Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.852
Filter
1.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39125620

ABSTRACT

Pulmonary arterial hypertension (PAH) is a chronic disorder characterized by excessive pulmonary vascular remodeling, leading to elevated pulmonary vascular resistance and right ventricle (RV) overload and failure. MicroRNA-146a (miR-146a) promotes vascular smooth muscle cell proliferation and vascular neointimal hyperplasia, both hallmarks of PAH. This study aimed to investigate the effects of miR-146a through pharmacological or genetic inhibition on experimental PAH and RV pressure overload animal models. Additionally, we examined the overexpression of miR-146a on human pulmonary artery smooth muscle cells (hPASMCs). Here, we showed that miR-146a genic expression was increased in the lungs of patients with PAH and the plasma of monocrotaline (MCT) rats. Interestingly, genetic ablation of miR-146a improved RV hypertrophy and systolic pressures in Sugen 5415/hypoxia (SuHx) and pulmonary arterial banding (PAB) mice. Pharmacological inhibition of miR-146a improved RV remodeling in PAB-wild type mice and MCT rats, and enhanced exercise capacity in MCT rats. However, overexpression of miR-146a did not affect proliferation, migration, and apoptosis in control-hPASMCs. Our findings show that miR-146a may play a significant role in RV function and remodeling, representing a promising therapeutic target for RV hypertrophy and, consequently, PAH.


Subject(s)
MicroRNAs , Pulmonary Arterial Hypertension , Pulmonary Artery , Ventricular Function, Right , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Rats , Humans , Mice , Male , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/metabolism , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Disease Models, Animal , Monocrotaline , Cell Proliferation/genetics , Myocytes, Smooth Muscle/metabolism , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/physiopathology , Hypertrophy, Right Ventricular/metabolism , Vascular Remodeling/genetics , Rats, Sprague-Dawley
2.
BMC Cardiovasc Disord ; 24(1): 381, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39044140

ABSTRACT

BACKGROUND: Metabolic abnormalities and immune inflammation are deeply involved in pulmonary vascular remodelling and the development of pulmonary hypertension (PH). However, the regulatory mechanisms of glycolysis in macrophages are still elusive. Cumulative evidence indicates that ß-catenin plays a crucial role in metabolic reprogramming. This study aimed to investigate the effect of ß-catenin on macrophage glycolysis in PH. METHODS: LPS-induced BMDMs were generated via in vitro experiments. A monocrotaline (MCT)-induced PH rat model was established, and the ß-catenin inhibitor XAV939 was administered in vivo. The role of ß-catenin in glycolysis was analysed. The degree of pulmonary vascular remodelling was measured. RESULTS: ß-catenin was significantly increased in both in vitro and in vivo models. In LPS-induced BMDMs, ß-catenin increased the levels of hexokinase 2 (HK2), phosphofructokinase (PFK), M2-pyruvate kinase (PKM2), lactate dehydrogenase (LDH), and lactate (LA) and the expression of inflammatory cytokines and promoted PASMC proliferation and migration in vitro. XAV939 decreased the level of glycolysis and downregulated the expression of inflammatory cytokines in vivo. MCT promoted pulmonary arterial structural remodelling and right ventricular hypertrophy, and XAV939 alleviated these changes. CONCLUSIONS: Our findings suggest that ß-catenin is involved in the development of PH by promoting glycolysis and the inflammatory response in macrophages. Inhibition of ß-catenin could improve the progression of PH.


Subject(s)
Disease Models, Animal , Glycolysis , Hypertension, Pulmonary , Macrophages , Monocrotaline , Pulmonary Artery , Rats, Sprague-Dawley , Vascular Remodeling , beta Catenin , Animals , Glycolysis/drug effects , beta Catenin/metabolism , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Male , Vascular Remodeling/drug effects , Macrophages/metabolism , Macrophages/drug effects , Pulmonary Artery/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Pulmonary Artery/pathology , Cell Proliferation/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Signal Transduction , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Hypertrophy, Right Ventricular/chemically induced , Inflammation Mediators/metabolism , Rats , Cell Movement/drug effects
3.
Exp Cell Res ; 441(1): 114154, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38996959

ABSTRACT

Platelet-derived growth factor (PDGF) is one of the most important cytokines associated with pulmonary vascular remodeling in pulmonary arterial hypertension (PAH). PDGF receptor (PDGFR) inhibition exerted therapeutic effects on PAH in clinical trials, but serious side effects warrant the withdrawal of existing drugs. In this study, a novel highly selective PDGFR inhibitor WQ-C-401 was developed, and its effects on PDGFR signaling pathway and pulmonary vascular remodeling in PAH were investigated. Cell proliferation assays and Western blot analysis of PDGFRα/ß phosphorylation showed that WQ-C-401 inhibited PDGFR-mediated cell proliferation assay and suppressed PDGFR phosphorylation in a concentration-dependent manner. DiscoverX's KinomeScanTM technology confirmed the good kinome selectivity of WQ-C-401 (S score (1) of PDGFR = (0.01)). In monocrotaline (MCT)-induced PAH rats, intragastric administration of WQ-C-401 (25, 50, 100 mg/kg/d) or imatinib (50 mg/kg/d, positive control) significantly decreased right ventricular systolic pressure (RVSP). Histological analysis demonstrated that WQ-C-401 inhibited pulmonary vascular remodeling by reducing muscularization and fibrosis, as well as alleviated right ventricular hypertrophy in MCT-treated rats. In addition, WQ-C-401 suppressed MCT-induced cell hyperproliferation and CD68+ macrophage infiltration around the pulmonary artery. In vitro, WQ-C-401 inhibited PDGF-BB-induced proliferation and migration of human pulmonary arterial smooth muscle cells (PASMCs). Moreover, Western blot analysis showed that WQ-C-401 concertration-dependently inhibited PDGF-BB-induced phosphorylation of ERK1/2 and PDGFRß Y751, decreased collagen Ⅰ synthesis and increased alpha smooth muscle actin (α-SMA) expression in PASMCs. Collectively, our results suggest that WQ-C-401 is a selective and potent PDGFR inhibitor which could be a promising drug for the therapeutics of PAH by preventing pulmonary vascular remodeling.


Subject(s)
Cell Proliferation , Monocrotaline , Pulmonary Arterial Hypertension , Rats, Sprague-Dawley , Vascular Remodeling , Animals , Vascular Remodeling/drug effects , Rats , Cell Proliferation/drug effects , Male , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/pathology , Humans , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/metabolism , Phosphorylation/drug effects , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/metabolism , Signal Transduction/drug effects , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/prevention & control , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors
4.
Can Respir J ; 2024: 6619471, 2024.
Article in English | MEDLINE | ID: mdl-39081335

ABSTRACT

Pulmonary hypertension (PH) is a progressive and complex pulmonary vascular disease with poor prognosis. The aim of this study was to provide a new understanding of the lung pathology of disease and a noninvasive method in monitoring the establishment of animal models for basic and clinical studies of PH, indeed to explore clinical application value of lung ultrasound for patients with PH. Totally 32 male SD rats were randomly divided into control group, MCT (monocrotaline) group, PDTC (pyrrolidine dithiocarbamate) group, and NS (normal saline) group. Rats in the MCT group, PDTC group, and NS group received single intraperitoneal injection of MCT, while the control group received the same dose of NS. Then, PDTC group and NS group received PDTC and NS daily for treatment at the end of the model. Each group received lung ultrasound examination and measurement of pulmonary arterial pressure (PAP). Then, the rats were sacrificed to take the lung specimens to being observed. The ultrasound and pathological results were analyzed with a semiquantitative score. With the pulmonary artery pressure increases, the MCT group had a higher pulmonary ultrasound score and pathological score compared with the control group (p < 0.05). After PDTC treatment, the pulmonary ultrasound score and the pathological score decline (p < 0.05). We investigated both lung ultrasound scores, and the pathological scores were positively correlated with mean pulmonary artery pressure (mPAP) (both r > 0.8, p < 0.0001). Moreover, lung ultrasound scores were positively correlated with pathological scores (r > 0.8, p < 0.0001). We elucidated lung ultrasound evaluation providing more evidence for the management of PH in the rat model. Moreover, lung ultrasound provided a noninvasive method in monitoring the establishment of animal models for basic and clinical studies of PH.


Subject(s)
Disease Models, Animal , Hypertension, Pulmonary , Lung , Monocrotaline , Rats, Sprague-Dawley , Ultrasonography , Animals , Monocrotaline/toxicity , Hypertension, Pulmonary/diagnostic imaging , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Male , Rats , Lung/diagnostic imaging , Lung/pathology , Ultrasonography/methods , Pulmonary Artery/diagnostic imaging , Pulmonary Artery/pathology , Thiocarbamates , Pyrrolidines
5.
Sci Rep ; 14(1): 14056, 2024 06 18.
Article in English | MEDLINE | ID: mdl-38890390

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive disease characterized by pulmonary vascular remolding and occlusion, leading to the elevated pulmonary arterial pressures, right ventricular hypertrophy, and eventual heart failure if left untreated. Understanding the molecular mechanisms underlying the development and progression of pulmonary hypertension (PH) is crucial for devising efficient therapeutic approaches for the disease. Lung homogenates were collected weekly and underwent RNA-sequencing in the monocrotaline (MCT)-induced PH rat model to explore genes associated with PH progression. Statistical analyses revealed 1038, 1244, and 3125 significantly altered genes (P < 0.05, abs (log2fold change) > log21.5) between control and MCT-exposed rats during the first, second, and third week, respectively. Pathway enrichment analyses revealed involvement of cell cycle and innate immune system for the upregulated genes, GPCR and VEGF signaling for the downregulated genes. Furthermore, qRT-PCR validated upregulation of representative genes associated with cell cycle including Cdc25c (cell division cycle 25C), Cdc45, Top2a (topoisomerase IIα), Ccna2 (cyclin A2) and Ccnb1 (cyclin B1). Western blot and immunofluorescence analysis confirmed increases in PCNA, Ccna2, Top2a, along with other proliferation markers in the lung tissue of MCT-treated rats. In summary, RNA sequencing data highlights the significance of cell proliferation in progression of rodent PH.


Subject(s)
Cell Proliferation , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Hypertension, Pulmonary , Monocrotaline , Animals , Rats , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Male , Rats, Sprague-Dawley , Transcriptome , Lung/pathology , Lung/metabolism
6.
Eur J Pharmacol ; 978: 176786, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38942264

ABSTRACT

Pulmonary hypertension (PH) is characterized by pulmonary vascular remodeling, which endothelial-to-mesenchymal transition (EndMT) being its main progressive phase. Wogonin, a flavonoid extracted from the root of Scutellaria baicalensis Georgi, hinders the abnormal proliferation of cells and has been employed in the treatment of several cardiopulmonary diseases. This study was designed to investigate how wogonin affected EndMT during PH. Monocrotaline (MCT) was used to induce PH in rats. Binding capacity of TGF-ß1 receptor to wogonin detected by molecular docking and molecular dynamics. EndMT model was established in pulmonary microvascular endothelial cells (PMVECs) by transforming growth factor beta-1 (TGF-ß1). The result demonstrated that wogonin (20 mg/kg/day) attenuated right ventricular systolic pressure (RVSP), right ventricular hypertrophy and pulmonary vascular thickness in PH rats. EndMT in the pulmonary vascular was inhibited after wogonin treatment as evidenced by the restored expression of CD31 and decreased expression of α-SMA. Wogonin has strong affinity for both TGFBRI and TGFBRII, and has a better binding stability for TGFBRI. In TGF-ß1-treated PMVECs, wogonin (0.3, 1, and 3 µM) exhibited significant inhibitory effects on this transformation process via down-regulating the expression of p-Smad2 and Snail, while up-regulating the expression of p-Smad1/5. Additionally, results of Western blot and fluorescence shown that the expression of α-SMA were decrease with increasing level of CD31 in PMVECs. In conclusion, our research showed that wogonin suppressed EndMT via the TGF-ß1/Smad pathway which may lead to its alleviated effect on PH. Wogonin may be a promising drug against PH.


Subject(s)
Endothelial Cells , Flavanones , Hypertension, Pulmonary , Signal Transduction , Transforming Growth Factor beta1 , Animals , Male , Rats , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial-Mesenchymal Transition , Flavanones/pharmacology , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Lung/drug effects , Lung/metabolism , Molecular Docking Simulation , Monocrotaline , Rats, Sprague-Dawley , Transforming Growth Factor beta1/metabolism , Vascular Remodeling/drug effects
7.
Am J Physiol Heart Circ Physiol ; 327(1): H000, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38819383

ABSTRACT

Nitric oxide (NO) inhalation improves pulmonary hemodynamics in participants with pulmonary arterial hypertension (PAH). Although it can reduce pulmonary vascular resistance (PVR) in PAH, its impact on the dynamic mechanics of pulmonary arteries and its potential difference between control and participants with PAH remain unclear. PA impedance provides a comprehensive description of PA mechanics. With an arterial model, PA impedance can be parameterized into peripheral pulmonary resistance (Rp), arterial compliance (Cp), characteristic impedance of the proximal arteries (Zc), and transmission time from the main PA to the reflection site. This study investigated the effects of inhaled NO on PA impedance and its associated parameters in control and monocrotaline-induced pulmonary arterial hypertension (MCT-PAH) male rats (6/group). Measurements were obtained at baseline and during NO inhalation at 40 and 80 ppm. In both groups, NO inhalation decreased PVR and increased the left atrial pressure. Notably, its impact on PA impedance was frequency dependent, as revealed by reduced PA impedance modulus in the low-frequency range below 10 Hz, with little effect on the high-frequency range. Furthermore, NO inhalation attenuated Rp, increased Cp, and prolonged transmission time without affecting Zc. It reduced Rp more pronouncedly in MCT-PAH rats, whereas it increased Cp and delayed transmission time more effectively in control rats. In conclusion, the therapeutic effects of inhaled NO on PA impedance were frequency dependent and may differ between the control and MCT-PAH groups, suggesting that the effect on the mechanics differs depending on the pathological state.NEW & NOTEWORTHY Nitric oxide inhalation decreased pulmonary arterial impedance in the low-frequency range (<10 Hz) with little impact on the high-frequency range. It reduced peripheral pulmonary resistance more pronouncedly in pulmonary hypertension rats, whereas it increased arterial compliance and transmission time in control rats. Its effect on the mechanics of the pulmonary arteries may differ depending on the pathological status.


Subject(s)
Nitric Oxide , Pulmonary Artery , Vascular Resistance , Animals , Male , Nitric Oxide/metabolism , Pulmonary Artery/physiopathology , Pulmonary Artery/drug effects , Administration, Inhalation , Vascular Resistance/drug effects , Monocrotaline , Rats , Rats, Sprague-Dawley , Disease Models, Animal , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/chemically induced , Arterial Pressure/drug effects
8.
Aging (Albany NY) ; 16(9): 8142-8154, 2024 05 08.
Article in English | MEDLINE | ID: mdl-38728253

ABSTRACT

The specific mechanism of 4-hydroxysesamin (4-HS), a modification of Sesamin, on right ventricular failure due to pulmonary hypertension (PH) is ominous. By creating a rat model of PH in vivo and a model of pulmonary artery smooth muscle cell (PASMC) hypoxia and inflammation in vitro, the current work aimed to investigate in depth the molecular mechanism of the protective effect of 4-HS. In an in vitro model of hypoxia PASMC, changes in cell proliferation and inflammatory factors were detected after treatment with 4-HS, followed by changes in the JNK/p38 MAPK signaling pathway as detected by Western blot signaling pathway. The findings demonstrated that 4-HS was able to minimize PASMC cell death, block the JNK/p38 MAPK signaling pathway, and resist the promoting effect of hypoxia on PASMC cell proliferation. Following that, we found that 4-HS could both mitigate the right ventricular damage brought on by MCT and had a protective impact on rats Monocrotaline (MCT)-induced PH in in vivo investigations. The key finding of this study is that 4-HS may protect against PH by inhibiting the JNK/p38 MAPK signaling pathway.


Subject(s)
Cell Proliferation , Hypertension, Pulmonary , MAP Kinase Signaling System , p38 Mitogen-Activated Protein Kinases , Animals , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/drug therapy , Rats , p38 Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Male , Cell Proliferation/drug effects , Ventricular Dysfunction, Right/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Lignans/pharmacology , Lignans/therapeutic use , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/metabolism , Heart Failure/metabolism , Rats, Sprague-Dawley , Monocrotaline , Disease Models, Animal
9.
Biochem Biophys Res Commun ; 723: 150159, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-38815488

ABSTRACT

Exercise has been recognized as an effective intervention in the treatment of pulmonary arterial hypertension (PAH), supported by numerous studies. However, the precise effects of exercise on pulmonary function remain to be fully elucidated. In this study, using a rat model of swimming exercise training and monocrotaline-induced PAH, we aimed to explore its impact on pulmonary morphology and function. Our investigations revealed that MCT-treated rats exhibited augmented mean pulmonary arterial pressure (MPAP) and pulmonary vascular remodeling, which can be attenuated by 4 weeks of swimming exercise training (60 min/day, 5 days/week). Notably, MCT-treated rats showed impaired pulmonary function, as manifested by decreased tidal volume and dynamic compliance, which were reversed by exercise training. Assessment of pulmonary substrate in PAH rats indicated a prominent pro-inflammatory substrate, evidenced by macrophage accumulation through quantitative immunohistological analysis of macrophage-like cell expression (CD68), and extracellular matrix remodeling, evaluated by Masson staining. Importantly, both the pro-inflammatory substrate and extracellular matrix remodeling were ameliorated by swimming exercise training. Additionally, serum biochemical analysis demonstrated elevated levels of low-density lipoprotein cholesterol and Apolipoprotein B following MCT treatment, which were reduced with exercise intervention. Moreover, exercise enhanced systemic insulin sensitivity in both MCT-treated and untreated rats. Notably, MCT and exercise treatment both decreased fasting blood glucose (FBG) levels in rats, whereas exercise training reinstated FBG levels to normal in MCT-treated rats. In summary, our study suggests that swimming exercise confers a pulmonary protective effect in MCT-induced PAH rats, highlighting the potential importance of exercise-based rehabilitation in the management of PAH.


Subject(s)
Hypertension, Pulmonary , Insulin Resistance , Monocrotaline , Physical Conditioning, Animal , Rats, Sprague-Dawley , Swimming , Animals , Monocrotaline/toxicity , Male , Rats , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/therapy , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Lung/pathology , Lung/metabolism , Vascular Remodeling
10.
J Transl Med ; 22(1): 502, 2024 May 26.
Article in English | MEDLINE | ID: mdl-38797830

ABSTRACT

BACKGROUND: Inflammation and dysregulated immunity play vital roles in idiopathic pulmonary arterial hypertension (IPAH), while the mechanisms that initiate and promote these processes are unclear. METHODS: Transcriptomic data of lung tissues from IPAH patients and controls were obtained from the Gene Expression Omnibus database. Weighted gene co-expression network analysis (WGCNA), differential expression analysis, protein-protein interaction (PPI) and functional enrichment analysis were combined with a hemodynamically-related histopathological score to identify inflammation-associated hub genes in IPAH. The monocrotaline-induced rat model of pulmonary hypertension was utilized to confirm the expression pattern of these hub genes. Single-cell RNA-sequencing (scRNA-seq) data were used to identify the hub gene-expressing cell types and their intercellular interactions. RESULTS: Through an extensive bioinformatics analysis, CXCL9, CCL5, GZMA and GZMK were identified as hub genes that distinguished IPAH patients from controls. Among these genes, pulmonary expression levels of Cxcl9, Ccl5 and Gzma were elevated in monocrotaline-exposed rats. Further investigation revealed that only CCL5 and GZMA were highly expressed in T and NK cells, where CCL5 mediated T and NK cell interaction with endothelial cells, smooth muscle cells, and fibroblasts through multiple receptors. CONCLUSIONS: Our study identified a new inflammatory pathway in IPAH, where T and NK cells drove heightened inflammation predominantly via the upregulation of CCL5, providing groundwork for the development of targeted therapeutics.


Subject(s)
Chemokine CCL5 , Familial Primary Pulmonary Hypertension , Killer Cells, Natural , RNA-Seq , Single-Cell Analysis , T-Lymphocytes , Animals , Humans , Chemokine CCL5/metabolism , Chemokine CCL5/genetics , Killer Cells, Natural/metabolism , Killer Cells, Natural/immunology , Familial Primary Pulmonary Hypertension/genetics , Familial Primary Pulmonary Hypertension/pathology , Familial Primary Pulmonary Hypertension/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/immunology , Male , Cell Communication/genetics , Rats, Sprague-Dawley , Lung/pathology , Rats , Gene Regulatory Networks , Monocrotaline , Protein Interaction Maps/genetics , Computational Biology
11.
Am J Physiol Lung Cell Mol Physiol ; 327(2): L250-L257, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38810241

ABSTRACT

In the field of pulmonary hypertension (PH), a well-established protocol to induce severe angioproliferation in rats (SuHx) involves combining the VEGF-R inhibitor Sugen 5416 (SU5416) with 3 wk of hypoxia (Hx). In addition, injecting monocrotaline (MCT) into rats can induce inflammation and shear stress in the pulmonary vasculature, leading to neointima-like remodeling. However, the SuHx protocol in mice is still controversial, with some studies suggesting it yields higher and reversible PH than Hx alone, possibly due to species-dependent hypoxic responses. To establish an alternative rodent model of PH, we hypothesized mice would be more sensitive to hemodynamic changes secondary to shear stress compared with Hx. We attempted to induce severe and irreversible PH in mice by combining SU5416 or monocrotaline pyrrole (MCTP) injection with pneumonectomy (PNx). However, our experiments showed SU5416 administered to mice at various time points after PNx did not result in severe PH. Similarly, mice injected with MCTP after PNx (MPNx) showed no difference in right ventricular systolic pressure or exacerbated pulmonary vascular remodeling compared with PNx alone. These findings collectively demonstrate that C57/B6 mice do not develop severe and persistent PH when PNx is combined with either SU5416 or MCTP.NEW & NOTEWORTHY We attempted to establish a mouse model of severe and irreversible pulmonary hypertension by substituting hypoxia with pulmonary overcirculation. To do so, we treated mice with either SU5416 or monocrotaline pyrrole after pneumonectomy and performed hemodynamic evaluations for PH. Despite this "two-hit" protocol, mice did not exhibit signs of severe pulmonary hypertension or exacerbated pulmonary vascular remodeling compared with PNx alone.


Subject(s)
Hypertension, Pulmonary , Indoles , Mice, Inbred C57BL , Monocrotaline , Pneumonectomy , Pyrroles , Animals , Monocrotaline/analogs & derivatives , Pyrroles/pharmacology , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/chemically induced , Indoles/pharmacology , Mice , Male , Disease Models, Animal , Hypoxia/pathology , Vascular Remodeling/drug effects , Lung/pathology , Lung/drug effects , Lung/metabolism , Hemodynamics/drug effects
12.
Sci Rep ; 14(1): 12431, 2024 05 30.
Article in English | MEDLINE | ID: mdl-38816406

ABSTRACT

Pulmonary arterial hypertension (PAH) is a fatal disease featured by high morbidity and mortality. Although Cordycepin is known for its anti-inflammatory, antioxidant and immune-enhancing effects, its role in PAH treatment and the underlying mechanisms remain unclear. The therapeutic effects of Cordycepin on rats with PAH were investigated using a monocrotaline (MCT)-induced rat model. The metabolic effects of Cordycepin were assessed based on the plasma metabolome. The potential mechanisms of Cordycepin in PAH treatment were investigated through transcriptome sequencing and validated in pulmonary artery smooth muscle cells (PASMC). Evaluations included hematoxylin and eosin staining for pulmonary vascular remodeling, CCK-8 assay, EDU, and TUNEL kits for cell viability, proliferation, and apoptosis, respectively, and western blot for protein expression. Cordycepin significantly reduced right ventricular systolic pressure (RVSP) and right ventricular hypertrophy index (RVHI) in PAH rats, and mitigated pulmonary vascular remodeling. Plasma metabolomics showed that Cordycepin could reverse the metabolic disorders in the lungs of MCT-induced PAH rats, particularly impacting linoleic acid and alpha-linolenic acid metabolism pathways. Transcriptomics revealed that the P53 pathway might be the primary pathway involved, and western blot results showed that Cordycepin significantly increased P53 and P21 protein levels in lung tissues. Integrated analysis of transcriptomics and metabolomics suggested that these pathways were mainly enriched in linoleic acid metabolism and alpha-linolenic acid metabolism pathway. In vitro experiments demonstrated that Cordycepin significantly inhibited the PDGFBB (PD)-induced abnormal proliferation and migration of PASMC and promoted PD-induced apoptosis. Meanwhile, Cordycepin enhanced the expression levels of P53 and P21 proteins in PD-insulted PASMC. However, inhibitors of P53 and P21 eliminated these effects of Cordycepin. Cordycepin may activate the P53-P21 pathway to inhibit abnormal proliferation and migration of PASMC and promote apoptosis, offering a potential approach for PAH treatment.


Subject(s)
Apoptosis , Cell Proliferation , Deoxyadenosines , Pulmonary Arterial Hypertension , Animals , Deoxyadenosines/pharmacology , Deoxyadenosines/therapeutic use , Rats , Male , Apoptosis/drug effects , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/pathology , Cell Proliferation/drug effects , Transcriptome/drug effects , Metabolomics , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Monocrotaline , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Rats, Sprague-Dawley , Disease Models, Animal , Vascular Remodeling/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Linoleic Acid/pharmacology , Hypertrophy, Right Ventricular/drug therapy , Hypertrophy, Right Ventricular/metabolism , Gene Expression Profiling
14.
An Acad Bras Cienc ; 96(3): e20230446, 2024.
Article in English | MEDLINE | ID: mdl-38655920

ABSTRACT

Pulmonary arterial hypertension (PAH) is characterized by right ventricular failure and diminished cardiac output, potentially leading to renal and bone impairments. In contrast, resistance exercise training (RT) offers cardiovascular and bone health benefits. This study aimed to assess the impacts of stable PAH induced by monocrotaline (MCT) and RT on renal morphometry, as well as bone morphometry and biomechanical properties in male Wistar rats. Four experimental groups, untrained control (UC, n=7), trained control (TC, n=7), untrained hypertensive (UH, n=7), trained hypertensive (TH, n=7), were defined. After the first MCT or saline injection (20 mg/kg), trained rats were submitted to a RT program (i.e., Ladder climbing), 5 times/week. Seven days later the rats received the second MCT or saline dose. After euthanasia, renal and femoral histomorphometry and femoral biomechanical properties were assessed. PAH reduced renal glomerular area and volume, which was prevented by the RT. While PAH did not harm the femoral morphometry, structural and mechanical properties, RT improved the femoral parameters (e.g., length, percentage of trabeculae and bone marrow, ultimte and yield loads). Experimental stable PAH promotes renal but not bone damages, whereas RT prevents renal deteriorations and improves the femoral morphological and biomechanical properties.


Subject(s)
Disease Models, Animal , Kidney , Monocrotaline , Physical Conditioning, Animal , Rats, Wistar , Resistance Training , Animals , Male , Physical Conditioning, Animal/physiology , Rats , Kidney/physiopathology , Kidney/pathology , Resistance Training/methods , Pulmonary Arterial Hypertension/physiopathology , Femur/pathology , Femur/physiopathology , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/chemically induced
15.
Free Radic Biol Med ; 219: 141-152, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38636714

ABSTRACT

Pulmonary hypertension (PH) is a devastating disease that lacks effective treatment options and is characterized by severe pulmonary vascular remodeling. Pulmonary arterial endothelial cell (PAEC) dysfunction drives the initiation and pathogenesis of pulmonary arterial hypertension. Canonical transient receptor potential (TRPC) channels, a family of Ca2+-permeable channels, play an important role in various diseases. However, the effect and mechanism of TRPCs on PH development have not been fully elucidated. Among the TRPC family members, TRPC4 expression was markedly upregulated in PAECs from hypoxia combined with SU5416 (HySu)-induced PH mice and monocrotaline (MCT)-treated PH rats, as well as in hypoxia-exposed PAECs, suggesting that TRPC4 in PAECs may participate in the occurrence and development of PH. In this study, we aimed to investigate whether TRPC4 in PAECs has an aggravating effect on PH and elucidate the molecular mechanisms. We observed that hypoxia treatment promoted PAEC apoptosis through a caspase-12/endoplasmic reticulum stress (ERS)-dependent pathway. Knockdown of TRPC4 attenuated hypoxia-induced apoptosis and caspase-3/caspase-12 activity in PAECs. Accordingly, adeno-associated virus (AAV) serotype 6-mediated pulmonary endothelial TRPC4 silencing (AAV6-Tie-shRNA-TRPC4) or TRPC4 antagonist suppressed PH progression as evidenced by reduced right ventricular systolic pressure (RVSP), pulmonary vascular remodeling, PAEC apoptosis and reactive oxygen species (ROS) production. Mechanistically, unbiased RNA sequencing (RNA-seq) suggested that TRPC4 deficiency suppressed the expression of the proapoptotic protein sushi domain containing 2 (Susd2) in hypoxia-exposed mouse PAECs. Moreover, TRPC4 activated hypoxia-induced PAEC apoptosis by promoting Susd2 expression. Therefore, inhibiting TRPC4 ameliorated PAEC apoptosis and hypoxic PH in animals by repressing Susd2 signaling, which may serve as a therapeutic target for the management of PH.


Subject(s)
Apoptosis , Endothelial Cells , Hypertension, Pulmonary , Hypoxia , TRPC Cation Channels , Animals , Mice , Rats , Cells, Cultured , Disease Models, Animal , Endoplasmic Reticulum Stress , Endothelial Cells/metabolism , Endothelial Cells/pathology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/genetics , Hypoxia/metabolism , Indoles , Mice, Inbred C57BL , Monocrotaline/toxicity , Pulmonary Artery/pathology , Pulmonary Artery/metabolism , Pyrroles , Rats, Sprague-Dawley , Signal Transduction , TRPC Cation Channels/metabolism , TRPC Cation Channels/genetics , Vascular Remodeling/genetics
16.
Methods Mol Biol ; 2803: 173-185, 2024.
Article in English | MEDLINE | ID: mdl-38676893

ABSTRACT

Pulmonary hypertension (PH) is a chronic and progressive disorder characterized by elevated mean pulmonary arterial pressure, pulmonary vascular remodeling, and the development of concentric laminar intimal fibrosis with plexiform lesions. While rodent models have been developed to study PH, they have certain deficiencies and do not entirely replicate the human disease due to the heterogeneity of PH pathology. Therefore, combined models are necessary to study PH. Recent studies have shown that altered pulmonary blood flow is a significant trigger in the development of vascular remodeling and neointimal lesions. One of the most promising rodent models for increased pulmonary flow is the combination of unilateral left pneumonectomy with a "second hit" of monocrotaline (MCT) or SU5416. The removal of one lung in this model forces blood to circulate only in the other lung and induces increased and turbulent pulmonary blood flow. This increased vascular flow leads to progressive remodeling and occlusion of small pulmonary arteries. The second hit by MCT or SU5416 leads to endothelial cell dysfunction, resulting in severe PH and the development of plexiform arteriopathy.


Subject(s)
Disease Models, Animal , Hypertension, Pulmonary , Indoles , Lung , Monocrotaline , Pyrroles , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/chemically induced , Animals , Rats , Humans , Lung/pathology , Pneumonectomy/methods , Vascular Remodeling , Pulmonary Artery/pathology , Mice
17.
J Cardiovasc Pharmacol ; 84(1): 101-109, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38573589

ABSTRACT

ABSTRACT: Myocardial infarction (MI) and pulmonary arterial hypertension (PAH) are 2 prevalent cardiovascular diseases. In both conditions, oxidative stress is associated with a worse prognosis. Pterostilbene (PTE), an antioxidant compound, has been studied as a possible therapy for cardiovascular diseases. This study aims to evaluate the effect of PTE on oxidative stress in the hearts of animals with MI and in the lungs of animals with PAH. Male Wistar rats were used in both models. In the MI model, the experimental groups were sham, MI, and MI + PTE. In the PAH model, the experimental groups were control, PAH, and PAH + PTE. Animals were exposed to MI through surgical ligation of the left coronary artery, or to PAH, by the administration of monocrotaline (60 mg/kg). Seven days after undergoing cardiac injury, the MI + PTE animals were treated with PTE (100 mg/kg day) for 8 days. After this, the heart was collected for molecular analysis. The PAH + PTE animals were treated with PTE (100 mg/kg day) for 14 days, beginning 7 days after PAH induction. After this, the lungs were collected for biochemical evaluation. We found that PTE administration attenuated the decrease in ejection fraction and improved left ventricle end-systolic volume in infarcted animals. In the PAH model, PTE improved pulmonary artery flow and decreased reactive oxygen species levels in the lung. PTE administration promoted protective effects in terms of oxidative stress in 2 experimental models of cardiac diseases: MI and PAH. PTE also improved cardiac function in infarcted rats and pulmonary artery flow in animals with PAH.


Subject(s)
Antioxidants , Disease Models, Animal , Lung , Myocardial Infarction , Myocardium , Oxidative Stress , Pulmonary Arterial Hypertension , Rats, Wistar , Stilbenes , Animals , Oxidative Stress/drug effects , Male , Myocardial Infarction/physiopathology , Myocardial Infarction/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Lung/drug effects , Lung/metabolism , Lung/physiopathology , Stilbenes/pharmacology , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Arterial Hypertension/metabolism , Antioxidants/pharmacology , Myocardium/metabolism , Myocardium/pathology , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Pulmonary Artery/metabolism , Ventricular Function, Left/drug effects , Rats , Reactive Oxygen Species/metabolism , Arterial Pressure/drug effects , Monocrotaline
18.
Microvasc Res ; 154: 104686, 2024 07.
Article in English | MEDLINE | ID: mdl-38614154

ABSTRACT

Pulmonary hypertension (PH) is a chronic, progressive condition in which respiratory muscle dysfunction is a primary contributor to exercise intolerance and dyspnea in patients. Contractile function, blood flow distribution, and the hyperemic response are altered in the diaphragm with PH, and we sought to determine whether this may be attributed, in part, to impaired vasoreactivity of the resistance vasculature. We hypothesized that there would be blunted endothelium-dependent vasodilation and impaired myogenic responsiveness in arterioles from the diaphragm of PH rats. Female Sprague-Dawley rats were randomized into healthy control (HC, n = 9) and monocrotaline-induced PH rats (MCT, n = 9). Endothelium-dependent and -independent vasodilation and myogenic responses were assessed in first-order arterioles (1As) from the medial costal diaphragm in vitro. There was a significant reduction in endothelium-dependent (via acetylcholine; HC, 78 ± 15% vs. MCT, 47 ± 17%; P < 0.05) and -independent (via sodium nitroprusside; HC, 89 ± 10% vs. MCT, 66 ± 10%; P < 0.05) vasodilation in 1As from MCT rats. MCT-induced PH also diminished myogenic constriction (P < 0.05) but did not alter passive pressure responses. The diaphragmatic weakness, impaired hyperemia, and blood flow redistribution associated with PH may be due, in part, to diaphragm vascular dysfunction and thus compromised oxygen delivery which occurs through both endothelium-dependent and -independent mechanisms.


Subject(s)
Diaphragm , Hypertension, Pulmonary , Rats, Sprague-Dawley , Vasodilation , Animals , Female , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/etiology , Arterioles/physiopathology , Diaphragm/physiopathology , Diaphragm/blood supply , Disease Models, Animal , Vasodilator Agents/pharmacology , Endothelium, Vascular/physiopathology , Vasoconstriction , Monocrotaline/toxicity , Rats
19.
Sci Rep ; 14(1): 8128, 2024 04 07.
Article in English | MEDLINE | ID: mdl-38584196

ABSTRACT

Fat loss predicts adverse outcomes in advanced heart failure (HF). Disrupted circadian clocks are a primary cause of lipid metabolic issues, but it's unclear if this disruption affects fat expenditure in HF. To address this issue, we investigated the effects of disruption of the BMAL1/REV-ERBα circadian rhythmic loop on adipose tissue metabolism in HF.50 Wistar rats were initially divided into control (n = 10) and model (n = 40) groups. The model rats were induced with HF via monocrotaline (MCT) injections, while the control group received equivalent solvent injections. After establishing the HF model, the model group was further subdivided into four groups: normal rhythm (LD), inverted rhythm (DL), lentivirus vector carrying Bmal1 short hairpin RNA (LV-Bmal1 shRNA), and empty lentivirus vector control (LV-Control shRNA) groups, each with 10 rats. The DL subgroup was exposed to a reversed light-dark cycle of 8 h: 16 h (dark: light), while the rest adhered to normal light-dark conditions (light: dark 12 h: 12 h). Histological analyses were conducted using H&E, Oil Red O, and Picrosirius red stains to examine adipose and liver tissues. Immunohistochemical staining, RT-qPCR, and Western blotting were performed to detect markers of lipolysis, lipogenesis, and beiging of white adipose tissue (WAT), while thermogenesis indicators were detected in brown adipose tissue (BAT). The LD group rats exhibited decreased levels of BMAL1 protein, increased levels of REV-ERBα protein, and disrupted circadian circuits in adipose tissue compared to controls. Additionally, HF rats showed reduced adipose mass and increased ectopic lipid deposition, along with smaller adipocytes containing lower lipid content and fibrotic adipose tissue. In the LD group WAT, expression of ATGL, HSL, PKA, and p-PKA proteins increased, alongside elevated mRNA levels of lipase genes (Hsl, Atgl, Peripilin) and FFA ß-oxidation genes (Cpt1, acyl-CoA). Conversely, lipogenic gene expression (Scd1, Fas, Mgat, Dgat2) decreased, while beige adipocyte markers (Cd137, Tbx-1, Ucp-1, Zic-1) and UCP-1 protein expression increased. In BAT, HF rats exhibited elevated levels of PKA, p-PKA, and UCP-1 proteins, along with increased expression of thermogenic genes (Ucp-1, Pparγ, Pgc-1α) and lipid transportation genes (Cd36, Fatp-1, Cpt-1). Plasma NT-proBNP levels were higher in LD rats, accompanied by elevated NE and IL-6 levels in adipose tissue. Remarkably, morphologically, the adipocytes in the DL and LV-Bmal1 shRNA groups showed reduced size and lower lipid content, while lipid deposition in the liver was more pronounced in these groups compared to the LD group. At the gene/protein level, the BMAL1/REV-ERBα circadian loop exhibited severe disruption in LV-Bmal1 shRNA rats compared to LD rats. Additionally, there was increased expression of lipase genes, FFA ß oxidation genes, and beige adipocyte markers in WAT, as well as higher expression of thermogenic genes and lipid transportation genes in BAT. Furthermore, plasma NT-proBNP levels and adipose tissue levels of NE and IL-6 were elevated in LV-Bmal1 shRNA rats compared with LD rats. The present study demonstrates that disruption of the BMAL1/REV-ERBα circadian rhythmic loop is associated with fat expenditure in HF. This result suggests that restoring circadian rhythms in adipose tissue may help counteract disorders of adipose metabolism and reduce fat loss in HF.


Subject(s)
ARNTL Transcription Factors , Heart Failure , Rats , Animals , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Monocrotaline , Health Expenditures , Interleukin-6/metabolism , Rats, Wistar , Circadian Rhythm/genetics , Adipose Tissue, Brown/metabolism , Heart Failure/genetics , Heart Failure/metabolism , Lipase/metabolism , RNA, Small Interfering/metabolism , Lipids
20.
Biomed Pharmacother ; 174: 116505, 2024 May.
Article in English | MEDLINE | ID: mdl-38574614

ABSTRACT

Pulmonary arterial hypertension (PAH) was a devastating disease characterized by artery remodeling, ultimately resulting in right heart failure. The aim of this study was to investigate the effects of canagliflozin (CANA), a sodium-glucose cotransporter 2 inhibitor (SGLT2i) with mild SGLT1 inhibitory effects, on rats with PAH, as well as its direct impact on pulmonary arterial smooth muscle cells (PASMCs). PAH rats were induced by injection of monocrotaline (MCT) (40 mg/kg), followed by four weeks of treatment with CANA (30 mg/kg/day) or saline alone. Pulmonary artery and right ventricular (RV) remodeling and dysfunction in PAH were alleviated with CANA, as assessed by echocardiography. Hemodynamic parameters and structural of pulmonary arteriole, including vascular wall thickness and wall area, were reduced by CANA. RV hypertrophy index, cardiomyocyte hypertrophy, and fibrosis were decreased with CANA treatment. PASMCs proliferation was inhibited by CANA under stimulation by platelet-derived growth factor (PDGF)-BB or hypoxia. Activation of AMP kinase (AMPK) was induced by CANA treatment in cultured PASMCs in a time- and concentration-dependent manner. These effects of CANA were attenuated when treatment with compound C, an AMPK inhibitor. Abundant expression of SGLT1 was observed in PASMCs and pulmonary arteries, while SGLT2 expression was undetectable. SGLT1 increased in response to PDGF-BB or hypoxia stimulation, while PASMCs proliferation was inhibited and beneficial effects of CANA were counteracted by knockdown of SGLT1. Our research demonstrated for the first time that CANA inhibited the proliferation of PASMCs by regulating SGLT1/AMPK signaling and thus exerted an anti-proliferative effect on MCT-induced PAH.


Subject(s)
Canagliflozin , Cell Proliferation , Myocytes, Smooth Muscle , Pulmonary Arterial Hypertension , Vascular Remodeling , Animals , Rats , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Canagliflozin/pharmacology , Cell Proliferation/drug effects , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Monocrotaline/adverse effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/metabolism , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/pathology , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sodium-Glucose Transporter 1/drug effects , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Vascular Remodeling/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL