Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177
Filter
1.
Biochim Biophys Acta Mol Cell Res ; 1871(4): 119704, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38462075

ABSTRACT

Pulmonary arterial hypertension (PAH) is characterized by increased pulmonary vascular resistance (PVR), right ventricular (RV) failure and premature death. Compounds with vasodilatory characteristics, such as ß-caryophyllene, could be promising therapeutics for PAH. This study aimed to determine the effects of free and nanoemulsified ß-caryophyllene in lung oxidative stress and heart function in PAH rats. Male Wistar rats (170 g, n = 6/group) were divided into four groups: control (CO), monocrotaline (MCT), monocrotaline + ß-caryophyllene (MCT-Bcar) and monocrotaline + nanoemulsion with ß-caryophyllene (MCT-Nano). PAH was induced by MCT (60 mg/kg i.p.), and 7 days later, treatment with ß-caryophyllene, either free or in a nanoemulsion (by gavage, 176 mg/kg/day) or vehicle was given for 14 days. Echocardiographic and hemodynamic measurements were performed, and after, the RV was collected for morphometry and the lungs for evaluation of oxidative stress, antioxidant enzymes, total sulfhydryl compounds, nitric oxide synthase (NOS) activity and endothelin-1 receptor expression. RV hypertrophy, increased PVR and RV systolic and diastolic pressures (RVSP and RVEDP, respectively) and increased mean pulmonary arterial pressure (mPAP) were observed in the MCT group. Treatment with both free and nanoemulsified ß-caryophyllene reduced RV hypertrophy, mPAP, RVSP and lipid peroxidation. The reduction in RVSP was more pronounced in the MCT-Nano group. Moreover, RVEDP decreased only in the MCT-Nano group. These treatments also increased superoxide dismutase, catalase and NOS activities and decreased endothelin-1 receptors expression. Both ß-caryophyllene formulations improved mPAP, PVR and oxidative stress parameters. However, ß-caryophyllene in a nanoemulsion was more effective in attenuating the effects of PAH.


Subject(s)
Hypertension, Pulmonary , Polycyclic Sesquiterpenes , Pulmonary Arterial Hypertension , Rats , Male , Animals , Pulmonary Arterial Hypertension/metabolism , Monocrotaline/toxicity , Monocrotaline/metabolism , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Rats, Wistar , Pulmonary Artery/metabolism , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/metabolism
2.
Can J Physiol Pharmacol ; 101(9): 447-454, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37581356

ABSTRACT

Oxidative stress is involved in increased pulmonary vascular resistance (PVR) and right ventricular (RV) hypertrophy, characteristics of pulmonary arterial hypertension (PAH). Copaiba oil, an antioxidant compound, could attenuate PAH damage. This study's aim was to determine the effects of copaiba oil on lung oxidative stress, PVR, and mean pulmonary arterial pressure (mPAP) in the monocrotaline (MCT) model of PAH. Male Wistar rats (170 g, n = 7/group) were divided into four groups: control, MCT, copaiba oil, and MCT + copaiba oil (MCT-O). PAH was induced by MCT (60 mg/kg i.p.) and, after 1 week, the treatment with copaiba oil (400 mg/kg/day gavage) was started for 14 days. Echocardiographic and hemodynamic measurements were performed. RV was collected for morphometric evaluations and lungs and the pulmonary artery were used for biochemical analysis. Copaiba oil significantly reduced RV hypertrophy, PVR, mPAP, and antioxidant enzyme activities in the MCT-O group. Moreover, increased nitric oxide synthase and decreased NADPH oxidase activities were observed in the MCT-O group. In conclusion, copaiba oil was able to improve the balance between nitric oxide and reactive oxygen species in lungs and the pulmonary artery and to reduce PVR, which could explain a decrease in RV hypertrophy in this PAH model.


Subject(s)
Hypertension, Pulmonary , Oils, Volatile , Pulmonary Arterial Hypertension , Rats , Male , Animals , Rats, Wistar , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Monocrotaline/adverse effects , Nitric Oxide , Antioxidants/pharmacology , Biological Availability , Lung , Pulmonary Artery , Familial Primary Pulmonary Hypertension , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/drug therapy , Oils, Volatile/pharmacology , Disease Models, Animal
3.
Naunyn Schmiedebergs Arch Pharmacol ; 396(12): 3775-3788, 2023 12.
Article in English | MEDLINE | ID: mdl-37338577

ABSTRACT

The TASK-1 channel belongs to the two-pore domain potassium channel family. It is expressed in several cells of the heart, including the right atrial (RA) cardiomyocytes and the sinus node, and TASK-1 channel has been implicated in the pathogenesis of atrial arrhythmias (AA). Thus, using the rat model of monocrotaline-induced pulmonary hypertension (MCT-PH), we explored the involvement of TASK-1 in AA. Four-week-old male Wistar rats were injected with 50 mg/kg of MCT to induce MCT-PH and isolated RA function was studied 14 days later. Additionally, isolated RA from six-week-old male Wistar rats were used to explore the ability of ML365, a selective blocker of TASK-1, to modulate RA function. The hearts developed right atrial and ventricular hypertrophy, inflammatory infiltrate and the surface ECG demonstrated increased P wave duration and QT interval, which are markers of MCT-PH. The isolated RA from the MCT animals showed enhanced chronotropism, faster contraction and relaxation kinetics, and a higher sensibility to extracellular acidification. However, the addition of ML365 to extracellular media was not able to restore the phenotype. Using a burst pacing protocol, the RA from MCT animals were more susceptible to develop AA, and simultaneous administration of carbachol and ML365 enhanced AA, suggesting the involvement of TASK-1 in AA induced by MCT. TASK-1 does not play a key role in the chronotropism and inotropism of healthy and diseased RA; however, it may play a role in AA in the MCT-PH model.


Subject(s)
Atrial Fibrillation , Hypertension, Pulmonary , Animals , Male , Rats , Heart Atria/pathology , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/pathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/pathology , Models, Theoretical , Monocrotaline/adverse effects , Rats, Wistar
4.
J Muscle Res Cell Motil ; 44(4): 299-309, 2023 12.
Article in English | MEDLINE | ID: mdl-37249732

ABSTRACT

Pulmonary arterial hypertension (PAH) leads to changes in the pump function of the heart and causes right-sided myocardial hypertrophy and heart failure. This study was the first to compare the contractile characteristics of the multicellular myocardial preparations of the right atrium (RA) and right ventricle (RV) of male rats from the control group (CON) and the group with monocrotaline (MCT)-induced hypertrophy at the molecular and multicellular levels. In both RA and RV in MCT-treated rats, the fraction of motile filaments and the maximum sliding velocity of actin and reconstituted thin filaments over myosin decreased, and the ratio of α-/ß-myosin heavy chains (MHC) shifted towards ß-MHC. In the RA strips and RV trabeculae, the maximum shortening velocity, the extent of muscle shortening, the amplitude of isometric stress, the amount of work decreased. PAH leads to a greater drop in right atrial contractility than that of the ventricle.


Subject(s)
Atrial Fibrillation , Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Rats , Male , Animals , Pulmonary Arterial Hypertension/complications , Heart Ventricles , Monocrotaline/toxicity , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/complications , Atrial Fibrillation/complications , Hypertrophy, Right Ventricular/chemically induced , Heart Atria , Disease Models, Animal
5.
Lung ; 200(5): 619-631, 2022 10.
Article in English | MEDLINE | ID: mdl-36107242

ABSTRACT

PURPOSE: It has been shown that activation of autophagy promotes the development of pulmonary arterial hypertension (PAH). Meanwhile, forkhead box M1 (FOXM1) has been found to induce autophagy in several types of cancer. However, it is still unclear whether FOXM1 mediates autophagy activation in PAH, and detailed mechanisms responsible for these processes are indefinite. METHOD: PAH was induced by a single intraperitoneal injection of monocrotaline (MCT) to rats. The right ventricle systolic pressure (RVSP), right ventricular hypertrophy index (RVHI), percentage of medial wall thickness (%MT), α-smooth muscle actin (α-SMA) staining, and Ki67 staining were performed to evaluate the development of PAH. The protein levels of FOXM1, phospho-focal adhesion kinase (p-FAK), FAK, and LC3B were determined by immunoblotting or immunohistochemistry. RESULTS: FOXM1 protein level and FAK activity were significantly increased in MCT-induced PAH rats, this was accompanied with the activation of autophagy. Pharmacological inhibition of FOXM1 or FAK suppressed MCT-induced autophagy activation, decreased RVSP, RVHI and %MT in MCT-induced PAH rats, and inhibited the proliferation of pulmonary arterial smooth muscle cells and pulmonary vessel muscularization in MCT-induced PAH rats. CONCLUSION: FOXM1 promotes the development of PAH by inducing FAK phosphorylation and subsequent activation of autophagy in MCT-treated rats.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Actins/metabolism , Animals , Autophagy , Disease Models, Animal , Familial Primary Pulmonary Hypertension , Forkhead Box Protein M1/metabolism , Forkhead Box Protein M1/therapeutic use , Hypertension, Pulmonary/drug therapy , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/metabolism , Ki-67 Antigen/metabolism , Monocrotaline/metabolism , Monocrotaline/toxicity , Phosphorylation , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Artery , Rats , Rats, Sprague-Dawley
6.
Clin Sci (Lond) ; 135(21): 2467-2481, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34676402

ABSTRACT

Pulmonary hypertension (PH) is a life-threatening disease characterized by vascular remodeling. Exploring new therapy target is urgent. The purpose of the present study is to investigate whether and how spliced x-box binding protein 1 (xbp1s), a key component of endoplasmic reticulum stress (ERS), contributes to the pathogenesis of PH. Forty male SD rats were randomly assigned to four groups: Control, Monocrotaline (MCT), MCT+AAV-CTL (control), and MCT+AAV-xbp1s. The xbp1s protein levels were found to be elevated in lung tissues of the MCT group. Intratracheal injection of adeno-associated virus serotype 1 carrying xbp1s shRNA (AAV-xbp1s) to knock down the expression of xbp1s effectively ameliorated the MCT-induced elevation of right ventricular systolic pressure (RVSP), total pulmonary resistance (TPR), right ventricular hypertrophy and medial wall thickness of muscularized distal pulmonary arterioles. The abnormally increased positive staining rates of proliferating cell nuclear antigen (PCNA) and Ki67 and decreased positive staining rates of terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) in pulmonary arterioles were also reversed in the MCT+AAV-xbp1s group. For mechanistic exploration, bioinformatics prediction of the protein network was performed on the STRING database, and further verification was performed by qRT-PCR, Western blots and co-immunoprecipitation (Co-IP). DNA damage-inducible transcript 3 (Ddit3) was identified as a downstream protein that interacted with xbp1s. Overexpression of Ddit3 restored the decreased proliferation, migration and cell viability caused by silencing of xbp1s. The protein level of Ddit3 was also highly consistent with xbp1s in the animal model. Taken together, our study demonstrated that xbp1s-Ddit3 may be a potential target to interfere with vascular remodeling in PH.


Subject(s)
Arterial Pressure , Hypertension, Pulmonary/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Transcription Factor CHOP/metabolism , Vascular Remodeling , X-Box Binding Protein 1/metabolism , Animals , Apoptosis , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Male , Monocrotaline , Muscle, Smooth, Vascular/physiopathology , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Rats, Sprague-Dawley , Signal Transduction , Transcription Factor CHOP/genetics , Ventricular Dysfunction, Right/chemically induced , Ventricular Dysfunction, Right/metabolism , Ventricular Dysfunction, Right/physiopathology , Ventricular Function, Right , X-Box Binding Protein 1/genetics
7.
Sci Rep ; 11(1): 18002, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34504231

ABSTRACT

Pulmonary hypertension (PH) initially results in compensatory right ventricular (RV) hypertrophy, but eventually in RV failure. This transition is poorly understood, but may be triggered by hypoxia. Measurements of RV oxygen tension (pO2) in PH are lacking. We hypothesized that RV hypoxia occurs in monocrotaline-induced PH in rats and that myo-inositol trispyrophosphate (ITPP), facilitating oxygen dissociation from hemoglobin, can relieve it. Rats received monocrotaline (PH) or saline (control) and 24 days later echocardiograms, pressure-volume loops were obtained and myocardial pO2 was measured using a fluorescent probe. In PH mean pulmonary artery pressure more than doubled (35 ± 5 vs. 15 ± 2 in control), RV was hypertrophied, though its contractility was augmented. RV and LV pO2 was 32 ± 5 and 15 ± 8 mmHg, respectively, in control rats. In PH RV pO2 was reduced to 18 ± 9 mmHg, while LV pO2 was unchanged. RV pO2 correlated with RV diastolic wall stress (negatively) and LV systolic pressure (positively). Acute ITPP administration did not affect RV or LV pO2 in control animals, but increased RV pO2 to 26 ± 5 mmHg without affecting LV pO2 in PH. RV oxygen balance is impaired in PH and as such can be an important target for PH therapy. ITPP may be one of such potential therapies.


Subject(s)
Cardiotonic Agents/pharmacology , Hypertension, Pulmonary/drug therapy , Hypertrophy, Right Ventricular/drug therapy , Hypoxia/drug therapy , Inositol Phosphates/pharmacology , Ventricular Dysfunction, Right/drug therapy , Animals , Cardiotonic Agents/administration & dosage , Disease Models, Animal , Hemoglobins/metabolism , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/chemically induced , Hypoxia/metabolism , Hypoxia/physiopathology , Male , Monocrotaline/administration & dosage , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Rats , Rats, Wistar , Treatment Outcome , Ventricular Dysfunction, Right/chemically induced , Ventricular Dysfunction, Right/metabolism , Ventricular Dysfunction, Right/physiopathology , Ventricular Function, Right/physiology
8.
Am J Physiol Heart Circ Physiol ; 321(5): H940-H947, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34559582

ABSTRACT

Right-sided heart failure is a common consequence of pulmonary arterial hypertension. Overloading the right ventricle results in right ventricular hypertrophy, which progresses to failure in a process characterized by impaired Ca2+ dynamics and force production that is linked with transverse (t)-tubule remodeling. This also unloads the left ventricle, which consequently atrophies. Experimental left-ventricular unloading can result in t-tubule remodeling, but it is currently unclear if this occurs in right-sided heart failure. In this work, we used a model of monocrotaline (MCT)-induced right heart failure in male rats, using confocal microscopy to investigate cellular remodeling of t-tubules, junctophilin-2 (JPH2), and ryanodine receptor-2 (RyR2). We examined remodeling across tissue anatomical regions of both ventricles: in trabeculae, papillary muscles, and free walls. Our analyses revealed that MCT hearts demonstrated a significant loss of t-tubule periodicity, disruption of the normal sarcomere striated pattern with JPH2 labeling, and also a disorganized striated pattern of RyR2, a feature not previously reported in right heart failure. Remodeling of JPH2 and RyR2 in the MCT heart was more pronounced in papillary muscles and trabeculae compared with free walls, particularly in the left ventricle. We find that these structures, commonly used as ex vivo muscle preparations, are more sensitive to the disease process.NEW & NOTEWORTHY In this work, we demonstrate that t-tubule remodeling occurs in the atrophied left ventricle as well as the overloaded right ventricle after right-side heart failure. Moreover, we identify that t-tubule remodeling in both ventricles is linked to sarcoplasmic reticulum remodeling as indicated by decreased labeling periodicity of both the Ca2+ release channel, RyR2, and the cardiac junction-forming protein, JPH2, that forms a link between the sarcoplasmic reticulum and sarcolemma. Studies developing treatments for right-sided heart failure should consider effects on both the right and left ventricle.


Subject(s)
Heart Failure/physiopathology , Heart Ventricles/physiopathology , Hypertrophy, Left Ventricular/physiopathology , Sarcomeres/pathology , Ventricular Function, Left , Ventricular Function, Right , Ventricular Remodeling , Animals , Calcium Signaling , Disease Models, Animal , Heart Failure/chemically induced , Heart Failure/metabolism , Heart Failure/pathology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Hypertrophy, Left Ventricular/chemically induced , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/pathology , Hypertrophy, Right Ventricular/physiopathology , Male , Membrane Proteins/metabolism , Monocrotaline , Rats, Wistar , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcomeres/metabolism
9.
Am J Physiol Heart Circ Physiol ; 321(2): H382-H389, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34142888

ABSTRACT

Pulmonary hypertension (PH) is associated with structural remodeling of pulmonary arteries (PAs) because of excessive proliferation of fibroblasts, endothelial cells, and smooth muscle cells (SMCs). The peptide hormone angiotensin II (ANG II) contributes to pulmonary vascular remodeling, in part, through its ability to trigger extracellular signal-regulated kinase (ERK1/2) activation. Here, we demonstrate that the ERK1/2 phosphatase, dual-specificity phosphatase 5 (DUSP5), functions as a negative regulator of ANG II-mediated SMC proliferation and PH. In contrast to wild-type controls, Dusp5 null mice infused with ANG II developed PH and right ventricular (RV) hypertrophy. PH in Dusp5 null mice was associated with thickening of the medial layer of small PAs, suggesting an in vivo role for DUSP5 as a negative regulator of ANG II-dependent SMC proliferation. Consistent with this, overexpression of DUSP5 blocked ANG II-mediated proliferation of cultured human pulmonary artery SMCs (hPASMCs) derived from patients with idiopathic PH or from failed donor controls. Collectively, the data support a role for DUSP5 as a feedback inhibitor of ANG II-mediated ERK signaling and PASMC proliferation and suggest that disruption of this circuit leads to adverse cardiopulmonary remodeling.NEW & NOTEWORTHY Dual-specificity phosphatases (DUSPs) serve critical roles in the regulation of mitogen-activated protein kinases, but their functions in the cardiovascular system remain poorly defined. Here, we provide evidence that DUSP5, which resides in the nucleus and specifically dephosphorylates extracellular signal-regulated kinase (ERK1/2), blocks pulmonary vascular smooth muscle cell proliferation. In response to angiotensin II infusion, mice lacking DUSP5 develop pulmonary hypertension and right ventricular cardiac hypertrophy. These findings illustrate DUSP5-mediated suppression of ERK signaling in the lungs as a protective mechanism.


Subject(s)
Cell Proliferation/genetics , Dual-Specificity Phosphatases/genetics , Heart Ventricles/metabolism , Hypertension, Pulmonary/genetics , Hypertrophy, Right Ventricular/genetics , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , Vascular Remodeling/genetics , Angiotensin II/pharmacology , Animals , Case-Control Studies , Cells, Cultured , Heart Ventricles/drug effects , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/physiopathology , MAP Kinase Signaling System , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Vasoconstrictor Agents/pharmacology
10.
Am J Physiol Heart Circ Physiol ; 320(4): H1526-H1534, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33577434

ABSTRACT

Use of electronic cigarettes is rapidly increasing among youth and young adults, but little is known regarding the long-term cardiopulmonary health impacts of these nicotine-containing devices. Our group has previously demonstrated that chronic, inhaled nicotine induces pulmonary hypertension (PH) and right ventricular (RV) remodeling in mice. These changes were associated with upregulated RV angiotensin-converting enzyme (ACE). Angiotensin II receptor blockers (ARBs) have been shown to reverse cigarette smoking-induced PH in rats. ACE inhibitor and ARB use in a large retrospective cohort of patients with PH is associated with improved survival. Here, we utilized losartan (an ARB specific for angiotensin II type 1 receptor) to further explore nicotine-induced PH. Male C57BL/6 mice received nicotine vapor for 12 h/day, and exposure was assessed using serum cotinine to achieve levels comparable to human smokers or electronic cigarette users. Mice were exposed to nicotine for 8 wk and a subset was treated with losartan via an osmotic minipump. Cardiac function was assessed using echocardiography and catheterization. Although nicotine exposure increased angiotensin II in the RV and lung, this finding was nonsignificant. Chronic, inhaled nicotine significantly increased RV systolic pressure and RV free wall thickness versus air control. These parameters were significantly lower in mice receiving both nicotine and losartan. Nicotine significantly increased RV internal diameter, with no differences seen between the nicotine and nicotine-losartan group. Neither nicotine nor losartan affected left ventricular structure or function. These findings provide the first evidence that antagonism of the angiotensin II type 1 receptor can ameliorate chronic, inhaled nicotine-induced PH and RV remodeling.NEW & NOTEWORTHY Chronic, inhaled nicotine causes pulmonary hypertension and right ventricular remodeling in mice. Treatment with losartan, an angiotensin II type 1 receptor antagonist, ameliorates nicotine-induced pulmonary hypertension and right ventricular remodeling. This novel finding provides preclinical evidence for the use of renin-angiotensin system-based therapies in the treatment of pulmonary hypertension, particularly in patients with a history of tobacco-product use.


Subject(s)
Arterial Pressure , E-Cigarette Vapor , Hypertension, Pulmonary/metabolism , Hypertrophy, Right Ventricular/metabolism , Nicotine , Pulmonary Artery/metabolism , Receptor, Angiotensin, Type 1/metabolism , Ventricular Function, Right , Ventricular Remodeling , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Arterial Pressure/drug effects , Disease Models, Animal , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/prevention & control , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/pathology , Hypertrophy, Right Ventricular/prevention & control , Inhalation Exposure , Losartan/pharmacology , Male , Mice, Inbred C57BL , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Receptor, Angiotensin, Type 1/drug effects , Signal Transduction , Time Factors , Ventricular Function, Right/drug effects , Ventricular Remodeling/drug effects
11.
Biomed Pharmacother ; 133: 111081, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33378977

ABSTRACT

OBJECTIVE: A growing evidence demonstrates that inflammation is a major contributor to the pathogenesis of pulmonary arterial hypertension (PAH). However, blocking inflammation has only been shown to be of minor clinical benefit due to a lack of understanding of the precise inflammation present in PAH. Thus, the present study aimed to investigate characteristics of inflammatory process in PAH induced by monocrotaline (MCT) in rats. METHODS: Adult male Sprague-Dawley rats received a single dose of MCT (50 mg/kg, ip), and the occurrence of PAH and inflammation biomarkers were measured at 3, 6, 9, 12, 15, 18, 21, 24, 27 and 30 days after MCT injection. RESULTS: From the 6th day after the injection of MCT, the mean pulmonary artery pressure gradually increased and doubled on the 30th day, accompanied by right ventricular hypertrophy and pulmonary arterial remodeling in a time-dependent manner. In the first 6 days after MCT treatment, only pro-inflammatory cytokines TNF-α, IL-1ß increased, which was defined as acute inflammatory phase, after that, both pro-inflammatory factors TNF-α, IL-1ß, IL-6, IL-12 and anti-inflammatory factors Arg1, IL-10, TGF-ß increased, which was defined as chronic inflammatory phase. The M1/M2 macrophage ratios in lung and alveolar lavage fluid were elevated on the 6th and 30th day, moreover, which were higher on the 6th than 30th day, and the PI3K/Akt signaling pathway increased along with the progression of PAH and correlated with pro-inflammatory proteins, which revealed also to some extent the characteristics of inflammation of PAH induced by MCT. CONCLUSION: The course of PAH induced by MCT injection is progressive with persistent inflammation, which is defined as acute inflammatory phase within 6 days after MCT treatment, after that, is defined as chronic inflammatory phase.


Subject(s)
Cytokines/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Pulmonary Arterial Hypertension/metabolism , Pulmonary Artery/metabolism , Vascular Remodeling , Animals , Arterial Pressure , Cytokines/genetics , Disease Models, Animal , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Inflammation/chemically induced , Inflammation/pathology , Inflammation/physiopathology , Macrophages/metabolism , Macrophages/pathology , Male , Monocrotaline , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Arterial Hypertension/pathology , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Rats, Sprague-Dawley , Signal Transduction , Time Factors
12.
Arq Bras Cardiol ; 115(3): 480-490, 2020 09.
Article in English, Portuguese | MEDLINE | ID: mdl-33027370

ABSTRACT

BACKGROUND: The monocrotaline (MCT)-induced pulmonary arterial hypertension model is one of the most reproduced today, presenting as a limitation the absence of plexiform lesions, typical manifestations of the severe disease in humans. OBJECTIVE: To evaluate the severity of MCT-induced pulmonary arteriopathy by pathological findings of lung and heart tissue samples, clinical course and 37-day survival. METHODS: Fifty male Wistar rats were divided into one of the four groups - control (CG) (n = 10) and three intervention (MCT) groups. The MCT groups received intraperitoneal injection (60 mg/kg) of MCT and remained exposed to the substance for 15 days (G15, n = 10), 30 days (G30, n = 10) and 37 days (G37, n = 20). At the end of each period, the animals were sacrificed, and pulmonary and cardiac tissues were collected for anatomopathological and morphometric analysis. The Kruskal-Wallis test was used, considering a level of significance of 5%. RESULTS: In the lungs of MCT animals, lesions related to pulmonary arteriopathy were found, including muscularization of the arterioles, hypertrophy of the middle layer and concentric neointimal lesions. Complex lesions were observed in MCT groups, described as plexiform and plexiform-like lesions. Right ventricular hypertrophy was evidenced by increased thickness and diameter of the cardiomyocytes and a significant increase in the right ventricular wall thickness (p <0.0000). CONCLUSION: The MCT model was able to generate moderate-severe pulmonary arteriopathy associated with secondary right ventricular hypertrophy. The 37-day survival rate was 50%. To our knowledge, this study was the first to note the presence of complex vascular lesions, similar to those observed in patients with severe pulmonary arterial hypertension, in an isolated MCT model. (Arq Bras Cardiol. 2020; 115(3):480-490).


FUNDAMENTO: O modelo de hipertensão arterial pulmonar induzida por monocrotalina (MCT) é um dos mais reproduzidos atualmente, apresentando como limitação a ausência de lesões plexiformes, manifestações típicas da doença grave em humanos. OBJETIVO: Avaliar a gravidade da arteriopatia pulmonar induzida por MCT por meio dos achados anatomopatológicos pulmonares e cardíacos, evolução clínica e sobrevida em 37 dias. MÉTODOS: Foram utilizados 50 ratos machos Wistar divididos em quatro grupos, sendo um controle (n = 10). Os três grupos restantes foram submetidos à inoculação de MCT (60 mg/kg i.p.) e ficaram sob o seu efeito por 15 (n = 10), 30 (n = 10) e 37 dias (n = 20). Ao final de cada período, os animais foram sacrificados, obtendo-se tecidos pulmonar e cardíaco para análise anatomopatológica e morfométrica. Empregou-se o teste Kruskal-Wallis, considerando nível de significância de 5%. RESULTADOS: Nos pulmões dos animais MCT foram constatadas lesões referentes à arteriopatia pulmonar, incluindo muscularização das arteríolas, hipertrofia da camada média e lesões neointimais concêntricas. Lesões complexas foram observadas nos grupos MCT, descritas como plexiforme e do "tipo" plexiforme (plexiform-like). A hipertrofia do ventrículo direito foi constatada pelo aumento da espessura e diâmetro dos cardiomiócitos e pelo aumento significativo da espessura da parede do ventrículo direito (p<0,0000). CONCLUSÃO: O modelo foi capaz de gerar arteriopatia pulmonar moderada-grave associada à hipertrofia do ventrículo direito secundária, com sobrevida de 50% em 37 dias. De nosso conhecimento, este estudo foi o primeiro a constatar a presença de lesões vasculares complexas, semelhantes às observadas em pacientes com hipertensão arterial pulmonar grave, em modelo isolado de MCT. (Arq Bras Cardiol. 2020; 115(3):480-490).


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Animals , Humans , Hypertension, Pulmonary/chemically induced , Hypertrophy, Right Ventricular/chemically induced , Male , Monocrotaline/toxicity , Rats , Rats, Wistar
13.
PLoS One ; 15(10): e0240692, 2020.
Article in English | MEDLINE | ID: mdl-33057388

ABSTRACT

Pulmonary arterial hypertension (PAH) is a lethal disease characterized by a progressive increase in pulmonary artery pressure due to an increase in vessel tone and occlusion of vessels. The endogenous vasodilator prostacyclin and its analogs are used as therapeutic agents for PAH. However, their pharmacological effects on occlusive vascular remodeling have not been elucidated yet. Selexipag is a recently approved, orally available and selective prostacyclin receptor agonist with a non-prostanoid structure. In this study, we investigated the pharmacological effects of selexipag on the pathology of chronic severe PAH in Sprague-Dawley and Fischer rat models in which PAH was induced by a combination of injection with the vascular endothelial growth factor receptor antagonist Sugen 5416 and exposure to hypoxia (SuHx). Oral administration of selexipag for three weeks significantly improved right ventricular systolic pressure and right ventricular (RV) hypertrophy in Sprague-Dawley SuHx rats. Selexipag attenuated the proportion of lung vessels with occlusive lesions and the medial wall thickness of lung arteries, corresponding to decreased numbers of Ki-67-positive cells and a reduced expression of collagen type 1 in remodeled vessels. Administration of selexipag to Fischer rats with SuHx-induced PAH reduced RV hypertrophy and mortality caused by RV failure. These effects were probably based on the potent prostacyclin receptor agonistic effect of selexipag on pulmonary vessels. Selexipag has been approved and is used in the clinical treatment of PAH worldwide. It is thought that these beneficial effects of prostacyclin receptor agonists on multiple aspects of PAH pathology contribute to the clinical outcomes in patients with PAH.


Subject(s)
Acetamides/therapeutic use , Hypoxia/complications , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/etiology , Pyrazines/therapeutic use , Receptors, Epoprostenol/agonists , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/therapeutic use , Acetamides/pharmacology , Animals , Cell Proliferation/drug effects , Collagen Type I/metabolism , Disease Models, Animal , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Hemodynamics/drug effects , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/physiopathology , Indoles , Lung/drug effects , Lung/pathology , Lung/physiopathology , Male , Pulmonary Arterial Hypertension/complications , Pulmonary Arterial Hypertension/physiopathology , Pyrazines/pharmacology , Pyrroles , Rats, Sprague-Dawley , Receptors, Epoprostenol/metabolism , Systole/drug effects , Vascular Remodeling/drug effects
14.
Arq. bras. cardiol ; 115(3): 480-490, out. 2020. graf
Article in English, Portuguese | LILACS, Sec. Est. Saúde SP | ID: biblio-1131304

ABSTRACT

Resumo Fundamento O modelo de hipertensão arterial pulmonar induzida por monocrotalina (MCT) é um dos mais reproduzidos atualmente, apresentando como limitação a ausência de lesões plexiformes, manifestações típicas da doença grave em humanos. Objetivo Avaliar a gravidade da arteriopatia pulmonar induzida por MCT por meio dos achados anatomopatológicos pulmonares e cardíacos, evolução clínica e sobrevida em 37 dias. Métodos Foram utilizados 50 ratos machos Wistar divididos em quatro grupos, sendo um controle (n = 10). Os três grupos restantes foram submetidos à inoculação de MCT (60 mg/kg i.p.) e ficaram sob o seu efeito por 15 (n = 10), 30 (n = 10) e 37 dias (n = 20). Ao final de cada período, os animais foram sacrificados, obtendo-se tecidos pulmonar e cardíaco para análise anatomopatológica e morfométrica. Empregou-se o teste Kruskal-Wallis, considerando nível de significância de 5%. Resultados Nos pulmões dos animais MCT foram constatadas lesões referentes à arteriopatia pulmonar, incluindo muscularização das arteríolas, hipertrofia da camada média e lesões neointimais concêntricas. Lesões complexas foram observadas nos grupos MCT, descritas como plexiforme e do "tipo" plexiforme (plexiform-like). A hipertrofia do ventrículo direito foi constatada pelo aumento da espessura e diâmetro dos cardiomiócitos e pelo aumento significativo da espessura da parede do ventrículo direito (p<0,0000). Conclusão O modelo foi capaz de gerar arteriopatia pulmonar moderada-grave associada à hipertrofia do ventrículo direito secundária, com sobrevida de 50% em 37 dias. De nosso conhecimento, este estudo foi o primeiro a constatar a presença de lesões vasculares complexas, semelhantes às observadas em pacientes com hipertensão arterial pulmonar grave, em modelo isolado de MCT. (Arq Bras Cardiol. 2020; 115(3):480-490)


Abstract Background The monocrotaline (MCT)-induced pulmonary arterial hypertension model is one of the most reproduced today, presenting as a limitation the absence of plexiform lesions, typical manifestations of the severe disease in humans. Objective To evaluate the severity of MCT-induced pulmonary arteriopathy by pathological findings of lung and heart tissue samples, clinical course and 37-day survival. Methods Fifty male Wistar rats were divided into one of the four groups - control (CG) (n = 10) and three intervention (MCT) groups. The MCT groups received intraperitoneal injection (60 mg/kg) of MCT and remained exposed to the substance for 15 days (G15, n = 10), 30 days (G30, n = 10) and 37 days (G37, n = 20). At the end of each period, the animals were sacrificed, and pulmonary and cardiac tissues were collected for anatomopathological and morphometric analysis. The Kruskal-Wallis test was used, considering a level of significance of 5%. Results In the lungs of MCT animals, lesions related to pulmonary arteriopathy were found, including muscularization of the arterioles, hypertrophy of the middle layer and concentric neointimal lesions. Complex lesions were observed in MCT groups, described as plexiform and plexiform-like lesions. Right ventricular hypertrophy was evidenced by increased thickness and diameter of the cardiomyocytes and a significant increase in the right ventricular wall thickness (p <0.0000). Conclusion The MCT model was able to generate moderate-severe pulmonary arteriopathy associated with secondary right ventricular hypertrophy. The 37-day survival rate was 50%. To our knowledge, this study was the first to note the presence of complex vascular lesions, similar to those observed in patients with severe pulmonary arterial hypertension, in an isolated MCT model. (Arq Bras Cardiol. 2020; 115(3):480-490)


Subject(s)
Humans , Animals , Male , Rats , Pulmonary Arterial Hypertension , Hypertension, Pulmonary/chemically induced , Monocrotaline/toxicity , Rats, Wistar , Hypertrophy, Right Ventricular/chemically induced
15.
BMC Pulm Med ; 20(1): 85, 2020 Apr 06.
Article in English | MEDLINE | ID: mdl-32252727

ABSTRACT

BACKGROUND: NTP42 is a novel antagonist of the thromboxane prostanoid receptor (TP), currently in development for the treatment of pulmonary arterial hypertension (PAH). PAH is a devastating disease with multiple pathophysiological hallmarks including excessive pulmonary vasoconstriction, vascular remodelling, inflammation, fibrosis, in situ thrombosis and right ventricular hypertrophy. Signalling through the TP, thromboxane (TX) A2 is a potent vasoconstrictor and mediator of platelet aggregation. It is also a pro-mitogenic, pro-inflammatory and pro-fibrotic agent. Moreover, the TP also mediates the adverse actions of the isoprostane 8-iso-prostaglandin F2α, a free-radical-derived product of arachidonic acid produced in abundance during oxidative injury. Mechanistically, TP antagonists should treat most of the hallmarks of PAH, including inhibiting the excessive vasoconstriction and pulmonary artery remodelling, in situ thrombosis, inflammation and fibrosis. This study aimed to investigate the efficacy of NTP42 in the monocrotaline (MCT)-induced PAH rat model, alongside current standard-of-care drugs. METHODS: PAH was induced by subcutaneous injection of 60 mg/kg MCT in male Wistar-Kyoto rats. Animals were assigned into groups: 1. 'No MCT'; 2. 'MCT Only'; 3. MCT + NTP42 (0.25 mg/kg BID); 4. MCT + Sildenafil (50 mg/kg BID), and 5. MCT + Selexipag (1 mg/kg BID), where 28-day drug treatment was initiated within 24 h post-MCT. RESULTS: From haemodynamic assessments, NTP42 reduced the MCT-induced PAH, including mean pulmonary arterial pressure (mPAP) and right systolic ventricular pressure (RSVP), being at least comparable to the standard-of-care drugs Sildenafil or Selexipag in bringing about these effects. Moreover, NTP42 was superior to Sildenafil and Selexipag in significantly reducing pulmonary vascular remodelling, inflammatory mast cell infiltration and fibrosis in MCT-treated animals. CONCLUSIONS: These findings suggest that NTP42 and antagonism of the TP signalling pathway have a relevant role in alleviating the pathophysiology of PAH, representing a novel therapeutic target with marked benefits over existing standard-of-care therapies.


Subject(s)
Antihypertensive Agents/pharmacology , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Artery/physiopathology , Receptors, Thromboxane/antagonists & inhibitors , Acetamides/pharmacology , Animals , Disease Models, Animal , Heart Ventricles/metabolism , Heart Ventricles/pathology , Hemodynamics/drug effects , Humans , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/drug therapy , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/pathology , Male , Monocrotaline , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/pathology , Pyrazines/pharmacology , Rats , Rats, Inbred WKY , Sildenafil Citrate/pharmacology , Vascular Remodeling/drug effects
16.
J Cardiovasc Pharmacol ; 75(6): 596-602, 2020 06.
Article in English | MEDLINE | ID: mdl-32168153

ABSTRACT

Chrysin (CH) is the main ingredient of many medicinal plants. Our previous study showed that CH could suppress hypoxia-induced pulmonary arterial smooth muscle cells proliferation and alleviate chronic hypoxia-induced pulmonary hypertension by targeting store-operated Ca entry (SOCE)-[Ca]i pathway. In this study, we investigated the effect of CH on monocrotaline-induced pulmonary hypertension (MCTPH) and the mechanism behind it. Results show that, in MCTPH model rats, (1) CH significantly reduced the enhancement of right ventricular pressure, right ventricular hypertrophy, and pulmonary vascular remodeling; (2) CH markedly suppressed the promotion of SOCE and [Ca]i in pulmonary arterial smooth muscle cells; and (3) CH obviously inhibited the MCT-upregulated proliferating cell nuclear antigen, TRPC1, TRPC4, and TRPC6 expression in distal pulmonary arteries. These results demonstrate that CH likely alleviates MCTPH by targeting TRPC1,4,6-SOCE-[Ca]i pathway.


Subject(s)
Antihypertensive Agents/pharmacology , Arterial Pressure/drug effects , Calcium Signaling/drug effects , Flavonoids/pharmacology , Hypertension, Pulmonary/drug therapy , Monocrotaline , Muscle, Smooth, Vascular/drug effects , Vascular Remodeling/drug effects , Animals , Disease Models, Animal , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Hypertrophy, Right Ventricular/prevention & control , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Rats, Sprague-Dawley , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/metabolism , Ventricular Function, Right/drug effects , Ventricular Pressure/drug effects , Ventricular Remodeling/drug effects
17.
J Cardiovasc Pharmacol ; 75(6): 545-555, 2020 06.
Article in English | MEDLINE | ID: mdl-32141989

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive and malignant disease characterized by pulmonary small arteries and right ventricle (RV) remodeling that can lead to severe RV dysfunction and death. The current therapeutic targets for RV dysfunction, which is strongly linked to mortality, are far from adequate. Therefore, we investigated the effect of ursolic acid (UA), a pentacyclic triterpenoid carboxylic acid, on PAH-induced RV remodeling and its underlying mechanism. We established a PAH model by injecting Sprague Dawley rats with monocrotaline (MCT, 60 mg/kg, ip), as verified by echocardiography and hemodynamic examination. Proteomic analysis was performed on RV samples using a Q Exactive high-field mass spectrometer, followed by KEGG enrichment analysis. The effect of 4 weeks of UA (50 mg/kg) treatment on RV remodeling was explored based on ultrasound, hemodynamic parameters, and histological changes, with the mechanism verified in vivo and in vitro by qRT-PCR and western blotting. RV hypertrophy, fibrosis, increased apoptosis, and abnormal metabolism were induced by MCT and suppressed by UA via a mechanism that changed the expression of key markers. UA also attenuated the Phenylephrine-induced hypertrophy of neonatal rat ventricular myocytes and upregulated peroxisome proliferator-activated receptor-alpha (PPARα), a key fatty acid metabolism regulator, and its downstream factor carnitine palmitoyl transferase 1b. In conclusion, UA exerts beneficial effects on PAH-induced RV dysfunction and remodeling by regulating PPARα-dependent fatty acid metabolism.


Subject(s)
Energy Metabolism/drug effects , Heart Ventricles/drug effects , Hypertrophy, Right Ventricular/prevention & control , Monocrotaline , Myocytes, Cardiac/drug effects , Pulmonary Arterial Hypertension/drug therapy , Triterpenes/pharmacology , Ventricular Function, Right/drug effects , Ventricular Remodeling/drug effects , Animals , Apoptosis/drug effects , Carnitine O-Palmitoyltransferase/metabolism , Cells, Cultured , Disease Models, Animal , Fatty Acids/metabolism , Fibrosis , Heart Ventricles/enzymology , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , PPAR alpha/metabolism , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/physiopathology , Rats, Sprague-Dawley , Ursolic Acid
18.
Int Heart J ; 61(1): 160-168, 2020 Jan 31.
Article in English | MEDLINE | ID: mdl-31956132

ABSTRACT

Pulmonary arterial hypertension (PAH) is a serious and fatal cardiovascular disorder characterized by increased pulmonary vascular resistance and progressive pulmonary vascular remodeling. The underlying pathological mechanisms of PAH are multi-factorial and multi-cellular. Alginate oligosaccharide (AOS), which is produced by depolymerizing alginate, shows better pharmacological activities and beneficial effects. The present study was undertaken to investigate the effects and potential mechanisms of AOS-mediated alleviation of pulmonary hypertension. Pulmonary hypertension was induced in Sprague-Dawley rats by a single intraperitoneal injection of monocrotaline (MCT; 60 mg/kg). Five weeks after the injection of MCT, AOS (5, 10, and 20 mg·kg-1·d-1) was injected intraperitoneally for another three weeks. The results showed that AOS prevented the development of MCT-induced pulmonary hypertension and right ventricular hypertrophy in a dose-dependent manner. AOS treatment also prevented MCT-induced pulmonary vascular remodeling via inhibition of the TGF-ß1/p-Smad2 signaling pathway. Furthermore, AOS treatment downregulated the expression of malondialdehyde, nicotinamide adenine dinucleotide phosphate oxidase, and pro-inflammatory cytokines, decreased macrophage infiltration, and upregulated the expression of anti-inflammatory cytokines. These findings indicate that AOS exerts anti-oxidative and anti-inflammatory effects in pulmonary arteries, which may contribute to the alleviation of pulmonary hypertension and pulmonary vascular remodeling.


Subject(s)
Alginates/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Hypertrophy, Right Ventricular/drug therapy , Monocrotaline/adverse effects , Pulmonary Arterial Hypertension/drug therapy , Alginates/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/metabolism , Injections, Intraperitoneal , Male , Malondialdehyde/metabolism , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Arterial Hypertension/metabolism , Random Allocation , Rats , Vascular Remodeling/drug effects
19.
Eur J Pharmacol ; 865: 172777, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31697933

ABSTRACT

Novel pharmacological approaches are needed to improve outcomes of patients with idiopathic pulmonary hypertension. Rho-associated protein kinase (ROCK) inhibitors have shown beneficial effects in preclinical models of pulmonary arterial hypertension (PAH), because of their role in the regulation of pulmonary artery vasoconstrictor tone and remodeling. We compared a ROCK inhibitor, Y-27632, for the first time with the dual endothelin receptor antagonist, macitentan, in a monocrotaline-induced rat pulmonary hypertension model. Different methods (echocardiography, hemodynamics, histology of right ventricle and pulmonary vessels, and circulating biomarkers) showed consistently that 100 mg/kg daily of Y-27632 and 10 mg/kg daily of macitentan slowed the progression of PAH both at the functional and structural levels. Treatments started on day 14 after monocrotaline injection and lasted 14 days. The findings of all experimental methods show that the selective ROCK inhibitor Y-27632 has more pronounced effects than macitentan, but a major limitation to its use is its marked peripheral vasodilating action.


Subject(s)
Amides/therapeutic use , Endothelin Receptor Antagonists/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pulmonary Arterial Hypertension/drug therapy , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , rho-Associated Kinases/antagonists & inhibitors , Animals , Heart Ventricles/pathology , Hemodynamics/drug effects , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/drug therapy , Male , Monocrotaline , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Arterial Hypertension/pathology , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Rats, Wistar
20.
Vascul Pharmacol ; 122-123: 106599, 2019.
Article in English | MEDLINE | ID: mdl-31629919

ABSTRACT

Targeting vascular remodeling in pulmonary arterial hypertension (PAH) remains a challenge given the lack of potent anti-remodeling abilities of the therapeutic drugs. Although sildenafil has been shown to ameliorate cardiopulmonary remodeling, that of tadalafil is questionable. Masitinib, a tyrosine kinase inhibitor appears safer and more potent than imatinib for treatment of malignancies, but its efficacy on PAH is unknown. Therefore, we investigated the anti-remodeling properties of masitinib (5, 15, 50 mg/kg) and tadalafil (5, 10 mg/kg) using a monocrotaline-induced rat model of PAH. The 14-day treatment with masitinib (15, 50 mg/kg) resulted in significantly decreased right ventricular (RV) systolic pressure (RVSP) and hypertrophy (RVH), and pulmonary vascular remodeling, whereas tadalafil showed weaker anti-remodeling properties. Besides, masitinib significantly blocked the mitogen-associated protein kinase (MAPK) pathway, and reduced phosphodiesterase (PDE)-5 mRNA expression in the lungs. By contrast, tadalafil did not significantly inhibit the MAPK pathway. Further, the 28-day treatment extension revealed that masitinib-treated rats (15 mg/kg) had significantly lower RVSP, and higher heart rate and serum cyclic guanosine monophosphate (cGMP) level, whereas those treated with tadalafil (10 mg/kg) showed insignificantly lower RVSP and higher cGMP level. Moreover, the RVH indices, heart rates, body weight gains, and survival rates of rats in both groups were comparable. Collectively, these results suggest that the treatment with a low-dose masitinib was non-inferior than tadalafil. A lower dose of masitinib may represent a novel approach to target both the cardiopulmonary remodeling and the dysregulated vasoconstriction in PAH.


Subject(s)
Antihypertensive Agents/pharmacology , Phosphodiesterase 5 Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Artery/drug effects , Tadalafil/pharmacology , Thiazoles/pharmacology , Vascular Remodeling/drug effects , Animals , Benzamides , Cyclic AMP/metabolism , Disease Models, Animal , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Hypertrophy, Right Ventricular/prevention & control , Mitogen-Activated Protein Kinases/metabolism , Molecular Targeted Therapy , Monocrotaline , Piperidines , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Pyridines , Signal Transduction , Ventricular Function, Right/drug effects , Ventricular Remodeling/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...