Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 284
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Oxid Med Cell Longev ; 2022: 3235102, 2022.
Article in English | MEDLINE | ID: mdl-35186183

ABSTRACT

Hypoxia-induced pulmonary arterial hypertension (HPAH) is due to hypoxia caused by vascular endothelial cell remolding and damage. Previous studies have suggested that CX3CL1 plays an important role in HPAH which is affected by oxidative stress. Ca2+ channel activation correlated with increasing NF-κB levels induced by ROS. Tanreqing injection (TRQ) is a traditional Chinese medicine (TCM) for acute upper respiratory tract infection and acute pneumonia. In the present study, we explored the effect of TRQ on human pulmonary artery smooth muscle cells (HPASMCs) undergoing hypoxia and feasible molecular mechanisms involved in. Cell proliferation was assayed using CCK8 kits. Immunofluorescence and western blotting along with ELISA assay were performed to investigate the effect of TRQ on hypoxia-induced ROS, Ca2+, hydroxyl free radicals, and the expression of Ca2+ channel protein TRPC1, CX3CR1, HIF-1α, NF-κBp65, and p-NF-κBp65 in HPASMCs. Human CX3CL1 and the inhibitor of TRPC1 as SKF96365 were used for further investigation. TRQ inhibited hypoxia-induced increasing cell adhesion, ROS, Ca2+, hydroxyl free radicals, CX3CR1, HIF-1α, NF-κBp65 activation, and even on TRPC1 expression in HPASMC which tended to be attenuated even reversed by CX3CL1. Our results suggested that TRQ might help to attenuate remodeling of HPASMC through inhibiting the ROS and TRPC1/CX3CL1 signaling pathway.


Subject(s)
Cell Hypoxia/drug effects , Chemokine CX3CL1/metabolism , Drugs, Chinese Herbal/therapeutic use , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/etiology , Myocytes, Smooth Muscle/drug effects , Pulmonary Artery/drug effects , TRPC Cation Channels/metabolism , Cell Proliferation , Drugs, Chinese Herbal/pharmacology , Humans , Hypertension, Pulmonary/physiopathology
2.
Physiol Rep ; 9(22): e15090, 2021 11.
Article in English | MEDLINE | ID: mdl-34816616

ABSTRACT

Pulmonary hypertension (PH) is a global health issue with a prevalence of 10% in ages >65 years. Right heart failure (RHF) is the main cause of death in PH. We have previously shown that monocrotaline (MCT)-induced PH and RHF are due to an increase in oxidative stress. In this study, probucol (PROB), a strong antioxidant with a lipid-lowering property, versus lovastatin (LOV), a strong lipid-lowering drug with some antioxidant effects, were evaluated for their effects on the MCT-induced RHF. Rats were treated (I.P.) with PROB (10 mg/kg ×12) or LOV (4 mg/kg ×12), daily 6 days before and 6 days after a single MCT injection (60 mg/kg). Serial echocardiography was performed and at 4-week post-MCT, lung wet-to-dry weight, hemodynamics, RV glutathione peroxidase (GSHPx), superoxide dismutase (SOD), catalase, lipid peroxidation, and myocardial as well as plasma lipids were examined. MCT increased RV systolic and diastolic pressures, wall thickness, RV end diastolic diameter, mortality, and decreased ejection fraction as well as pulmonary artery acceleration time. These changes were mitigated by PROB while LOV had no effect. Furthermore, PROB prevented lipid peroxidation, lowered lipids, and increased GSHPx and SOD in RV myocardium. LOV did decrease the lipids but had no effect on antioxidants and lipid peroxidation. A reduction in oxidative stress and not the lipid-lowering effect of PROB may explain the prevention of MCT-induced PH, RHF, and mortality. Thus targeting of oxidative stress as an adjuvant therapy is suggested.


Subject(s)
Anticholesteremic Agents/pharmacology , Antioxidants/pharmacology , Heart Failure/metabolism , Heart/drug effects , Hypertension, Pulmonary/metabolism , Lipid Peroxidation/drug effects , Lovastatin/pharmacology , Myocardium/metabolism , Oxidative Stress/drug effects , Probucol/pharmacology , Animals , Catalase/drug effects , Catalase/metabolism , Echocardiography , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/metabolism , Heart Failure/chemically induced , Heart Failure/drug therapy , Heart Failure/physiopathology , Hemodynamics , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Lung/drug effects , Monocrotaline/toxicity , Organ Size/drug effects , Rats , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism , Ventricular Dysfunction, Right/chemically induced , Ventricular Dysfunction, Right/drug therapy , Ventricular Dysfunction, Right/metabolism , Ventricular Dysfunction, Right/physiopathology
3.
Am J Physiol Heart Circ Physiol ; 321(2): H318-H338, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34142886

ABSTRACT

Chronic thromboembolic pulmonary hypertension (CTEPH) is caused by recurrent or unresolved pulmonary thromboemboli, leading to perfusion defects and increased arterial wave reflections. CTEPH treatment aims to reduce pulmonary arterial pressure and reestablish adequate lung perfusion, yet patients with distal lesions are inoperable by standard surgical intervention. Instead, these patients undergo balloon pulmonary angioplasty (BPA), a multisession, minimally invasive surgery that disrupts the thromboembolic material within the vessel lumen using a catheter balloon. However, there still lacks an integrative, holistic tool for identifying optimal target lesions for treatment. To address this insufficiency, we simulate CTEPH hemodynamics and BPA therapy using a multiscale fluid dynamics model. The large pulmonary arterial geometry is derived from a computed tomography (CT) image, whereas a fractal tree represents the small vessels. We model ring- and web-like lesions, common in CTEPH, and simulate normotensive conditions and four CTEPH disease scenarios; the latter includes both large artery lesions and vascular remodeling. BPA therapy is simulated by simultaneously reducing lesion severity in three locations. Our predictions mimic severe CTEPH, manifested by an increase in mean proximal pulmonary arterial pressure above 20 mmHg and prominent wave reflections. Both flow and pressure decrease in vessels distal to the lesions and increase in unobstructed vascular regions. We use the main pulmonary artery (MPA) pressure, a wave reflection index, and a measure of flow heterogeneity to select optimal target lesions for BPA. In summary, this study provides a multiscale, image-to-hemodynamics pipeline for BPA therapy planning for patients with inoperable CTEPH. NEW & NOTEWORTHY This article presents novel computational framework for predicting pulmonary hemodynamics in chronic thromboembolic pulmonary hypertension. The mathematical model is used to identify the optimal target lesions for balloon pulmonary angioplasty, combining simulated pulmonary artery pressure, wave intensity analysis, and a new quantitative metric of flow heterogeneity.


Subject(s)
Hemodynamics , Hypertension, Pulmonary/physiopathology , Pulmonary Artery/physiopathology , Pulmonary Embolism/physiopathology , Angioplasty, Balloon , Chronic Disease , Humans , Hypertension, Pulmonary/diagnostic imaging , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/therapy , Models, Cardiovascular , Models, Theoretical , Pulmonary Artery/diagnostic imaging , Pulmonary Embolism/complications , Pulmonary Embolism/diagnostic imaging , Pulmonary Embolism/therapy
4.
Int J Mol Sci ; 22(6)2021 Mar 13.
Article in English | MEDLINE | ID: mdl-33805714

ABSTRACT

Trifluoperazine (TFP), an antipsychotic drug approved by the Food and Drug Administration, has been show to exhibit anti-cancer effects. Pulmonary arterial hypertension (PAH) is a devastating disease characterized by a progressive obliteration of small pulmonary arteries (PAs) due to exaggerated proliferation and resistance to apoptosis of PA smooth muscle cells (PASMCs). However, the therapeutic potential of TFP for correcting the cancer-like phenotype of PAH-PASMCs and improving PAH in animal models remains unknown. PASMCs isolated from PAH patients were exposed to different concentrations of TFP before assessments of cell proliferation and apoptosis. The in vivo therapeutic potential of TFP was tested in two preclinical models with established PAH, namely the monocrotaline and sugen/hypoxia-induced rat models. Assessments of hemodynamics by right heart catheterization and histopathology were conducted. TFP showed strong anti-survival and anti-proliferative effects on cultured PAH-PASMCs. Exposure to TFP was associated with downregulation of AKT activity and nuclear translocation of forkhead box protein O3 (FOXO3). In both preclinical models, TFP significantly lowered the right ventricular systolic pressure and total pulmonary resistance and improved cardiac function. Consistently, TFP reduced the medial wall thickness of distal PAs. Overall, our data indicate that TFP could have beneficial effects in PAH and support the view that seeking new uses for old drugs may represent a fruitful approach.


Subject(s)
Cardiovascular Agents/pharmacology , Gene Expression Regulation/drug effects , Hypertension, Pulmonary/drug therapy , Hypoxia/prevention & control , Myocytes, Smooth Muscle/drug effects , Trifluoperazine/pharmacology , Animals , Antipsychotic Agents/pharmacology , Cell Proliferation/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Repositioning , Female , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Hemodynamics/drug effects , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Hypoxia/chemically induced , Hypoxia/genetics , Hypoxia/physiopathology , Indoles/administration & dosage , Monocrotaline/administration & dosage , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pyrroles/administration & dosage , Rats , Rats, Sprague-Dawley , Survivin/genetics , Survivin/metabolism
5.
Transl Res ; 233: 127-143, 2021 07.
Article in English | MEDLINE | ID: mdl-33691194

ABSTRACT

Ophiocordyceps sinensis (OCS), an entomopathogenic fungus, is known to exert antiproliferative and antitissue remodeling effects. Vascular remodeling and vasoconstriction play critical roles in the development of pulmonary hypertension (PH). The therapeutic potential of OCS for PH was investigated using rodent PH models, and cultured pulmonary artery endothelial and smooth muscle cells (PAECs and PASMCs), with a focus on the involvement of TRPM7. OCS ameliorated the development of PH, right ventricular hypertrophy and dysfunction in the monocrotaline-induced PH rats. The genetic knockout of TRPM7 attenuated the development of PH in mice with monocrotaline pyrrole-induced PH. TRPM7 was associated with medial hypertrophy and the plexiform lesions in rats and humans with PH. OCS suppressed proliferation of PASMCs derived from the PH patients. Ethanol extracts of OCS inhibited TRPM7-like current, TGF-ß2-induced endothelial-mesenchymal transition, IL-6-induced STAT3 phosphorylation, and PDGF-induced Akt phosphorylation in PAECs or PASMCs. These inhibitory effects were recapitulated by either siRNA-mediated TRPM7 knockdown or treatment with TRPM7 antagonist FTY-720. OCS and FTY-720 induced vasorelaxation in the isolated normal human pulmonary artery. As a result, the present study proposes the therapeutic potential of OCS for the treatment of PH. The inhibition of TRPM7 is suggested to underlie the therapeutic effect of OCS.


Subject(s)
Cordyceps/physiology , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/therapy , TRPM Cation Channels/antagonists & inhibitors , Animals , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Fingolimod Hydrochloride/pharmacology , Gene Knockdown Techniques , Humans , Hypertension, Pulmonary/pathology , Male , Medicine, Chinese Traditional , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , TRPM Cation Channels/deficiency , TRPM Cation Channels/genetics , TRPM Cation Channels/physiology , Translational Research, Biomedical , Vasodilation
6.
Medicine (Baltimore) ; 100(8): e24896, 2021 Feb 26.
Article in English | MEDLINE | ID: mdl-33663121

ABSTRACT

INTRODUCTION: Hemodynamically-instable ventricular arrhythmias (VAs) are rare in patients with pulmonary hypertension (PH). To the best of our knowledge, only 1 case has been reported so far. Moreover, the pathogenesis of this kind of arrhythmia remains obscured and its treatment is challenging. Here we report another case and presented the substrate for VAs initiation and therapeutic effect of radiofrequency ablation. PATIENT CONCERNS: This is a 57-year-old man who presented paroxysmal palpitation associated with presyncope at rest. Surface electrocardiogram (ECG) revealed frequent ventricular premature contractions and non-sustained ventricular tachycardia when symptoms occurred. He also had a history of severe PH which was secondary to atrial septal defect and partial anomalous pulmonary venous drainage and suffered from obvious dyspnea when climbing stairs World Health Organization Class III (WHO Class III). DIAGNOSIS: Hemodynamically-instable VAs associated with severe PH. INTERVENTION: Echocardiography revealed enlargement of right ventricle (right ventricle [RV]: 43 mm). Electrophysiological examination showed the origin of VAs is next to a small low-voltage zone of RV. Radiofrequency delivery at the origin successfully terminated VAs without occurrence of complication. OUTCOME: The patient was free from arrhythmias and got an improvement of exercise tolerance, just with mild dyspnea when climbing stairs World Health Organization Class II (WHO class II), during six-month follow up. LESSONS: This case suggests the low-voltage zone of remodeled RV, which may be secondary to increased pulmonary artery pressure, serves as the substrate for VAs initiation in patient with PH. Radiofrequency ablation can successfully terminate VAs and the termination of VAs can significantly improve the patient's impaired exercise tolerance.


Subject(s)
Hypertension, Pulmonary/complications , Radiofrequency Ablation/methods , Ventricular Premature Complexes/complications , Echocardiography , Electrophysiologic Techniques, Cardiac/methods , Heart Ventricles/diagnostic imaging , Heart Ventricles/physiopathology , Humans , Hypertension, Pulmonary/physiopathology , Male , Middle Aged , Ventricular Premature Complexes/physiopathology , Ventricular Premature Complexes/therapy , Ventricular Remodeling
7.
Am J Physiol Heart Circ Physiol ; 320(3): H980-H990, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33416457

ABSTRACT

Perinatal hypoxia induces permanent structural and functional changes in the lung and its pulmonary circulation that are associated with the development of pulmonary hypertension (PH) in later life. The mechanistic target of the rapamycin (mTOR) pathway is vital for fetal lung development and is implicated in hypoxia-associated PH, yet its involvement in the developmental programming of PH remains unclear. Pregnant C57/BL6 dams were placed in hyperbaric (760 mmHg) or hypobaric chambers during gestation (505 mmHg, day 15 through postnatal day 4) or from weaning through adulthood (420 mmHg, postnatal day 21 through 8 wk). Pulmonary hemodynamics and right ventricular systolic pressure (RVSP) were measured at 8 wk. mTOR pathway proteins were assessed in fetal (day 18.5) and adult lung (8 wk). Perinatal hypoxia induced PH during adulthood, even in the absence of a sustained secondary hypoxic exposure, as indicated by reduced pulmonary artery acceleration time (PAAT) and peak flow velocity through the pulmonary valve, as well as greater RVSP, right ventricular (RV) wall thickness, and RV/left ventricular (LV) weight. Such effects were independent of increased blood viscosity. In fetal lung homogenates, hypoxia reduced the expression of critical downstream mTOR targets, most prominently total and phosphorylated translation repressor protein (4EBP1), as well as vascular endothelial growth factor, a central regulator of angiogenesis in the fetal lung. In contrast, adult offspring of hypoxic dams tended to have elevated p4EBP1 compared with controls. Our data suggest that inhibition of mTORC1 activity in the fetal lung as a result of gestational hypoxia may interrupt pulmonary vascular development and thereby contribute to the developmental programming of PH.NEW & NOTEWORTHY We describe the first study to evaluate a role for the mTOR pathway in the developmental programming of pulmonary hypertension. Our findings suggest that gestational hypoxia impairs mTORC1 activation in the fetal lung and may impede pulmonary vascular development, setting the stage for pulmonary vascular disease in later life.


Subject(s)
Fetal Hypoxia/complications , Hypertension, Pulmonary/etiology , Lung/blood supply , Lung/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Neovascularization, Physiologic , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/metabolism , Disease Models, Animal , Female , Fetal Hypoxia/metabolism , Fetal Hypoxia/physiopathology , Gestational Age , Hemodynamics , Hyperbaric Oxygenation , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Mice, Inbred C57BL , Phosphorylation , Pregnancy , Prenatal Exposure Delayed Effects , Pulmonary Circulation , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Ventricular Function, Right , Ventricular Pressure
8.
J Pharmacol Sci ; 144(4): 237-244, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33070843

ABSTRACT

Hypoxic pulmonary hypertension (HPH) is a progressive and irreversible disease that reduces survival. Echinacoside is a phenylethanoid glycoside from Tibetan herbs known for its vasorelaxant effect and for inhibiting the proliferation of rat pulmonary arterial smooth muscle cells. This study aimed to investigate the effect of echinacoside on HPH. Sprague Dawley rats were housed in a hypobaric hypoxia chamber (4500 m) for 28 days to obtain the HPH model. Echinacoside (3.75, 7.5, 15, 30 and 40 mg/kg) was administered by intraperitoneal injection from the 1st to the 28th day. The mean pulmonary artery pressure (mPAP), right ventricular hypertrophy index, hemoglobin, hematocrit, red blood cell concentration and morphological change of pulmonary arteries were evaluated. Vascular perfusion assay was used to assess the pulmonary artery function. Echinacoside reduced mPAP, hemoglobin, hematocrit, right ventricular hypertrophy index and mean wall thickness% of pulmonary arteries in HPH rats. It significantly increased maximum vasoconstriction percentage of pulmonary arteries induced by noradrenaline in a dose-dependent manner. In addition, it improved the responsiveness of pulmonary arteries to acetylcholine and sodium nitroprusside. Therefore, Echinacoside might be an effective treatment against HPH, since it regulated pulmonary artery endothelium and smooth muscle layer function and improved the remodeling of pulmonary artery.


Subject(s)
Glycosides/administration & dosage , Glycosides/pharmacology , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/etiology , Hypoxia/complications , Phytotherapy , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Vascular Remodeling/drug effects , Vasoconstriction/drug effects , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Glycosides/therapeutic use , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/prevention & control , In Vitro Techniques , Injections, Intraperitoneal , Male , Rats, Sprague-Dawley , Vasodilator Agents
9.
J Int Med Res ; 48(5): 300060520914218, 2020 May.
Article in English | MEDLINE | ID: mdl-32419546

ABSTRACT

OBJECTIVES: Danshensu is a traditional Chinese medicine that is used for treatment of cardiovascular diseases. We previously demonstrated its preventive effect against early-stage hypoxic pulmonary hypertension (HPH) in a rat model. To determine whether danshensu treatment might be useful for patients with chronic HPH, we examined its therapeutic effect in rats with prolonged HPH. METHODS: Adult Sprague-Dawley rats received danshensu (80, 160, and 320 mg/kg) during or after hypoxia exposure to assess preventive and therapeutic effects, respectively. Right ventricle systolic pressure (RVSP), right ventricle hypertrophy index (RVHI), and mean left carotid artery pressure (mCAP) were measured in each group. Western blotting was used to assess transforming growth factor (TGF)-ß expression levels in rats and cultured cells exposed to hypoxia. RESULTS: Preventive danshensu treatment significantly reduced the elevation of RVSP and RVHI in rats exposed to hypoxia, whereas therapeutic danshensu treatment did not; mCAP did not change in any treatment group. The increased expression levels of TGF-ß induced by hypoxia were inhibited by preventive danshensu treatment, but not by therapeutic danshensu treatment. CONCLUSIONS: Although danshensu treatment could prevent HPH, it had no obvious therapeutic effect after development of HPH. Therefore, danshensu might be suitable for clinical treatment of early-stage HPH.


Subject(s)
Drugs, Chinese Herbal/administration & dosage , Hypertension, Pulmonary/prevention & control , Hypertrophy, Right Ventricular/prevention & control , Hypoxia/complications , Lactates/administration & dosage , Animals , Blood Pressure/physiology , Carotid Arteries/drug effects , Carotid Arteries/physiopathology , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/drug therapy , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/drug therapy , Male , Primary Cell Culture , Pulmonary Artery/cytology , Rats , Rats, Sprague-Dawley , Time Factors , Time-to-Treatment
10.
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
11.
Medicina (Kaunas) ; 56(3)2020 Mar 16.
Article in English | MEDLINE | ID: mdl-32188043

ABSTRACT

Background and objectives: Pulmonary hypertension (PH) is characterized by the vasoconstriction and abnormally proliferative vascular cells. The available allopathic treatment options for PH are still not able to cure the disease. Alternative medicine is becoming popular and drawing the attention of the general public and scientific communities. The entomogenous fungus Yarsagumba (Cordyceps sinensis) and its biologically active ingredient cordycepin may represent the therapeutic option for this incurable disease, owing to their anti-inflammatory, vasodilatory and anti-oxidative effects. Methods: In this study, we investigated whether Yarsagumba extract and cordycepin possess anti-proliferative and vasorelaxant properties in the context of PH, using 5-bromo-2'-deoxyuridine assay and isolated mice lungs, respectively. Results: Our results revealed that Yarsagumba extract and its bioactive compound cordycepin significantly attenuated the proliferation of human pulmonary artery smooth muscle cells derived from donor and PH subjects. In isolated murine lungs, only Yarsagumba extract, but not cordycepin, resulted in vasodilatation, indicating the probable existence of other bioactive metabolites present in Yarsagumba that may be responsible for this outcome. Conclusion: Future comprehensive in vivo and in vitro research is crucially needed to discover the profound mechanistic insights with regard to this promising therapeutic potency of Yarsagumba extract and to provide further evidence as to whether it can be used as a strategy for the treatment of PH.


Subject(s)
Antifungal Agents/pharmacology , Biocompatible Materials/pharmacology , Deoxyadenosines/pharmacology , Hypertension, Pulmonary/drug therapy , Vasodilator Agents/pharmacology , Animals , Antifungal Agents/administration & dosage , Biocompatible Materials/administration & dosage , Case-Control Studies , Cell Proliferation/drug effects , Cordyceps/chemistry , Cordyceps/metabolism , Deoxyadenosines/administration & dosage , Humans , Hypertension, Pulmonary/physiopathology , Lung/blood supply , Lung/physiopathology , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Oxidative Stress/drug effects , Placebos/administration & dosage , Vasodilator Agents/administration & dosage
12.
Poult Sci ; 99(2): 974-980, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32036989

ABSTRACT

The effect of orally administered hawthorn flavonoid extract (HFE) on growth, electrocardiographic waves, and cardiac parameters of pulmonary hypertensive chickens reared at high altitude (2,100 m above sea level) was examined. A total of 225 one-day-old, mixed broiler chicks (3 treatments with 5 replicates and 15 chicks per each, totally 75 birds/treatment) were assigned to 3 experimental groups: 0, 0.1, and 0.2 ml of HFE per 1 L of drinking water. Birds were administered the drinking water HFE treatments for 42 D. At an age of 28 and 42 D, electrocardiograms were undertaken and cardiac parameters such as the RV:TV, RV:BW, and TV:BW, and indicators of PHS on selected birds were measured. The final BW of chickens receiving the HFE at 0.2 ml/L was greater (2,579 ± 64 g) than that of birds receiving 0.1 ml/L (2,497 ± 62 g) and 0 ml/L (2,323 ± 57 g). Therefore, no supplemented group had a lower final BW than others (P < 0.05). Amplitudes of S and T waves in 0.1- and 0.2-ml/L HFE consumed groups at 28 and 42 D of age decreased compared with that in the control group (P < 0.05). The HFE reduced the heart weight and RV:TV, RV:BW, and TV:BW ratios when supplemented in drinking water at 0.1 and 0.2 mL/L compared with 0 mL/L (P < 0.05). In conclusion, supplementation of HFE in drinking water can reduce the PHS and incidence of cardiac disorders. Owing to the positive effect of HFE on cardiac parameters that mediated through flavonoids bioactive compounds, this product can be used to prevent complications of pulmonary hypertension and disarray of electrocardiographic waves in broiler chickens reared at high altitude.


Subject(s)
Chickens/physiology , Crataegus/chemistry , Electrocardiography/veterinary , Flavonoids/pharmacology , Hypertension, Pulmonary/veterinary , Plant Extracts/pharmacology , Poultry Diseases/physiopathology , Animal Feed/analysis , Animals , Chickens/growth & development , Diet/veterinary , Dietary Supplements/analysis , Dose-Response Relationship, Drug , Flavonoids/administration & dosage , Hypertension, Pulmonary/physiopathology , Phytochemicals/administration & dosage , Phytochemicals/pharmacology , Plant Extracts/administration & dosage , Random Allocation
13.
Intern Emerg Med ; 15(4): 573-585, 2020 06.
Article in English | MEDLINE | ID: mdl-32040829

ABSTRACT

Anaemia is a highly prevalent condition, which negatively impacts on patients' cardiovascular performance and quality of life. Anaemia is mainly caused by disturbances of iron homeostasis. While absolute iron deficiency mostly as a consequence of chronic blood loss or insufficient dietary iron absorption results in the emergence of iron deficiency anaemia, inflammation-driven iron retention in innate immune cells and blockade of iron absorption leads to the development of anaemia of chronic disease. Both, iron deficiency and anaemia have been linked to the clinical course of pulmonary hypertension. Various mechanistic links between iron homeostasis, anaemia, and pulmonary hypertension have been described and current treatment guidelines suggest regular iron status assessment and the implementation of iron supplementation strategies in these patients. The pathophysiology, diagnostic assessment as well as current and future treatment options concerning iron deficiency with or without anaemia in individuals suffering from pulmonary hypertension are discussed within this review.


Subject(s)
Anemia, Iron-Deficiency/diagnosis , Anemia, Iron-Deficiency/physiopathology , Hypertension, Pulmonary/physiopathology , Iron Deficiencies , Chronic Disease , Homeostasis , Humans
14.
BMJ Open ; 10(2): e028831, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32034015

ABSTRACT

INTRODUCTION: Management of inoperable chronic thromboembolic pulmonary hypertension (CTEPH) remains a clinical challenge. Currently, medical treatment involving pulmonary vasodilators (such as soluble guanylate-cyclase stimulators) is recommended, primarily for ameliorating symptoms. More recently, balloon pulmonary angioplasty (BPA) has been developed as alternative treatment for inoperable CTEPH. This study aimed to compare the efficacy and safety of BPA and riociguat (a soluble guanylate-cyclase stimulator) as treatments for inoperable CTEPH. METHODS AND ANALYSIS: This study is a multicentre randomised controlled trial. Subjects with inoperable CTEPH were randomised (1:1) into either a BPA or riociguat group, and observed for 12 months after initiation of treatment. The primary endpoint will be the change in mean pulmonary arterial pressure from baseline to 12 months after initiation of treatment. For primary analysis, we will estimate the least square means difference and 95% CI for the change of pulmonary arterial pressure between the groups at 12 months using the analysis of covariance adjusted for allocation factors. ETHICS AND DISSEMINATION: This study and its protocols were approved by the institutional review board of Keio University School of Medicine and each participating institution. Written informed consent was obtained from all participants. Results will be disseminated at medical conferences and in journal publications. TRIAL REGISTRATION NUMBER: University Hospital Medical Information Network Clinical Trial Registry (UMIN000019549); Pre-results.


Subject(s)
Angioplasty, Balloon/methods , Antihypertensive Agents/therapeutic use , Hypertension, Pulmonary/therapy , Pulmonary Embolism/therapy , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Adult , Combined Modality Therapy , Female , Humans , Hypertension, Pulmonary/physiopathology , Male , Middle Aged , Pulmonary Embolism/physiopathology , Quality of Life , Treatment Outcome
15.
Paediatr Respir Rev ; 34: 37-45, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31060947

ABSTRACT

This review summarizes current understanding of mitochondrial bioenergetic dysfunction applicable to mechanisms of lung diseases and outlines challenges and future directions in this rapidly emerging field. Although the role of mitochondria extends beyond the term of cellular "powerhouse", energy generation remains the most fundamental function of these organelles. It is not counterintuitive to propose that intact energy supply is important for favorable cellular fate following pulmonary insult. In this review, the discussion of mitochondrial dysfunction focuses on those molecular mechanisms that alter cellular bioenergetics in the lungs: (a) inhibition of mitochondrial respiratory chain, (b) mitochondrial leak and uncoupling, (c) alteration of mitochondrial Ca2+ handling, (d) mitochondrial production of reactive oxygen species and self-oxidation. The discussed lung diseases were selected according to their pathological nature and relevance to pediatrics: Acute lung injury (ALI), defined as acute parenchymal lung disease associated with cellular demise and inflammation (Acute Respiratory Distress Syndrome, ARDS, Pneumonia), alveolar developmental failure (Bronchopulmonary Dysplasia, BPD or chronic lung disease in premature infants), obstructive airway diseases (Bronchial asthma) and vascular remodeling affecting pulmonary circulation (Pulmonary Hypertension, PH). The analysis highlights primary mechanisms of mitochondrial bioenergetic dysfunction contributing to the disease-specific pulmonary insufficiency and proposes potential therapeutic targets.


Subject(s)
Energy Metabolism , Lung Diseases/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Acute Lung Injury/metabolism , Acute Lung Injury/physiopathology , Asthma/metabolism , Asthma/physiopathology , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/physiopathology , Calcium/metabolism , Cell Respiration , Electron Transport , Humans , Hyperoxia/metabolism , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Lung Diseases/physiopathology , Pneumonia/metabolism , Pneumonia/physiopathology , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/physiopathology , Vascular Remodeling
16.
Trials ; 20(1): 725, 2019 Dec 16.
Article in English | MEDLINE | ID: mdl-31842950

ABSTRACT

BACKGROUND: Tetramethylpyrazine (TMP), an active ingredient in the traditional Chinese herbal medicine Rhizoma Chuanxiong, has been used clinically for the prevention and treatment of cardiovascular disease. The benefits of TMP are largely attributed to its anti-oxidative and vasodilative properties. However, the efficacy of TMP in the treatment of pulmonary hypertension (PH) is unknown. We hypothesized that TMP may have a therapeutic effect in patients with PH. METHODS/DESIGN: A randomized, single-blinded, clinical study with a TMP treatment group and a control group will be conducted to evaluate the efficacy and safety of TMP intervention in patients with PH. The recruitment target is 120 subjects meeting the following criteria: (i) at rest and at sea level, mean pulmonary artery pressure above 20 mmHg and pulmonary capillary wedge pressure below 15 mmHg; (ii) type 1 or 4 PH in the stable phase; (iii) age 15-70 years; (iv) 6-min walk distance between 100 and 450 m; (v) World Health Organization (WHO) functional classification of pulmonary hypertension of II, III, or IV. Subjects will be assigned randomly into two groups at a ratio of 1:2 (control:TMP). Both groups will receive routine treatment, and the treatment group will also receive oral TMP (100 mg) three times a day for 16 weeks. All patients will be followed up for 4, 8, 12, and 16 weeks; symptoms and patient compliance will be recorded. DISCUSSION: We aimed to determine the efficacy and safety of TMP for the treatment of PH. TRIAL REGISTRATION: Chinese Clinical Trial Register, ChiCTR1800018664. Registered on 2 October 2018.


Subject(s)
Antihypertensive Agents/therapeutic use , Arterial Pressure/drug effects , Hypertension, Pulmonary/drug therapy , Pyrazines/therapeutic use , Vasodilator Agents/therapeutic use , Adolescent , Adult , Aged , Antihypertensive Agents/adverse effects , China , Exercise Tolerance/drug effects , Female , Humans , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/physiopathology , Male , Middle Aged , Pyrazines/adverse effects , Randomized Controlled Trials as Topic , Recovery of Function , Single-Blind Method , Time Factors , Treatment Outcome , Vasodilator Agents/adverse effects , Young Adult
17.
J Cardiovasc Pharmacol ; 74(5): 426-435, 2019 11.
Article in English | MEDLINE | ID: mdl-31725079

ABSTRACT

Chrysin (CH), the main ingredient of many medicinal plants, has been reported to be a very potent flavonoid possessing a large number of pharmacological activities. Recent studies have shown that CH significantly improves hemodynamic parameters such as right ventricular pressure, right ventricular hypertrophy, and pulmonary vascular remodeling in a rat model of chronic hypoxia-induced pulmonary hypertension (CHPH). These improvements are through the inhibition of NOX4 expression, reactive oxygen species and malondialdehyde production, pulmonary arterial smooth muscle cell (PASMC) proliferation, and collagen accumulation. In this study, we investigated another mechanism by which CH alleviates CHPH by regulating intracellular calcium concentrations ([Ca]i) in PASMCs, as well as the underlying signaling pathway. The results show that (1) in CHPH model rats, CH substantially attenuated elevated right ventricular pressure, right ventricular hypertrophy, and pulmonary vascular remodeling; (2) in cultured rat distal PASMCs, CH inhibited the hypoxia-triggered promotion of cell proliferation, store-operated Ca entry and [Ca]i; and (3) CH significantly suppressed the hypoxia-upregulated HIF-1α, BMP4, TRPC1, and TRPC6 expression in distal pulmonary arteries (PAs) and cultured rat distal PASMCs. These results indicate that CH likely exerts its CHPH protective activity by regulating [Ca]i, which may result from the downregulation of HIF-1α, BMP4, TRPC1, and TRPC in PASMCs.


Subject(s)
Antihypertensive Agents/pharmacology , Calcium Signaling/drug effects , Calcium/metabolism , Flavonoids/pharmacology , Hypertension, Pulmonary/prevention & control , Hypoxia/drug therapy , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Animals , Bone Morphogenetic Protein 4/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Hypoxia/complications , Hypoxia/metabolism , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Rats, Sprague-Dawley , TRPC Cation Channels/metabolism , Vascular Remodeling/drug effects , Ventricular Function, Right/drug effects , Ventricular Pressure/drug effects , Ventricular Remodeling/drug effects
18.
J. pediatr. (Rio J.) ; 95(5): 593-599, Sept.-Oct. 2019. tab, graf
Article in English | LILACS | ID: biblio-1040360

ABSTRACT

Abstract Objective: The purpose of this study was to illustrate the association between vascular endothelial growth factor level and pulmonary artery hypertension in children with β-thalassemia major. Method: This case-control study was conducted on 116 children with β-thalassemia major; 58 of them had pulmonary artery hypertension. They were compared to 58 healthy children who were age and sex-matched (control group). Serum levels of vascular endothelial growth factor and echocardiographic assessment were done for all children. Results: Vascular endothelial growth factor serum level was significantly higher in children with β-thalassemia major with pulmonary artery hypertension than in those without pulmonary artery hypertension, as well as in control groups (p < 0.001). Vascular endothelial growth factor serum level had a significant positive correlation with pulmonary artery pressure and serum ferritin, as well as a significant negative correlation with the duration of chelation therapy. Logistic regression analysis revealed that elevated vascular endothelial growth factor (Odd Ratio = 1.5; 95% Confidence Interval, 1.137-2.065; p = 0.005) was an independent risk factor of pulmonary artery hypertension in such children. Vascular endothelial growth factor serum level at a cutoff point of >169 pg/mL had 93.1% sensitivity and 93.1% specificity for the presence of pulmonary artery hypertension in children with β-thalassemia major. Conclusion: Elevated vascular endothelial growth factor serum level is associated with pulmonary artery hypertension in children with β-thalassemia.


Resumo: Objetivo: A finalidade deste estudo foi exemplificar a associação entre o nível de fator de crescimento endotelial vascular e a hipertensão arterial pulmonar em crianças com talassemia beta maior. Método: Este estudo caso-controle foi realizado em 116 crianças com talassemia beta maior; 58 das quais apresentaram hipertensão arterial pulmonar em comparação com 58 crianças saudáveis pareadas por idade e sexo (grupo de controle). Os níveis séricos do fator de crescimento endotelial vascular e a avaliação ecocardiográfica foram realizados em todas as crianças. Resultados: O nível sérico do fator de crescimento endotelial vascular foi significativamente maior em crianças com talassemia beta maior com hipertensão arterial pulmonar que as crianças sem hipertensão arterial pulmonar e os grupos de controle (p < 0,001). O nível sérico do fator de crescimento endotelial vascular apresentou uma correlação positiva significativa com a pressão arterial pulmonar e a ferritina sérica e correlação negativa significativa com a duração da terapia de quelação. A análise de regressão logística revelou que o fator de crescimento endotelial vascular elevado (RC = 1,5; IC de 95%: 1,137-2,065; p = 0,005) foi um fator de risco independente de hipertensão arterial pulmonar nessas crianças. O nível sérico do fator de crescimento endotelial vascular no ponto de corte > 169 (pg/mL) apresentou 93,1% de sensibilidade e 93,1% de especificidade na presença de hipertensão arterial pulmonar em crianças com talassemia beta maior. Conclusão: O nível sérico do fator de crescimento endotelial vascular elevado está associado à hipertensão arterial pulmonar em crianças com talassemia beta.


Subject(s)
Humans , Male , Female , Child , Adolescent , beta-Thalassemia/blood , Vascular Endothelial Growth Factor A/blood , Hypertension, Pulmonary/blood , Reference Values , Splenectomy , Time Factors , Echocardiography, Doppler , Case-Control Studies , Risk Factors , ROC Curve , Analysis of Variance , beta-Thalassemia/physiopathology , Age of Onset , Statistics, Nonparametric , Hypertension, Pulmonary/physiopathology
19.
Int J Cardiovasc Imaging ; 35(8): 1509-1524, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31049753

ABSTRACT

Multi-energy computed tomography (MECT) refers to acquisition of CT data at multiple energy levels (typically two levels) using different technologies such as dual-source, dual-layer and rapid tube voltage switching. In addition to conventional/routine diagnostic images, MECT provides additional image sets including iodine maps, virtual non-contrast images, and virtual monoenergetic images. These image sets provide tissue/material characterization beyond what is possible with conventional CT. MECT provides invaluable additional information in the evaluation of pulmonary vasculature, primarily by the assessment of pulmonary perfusion. This functional information provided by the MECT is complementary to the morphological information from a conventional CT angiography. In this article, we review the technique and applications of MECT in the evaluation of pulmonary vasculature.


Subject(s)
Computed Tomography Angiography/methods , Hemodynamics , Hypertension, Pulmonary/diagnostic imaging , Multidetector Computed Tomography/methods , Perfusion Imaging/methods , Pulmonary Artery/diagnostic imaging , Pulmonary Circulation , Pulmonary Embolism/diagnostic imaging , Humans , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/therapy , Predictive Value of Tests , Prognosis , Pulmonary Artery/physiopathology , Pulmonary Embolism/physiopathology , Pulmonary Embolism/therapy , Radiographic Image Interpretation, Computer-Assisted
20.
Br J Anaesth ; 122(4): 437-447, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30857600

ABSTRACT

BACKGROUND: A pulmonary hypertensive crisis (PHC) can be a life-threatening condition. We established a PHC model by exposing rats with monocrotaline (MCT)-induced pulmonary hypertension to acute hypoxia, and investigated the effects of vasopressin, phenylephrine, and norepinephrine on the PHC. METHODS: Four weeks after MCT 60 mg kg-1 administration i.v., right ventricular systolic pressure (RVSP), systolic BP (SBP), mean BP (MBP), cardiac index (CI), and pulmonary vascular resistance index (PVRI) were measured. PHC defined as an RVSP exceeding or equal to SBP was induced by changing the fraction of inspiratory oxygen to 0.1. Rats were subsequently treated by vasopressin, phenylephrine, or norepinephrine, followed by assessment of systemic haemodynamics, isometric tension of femoral and pulmonary arteries, cardiac function, blood gas composition, and survival. RESULTS: PHC was associated with increased RV dilatation and paradoxical septal motion. Vasopressin increased MBP [mean (standard error)] from 52.6 (3.8) to 125.0 (8.9) mm Hg and CI from 25.4 (2.3) to 40.6 (1.8) ml min-1 100 g-1 while decreasing PVRI. Vasopressin also improved RV dilatation, oxygenation, and survival in PHC. In contrast, phenylephrine increased MBP from 54.8 (2.3) to 96.8 (3.2) mm Hg without improving cardiac pump function. Norepinephrine did not alter MBP. Vasopressin contracted femoral but not pulmonary arteries, whereas phenylephrine contracted both arterial beds. Hence, improvements with vasopressin in PHC might be associated with decreased PVRI and selective systemic vasoconstriction. CONCLUSIONS: In this rat model of a PHC, vasopressin, but not phenylephrine or norepinephrine, resulted in better haemodynamic and vascular recovery.


Subject(s)
Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/etiology , Hypoxia/complications , Vasoconstrictor Agents/therapeutic use , Vasopressins/therapeutic use , Acute Disease , Animals , Drug Evaluation, Preclinical/methods , Echocardiography , Hemodynamics/drug effects , Hypertension, Pulmonary/diagnostic imaging , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Male , Monocrotaline , Nordefrin/pharmacology , Oxygen/blood , Partial Pressure , Phenylephrine/pharmacology , Rats, Sprague-Dawley , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasopressins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL