Natriuretic Peptide, Brain/metabolism , Rats, Sprague-Dawley , Risk Factors , Streptozocin , Systole/physiology , Vascular Remodeling/physiology
12.
Am J Physiol Lung Cell Mol Physiol ; 319(4): L627-L640, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32726132

ABSTRACT

Vitamin D (VitD) receptor regulates the expression of several genes involved in signaling pathways affected in pulmonary hypertension (PH). VitD deficiency is highly prevalent in PH, and low levels are associated with poor prognosis. We investigated if VitD deficiency may predispose to or exacerbate PH. Male Wistar rats were fed with a standard or a VitD-free diet for 5 wk. Next, rats were further divided into controls or PH, which was induced by a single dose of Su-5416 (20 mg/kg) and exposure to hypoxia (10% O2) for 2 wk. VitD deficiency had no effect on pulmonary pressure in normoxic rats, indicating that, by itself, it does not trigger PH. However, it induced several moderate but significant changes characteristic of PH in the pulmonary arteries, such as increased muscularization, endothelial dysfunction, increased survivin, and reduced bone morphogenetic protein (Bmp) 4, Bmp6, DNA damage-inducible transcript 4, and K+ two-pore domain channel subfamily K member 3 (Kcnk3) expression. Myocytes isolated from pulmonary arteries from VitD-deficient rats had a reduced whole voltage-dependent potassium current density and acid-sensitive (TASK-like) potassium currents. In rats with PH induced by Su-5416 plus hypoxia, VitD-free diet induced a modest increase in pulmonary pressure, worsened endothelial function, increased the hyperreactivity to serotonin, arterial muscularization, decreased total and TASK-1 potassium currents, and further depolarized the pulmonary artery smooth muscle cell membrane. In human pulmonary artery smooth muscle cells from controls and patients with PH, the active form of VitD calcitriol significantly increased KCNK3 mRNA expression. Altogether, these data strongly suggest that the deficit in VitD induces pulmonary vascular dysfunction.


Subject(s)
Hypertension, Pulmonary/metabolism , Muscle, Smooth, Vascular/metabolism , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Vitamin D Deficiency/metabolism , Animals , Humans , Lung/metabolism , Lung/physiopathology , Male , Membrane Potentials/physiology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism , Rats, Wistar , Vitamin D/metabolism
13.
J Physiol ; 597(4): 1185-1197, 2019 02.
Article in English | MEDLINE | ID: mdl-29717493

ABSTRACT

KEY POINTS: The expression of miR-1 is increased in lungs from the Hyp/Su5416 PAH rat model. Pulmonary artery smooth muscle cells from this animal model are more depolarized and show decreased expression and activity of voltage-dependent potassium channel (Kv)1.5. miR-1 directly targets Kv1.5 channels, reduces Kv1.5 activity and induces membrane depolarization. Antagomir-1 prevents Kv1.5 channel downregulation and the depolarization induced by hypoxia/Su5416 exposition. ABSTRACT: Impairment of the voltage-dependent potassium channel (Kv) plays a central role in the development of cardiovascular diseases, including pulmonary arterial hypertension (PAH). MicroRNAs are non-coding RNAs that regulate gene expression by binding to the 3'-untranslated region region of specific mRNAs. The present study aimed to analyse the effects of miR-1 on Kv channel function in pulmonary arteries (PA). Kv channel activity was studied in PA from healthy animals transfected with miR-1 or scrambled-miR. Kv currents were studied using the whole-cell configuration of the patch clamp technique. The characterization of the Kv1.5 currents was performed with the selective inhibitor DPO-1. miR-1 expression was increased and Kv1.5 channels were decreased in lungs from a rat model of PAH induced by hypoxia and Su5416. miR-1 transfection increased cell capacitance, reduced Kv1.5 currents and induced membrane depolarization in isolated pulmonary artery smooth muscle cells. A luciferase reporter assay indicated that KCNA5, which encodes Kv1.5 channels, is a direct target gene of miR-1. Incubation of PA with Su5416 and hypoxia (3% O2 ) increased miR-1 and induced a decline in Kv1.5 currents, which was prevented by antagomiR-1. In conclusion, these data indicate that miR-1 induces pulmonary artery smooth muscle cell hypertrophy and reduces the activity and expression of Kv channels, suggesting a pathophysiological role in PAH.


Subject(s)
Hypertension, Pulmonary/metabolism , Kv1.5 Potassium Channel/metabolism , MicroRNAs/metabolism , Pulmonary Artery/metabolism , Action Potentials , Animals , COS Cells , Cell Hypoxia , Chlorocebus aethiops , Down-Regulation , Hypertension, Pulmonary/etiology , Indoles/toxicity , Kv1.5 Potassium Channel/genetics , Male , MicroRNAs/genetics , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Potassium Channel Blockers/pharmacology , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Pyrroles/toxicity , Rats , Rats, Wistar
14.
Int J Mol Sci ; 20(2)2019 Jan 18.
Article in English | MEDLINE | ID: mdl-30669371

ABSTRACT

In addition to playing a role as a structural component of cellular membranes, ceramide is now clearly recognized as a bioactive lipid implicated in a variety of physiological functions. This review aims to provide updated information on the role of ceramide in the regulation of vascular tone. Ceramide may induce vasodilator or vasoconstrictor effects by interacting with several signaling pathways in endothelial and smooth muscle cells. There is a clear, albeit complex, interaction between ceramide and redox signaling. In fact, reactive oxygen species (ROS) activate different ceramide generating pathways and, conversely, ceramide is known to increase ROS production. In recent years, ceramide has emerged as a novel key player in oxygen sensing in vascular cells and mediating vascular responses of crucial physiological relevance such as hypoxic pulmonary vasoconstriction (HPV) or normoxic ductus arteriosus constriction. Likewise, a growing body of evidence over the last years suggests that exaggerated production of vascular ceramide may have detrimental effects in a number of pathological processes including cardiovascular and lung diseases.


Subject(s)
Blood Vessels/drug effects , Blood Vessels/physiology , Ceramides/pharmacology , Animals , Biomarkers , Ceramides/blood , Ceramides/metabolism , Disease Susceptibility , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Energy Metabolism/drug effects , Humans , Mechanotransduction, Cellular/drug effects , Metabolic Networks and Pathways/drug effects , Oxidation-Reduction , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Vasomotor System/drug effects
15.
Am J Physiol Lung Cell Mol Physiol ; 315(5): L711-L723, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30136611

ABSTRACT

Human immunodeficiency virus (HIV) infection is an established risk factor for pulmonary arterial hypertension (PAH); however, the pathogenesis of HIV-related PAH remains unclear. Since K+ channel dysfunction is a common marker in most forms of PAH, our aim was to analyze whether the expression of HIV proteins is associated with impairment of K+ channel function in the pulmonary vascular bed. HIV transgenic mice (Tg26) expressing seven of the nine HIV viral proteins and wild-type (WT) mice were used. Hemodynamic assessment was performed by echocardiography and catheterization. Vascular reactivity was studied in endothelium-intact pulmonary arteries. K+ currents were recorded in freshly isolated pulmonary artery smooth muscle cells (PASMC) using the patch-clamp technique. Gene expression was assessed using quantitative RT-PCR. PASMC from Tg26 mice had reduced K+ currents and were more depolarized than those from WT. Whereas voltage-gated K+ channel 1.5 (Kv1.5) currents were preserved, pH-sensitive noninactivating background currents ( IKN) were nearly abolished in PASMC from Tg26 mice. Tg26 mice had reduced lung expression of Kv7.1 and Kv7.4 channels and decreased responses to the Kv7.1 channel activator L-364,373 assessed by vascular reactivity and patch-clamp experimental approaches. Although we found pulmonary vascular remodeling and endothelial dysfunction in Tg26 mice, this was not accompanied by changes in hemodynamic parameters. In conclusion, the expression of HIV proteins in vivo impairs pH-sensitive IKN and Kv7 currents. This negative impact of HIV proteins in K+ channels was not sufficient to induce PAH, at least in mice, but may play a permissive or accessory role in the pathophysiology of HIV-associated PAH.


Subject(s)
HIV-1/genetics , Human Immunodeficiency Virus Proteins/metabolism , Hypertrophy, Right Ventricular/pathology , Muscle, Smooth, Vascular/pathology , Potassium Channels, Voltage-Gated/metabolism , Pulmonary Artery/pathology , Transgenes/physiology , Animals , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/virology , Human Immunodeficiency Virus Proteins/genetics , Humans , Hypertrophy, Right Ventricular/metabolism , Male , Membrane Potentials , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/metabolism , Potassium Channels, Voltage-Gated/genetics , Pulmonary Artery/metabolism , Vasoconstriction
16.
Thorax ; 73(6): 519-529, 2018 06.
Article in English | MEDLINE | ID: mdl-29440315

ABSTRACT

BACKGROUND: Pulmonary hypertension (PH) is a common disorder in patients with idiopathic pulmonary fibrosis (IPF) and portends a poor prognosis. Recent studies using vasodilators approved for PH have failed in improving IPF mainly due to ventilation (V)/perfusion (Q) mismatching and oxygen desaturation. Janus kinase type 2 (JAK2) is a non-receptor tyrosine kinase activated by a broad spectrum of profibrotic and vasoactive mediators, but its role in PH associated to PH is unknown. OBJECTIVE: The study of JAK2 as potential target to treat PH in IPF. METHODS AND RESULTS: JAK2 expression was increased in pulmonary arteries (PAs) from IPF (n=10; 1.93-fold; P=0.0011) and IPF+PH (n=9; 2.65-fold; P<0.0001) compared with PA from control subjects (n=10). PA remodelling was evaluated in human pulmonary artery endothelial cells (HPAECs) and human pulmonary artery smooth muscle cells (HPASMCs) from patients with IPF in vitro treated with the JAK2 inhibitor JSI-124 or siRNA-JAK2 and stimulated with transforming growth factor beta. Both JSI-124 and siRNA-JAK2 inhibited the HPAEC to mesenchymal transition and the HPASMCs to myofibroblast transition and proliferation. JAK2 inhibition induced small PA relaxation in precision-cut lung slice experiments. PA relaxation was dependent of the large conductance calcium-activated potassium channel (BKCa). JAK2 inhibition activated BKCa channels and reduced intracellular Ca2+. JSI-124 1 mg/kg/day, reduced bleomycin-induced lung fibrosis, PA remodelling, right ventricular hypertrophy, PA hypertension and V/Q mismatching in rats. The animal studies followed the ARRIVE guidelines. CONCLUSIONS: JAK2 participates in PA remodelling and tension and may be an attractive target to treat IPF associated to PH.


Subject(s)
Hypertension, Pulmonary/drug therapy , Idiopathic Pulmonary Fibrosis/drug therapy , Janus Kinase 2/antagonists & inhibitors , Triterpenes/pharmacology , Vascular Remodeling/drug effects , Animals , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Endothelial Cells , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Janus Kinase 2/metabolism , Myocytes, Smooth Muscle , Phenotype , RNA, Small Interfering/pharmacology , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Signal Transduction , Transforming Growth Factor beta/pharmacology
17.
Arch Biochem Biophys ; 646: 107-112, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29580946

ABSTRACT

Herein we describe, based on some bibliometric data, how the field of research on flavonoids has evolved over the last 25 years. The number of papers on flavonoids has risen in an exponential manner over these years, much faster than other fields on food constituents. This increase appears to be related to the contemporary explosion of interest in healthy foods, supplements and nutraceuticals. It was also probably triggered by large epidemiological studies on fruits and vegetables, and particularly on flavonoids, consumption and incidence of cancer, stroke and coronary heart disease. The widely distributed flavonols constitute the flavonoid subgroup upon which the greatest interest has been focused, followed by flavanols and more recently by anthocyanidins and other related polyphenols such as resveratrol. Research on isoflavones rapidly emerged in the 1990s but plateaued in the 2000s. In the 1990s flavonoids were mainly considered as the active components of medicinal plants, while from 2000 onward, they switched to be mainly regarded as bioactive food ingredients. We envision a continuation in the growth of research for the coming decade focused on clearly demonstrating the importance of flavonoids for human health.


Subject(s)
Flavonoids , Health/trends , Pharmaceutical Research/trends , Animals , Beverages , Dietetics/trends , Epidemiology/history , Flavonoids/chemistry , Flavonoids/history , Food , History, 19th Century , History, 20th Century , History, 21st Century , Humans , Pharmaceutical Research/history
19.
Thorax ; 72(5): 460-471, 2017 May.
Article in English | MEDLINE | ID: mdl-27701117

ABSTRACT

BACKGROUND: Pulmonary hypertension (PH) is frequently observed in patients with acute respiratory distress syndrome (ARDS) and it is associated with an increased risk of mortality. Both acid sphingomyelinase (aSMase) activity and interleukin 6 (IL-6) levels are increased in patients with sepsis and correlate with worst outcomes, but their role in pulmonary vascular dysfunction pathogenesis has not yet been elucidated. Therefore, the aim of this study was to determine the potential contribution of aSMase and IL-6 in the pulmonary vascular dysfunction induced by lipopolysaccharide (LPS). METHODS: Rat or human pulmonary arteries (PAs) or their cultured smooth muscle cells (SMCs) were exposed to LPS, SMase or IL-6 in the absence or presence of a range of pharmacological inhibitors. The effects of aSMase inhibition in vivo with D609 on pulmonary arterial pressure and inflammation were assessed following intratracheal administration of LPS. RESULTS: LPS increased ceramide and IL-6 production in rat pulmonary artery smooth muscle cells (PASMCs) and inhibited pulmonary vasoconstriction induced by phenylephrine or hypoxia (HPV), induced endothelial dysfunction and potentiated the contractile responses to serotonin. Exogenous SMase and IL-6 mimicked the effects of LPS on endothelial dysfunction, HPV failure and hyperresponsiveness to serotonin in PA; whereas blockade of aSMase or IL-6 prevented LPS-induced effects. Finally, administration of the aSMase inhibitor D609 limited the development of endotoxin-induced PH and ventilation-perfusion mismatch. The protective effects of D609 were validated in isolated human PAs. CONCLUSIONS: Our data indicate that aSMase and IL-6 are not simply biomarkers of poor outcomes but pathogenic mediators of pulmonary vascular dysfunction in ARDS secondary to Gram-negative infections.


Subject(s)
Hypertension, Pulmonary/metabolism , Interleukin-6/metabolism , Interleukin-6/pharmacology , Pulmonary Artery/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelin Phosphodiesterase/pharmacology , Animals , Bridged-Ring Compounds/pharmacology , Cells, Cultured , Ceramidases/metabolism , Disease Models, Animal , Fluorescent Antibody Technique , Humans , Interleukin-1beta/metabolism , Lipopolysaccharides , Male , Muscle, Smooth, Vascular/cytology , Norbornanes , Rats , Rats, Wistar , Thiocarbamates , Thiones/pharmacology , Vasoconstriction/drug effects
20.
Am J Physiol Heart Circ Physiol ; 312(2): H189-H200, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27881385

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily of ligand-activated transcription factors, which is composed of three members encoded by distinct genes: PPARα, PPARß/δ, and PPARγ. The biological actions of PPARα and PPARγ and their potential as a cardiovascular therapeutic target have been extensively reviewed, whereas the biological actions of PPARß/δ and its effectiveness as a therapeutic target in the treatment of hypertension remain less investigated. Preclinical studies suggest that pharmacological PPARß/δ activation induces antihypertensive effects in direct [spontaneously hypertensive rat (SHR), ANG II, and DOCA-salt] and indirect (dyslipemic and gestational) models of hypertension, associated with end-organ damage protection. This review summarizes mechanistic insights into the antihypertensive effects of PPARß/δ activators, including molecular and functional mechanisms. Pharmacological PPARß/δ activation induces genomic actions including the increase of regulators of G protein-coupled signaling (RGS), acute nongenomic vasodilator effects, as well as the ability to improve the endothelial dysfunction, reduce vascular inflammation, vasoconstrictor responses, and sympathetic outflow from central nervous system. Evidence from clinical trials is also examined. These preclinical and clinical outcomes of PPARß/δ ligands may provide a basis for the development of therapies in combating hypertension.


Subject(s)
Hypertension/physiopathology , PPAR delta/physiology , PPAR-beta/physiology , Vasodilation/physiology , Animals , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Blood Pressure/drug effects , Blood Pressure/physiology , Endothelium, Vascular/physiopathology , Fatty Acids/metabolism , Gene Expression Regulation , Humans , Hypertension/drug therapy , Inflammation , PPAR delta/agonists , PPAR delta/metabolism , PPAR-beta/agonists , PPAR-beta/metabolism , Phenoxyacetates/pharmacology , Phenoxyacetates/therapeutic use , RGS Proteins/drug effects , RGS Proteins/genetics , Rats , Rats, Inbred SHR , Sympathetic Nervous System/physiopathology , Thiazoles/pharmacology , Thiazoles/therapeutic use , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasodilation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL