Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Arterioscler Thromb Vasc Biol ; 44(6): e172-e195, 2024 Jun.
Article En | MEDLINE | ID: mdl-38572649

BACKGROUND: Pulmonary hypertension (PH) is a progressive and life-threatening disease characterized by pulmonary vascular remodeling, which involves aberrant proliferation and apoptosis resistance of the pulmonary arterial smooth muscle cells (PASMCs), resembling the hallmark characteristics of cancer. In cancer, the HMGB2 (high-mobility group box 2) protein promotes the pro-proliferative/antiapoptotic phenotype. However, the function of HMGB2 in PH remains uninvestigated. METHODS: Smooth muscle cell (SMC)-specific HMGB2 knockout or HMGB2-OE (HMGB2 overexpression) mice and HMGB2 silenced rats were used to establish hypoxia+Su5416 (HySu)-induced PH mouse and monocrotaline-induced PH rat models, respectively. The effects of HMGB2 and its underlying mechanisms were subsequently elucidated using RNA-sequencing and cellular and molecular biology analyses. Serum HMGB2 levels were measured in the controls and patients with pulmonary arterial (PA) hypertension. RESULTS: HMGB2 expression was markedly increased in the PAs of patients with PA hypertension and PH rodent models and was predominantly localized in PASMCs. SMC-specific HMGB2 deficiency or silencing attenuated PH development and pulmonary vascular remodeling in hypoxia+Su5416-induced mice and monocrotaline-treated rats. SMC-specific HMGB2 overexpression aggravated hypoxia+Su5416-induced PH. HMGB2 knockdown inhibited PASMC proliferation in vitro in response to PDGF-BB (platelet-derived growth factor-BB). In contrast, HMGB2 protein stimulation caused the hyperproliferation of PASMCs. In addition, HMGB2 promoted PASMC proliferation and the development of PH by RAGE (receptor for advanced glycation end products)/FAK (focal adhesion kinase)-mediated Hippo/YAP (yes-associated protein) signaling suppression. Serum HMGB2 levels were significantly increased in patients with PA hypertension, and they correlated with disease severity, predicting worse survival. CONCLUSIONS: Our findings indicate that targeting HMGB2 might be a novel therapeutic strategy for treating PH. Serum HMGB2 levels could serve as a novel biomarker for diagnosing PA hypertension and determining its prognosis.


Disease Models, Animal , HMGB2 Protein , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Pulmonary Artery , Vascular Remodeling , Animals , HMGB2 Protein/genetics , HMGB2 Protein/metabolism , Humans , Male , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Pulmonary Artery/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Rats , Mice , Cell Proliferation , Severity of Illness Index , Signal Transduction , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/physiopathology , Rats, Sprague-Dawley , Female , Cells, Cultured , Middle Aged , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology
2.
Acta Diabetol ; 61(5): 643-655, 2024 May.
Article En | MEDLINE | ID: mdl-38383671

AIMS: Inflammation is central to the pathogenesis of metabolic syndrome (MetS). Leukocyte cell-derived chemotaxin 2 (LECT2) is constitutively secreted in response to inflammatory stimuli and oxidative stress contributing to tissue or systemic inflammation. We explored the relationship between LECT2 levels and MetS severity in humans and mice. METHODS: Serum LECT2 levels were measured in 210 participants with MetS and 114 without MetS (non-MetS). LECT2 expression in the liver and adipose tissue was also examined in mice fed a high-fat diet (HFD) and genetically obese (ob/ob) mice. RESULTS: Serum LECT2 levels were significantly higher in MetS participants than in non-MetS participants (7.47[3.36-17.14] vs. 3.74[2.61-5.82], P < 0.001). Particularly, serum LECT2 levels were significantly elevated in participants with hypertension, central obesity, diabetes mellitus (DM), hyperglycaemia, elevated triglyceride (TG) levels, and reduced high-density lipoprotein cholesterol (HDL-C) levels compared to those in participants without these conditions. Pearson's correlation analysis showed that serum LECT2 levels were positively associated with conventional risk factors in all patients. Moreover, LECT2 was positively associated with the number of MetS components (r = 0.355, P < 0.001), indicating that higher serum LECT2 levels reflected MetS severity. Multivariate regression analysis revealed that a one standard deviation increase in LECT2 was associated with an odds ratio of 1.52 (1.01-2.29, P = 0.044) for MetS prevalence after adjusting for age, sex, body mass index, waist circumference, smoking status, white blood cell count, fasting blood glucose, TG, total cholesterol, HDL-C, blood urea nitrogen, and alanine aminotransferase. Receiver operating characteristic curve analysis confirmed the strong predictive ability of serum LECT2 levels for MetS. The optimum serum LECT2 cut-off value was 9.05. The area under the curve was 0.73 (95% confidence interval 0.68-0.78, P < 0.001), with a sensitivity and specificity of 45.71% and 95.61%, respectively. Additionally, LECT2 expression levels were higher at baseline and dramatically enhanced in metabolic organs (e.g. the liver) and adipose tissue in HFD-induced obese mice and ob/ob mice. CONCLUSIONS: Increased LECT2 levels were significantly and independently associated with the presence and severity of MetS, indicating that LECT2 could be used as a novel biomarker and clinical predictor of MetS.


Intercellular Signaling Peptides and Proteins , Metabolic Syndrome , Adult , Animals , Female , Humans , Male , Mice , Diet, High-Fat , Intercellular Signaling Peptides and Proteins/blood , Metabolic Syndrome/blood , Metabolic Syndrome/diagnosis , Metabolic Syndrome/metabolism , Mice, Inbred C57BL , Mice, Obese , Prevalence
3.
Circ Res ; 134(1): 9-29, 2024 01 05.
Article En | MEDLINE | ID: mdl-38047378

BACKGROUND: T cells are central to the immune responses contributing to hypertension. LGMN (legumain) is highly expressed in T cells; however, its role in the pathogenesis of hypertension remains unclear. METHODS: Peripheral blood samples were collected from patients with hypertension, and cluster of differentiation (CD)4+ T cells were sorted for gene expression and Western blotting analysis. TLGMNKO (T cell-specific LGMN-knockout) mice (Lgmnf/f/CD4Cre), regulatory T cell (Treg)-specific LGMN-knockout mice (Lgmnf/f/Foxp3YFP Cre), and RR-11a (LGMN inhibitor)-treated C57BL/6 mice were infused with Ang II (angiotensin II) or deoxycorticosterone acetate/salt to establish hypertensive animal models. Flow cytometry, 4-dimensional label-free proteomics, coimmunoprecipitation, Treg suppression, and in vivo Treg depletion or adoptive transfer were used to delineate the functional importance of T-cell LGMN in hypertension development. RESULTS: LGMN mRNA expression was increased in CD4+ T cells isolated from hypertensive patients and mice, was positively correlated with both systolic and diastolic blood pressure, and was negatively correlated with serum IL (interleukin)-10 levels. TLGMNKO mice exhibited reduced Ang II-induced or deoxycorticosterone acetate/salt-induced hypertension and target organ damage relative to wild-type (WT) mice. Genetic and pharmacological inhibition of LGMN blocked Ang II-induced or deoxycorticosterone acetate/salt-induced immunoinhibitory Treg reduction in the kidneys and blood. Anti-CD25 antibody depletion of Tregs abolished the protective effects against Ang II-induced hypertension in TLGMNKO mice, and LGMN deletion in Tregs prevented Ang II-induced hypertension in mice. Mechanistically, endogenous LGMN impaired Treg differentiation and function by directly interacting with and facilitating the degradation of TRAF6 (tumor necrosis factor receptor-associated factor 6) via chaperone-mediated autophagy, thereby inhibiting NF-κB (nuclear factor kappa B) activation. Adoptive transfer of LGMN-deficient Tregs reversed Ang II-induced hypertension, whereas depletion of TRAF6 in LGMN-deficient Tregs blocked the protective effects. CONCLUSIONS: LGMN deficiency in T cells prevents hypertension and its complications by promoting Treg differentiation and function. Specifically targeting LGMN in Tregs may be an innovative approach for hypertension treatment.


Hypertension , TNF Receptor-Associated Factor 6 , Animals , Humans , Mice , Acetates/adverse effects , Acetates/metabolism , Angiotensin II/toxicity , Angiotensin II/metabolism , CD4-Positive T-Lymphocytes/metabolism , Desoxycorticosterone/adverse effects , Desoxycorticosterone/metabolism , Hypertension/chemically induced , Hypertension/genetics , Hypertension/prevention & control , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory , TNF Receptor-Associated Factor 6/metabolism
4.
Respir Res ; 24(1): 263, 2023 Nov 01.
Article En | MEDLINE | ID: mdl-37915044

BACKGROUND: Pulmonary arterial hypertension (PAH), Group 1 pulmonary hypertension (PH), is a type of pulmonary vascular disease characterized by abnormal contraction and remodeling of the pulmonary arterioles, manifested by pulmonary vascular resistance (PVR) and increased pulmonary arterial pressure, eventually leading to right heart failure or even death. The mechanisms involved in this process include inflammation, vascular matrix remodeling, endothelial cell apoptosis and proliferation, vasoconstriction, vascular smooth muscle cell proliferation and hypertrophy. In this study, we review the mechanisms of action of prostaglandins and their receptors in PAH. MAIN BODY: PAH-targeted therapies, such as endothelin receptor antagonists, phosphodiesterase type 5 inhibitors, activators of soluble guanylate cyclase, prostacyclin, and prostacyclin analogs, improve PVR, mean pulmonary arterial pressure, and the six-minute walk distance, cardiac output and exercise capacity and are licensed for patients with PAH; however, they have not been shown to reduce mortality. Current treatments for PAH primarily focus on inhibiting excessive pulmonary vasoconstriction, however, vascular remodeling is recalcitrant to currently available therapies. Lung transplantation remains the definitive treatment for patients with PAH. Therefore, it is imperative to identify novel targets for improving pulmonary vascular remodeling in PAH. Studies have confirmed that prostaglandins and their receptors play important roles in the occurrence and development of PAH through vasoconstriction, vascular smooth muscle cell proliferation and migration, inflammation, and extracellular matrix remodeling. CONCLUSION: Prostacyclin and related drugs have been used in the clinical treatment of PAH. Other prostaglandins also have the potential to treat PAH. This review provides ideas for the treatment of PAH and the discovery of new drug targets.


Prostaglandins , Pulmonary Arterial Hypertension , Humans , Pulmonary Arterial Hypertension/drug therapy , Receptors, Prostaglandin , Vascular Remodeling , Familial Primary Pulmonary Hypertension , Epoprostenol/therapeutic use , Prostaglandins I , Inflammation/drug therapy , Pulmonary Artery
5.
Biomed Pharmacother ; 165: 115118, 2023 Sep.
Article En | MEDLINE | ID: mdl-37437373

Cardiovascular disease (CVD) is the most fatal disease that causes sudden death, and inflammation contributes substantially to its occurrence and progression. The prevalence of CVD increases as the population ages, and the pathophysiology is complex. Anti-inflammatory and immunological modulation are the potential methods for CVD prevention and treatment. High-Mobility Group (HMG) chromosomal proteins are one of the most abundant nuclear nonhistone proteins which act as inflammatory mediators in DNA replication, transcription, and repair by producing cytokines and serving as damage-associated molecular patterns in inflammatory responses. The most common and well-studied HMG proteins are those with an HMGB domain, which participate in a variety of biological processes. HMGB1 and HMGB2 were the first members of the HMGB family to be identified and are present in all investigated eukaryotes. Our review is primarily concerned with the involvement of HMGB1 and HMGB2 in CVD. The purpose of this review is to provide a theoretical framework for diagnosing and treating CVD by discussing the structure and function of HMGB1 and HMGB2.


Cardiovascular Diseases , HMGB1 Protein , Humans , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , HMGB2 Protein/genetics , HMGB2 Protein/metabolism , HMGB Proteins/chemistry , HMGB Proteins/metabolism , Biomarkers
6.
Int Immunopharmacol ; 119: 110198, 2023 Jun.
Article En | MEDLINE | ID: mdl-37087872

The contractile-syntheticphenotypicconversion of vascular smooth muscle cells (VSMCs) plays a key role in atherosclerosis, vascular restenosis, and hypertension. Our previous study explored the correlation between high mobility group box protein (HMGB) 1 and HMGB2 and neointimal hyperplasia after vascular injury. In the present study, we explore whether inflachromene (ICM), a novel inhibitor of the expression of both HMGB1 and HMGB2, modulates phenotypic changes in VSMCs and the mechanisms involved. Mice treated with ICM after carotid artery wire injury showed a decrease in excessive neointimal hyperplasia compared with that in the vehicle groups. In cultured VSMCs, pretreatment with ICM suppressed the angiotensin II (Ang II)-induced phenotypic conversion, proliferation, and migration. We discovered that ICM reduced the Ang II-induced upregulation of the expression of HMGB1 and HMGB2 and inhibited their shuttling between the nucleus and the cytosol. Mechanistically, Ang II-treated VSMCs exhibited higher levels of Toll-like receptor 4 (TLR4) and nuclear factor-κB (NF-κB) phosphorylation, which were attenuated by ICM. In addition, the NF-κB inhibitor Bay-117082 abolished the recombinant HMGB1-mediated VSMC phenotypic conversion, proliferation, and migration. Furthermore, ICM ameliorated the Ang II-induced increases in NAD[P]H oxidase expression, thereby attenuating the Ang II-induced proliferation and migration. These results reveal that ICM pretreatment attenuates Ang II-induced VSMC dedifferentiation, proliferation, and migration may by regulating the TLR4-NF-kB pathway. Thus, ICM is a potential therapy and preventive treatment for vascular proliferative diseases.


HMGB1 Protein , NF-kappa B , Mice , Animals , NF-kappa B/metabolism , Hyperplasia/metabolism , Toll-Like Receptor 4/metabolism , Cell Proliferation , HMGB1 Protein/metabolism , HMGB2 Protein/metabolism , Angiotensin II/metabolism , Cells, Cultured , Myocytes, Smooth Muscle/metabolism
7.
J Adv Res ; 41: 205-218, 2022 11.
Article En | MEDLINE | ID: mdl-36328749

INTRODUCTION: Vascular smooth muscle cell (VSMC) senescence in the vasculature results in vascular aging as well as age-related diseases, while metformin improves the inflamm-aging profile by enhancing autophagy. However, metformin's impact on VSMC senescence is largely undefined. OBJECTIVES: To test the hypothesis that metformin exerts an anti-senescence role by restoring autophagic activity in VSMCs and vascular tissues. METHODS: Animal models established by angiotensin II (Ang II) induction and physiological aging and senescent primary VSMCs from the aortas of elderly patients were treated with metformin. Cellular and vascular senescence were assessed by measuring the amounts of senescence-associated ß-galactosidase and senescence markers, including p21 and p53. Autophagy levels were assessed by autophagy-related protein expression, transmission electron microscope, and autolysosome staining. In order to explore the underlying mechanism of the anti-senescence effects of metformin, 4D label-free quantitative proteomics and bioinformatic analyses were conducted, with subsequent experiments validating these findings. RESULTS: Ang II-dependent senescence was suppressed by metformin in VSMCs and vascular tissues. Metformin also significantly improved arterial stiffness and alleviated structural changes in aged arteries, reduced senescence-associated secretory phenotype (SASP), and improved proliferation and migration of senescent VSMCs. Mechanistically, the proteomic analysis indicated that autophagy might contribute to metformin's anti-senescence effects. Reduced autophagic flux was observed in Ang II-induced cellular and vascular senescence; this reduction was reversed by metformin. Specifically, metformin enhanced the autophagic flux at the autophagosome-lysosome fusion level, whereas blockade of autophagosome-lysosome fusion inhibited the anti-senescence effects of metformin. CONCLUSIONS: Metformin prevents VSMC and vascular senescence by promoting autolysosome formation.


Metformin , Muscle, Smooth, Vascular , Animals , Muscle, Smooth, Vascular/metabolism , Cellular Senescence/physiology , Metformin/pharmacology , Metformin/metabolism , Proteomics , Tumor Suppressor Protein p53/metabolism , Oxidative Stress , Angiotensin II/metabolism , Angiotensin II/pharmacology , Autophagy
8.
Cell Death Dis ; 11(11): 1014, 2020 11 26.
Article En | MEDLINE | ID: mdl-33243972

The prognostic impact of extracellular matrix (ECM) modulation and its regulatory mechanism post-acute myocardial infarction (AMI), require further clarification. Herein, we explore the predictive role of legumain-which showed the ability in ECM degradation-in an AMI patient cohort and investigate the underlying mechanisms. A total of 212 AMI patients and 323 healthy controls were enrolled in the study. Moreover, AMI was induced in mice by permanent ligation of the left anterior descending artery and fibroblasts were adopted for mechanism analysis. Based on the cut-off value for the receiver-operating characteristics curve, AMI patients were stratified into low (n = 168) and high (n = 44) plasma legumain concentration (PLG) groups. However, PLG was significantly higher in AMI patients than that in the healthy controls (median 5.9 µg/L [interquartile range: 4.2-9.3 µg/L] vs. median 4.4 µg/L [interquartile range: 3.2-6.1 µg/L], P < 0.001). All-cause mortality was significantly higher in the high PLG group compared to that in the low PLG group (median follow-up period, 39.2 months; 31.8% vs. 12.5%; P = 0.002). Multivariate Cox regression analysis showed that high PLG was associated with increased all-cause mortality after adjusting for clinical confounders (HR = 3.1, 95% confidence interval (CI) = 1.4-7.0, P = 0.005). In accordance with the clinical observations, legumain concentration was also increased in peripheral blood, and infarcted cardiac tissue from experimental AMI mice. Pharmacological blockade of legumain with RR-11a, improved cardiac function, decreased cardiac rupture rate, and attenuated left chamber dilation and wall thinning post-AMI. Hence, plasma legumain concentration is of prognostic value in AMI patients. Moreover, legumain aggravates cardiac remodelling through promoting ECM degradation which occurs, at least partially, via activation of the MMP-2 pathway.


Cysteine Endopeptidases/adverse effects , Echocardiography/methods , Myocardial Infarction/chemically induced , Acute Disease , Animals , Humans , Male , Mice , Middle Aged , Myocardial Infarction/mortality , Prognosis , Risk Factors
9.
Cardiovasc Ther ; 2020: 9059562, 2020.
Article En | MEDLINE | ID: mdl-32874203

BACKGROUND: Information regarding the impact of cardiovascular (CV) conditions on disease progression among patients with mild coronavirus disease 2019 (COVID-19) is limited. METHODS: This study evaluated the association of underlying CV conditions with disease progression in patients with mild COVID-19. The primary outcome was the need to be transferred to the designated hospital for intensive care due to COVID-19 disease progression. The patients were divided into with and without CV conditions as well as stable and intensive care groups. RESULTS: Of the 332 patients with mild COVID-19, the median age was 51 years (IQR, 40-59 years), and 200 (61.2%) were female. Of the 48 (14.5%) patients with CV conditions, 23 (47.9%) progressed to severe disease status and required intensive care. Compared with patients without CV conditions, patients with CV conditions were older and more likely to have fatigue, chest tightness, and myalgia. The rate of requiring intensive care was significantly higher among patients with CV conditions than in patients without CV conditions (47.92% vs. 12.4%; P < 0.001). In subgroup analysis, the rate of requiring intensive care was also higher among patients with either hypertension or coronary heart disease (CHD) than in patients without hypertension or CHD. The multivariable regression model showed that CV condition served as an independent risk factor for intensive care (odds ratio (OR), 2.652 (95% CI, 1.019-6.899)) after adjustment for various cofounders. CONCLUSIONS: Patients with mild COVID-19 complicating CV conditions are susceptible to develop severe disease status and requirement for intensive care.


Betacoronavirus , Cardiovascular Diseases/complications , Coronavirus Infections/complications , Critical Care , Pneumonia, Viral/complications , Adult , COVID-19 , Disease Progression , Female , Humans , Male , Middle Aged , Pandemics , SARS-CoV-2
10.
Circ Res ; 127(10): 1323-1336, 2020 10 23.
Article En | MEDLINE | ID: mdl-32912104

RATIONALE: Pulmonary arterial hypertension (PAH) is characterized by progressive pulmonary vascular remodeling, accompanied by varying degrees of perivascular inflammation. Niacin, a commonly used lipid-lowering drug, possesses vasodilating and proresolution effects by promoting the release of prostaglandin D2 (PGD2). However, whether or not niacin confers protection against PAH pathogenesis is still unknown. OBJECTIVE: This study aimed to determine whether or not niacin attenuates the development of PAH and, if so, to elucidate the molecular mechanisms underlying its effects. METHODS AND RESULTS: Vascular endothelial growth factor receptor inhibitor SU5416 and hypoxic exposure were used to induce pulmonary hypertension (PH) in rodents. We found that niacin attenuated the development of this hypoxia/SU5416-induced PH in mice and suppressed progression of monocrotaline-induced and hypoxia/SU5416-induced PH in rats through the reduction of pulmonary artery remodeling. Niacin boosted PGD2 generation in lung tissue, mainly through H-PGDS (hematopoietic PGD2 synthases). Deletion of H-PGDS, but not lipocalin-type PGDS, exacerbated the hypoxia/SU5416-induced PH in mice and abolished the protective effects of niacin against PAH. Moreover, H-PGDS was expressed dominantly in infiltrated macrophages in lungs of PH mice and patients with idiopathic PAH. Macrophage-specific deletion of H-PGDS markedly decreased PGD2 generation in lungs, aggravated hypoxia/SU5416-induced PH in mice, and attenuated the therapeutic effect of niacin on PAH. CONCLUSIONS: Niacin treatment ameliorates the progression of PAH through the suppression of vascular remodeling by stimulating H-PGDS-derived PGD2 release from macrophages.


Antihypertensive Agents/pharmacology , Hypertension, Pulmonary/drug therapy , Hypolipidemic Agents/pharmacology , Macrophages/drug effects , Niacin/pharmacology , Animals , Antihypertensive Agents/therapeutic use , Cells, Cultured , Humans , Hypertension, Pulmonary/metabolism , Hypolipidemic Agents/therapeutic use , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Lung/drug effects , Lung/metabolism , Macrophages/metabolism , Mice , Niacin/therapeutic use , Prostaglandin D2/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Rats
11.
J Mol Cell Cardiol ; 139: 1-13, 2020 02.
Article En | MEDLINE | ID: mdl-31958462

OBJECTIVE: Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) are related to in-stent-restenosis (ISR) following percutaneous coronary intervention (PCI). Osteoprotegerin (OPG) has been implicated in various vascular diseases. However, the effects of OPG on ISR and the underlying mechanism remained elusive. We here investigated the association between OPG and ISR, and to demonstrate the role and potential mechanisms of OPG in neointimal hyperplasia. APPROACH AND RESULTS: From 2962 patients who received coronary angiography and follow-up coronary angiography at approximately one year, 291 patients were diagnosed with ISR, and another 291 gender- and age- matched patients without ISR were selected as controls. Serum OPG levels were significantly increased in patients with ISR. Multivariable logistic regression analysis indicated that OPG level was independently associated with the increased risk of ISR. In a mouse femoral artery wire injury model, upregulated OPG was evidenced in vascular tissue after injury. OPG deletion attenuated the vascular injury-induced neointimal hyperplasia and related gene expression in mice. OPG promoted neointimal hyperplasia and human aortic smooth muscle cell (hASMC) proliferation and migration through activation of yes-associated protein (YAP), a major downstream effector of the Hippo signaling pathway, whereas knockdown or inhibition of YAP in hASMCs blunted OPG-induced above effects. Moreover, we found that OPG, as a ligand for integrin αVß3, mediated phosphorylation of focal adhesion kinase (FAK) and actin cytoskeleton reorganization, resulting in YAP dephosphorylation in hASMCs. OPG-dependent YAP and VSMC activation was prevented by treatment with αVß3-blocking antibodies and inhibitors of FAK and actin stress fibers. CONCLUSIONS: Increased serum OPG levels are associated with increased risk of ISR following PCI and OPG could promote neointimal hyperplasia in response to injury through integrin αVß3 mediated FAK and YAP activation, indicating OPG/YAP inhibition might serve as an attractive novel target for the prevention of ISR after PCI.


Coronary Restenosis/complications , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrin alphaVbeta3/metabolism , Neointima/complications , Neointima/pathology , Osteoprotegerin/metabolism , Signal Transduction , Stents/adverse effects , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Aged , Animals , Cell Movement , Cell Proliferation , Coronary Restenosis/blood , Disease Progression , Female , Femoral Artery/metabolism , Femoral Artery/pathology , Humans , Hyperplasia , Incidence , Logistic Models , Male , Mice, Inbred C57BL , Multivariate Analysis , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Neointima/blood , Osteoprotegerin/blood , Osteoprotegerin/deficiency , Phosphorylation/drug effects , Severity of Illness Index , Up-Regulation , Verteporfin/pharmacology
12.
Am J Respir Crit Care Med ; 201(10): 1263-1276, 2020 05 15.
Article En | MEDLINE | ID: mdl-31917615

Rationale: Vascular remodeling, including smooth muscle cell hypertrophy and proliferation, is the key pathological feature of pulmonary arterial hypertension (PAH). Prostaglandin I2 analogs (beraprost, iloprost, and treprostinil) are effective in the treatment of PAH. Of note, the clinically favorable effects of treprostinil in severe PAH may be attributable to concomitant activation of DP1 (D prostanoid receptor subtype 1).Objectives: To study the role of DP1 in the progression of PAH and its underlying mechanism.Methods: DP1 levels were examined in pulmonary arteries of patients and animals with PAH. Multiple genetic and pharmacologic approaches were used to investigate DP1-mediated signaling in PAH.Measurements and Main Results: DP1 expression was downregulated in hypoxia-treated pulmonary artery smooth muscle cells and in pulmonary arteries from rodent PAH models and patients with idiopathic PAH. DP1 deletion exacerbated pulmonary artery remodeling in hypoxia-induced PAH, whereas pharmacological activation or forced expression of the DP1 receptor had the opposite effect in different rodent models. DP1 deficiency promoted pulmonary artery smooth muscle cell hypertrophy and proliferation in response to hypoxia via induction of mTORC1 (mammalian target of rapamycin complex 1) activity. Rapamycin, an inhibitor of mTORC1, alleviated the hypoxia-induced exacerbation of PAH in DP1-knockout mice. DP1 activation facilitated raptor dissociation from mTORC1 and suppressed mTORC1 activity through PKA (protein kinase A)-dependent phosphorylation of raptor at Ser791. Moreover, treprostinil treatment blocked the progression of hypoxia-induced PAH in mice in part by targeting the DP1 receptor.Conclusions: DP1 activation attenuates hypoxia-induced pulmonary artery remodeling and PAH through PKA-mediated dissociation of raptor from mTORC1. These results suggest that the DP1 receptor may serve as a therapeutic target for the management of PAH.


Hypoxia/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Pulmonary Arterial Hypertension/genetics , Receptors, Immunologic/genetics , Receptors, Prostaglandin/genetics , Vascular Remodeling/genetics , Animals , Antihypertensive Agents/pharmacology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Down-Regulation , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Humans , Hypertrophy , Immunosuppressive Agents/pharmacology , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mice , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/metabolism , Pulmonary Artery , RNA, Messenger/metabolism , Rats , Sirolimus/pharmacology
13.
J Cardiovasc Pharmacol ; 74(4): 306-307, 2019 10.
Article En | MEDLINE | ID: mdl-31356557

Despite considerable progress in the field of heart failure about drugs and device therapy, the mortality rate of patients with heart failure remains high. Studies have shown that thromboembolism and stroke are associated with high mortality in patients with heart failure. Although warfarin therapy reduces the rate of ischemic stroke in patients with heart failure, the overall benefit from warfarin in this population seems to be offset by the increased bleeding risk. Thus, whether patients with chronic heart failure might benefit from anticoagulation, especially in patients with sinus rhythm, is still controversial. Rivaroxaban, a new oral anticoagulant, is a selective direct factor Xa inhibitor that is used to reduce thrombin generation, which may bring hope to anticoagulation in patients with heart failure. However, the COMPASS trial and recently published COMMANDER HF trial presented different results. By carefully analyzing 2 clinical trials, we think several factors might explain this different outcome.


Factor Xa Inhibitors/therapeutic use , Heart Failure/drug therapy , Rivaroxaban/therapeutic use , Stroke/prevention & control , Thromboembolism/prevention & control , Chronic Disease , Clinical Decision-Making , Evidence-Based Medicine , Factor Xa Inhibitors/adverse effects , Heart Failure/diagnosis , Heart Failure/mortality , Heart Failure/physiopathology , Hemorrhage/chemically induced , Humans , Patient Selection , Randomized Controlled Trials as Topic , Risk Assessment , Risk Factors , Rivaroxaban/adverse effects , Stroke/diagnosis , Stroke/mortality , Stroke/physiopathology , Thromboembolism/diagnosis , Thromboembolism/mortality , Thromboembolism/physiopathology , Treatment Outcome
14.
J Vis Exp ; (147)2019 05 04.
Article En | MEDLINE | ID: mdl-31107460

The mouse is the foremost mammalian model for studying human disease and human health. However, blood sample collection from mice is challenging in research work. Tail blood collection is a popular method when a small amount of blood sample is needed. Orbital artery could be considered if a large amount of blood is needed but this blood collection method has ethical issues. Formerly, we demonstrated the feasibility and safety of blood sample collection through subclavian vein puncture in rats, and here we investigate whether this method could be used in mice. We report that this method is safe and practical for blood collection in mice. Blood collection through the subclavian vein puncture in mice can be a convenient method in daily research works.


Phlebotomy/methods , Punctures , Subclavian Vein , Animals , Female , Male , Mice
15.
Arterioscler Thromb Vasc Biol ; 39(4): e130-e145, 2019 04.
Article En | MEDLINE | ID: mdl-30676070

Objective- Macrophages participate in the pathogenesis of pulmonary arterial hypertension (PAH). Lgmn (Legumain), a newly discovered cysteine proteinase belonging to the C13 peptidase family, is primarily expressed in macrophages; however, its roles in PAH remain unknown. Approach and Results- Herein, Lgmn was upregulated in lung tissues of PAH mice subjected to hypoxia plus SU5416 and PAH rats challenged with monocrotaline. Global Lgmn ablation and macrophage-specific ablation alleviated PAH compared with wild-type mice, evident from a reduction in right ventricular systolic pressure, the ratio of the right ventricular wall to the left ventricular wall plus the septum, the pulmonary vascular media thickness, and pulmonary vascular muscularization. Increased expression of ECM (extracellular matrix) proteins was correlated with MMP (matrix metalloproteinase)-2 activation and TGF (transforming growth factor)-ß1 signaling in the PAs. Although Lgmn did not affect inflammatory cell infiltration and PA smooth muscle cell proliferation, it drove increased the synthesis of ECM proteins via MMP-2 activation. MMP-2 hydrolyzed the TGF-ß1 precursor to the active form. An Lgmn-specific inhibitor markedly ameliorated PAH. Clinically, serum Lgmn levels were closely associated with the severity of idiopathic PAH. Conclusions- Our results indicate that Lgmn inhibition could be an effective strategy for preventing or delaying PAH.


Cysteine Endopeptidases/physiology , Hypertension, Pulmonary/enzymology , Macrophages/enzymology , Matrix Metalloproteinase 2/physiology , Transforming Growth Factor beta1/physiology , Animals , Caspase Inhibitors/pharmacology , Cysteine Endopeptidases/deficiency , Extracellular Matrix Proteins/metabolism , Female , Follow-Up Studies , Humans , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/prevention & control , Hypoxia/enzymology , Indoles/toxicity , Inflammation , Lung/metabolism , Male , Mice , Middle Aged , Monocrotaline/toxicity , Pyrroles/toxicity , Rats , Severity of Illness Index , Signal Transduction , Vascular Remodeling/physiology
16.
Cardiovasc Res ; 113(6): 586-597, 2017 May 01.
Article En | MEDLINE | ID: mdl-28407046

AIMS: Extracellular matrix (ECM) proteins accumulation contributes to the progression of pulmonary arterial hypertension (PAH), a rare and fatal cardiovascular condition defined by high pulmonary arterial pressure, whether primary, idiopathic, or secondary to other causes. The receptor for advanced glycation end products (RAGE) is constitutively expressed in the lungs and plays an important role in ECM deposition. Nonetheless, the mechanisms by which RAGE mediates ECM deposition/formation in pulmonary arteries and its roles in PAH progression remain unclear. METHODS AND RESULTS: Expression of RAGE and its activating ligands, S100/calgranulins and high mobility group box 1 (HMGB1), were increased in both human and mouse pulmonary arterial smooth muscle cells (PASMCs) under hypoxic conditions and were also strikingly upregulated in pulmonary arteries in hypoxia plus SU5416 (HySu)-induced PAH in mice. RAGE deletion alleviated pulmonary arterial pressure and restrained extracellular matrix accumulation in pulmonary arteries in HySu-induced PAH murine model. Moreover, blocking RAGE activity with a neutralizing antibody in human PASMCs, or RAGE deficiency in mouse PASMCs exposed to hypoxia, suppressed the expression of fibrotic proteins by reducing TGF-ß1 expression. RAGE reconstitution in deficient mouse PASMCs restored hypoxia-stimulated TGF-ß1 production via ERK1/2 and p38 MAPK pathway activation and subsequently increased ECM protein expression. Interestingly, HMGB1 acting on RAGE, not toll-like receptor 4 (TLR4), induced ECM deposition in PASMCs. Finally, in both idiopathic PAH patients and HySu-induced PAH mice, soluble RAGE (sRAGE) levels in serum were significantly elevated compared to those in controls. CONCLUSIONS: Activation of RAGE facilitates the development of hypoxia-induced pulmonary hypertension by increase of ECM deposition in pulmonary arteries. Our results indicate that sRAGE may be a potential biomarker for PAH diagnosis and disease severity, and that RAGE may be a promising target for PAH treatment.


Extracellular Matrix Proteins/metabolism , Hypertension, Pulmonary/chemically induced , Indoles , Pulmonary Artery/metabolism , Pyrroles , Receptor for Advanced Glycation End Products/metabolism , Vascular Remodeling , Animals , Case-Control Studies , Cell Hypoxia , Cells, Cultured , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Ligands , Male , Middle Aged , Phosphorylation , Pulmonary Artery/pathology , RNA Interference , Receptor for Advanced Glycation End Products/genetics , S100 Calcium Binding Protein beta Subunit/genetics , S100 Calcium Binding Protein beta Subunit/metabolism , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Circ Cardiovasc Genet ; 10(1)2017 Feb.
Article En | MEDLINE | ID: mdl-28077433

BACKGROUND: Pulmonary arterial remodeling characterized by increased vascular smooth muscle proliferation is commonly seen in life-threatening disease, pulmonary arterial hypertension (PAH). Clinical studies have suggested a correlation between osteoprotegerin serum levels and PAH severity. Here, we aimed to invhestigate vascular osteoprotegerin expression and its effects on pulmonary arterial smooth muscle cell proliferation in vitro and in vivo, as well as examine the signal transduction pathways mediating its activity. METHODS AND RESULTS: Serum osteoprotegerin levels were significantly elevated in patients with PAH and correlated with disease severity as determined by the World Health Organization (WHO) functional classifications and 6-minute walking distance tests. Similarly, increased osteoprotegerin expression was observed in the pulmonary arteries of hypoxia plus SU5416- and monocrotaline-induced PAH animal models. Moreover, osteoprotegerin disruption attenuated hypoxia plus SU5416-induced PAH progression by reducing pulmonary vascular remodeling, whereas lentiviral osteoprotegerin reconstitution exacerbated PAH by increasing pulmonary arterial smooth muscle cell proliferation. Furthermore, pathway analysis revealed that osteoprotegerin induced pulmonary arterial smooth muscle cell proliferation by interacting with integrin αvß3 to elicit downstream focal adhesion kinase and AKT pathway activation. CONCLUSIONS: Osteoprotegerin facilitates PAH pathogenesis by regulating pulmonary arterial smooth muscle cell proliferation, suggesting that it may be a potential biomarker and therapeutic target in this disease.


Arterial Pressure , Focal Adhesion Kinase 1/metabolism , Hypertension, Pulmonary/prevention & control , Hypoxia/complications , Indoles , Muscle, Smooth, Vascular/enzymology , Osteoprotegerin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyrroles , Signal Transduction , Animals , Case-Control Studies , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Female , Focal Adhesion Kinase 1/genetics , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/physiopathology , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , Male , Mice, Knockout , Middle Aged , Monocrotaline , Muscle, Smooth, Vascular/physiopathology , Osteoprotegerin/blood , Osteoprotegerin/deficiency , Osteoprotegerin/genetics , Pulmonary Artery/enzymology , Pulmonary Artery/physiopathology , RNA Interference , Rats, Sprague-Dawley , Severity of Illness Index , Transfection , Vascular Remodeling , Walk Test
18.
Pulm Pharmacol Ther ; 33: 39-46, 2015 Aug.
Article En | MEDLINE | ID: mdl-26086178

Pulmonary hypertension (PH) is a rapidly progressive disease that eventually leads to right heart failure and death. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its receptors (TRAIL-Rs) play an important role in the survival, migration, and proliferation of vascular smooth muscle cells. However, the association between serum TRAIL levels and PH is unknown. In this study, we assayed the serum soluble TRAIL (sTRAIL) levels in 78 patients with PH and 80 controls. The sTRAIL concentrations were elevated in the PH patients compared with the controls (138.76 ± 6.60 pg/mL vs. 80.14 ± 3.38 pg/mL, p < 0.0001). The presence of sTRAIL levels of >103 pg/mL could discriminate PH patients from healthy individuals, with a sensitivity of 75.6% and specificity of 81.2%. Moreover, elevated sTRAIL concentrations were associated with eventual pathological complications; this is consistent with the finding that sTRAIL levels decreased in patients who responded to treatment. In a hypoxia-induced PH mouse model, sTRAIL levels were significantly higher compared with those in normoxia mice, and clearly decreased when the mice were treated with treprostinil. The sTRAIL levels were positively correlated with right ventricular systolic pressure and the index of right ventricular hypertrophy. In conclusion, serum sTRAIL could be a biomarker for diagnosis and effective therapy for PH patients.


Hypertension, Pulmonary/physiopathology , Hypoxia/complications , TNF-Related Apoptosis-Inducing Ligand/blood , Animals , Case-Control Studies , Disease Models, Animal , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Female , Humans , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/drug therapy , Hypoxia/physiopathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Prospective Studies , Sensitivity and Specificity , Severity of Illness Index
19.
J Clin Invest ; 125(3): 1228-42, 2015 Mar 02.
Article En | MEDLINE | ID: mdl-25664856

Pulmonary arterial hypertension (PAH) is commonly associated with chronic hypoxemia in disorders such as chronic obstructive pulmonary disease (COPD). Prostacyclin analogs are widely used in the management of PAH patients; however, clinical efficacy and long-term tolerability of some prostacyclin analogs may be compromised by concomitant activation of the E-prostanoid 3 (EP3) receptor. Here, we found that EP3 expression is upregulated in pulmonary arterial smooth muscle cells (PASMCs) and human distal pulmonary arteries (PAs) in response to hypoxia. Either pharmacological inhibition of EP3 or Ep3 deletion attenuated both hypoxia and monocrotaline-induced pulmonary hypertension and restrained extracellular matrix accumulation in PAs in rodent models. In a murine PAH model, Ep3 deletion in SMCs, but not endothelial cells, retarded PA medial thickness. Knockdown of EP3α and EP3ß, but not EP3γ, isoforms diminished hypoxia-induced TGF-ß1 activation. Expression of either EP3α or EP3ß in EP3-deficient PASMCs restored TGF-ß1 activation in response to hypoxia. EP3α/ß activation in PASMCs increased RhoA-dependent membrane type 1 extracellular matrix metalloproteinase (MMP) translocation to the cell surface, subsequently activating pro-MMP-2 and promoting TGF-ß1 signaling. Activation or disruption of EP3 did not influence PASMC proliferation. Together, our results indicate that EP3 activation facilitates hypoxia-induced vascular remodeling and pulmonary hypertension in mice and suggest EP3 inhibition as a potential therapeutic strategy for pulmonary hypertension.


Hypertension, Pulmonary/metabolism , Receptors, Prostaglandin E, EP3 Subtype/genetics , Transforming Growth Factor beta1/physiology , rho GTP-Binding Proteins/metabolism , Animals , Cell Hypoxia , Cells, Cultured , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Hypertension, Pulmonary/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Artery/metabolism , Rats, Sprague-Dawley , Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Signal Transduction , Sulfonamides/pharmacology , Vascular Remodeling , rhoA GTP-Binding Protein
20.
J Biol Chem ; 289(17): 11681-11694, 2014 Apr 25.
Article En | MEDLINE | ID: mdl-24619416

Inadequate vitamin D status has been linked to increased risk of type 2 diabetes and cardiovascular disease. Inducible cyclooxygenase (COX) isoform COX-2 has been involved in the pathogenesis of such chronic inflammatory diseases. We found that the active form of vitamin D, 1,25(OH)2D produces dose-dependent inhibition of COX-2 expression in murine macrophages under both basal and LPS-stimulated conditions and suppresses proinflammatory mediators induced by LPS. Administration of 1,25(OH)2D significantly alleviated local inflammation in a carrageenan-induced paw edema mouse model. Strikingly, the phosphorylation of both Akt and its downstream target IκBα in macrophages were markedly suppressed by 1,25(OH)2D in the presence and absence of LPS stimulation through up-regulation of THEM4 (thioesterase superfamily member 4), an Akt modulator protein. Knockdown of both vitamin D receptor and THEM4 attenuated the inhibitory effect of 1,25(OH)2D on COX-2 expression in macrophages. A functional vitamin D-responsive element in the THEM4 promoter was identified by chromatin immunoprecipitation and luciferase reporter assay. Our results indicate that vitamin D restrains macrophage-mediated inflammatory processes by suppressing the Akt/NF-κB/COX-2 pathway, suggesting that vitamin D supplementation might be utilized for adjunctive therapy for inflammatory disease.


Calcitriol/pharmacology , Cyclooxygenase 2/genetics , Gene Expression Regulation, Enzymologic/drug effects , Inflammation/prevention & control , Thiolester Hydrolases/metabolism , Animals , Base Sequence , Carrageenan/toxicity , Cell Line , DNA Primers , Enzyme-Linked Immunosorbent Assay , Fatty Acids, Nonesterified/metabolism , Humans , Inflammation/chemically induced , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Macrophages/enzymology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Prostaglandins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
...