Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Am J Respir Crit Care Med ; 201(10): 1263-1276, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31917615

RESUMEN

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.


Asunto(s)
Hipoxia/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Hipertensión Arterial Pulmonar/genética , Receptores Inmunológicos/genética , Receptores de Prostaglandina/genética , Remodelación Vascular/genética , Animales , Antihipertensivos/farmacología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulación hacia Abajo , Epoprostenol/análogos & derivados , Epoprostenol/farmacología , Humanos , Hipertrofia , Inmunosupresores/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Ratones , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Hipertensión Arterial Pulmonar/tratamiento farmacológico , Hipertensión Arterial Pulmonar/metabolismo , Arteria Pulmonar , ARN Mensajero/metabolismo , Ratas , Sirolimus/farmacología
2.
Arterioscler Thromb Vasc Biol ; 39(4): e130-e145, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30676070

RESUMEN

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.


Asunto(s)
Cisteína Endopeptidasas/fisiología , Hipertensión Pulmonar/enzimología , Macrófagos/enzimología , Metaloproteinasa 2 de la Matriz/fisiología , Factor de Crecimiento Transformador beta1/fisiología , Animales , Inhibidores de Caspasas/farmacología , Cisteína Endopeptidasas/deficiencia , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Hipertensión Pulmonar/sangre , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/prevención & control , Hipoxia/enzimología , Indoles/toxicidad , Inflamación , Pulmón/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Monocrotalina/toxicidad , Pirroles/toxicidad , Ratas , Índice de Severidad de la Enfermedad , Transducción de Señal , Remodelación Vascular/fisiología
3.
Blood ; 124(10): 1610-21, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-25030064

RESUMEN

Cyclooxygenases (COXs) and their prostanoid products play important roles in a diverse range of physiological processes, including in the immune system. Here, we provide evidence that COX-1 is an essential regulator in early stages of B-cell development. COX-1-deficient mice displayed systematic reduction in total B cells, which was attributed to the arrest of early B-cell development from pro-B to pre-B stage. We further demonstrated that this defect was mediated through downregulation of the Janus kinase/signal transducer and activator of transcription 5 (JAK/STAT5) signaling and its target genes, including Pax5, in COX-1(-/-) mice. Mechanistic studies revealed that COX-1-derived thromboxane A2 (TxA2) could regulate JAK3/STAT5 signaling through the cyclic adenosine monophosphate-protein kinase A pathway, via binding with its receptor thromboxane A2 receptor (TP). Administration of the TP agonist could rescue the defective B-cell development and JAK/STAT5 signaling activity in COX-1-deficient mice. Moreover, administration of low-dose aspirin caused a significant reduction in total B cells in peripheral blood of healthy human volunteers, coincidentally with reduced TxA2 production and downregulation of JAK/STAT5 signaling. Taken together, our results demonstrate that COX-1-derived TxA2 plays a critical role in the stage transition of early B-cell development through regulation of JAK/STAT5 signaling and indicate a potential immune-suppressive effect of low-dose aspirin in humans.


Asunto(s)
Linfocitos B/fisiología , Diferenciación Celular , Ciclooxigenasa 1/metabolismo , Proteínas de la Membrana/metabolismo , Tromboxano A2/fisiología , Animales , Diferenciación Celular/genética , Células Cultivadas , Ciclooxigenasa 1/genética , Humanos , Quinasas Janus/metabolismo , Leucopoyesis/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/genética , Tromboxano A2/metabolismo
4.
J Biol Chem ; 289(17): 11681-11694, 2014 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-24619416

RESUMEN

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.


Asunto(s)
Calcitriol/farmacología , Ciclooxigenasa 2/genética , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Inflamación/prevención & control , Tioléster Hidrolasas/metabolismo , Animales , Secuencia de Bases , Carragenina/toxicidad , Línea Celular , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Ácidos Grasos no Esterificados/metabolismo , Humanos , Inflamación/inducido químicamente , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Prostaglandinas/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Pulm Pharmacol Ther ; 33: 39-46, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26086178

RESUMEN

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.


Asunto(s)
Hipertensión Pulmonar/fisiopatología , Hipoxia/complicaciones , Ligando Inductor de Apoptosis Relacionado con TNF/sangre , Animales , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Epoprostenol/análogos & derivados , Epoprostenol/farmacología , Femenino , Humanos , Hipertensión Pulmonar/sangre , Hipertensión Pulmonar/tratamiento farmacológico , Hipoxia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Estudios Prospectivos , Sensibilidad y Especificidad , Índice de Severidad de la Enfermedad
6.
J Allergy Clin Immunol ; 134(5): 1163-74.e16, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24948368

RESUMEN

BACKGROUND: Myeloid-derived suppressor cells (MDSCs) have recently been implicated in the pathogenesis of asthma, but their regulation in patients with aspirin-intolerant asthma (AIA) remains unclear. OBJECTIVE: We sought to characterize MDSC accumulation and pathogenic functions in allergic airway inflammation mediated by COX-1 deficiency or aspirin treatment in mice. METHODS: Allergic airway inflammation was induced in mice by means of ovalbumin challenge. The distribution and function of MDSCs in mice were analyzed by using flow cytometry and pharmacologic/gene manipulation approaches. RESULTS: CD11b(+)Gr1(high)Ly6G(+)Ly6C(int) MDSCs (polymorphonuclear MDSCs [PMN-MDSCs]) recruited to the lungs are negatively correlated with airway inflammation in allergen-challenged mice. Aspirin-treated and COX-1 knockout (KO) mice showed significantly lower accumulation of PMN-MDSCs in the inflamed lung and immune organs accompanied by increased TH2 airway responses. The TH2-suppressive function of PMN-MDSCs was notably impaired by COX-1 deletion or inhibition, predominantly through downregulation of arginase-1. COX-1-derived prostaglandin E2 promoted PMN-MDSC generation in bone marrow through E prostanoid 2 and 4 receptors (EP2 and EP4), whereas the impaired arginase-1 expression in PMN-MDSCs in COX-1 KO mice was mediated by dysregulation of the prostaglandin E2/EP4/cyclic AMP/protein kinase A pathway. EP4 agonist administration alleviated allergy-induced airway hyperresponsiveness in COX-1 KO mice. Moreover, the immunosuppressive function of PMN-MDSCs from patients with AIA was dramatically decreased compared with that from patients with aspirin-tolerant asthma. CONCLUSION: The immunosuppressive activity of PMN-MDSCs was diminished in both allergen-challenged COX-1 KO mice and patients with AIA, probably through an EP4-mediated signaling pathway, indicating that activation of PMN-MDSCs might be a promising therapeutic strategy for asthma, particularly AIA.


Asunto(s)
Asma Inducida por Aspirina/inmunología , Tolerancia Inmunológica , Células Mieloides/inmunología , Transducción de Señal/inmunología , Alérgenos/toxicidad , Animales , Antiinflamatorios no Esteroideos/efectos adversos , Antiinflamatorios no Esteroideos/farmacología , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Arginasa/genética , Arginasa/inmunología , Aspirina/efectos adversos , Aspirina/farmacología , Asma Inducida por Aspirina/genética , Asma Inducida por Aspirina/patología , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/inmunología , Ciclooxigenasa 1/genética , Ciclooxigenasa 1/inmunología , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , Células Mieloides/patología , Subtipo EP2 de Receptores de Prostaglandina E/genética , Subtipo EP2 de Receptores de Prostaglandina E/inmunología , Subtipo EP4 de Receptores de Prostaglandina E/genética , Subtipo EP4 de Receptores de Prostaglandina E/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Células Th2/inmunología , Células Th2/patología
7.
Cell Death Dis ; 15(4): 263, 2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38615011

RESUMEN

Abnormal cardiac fibrosis is the main pathological change of post-myocardial infarction (MI) heart failure. Although the E3 ubiquitin ligase FBXL8 is a key regulator in the cell cycle, cell proliferation, and inflammation, its role in post-MI ventricular fibrosis and heart failure remains unknown. FBXL8 was primarily expressed in cardiac fibroblasts (CFs) and remarkably decreased in CFs treated by TGFß and heart subjected to MI. The echocardiography and histology data suggested that adeno-associated viruses (AAV9)-mediated FBXL8 overexpression had improved cardiac function and ameliorated post-MI cardiac fibrosis. In vitro, FBXL8 overexpression prevented TGFß-induced proliferation, migration, contraction, and collagen secretion in CFs, while knockdown of FBXL8 demonstrated opposite effects. Mechanistically, FBXL8 interacted with Snail1 to promote Snail1 degradation through the ubiquitin-proteasome system and decreased the activation of RhoA. Moreover, the FBXL8ΔC3 binding domain was indispensable for Snail1 interaction and degradation. Ectopic Snail1 expression partly abolished the effects mediated by FBXL8 overexpression in CFs treated by TGFß. These results characterized the role of FBXL8 in regulating the ubiquitin-mediated degradation of Snail1 and revealed the underlying molecular mechanism of how MI up-regulated the myofibroblasts differentiation-inducer Snail1 and suggested that FBXL8 may be a potential curative target for improving post-MI cardiac function.


Asunto(s)
Insuficiencia Cardíaca , Infarto del Miocardio , Humanos , Complejo de la Endopetidasa Proteasomal , Infarto del Miocardio/genética , Factor de Crecimiento Transformador beta , Ubiquitinas
8.
Cardiovasc Toxicol ; 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951468

RESUMEN

Radix Paeoniae Rubra. (Chishao, RPR) and Cortex Moutan. (Mudanpi, CM) are a pair of traditional Chinese medicines that play an important role in the treatment of atherosclerosis (AS). The main objective of this study was to identify potential synergetic function and underlying mechanisms of RPR-CM in the treatment of AS. The main active ingredients, targets of RPR-CM and AS-related genes were obtained from public databases. A Venn diagram was utilized to screen the common targets of RPR-CM in treating AS. The protein-protein interaction network was established based on STRING database. Biological functions and pathways of potential targets were analyzed through Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses. Cytoscape was used to construct the drug-compound-target-signal pathway network. Molecular docking was performed to verify the binding ability of the bioactive ingredients and the target proteins. The endothelial inflammation model was constructed with human umbilical vein endothelial cells stimulated with ox-LDL, and the function of RPR-CM in treating AS was verified by CCK-8 assay, enzyme-linked immunosorbent assay, and qPCR. In this study, 12 active components and 401 potential target genes of RPR-CM were identified, among which quercetin, kaempferol and baicalein were considered to be the main active components. A total of 1903 AS-related genes were identified through public databases and four GEO datasets (GSE57691, GSE72633, GSE6088 and GSE199819). There are 113 common target genes of RPR-CM in treating AS. PPI network analysis identified 17 genes in cluster 1 as the core targets. Bioinformatics analysis showed that RPR-CM in AS treatment was associated with multiple downstream biological processes and signal pathways. PTGS2, JUN, CASP3, TNF, IL1B, IL6, FOS, STAT1 were identified as the core targets of RPR-CM, and molecular docking showed that the main bioactive components of RPR-CM had good binding ability with the core targets. RPR-CM extract significantly inhibited the levels of inflammatory factors TNF-α, IL-6, IL-1ß, MCP-1, VCAM-1 and ICAM-1 in HUVECs, and inhibited endothelial inflammation. This study revealed the active ingredients of RPR-CM, and identified the key downstream targets and signaling pathways in the treatment of AS, providing theoretical basis for the application of RPR-CM in prevention and treatment of AS.

9.
Cardiovasc Res ; 114(5): 703-712, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29415171

RESUMEN

Aims: Cardiac myofibroblasts (CMFs) play a crucial role in the progression of pathological fibrotic cardiac remodelling. The expression of osteoglycin (OGN) is increased in diseased hearts; however, the role of OGN in pathological cardiac remodelling is not understood. Here, we sought to determine the effect of OGN on cardiac interstitial fibrosis and investigate the molecular mechanisms of OGN in CMF activation and matrix production. Methods and results: We found that OGN expression was significantly upregulated in mouse hearts in response to chronic 14-day angiotensin II (Ang II) infusion. Mice lacking OGN (OGN-/-) exhibited enhanced cardiac interstitial fibrosis and significantly more severe cardiac dysfunction following Ang II infusion compared to wild-type mice. OGN deficiency did not alter blood pressure, nor had effect on transforming growth factor-beta signalling activation, but presented with increased proliferative activity in hearts. In vitro studies with isolated CMFs revealed that OGN deficiency significantly increased proliferation and migration and enhanced the transactivation of epidermal growth factor receptor (EGFR) signalling by Ang II. On the other hand, OGN overexpression in CMFs decreased their proliferation and migration via reducing EGFR activation. Overexpression of OGN also suppressed the shedding of membrane anchored EGFR ligand. Moreover, OGN was found to interact with a lysophosphatidic acid (LPA) receptor isoform 3 and thus to attenuate EGFR transactivation through blocking cell surface translocation of membrane type 1 matrix metalloproteinase (MT1-MMP) and subsequent pro-MMP-2 activation in a Ras homolog gene family, member A (RhoA)/Rho-associated, coiled-coil containing protein kinase (ROCK)-dependent manner. Conclusion: These findings suggest that OGN negatively regulates cardiac fibrotic remodelling by attenuating CMF proliferation and migration through LPA3-mediated and Rho/ROCK-dependent inhibition of MT1-MMP translocation, MMP2 activation and EGFR transactivation.


Asunto(s)
Cardiomegalia/enzimología , Proliferación Celular , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Miofibroblastos/enzimología , Receptores del Ácido Lisofosfatídico/metabolismo , Remodelación Ventricular , Angiotensina II , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Movimiento Celular , Células Cultivadas , Modelos Animales de Enfermedad , Receptores ErbB/metabolismo , Fibrosis , Hipertensión/inducido químicamente , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Metaloproteinasa 14 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Miofibroblastos/patología , Receptor Cross-Talk , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA
10.
J Exp Med ; 215(8): 2175-2195, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-29970474

RESUMEN

Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by progressive pulmonary artery (PA) remodeling. T helper 2 cell (Th2) immune response is involved in PA remodeling during PAH progression. Here, we found that CRTH2 (chemoattractant receptor homologous molecule expressed on Th2 cell) expression was up-regulated in circulating CD3+CD4+ T cells in patients with idiopathic PAH and in rodent PAH models. CRTH2 disruption dramatically ameliorated PA remodeling and pulmonary hypertension in different PAH mouse models. CRTH2 deficiency suppressed Th2 activation, including IL-4 and IL-13 secretion. Both CRTH2+/+ bone marrow reconstitution and CRTH2+/+ CD4+ T cell adoptive transfer deteriorated hypoxia + ovalbumin-induced PAH in CRTH2-/- mice, which was reversed by dual neutralization of IL-4 and IL-13. CRTH2 inhibition alleviated established PAH in mice by repressing Th2 activity. In culture, CRTH2 activation in Th2 cells promoted pulmonary arterial smooth muscle cell proliferation through activation of STAT6. These results demonstrate the critical role of CRTH2-mediated Th2 response in PAH pathogenesis and highlight the CRTH2 receptor as a potential therapeutic target for PAH.


Asunto(s)
Hipertensión Pulmonar/inmunología , Activación de Linfocitos/inmunología , Receptores Inmunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Células Th2/inmunología , Traslado Adoptivo , Adulto , Animales , Anticuerpos/farmacología , Presión Sanguínea/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Proliferación Celular/efectos de los fármacos , Quimera , Enfermedad Crónica , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Humanos , Hipertensión Pulmonar/fisiopatología , Hipoxia/complicaciones , Hipoxia/fisiopatología , Inmunidad/efectos de los fármacos , Indoles , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Masculino , Ratones , Ovalbúmina , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Pirroles , Receptores Inmunológicos/deficiencia , Receptores de Prostaglandina/deficiencia , Factor de Transcripción STAT6/metabolismo , Regulación hacia Arriba/efectos de los fármacos
11.
Circ Cardiovasc Genet ; 10(1)2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28077433

RESUMEN

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.


Asunto(s)
Presión Arterial , Quinasa 1 de Adhesión Focal/metabolismo , Hipertensión Pulmonar/prevención & control , Hipoxia/complicaciones , Indoles , Músculo Liso Vascular/enzimología , Osteoprotegerina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirroles , Transducción de Señal , Animales , Estudios de Casos y Controles , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Quinasa 1 de Adhesión Focal/genética , Humanos , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/enzimología , Hipertensión Pulmonar/fisiopatología , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Masculino , Ratones Noqueados , Persona de Mediana Edad , Monocrotalina , Músculo Liso Vascular/fisiopatología , Osteoprotegerina/sangre , Osteoprotegerina/deficiencia , Osteoprotegerina/genética , Arteria Pulmonar/enzimología , Arteria Pulmonar/fisiopatología , Interferencia de ARN , Ratas Sprague-Dawley , Índice de Severidad de la Enfermedad , Transfección , Remodelación Vascular , Prueba de Paso
12.
Cardiovasc Res ; 113(6): 586-597, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28407046

RESUMEN

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.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Hipertensión Pulmonar/inducido químicamente , Indoles , Arteria Pulmonar/metabolismo , Pirroles , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Remodelación Vascular , Animales , Estudios de Casos y Controles , Hipoxia de la Célula , Células Cultivadas , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/metabolismo , Ligandos , Masculino , Persona de Mediana Edad , Fosforilación , Arteria Pulmonar/patología , Interferencia de ARN , Receptor para Productos Finales de Glicación Avanzada/genética , Subunidad beta de la Proteína de Unión al Calcio S100/genética , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
J Clin Invest ; 125(3): 1228-42, 2015 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-25664856

RESUMEN

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.


Asunto(s)
Hipertensión Pulmonar/metabolismo , Subtipo EP3 de Receptores de Prostaglandina E/genética , Factor de Crecimiento Transformador beta1/fisiología , Proteínas de Unión al GTP rho/metabolismo , Animales , Hipoxia de la Célula , Células Cultivadas , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Hipertensión Pulmonar/fisiopatología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Arteria Pulmonar/metabolismo , Ratas Sprague-Dawley , Subtipo EP3 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP3 de Receptores de Prostaglandina E/metabolismo , Transducción de Señal , Sulfonamidas/farmacología , Remodelación Vascular , Proteína de Unión al GTP rhoA
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA