Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 1.995
Filtrar
1.
Apoptosis ; 29(9-10): 1648-1662, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38635022

RESUMEN

Hypoxic pulmonary hypertension (HPH) is a pathophysiological syndrome in which pulmonary vascular pressure increases under hypoxic stimulation and there is an urgent need to develop emerging therapies for the treatment of HPH. LncRNA MIR210HG is a long non-coding RNA closely related to hypoxia and has been widely reported in a variety of tumor diseases. But its mechanism in hypoxic pulmonary hypertension is not clear. In this study, we identified for the first time the potential effect of MIR210HG on disease progression in HPH. Furthermore, we investigated the underlying mechanism through which elevated levels of MIR210HG promotes the transition from a contractile phenotype to a synthetic phenotype in PASMCs under hypoxia via activation of autophagy-dependent ferroptosis pathway. While overexpression of HIF-2α in PASMCs under hypoxia significantly reversed the phenotypic changes induced by MIR210HG knockdown. We further investigated the potential positive regulatory relationship between STAT3 and the transcription of MIR210HG in PASMCs under hypoxic conditions. In addition, we established both in vivo and in vitro models of HPH to validate the differential expression of specific markers associated with hypoxia. Our findings suggest a potential mechanism of LncRNA MIR210HG in the progression of HPH and offer potential targets for disease intervention and treatment.


Asunto(s)
Autofagia , Ferroptosis , Miocitos del Músculo Liso , Fenotipo , Arteria Pulmonar , ARN Largo no Codificante , Animales , Humanos , Masculino , Ratones , Ratas , Autofagia/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Hipoxia de la Célula/genética , Ferroptosis/genética , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Hipoxia/genética , Hipoxia/metabolismo , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Arteria Pulmonar/citología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Transducción de Señal , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética
2.
J Biomech Eng ; 146(11)2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-38980683

RESUMEN

This study was undertaken to develop a mathematical model of the long-term in vivo remodeling processes in postimplanted pulmonary artery (PA) conduits. Experimental results from two extant ovine in vivo studies, wherein polyglycolic-acid (PGA)/poly-L-lactic acid tubular conduits were constructed, cell seeded, incubated for 4 weeks, and then implanted in mature sheep to obtain the remodeling data for up to two years. Explanted conduit analysis included detailed novel structural and mechanical studies. Results in both studies indicated that the in vivo conduits remained dimensionally stable up to 80 weeks, so that the conduits maintained a constant in vivo stress and deformation state. In contrast, continued remodeling of the constituent collagen fiber network as evidenced by an increase in effective tissue uniaxial tangent modulus, which then stabilized by one year postimplant. A mesostructural constitute model was then applied to extant planar biaxial mechanical data and revealed several interesting features, including an initial pronounced increase in effective collagen fiber modulus, paralleled by a simultaneous shift toward longer, more uniformly length-distributed collagen fibers. Thus, while the conduit remained dimensionally stable, its internal collagen fibrous structure and resultant mechanical behaviors underwent continued remodeling that stabilized by one year. A time-evolving structural mixture-based mathematical model specialized for this unique form of tissue remodeling was developed, with a focus on time-evolving collagen fiber stiffness as the driver for tissue-level remodeling. The remodeling model was able to fully reproduce (1) the observed tissue-level increases in stiffness by time-evolving simultaneous increases in collagen fiber modulus and lengths, (2) maintenance of the constant collagen fiber angular dispersion, and (3) stabilization of the remodeling processes at one year. Collagen fiber remodeling geometry was directly verified experimentally by histological analysis of the time-evolving collagen fiber crimp, which matches model predictions very closely. Interestingly, the remodeling model indicated that the basis for tissue homeostasis was maintenance of the collagen fiber ensemble stress for all orientations, and not individual collagen fiber stresses. Unlike other growth and remodeling models that traditionally treat changes in the external boundary conditions (e.g., changes in blood pressure) as the primary input stimuli, the driver herein is changes to the internal constituent collagen fiber themselves due to cellular mediated cross-linking.


Asunto(s)
Colágeno , Arteria Pulmonar , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Animales , Colágeno/metabolismo , Colágeno/química , Ovinos , Ingeniería de Tejidos , Modelos Biológicos , Ácido Poliglicólico/química , Prótesis Vascular , Andamios del Tejido/química
3.
Int J Mol Sci ; 25(17)2024 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-39273443

RESUMEN

Vascular smooth muscle cells (SMCs) can transition between a quiescent contractile or "differentiated" phenotype and a "proliferative-dedifferentiated" phenotype in response to environmental cues, similar to what in occurs in the wound healing process observed in fibroblasts. When dysregulated, these processes contribute to the development of various lung and cardiovascular diseases such as Chronic Obstructive Pulmonary Disease (COPD). Long non-coding RNAs (lncRNAs) have emerged as key modulators of SMC differentiation and phenotypic changes. In this study, we examined the expression of lncRNAs in primary human pulmonary artery SMCs (hPASMCs) during cell-to-cell contact-induced SMC differentiation. We discovered a novel lncRNA, which we named Differentiation And Growth Arrest-Related lncRNA (DAGAR) that was significantly upregulated in the quiescent phenotype with respect to proliferative SMCs and in cell-cycle-arrested MRC5 lung fibroblasts. We demonstrated that DAGAR expression is essential for SMC quiescence and its knockdown hinders SMC differentiation. The treatment of quiescent SMCs with the pro-inflammatory cytokine Tumor Necrosis Factor (TNF), a known inducer of SMC dedifferentiation and proliferation, elicited DAGAR downregulation. Consistent with this, we observed diminished DAGAR expression in pulmonary arteries from COPD patients compared to non-smoker controls. Through pulldown experiments followed by mass spectrometry analysis, we identified several proteins that interact with DAGAR that are related to cell differentiation, the cell cycle, cytoskeleton organization, iron metabolism, and the N-6-Methyladenosine (m6A) machinery. In conclusion, our findings highlight DAGAR as a novel lncRNA that plays a crucial role in the regulation of cell proliferation and SMC differentiation. This paper underscores the potential significance of DAGAR in SMC and fibroblast physiology in health and disease.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Fibroblastos , Miocitos del Músculo Liso , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Fibroblastos/metabolismo , Diferenciación Celular/genética , Miocitos del Músculo Liso/metabolismo , Proliferación Celular/genética , Arteria Pulmonar/metabolismo , Arteria Pulmonar/citología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/citología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/patología , Células Cultivadas
4.
Molecules ; 29(11)2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38893423

RESUMEN

A chemical investigation of Anthriscus sylvestris roots led to the isolation and characterization of two new nitrogen-containing phenylpropanoids (1-2) and two new phenol glycosides (8-9), along with fifteen known analogues. Structure elucidation was based on HRESIMS, 1D and 2D NMR spectroscopy, and electronic circular dichroism (ECD). In addition, compounds 3, 6, 9-10, 12, and 17 exhibited inhibitory effects against the abnormal proliferation of pulmonary arterial smooth muscle cells with IC50 values ranging from 10.7 ± 0.6 to 57.1 ± 1.1 µM.


Asunto(s)
Proliferación Celular , Miocitos del Músculo Liso , Raíces de Plantas , Arteria Pulmonar , Raíces de Plantas/química , Proliferación Celular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Animales , Estructura Molecular , Extractos Vegetales/farmacología , Extractos Vegetales/química , Glicósidos/farmacología , Glicósidos/química , Glicósidos/aislamiento & purificación , Ratas , Espectroscopía de Resonancia Magnética
5.
Molecules ; 29(10)2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38792081

RESUMEN

Prolylcarboxypeptidase (PRCP, PCP, Lysosomal Pro-X-carboxypeptidase, Angiotensinase C) controls angiotensin- and kinin-induced cell signaling. Elevation of PRCP appears to be activated in chronic inflammatory diseases [cardiovascular disease (CVD), diabetes] in proportion to severity. Vascular endothelial cell senescence and mitochondrial dysfunction have consistently been shown in models of CVD in aging. Cellular senescence, a driver of age-related dysfunction, can differentially alter the expression of lysosomal enzymes due to lysosomal membrane permeability. There is a lack of data demonstrating the effect of age-related dysfunction on the expression and function of PRCP. To explore the changes in PRCP, the PRCP-dependent prekallikrein (PK) pathway was characterized in early- and late-passage human pulmonary artery endothelial cells (HPAECs). Detailed kinetic analysis of cells treated with high molecular weight kininogen (HK), a precursor of bradykinin (BK), and PK revealed a mechanism by which senescent HPAECs activate the generation of kallikrein upon the assembly of the HK-PK complex on HPAECs in parallel with an upregulation of PRCP and endothelial nitric oxide (NO) synthase (eNOS) and NO formation. The NO production and expression of both PRCP and eNOS increased in early-passage HPAECs and decreased in late-passage HPAECs. Low activity of PRCP in late-passage HPAECs was associated with rapid decreased telomerase reverse transcriptase mRNA levels. We also found that, with an increase in the passage number of HPAECs, reduced PRCP altered the respiration rate. These results indicated that aging dysregulates PRCP protein expression, and further studies will shed light into the complexity of the PRCP-dependent signaling pathway in aging.


Asunto(s)
Biomarcadores , Carboxipeptidasas , Senescencia Celular , Células Endoteliales , Humanos , Células Endoteliales/metabolismo , Biomarcadores/metabolismo , Carboxipeptidasas/metabolismo , Carboxipeptidasas/genética , Precalicreína/metabolismo , Precalicreína/genética , Bradiquinina/farmacología , Bradiquinina/metabolismo , Arteria Pulmonar/metabolismo , Arteria Pulmonar/citología , Células Cultivadas , Quininógeno de Alto Peso Molecular/metabolismo , Transducción de Señal , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Calicreínas/metabolismo , Calicreínas/genética
6.
Am J Physiol Cell Physiol ; 323(4): C959-C973, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-35968892

RESUMEN

Mechanosensitive cation channels and Ca2+ influx through these channels play an important role in the regulation of endothelial cell functions. Transient receptor potential canonical channel 6 (TRPC6) is a diacylglycerol-sensitive nonselective cation channel that forms receptor-operated Ca2+ channels in a variety of cell types. Piezo1 is a mechanosensitive cation channel activated by membrane stretch and shear stress in lung endothelial cells. In this study, we report that TRPC6 and Piezo1 channels both contribute to membrane stretch-mediated cation currents and Ca2+ influx or increase in cytosolic-free Ca2+ concentration ([Ca2+]cyt) in human pulmonary arterial endothelial cells (PAECs). The membrane stretch-mediated cation currents and increase in [Ca2+]cyt in human PAECs were significantly decreased by GsMTX4, a blocker of Piezo1 channels, and by BI-749327, a selective blocker of TRPC6 channels. Extracellular application of 1-oleoyl-2-acetyl-sn-glycerol (OAG), a membrane permeable analog of diacylglycerol, rapidly induced whole cell cation currents and increased [Ca2+]cyt in human PAECs and human embryonic kidney (HEK)-cells transiently transfected with the human TRPC6 gene. Furthermore, membrane stretch with hypo-osmotic or hypotonic solution enhances the cation currents in TRPC6-transfected HEK cells. In HEK cells transfected with the Piezo1 gene, however, OAG had little effect on the cation currents, but membrane stretch significantly enhanced the cation currents. These data indicate that, while both TRPC6 and Piezo1 are involved in generating mechanosensitive cation currents and increases in [Ca2+]cyt in human PAECs undergoing mechanical stimulation, only TRPC6 (but not Piezo1) is sensitive to the second messenger diacylglycerol. Selective blockers of these channels may help develop novel therapies for mechanotransduction-associated pulmonary vascular remodeling in patients with pulmonary arterial hypertension.


Asunto(s)
Células Endoteliales , Canales Iónicos , Mecanorreceptores , Canal Catiónico TRPC6 , Calcio/metabolismo , Cationes/metabolismo , Diglicéridos/metabolismo , Diglicéridos/farmacología , Células Endoteliales/metabolismo , Humanos , Soluciones Hipotónicas/metabolismo , Soluciones Hipotónicas/farmacología , Canales Iónicos/genética , Canales Iónicos/metabolismo , Mecanorreceptores/metabolismo , Mecanotransducción Celular/genética , Mecanotransducción Celular/fisiología , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Canal Catiónico TRPC6/genética , Canal Catiónico TRPC6/metabolismo
7.
J Biol Chem ; 296: 100599, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33781742

RESUMEN

Sphingosine-1-phosphate (S1P), a natural multifunctional phospholipid, is highly increased in plasma from patients with pulmonary arterial hypertension and mediates proliferation of pulmonary artery smooth muscle cells (PASMCs) by activating the Notch3 signaling pathway. However, the mechanisms underpinning S1P-mediated induction of PASMCs proliferation remain unclear. In this study, using biochemical and molecular biology approaches, RNA interference and gene expression analyses, 5'-ethynyl-2'-deoxyuridine incorporation assay, and 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide assay, we demonstrated that S1P promoted the activation of signal transducers and activators of transcription 3 (STAT3) through sphingosine-1-phosphate receptor 2 (S1PR2), and subsequently upregulated the expression of the microRNA miR-135b, which further reduced the expression of E3 ubiquitin ligase ß-transduction repeat-containing protein and led to a reduction in yes-associated protein (YAP) ubiquitinated degradation in PASMCs. YAP is the core effector of the Hippo pathway and mediates the expression of particular genes. The accumulation of YAP further increased the expression and activation of Notch3 and ultimately promoted the proliferation of PASMCs. In addition, we showed that preblocking S1PR2, prior silencing of STAT3, miR-135b, or YAP, and prior inhibition of Notch3 all attenuated S1P-induced PASMCs proliferation. Taken together, our study indicates that S1P stimulates PASMCs proliferation by activation of the S1PR2/STAT3/miR-135b/ß-transduction repeat-containing protein/YAP/Notch3 pathway, and our data suggest that targeting this cascade might have potential value in ameliorating PASMCs hyperproliferation and benefit pulmonary arterial hypertension.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisofosfolípidos/farmacología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Arteria Pulmonar/citología , Receptor Notch3/metabolismo , Esfingosina/análogos & derivados , Animales , Proliferación Celular/efectos de los fármacos , Masculino , Miocitos del Músculo Liso/metabolismo , Ratas , Ratas Sprague-Dawley , Esfingosina/farmacología , Proteínas Señalizadoras YAP
8.
Nat Methods ; 16(1): 103-110, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30559434

RESUMEN

We present deep-learning-enabled super-resolution across different fluorescence microscopy modalities. This data-driven approach does not require numerical modeling of the imaging process or the estimation of a point-spread-function, and is based on training a generative adversarial network (GAN) to transform diffraction-limited input images into super-resolved ones. Using this framework, we improve the resolution of wide-field images acquired with low-numerical-aperture objectives, matching the resolution that is acquired using high-numerical-aperture objectives. We also demonstrate cross-modality super-resolution, transforming confocal microscopy images to match the resolution acquired with a stimulated emission depletion (STED) microscope. We further demonstrate that total internal reflection fluorescence (TIRF) microscopy images of subcellular structures within cells and tissues can be transformed to match the results obtained with a TIRF-based structured illumination microscope. The deep network rapidly outputs these super-resolved images, without any iterations or parameter search, and could serve to democratize super-resolution imaging.


Asunto(s)
Aprendizaje Profundo , Microscopía Confocal/métodos , Microscopía Fluorescente/métodos , Animales , Bovinos , Células Endoteliales/citología , Células HeLa , Humanos , Arteria Pulmonar/citología , Fracciones Subcelulares/ultraestructura
9.
Circ Res ; 127(9): 1138-1152, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-32752980

RESUMEN

RATIONALE: POSTN (Periostin) is an ECM (extracellular matrix) protein involved in tissue remodeling in response to injury and a contributing factor in tumorigenesis, suggesting that POSTN plays a role in the pathogenesis of pulmonary hypertension (PH). OBJECTIVE: We aimed to gain insight into the mechanistic contribution of POSTN in experimental mouse models of PH and correlate these findings with PH in humans. METHODS AND RESULTS: We used genetic epistasis approaches in human pulmonary artery endothelial cells (hPAECs), human pulmonary artery smooth muscle cells, and experimental mouse models of PH (Sugen 5416/hypoxia or chronic hypoxia) to discern the role of POSTN and its relationship to HIF (hypoxia-inducible factor)-1α signaling. We found that POSTN expression was correlated with the extent of PH in mouse models and in humans. Decreasing POSTN improved hemodynamic and cardiac responses in PH mice, blunted the release of growth factors and HIF-1α, and reversed the downregulated BMPR (bone morphogenetic protein receptor)-2 expression in hPAECs from patients with PH, whereas increasing POSTIN had the opposite effects and induced a hyperproliferative and promigratory phenotype in both hPAECs and human pulmonary artery smooth muscle cells. Overexpression of POSTN-induced activation of HIFs and increased the production of ET (endothelin)-1 and VEGF (vascular endothelial growth factor) in hPAECs. SiRNA-mediated knockdown of HIF-1α abolished the proangiogenic effect of POSTN. Blockade of TrkB (tyrosine kinase receptor B) attenuated the effect of POSTN on HIF-1α expression, while inhibition of HIF-1α reduced the expression of POSTN and TrkB. These results suggest that hPAECs produce POSTN via a HIF-1α-dependent mechanism. CONCLUSIONS: Our study reveals that POSTN expression is increased in human and animal models of PH and fosters PH development via a positive feedback loop between HIF-1α and POSTN during hypoxia. We propose that manipulating POSTIN expression may be an efficacious therapeutic target in the treatment of PH. Our results also suggest that POSTN may serve as a biomarker to estimate the severity of PH.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Hipertensión Pulmonar/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Animales , Biomarcadores/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Hipoxia de la Célula , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/fisiología , Endotelina-1/metabolismo , Humanos , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/terapia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Indoles , Glicoproteínas de Membrana/antagonistas & inhibidores , Ratones , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/fisiología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Arteria Pulmonar/citología , Pirroles , Receptor trkB/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
PLoS Comput Biol ; 17(4): e1008930, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33878108

RESUMEN

In this work, non-invasive high-spatial resolution three-dimensional (3D) X-ray micro-computed tomography (µCT) of healthy mouse lung vasculature is performed. Methodologies are presented for filtering, segmenting, and skeletonizing the collected 3D images. Novel methods for the removal of spurious branch artefacts from the skeletonized 3D image are introduced, and these novel methods involve a combination of distance transform gradients, diameter-length ratios, and the fast marching method (FMM). These new techniques of spurious branch removal result in the consistent removal of spurious branches without compromising the connectivity of the pulmonary circuit. Analysis of the filtered, skeletonized, and segmented 3D images is performed using a newly developed Vessel Network Extraction algorithm to fully characterize the morphology of the mouse pulmonary circuit. The removal of spurious branches from the skeletonized image results in an accurate representation of the pulmonary circuit with significantly less variability in vessel diameter and vessel length in each generation. The branching morphology of a full pulmonary circuit is characterized by the mean diameter per generation and number of vessels per generation. The methods presented in this paper lead to a significant improvement in the characterization of 3D vasculature imaging, allow for automatic separation of arteries and veins, and for the characterization of generations containing capillaries and intrapulmonary arteriovenous anastomoses (IPAVA).


Asunto(s)
Algoritmos , Procesamiento de Imagen Asistido por Computador/métodos , Pulmón/irrigación sanguínea , Tomografía Computarizada por Rayos X/métodos , Animales , Ratones Endogámicos C57BL , Arteria Pulmonar/citología , Venas Pulmonares/citología
12.
J Immunol ; 204(6): 1474-1485, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31996456

RESUMEN

An increasing body of evidence suggests that bone marrow-derived myeloid cells play a critical role in the pathophysiology of pulmonary hypertension (PH). However, the true requirement for myeloid cells in PH development has not been demonstrated, and a specific disease-promoting myeloid cell population has not been identified. Using bone marrow chimeras, lineage labeling, and proliferation studies, we determined that, in murine hypoxia-induced PH, Ly6Clo nonclassical monocytes are recruited to small pulmonary arteries and differentiate into pulmonary interstitial macrophages. Accumulation of these nonclassical monocyte-derived pulmonary interstitial macrophages around pulmonary vasculature is associated with increased muscularization of small pulmonary arteries and disease severity. To determine if the sensing of hypoxia by nonclassical monocytes contributes to the development of PH, mice lacking expression of hypoxia-inducible factor-1α in the Ly6Clo monocyte lineage were exposed to hypoxia. In these mice, vascular remodeling and PH severity were significantly reduced. Transcriptome analyses suggest that the Ly6Clo monocyte lineage regulates PH through complement, phagocytosis, Ag presentation, and chemokine/cytokine pathways. Consistent with these murine findings, relative to controls, lungs from pulmonary arterial hypertension patients displayed a significant increase in the frequency of nonclassical monocytes. Taken together, these findings show that, in response to hypoxia, nonclassical monocytes in the lung sense hypoxia, infiltrate small pulmonary arteries, and promote vascular remodeling and development of PH. Our results demonstrate that myeloid cells, specifically cells of the nonclassical monocyte lineage, play a direct role in the pathogenesis of PH.


Asunto(s)
Hipertensión Pulmonar/inmunología , Hipoxia/complicaciones , Macrófagos Alveolares/inmunología , Monocitos/inmunología , Remodelación Vascular/inmunología , Animales , Antígenos Ly/metabolismo , Trasplante de Médula Ósea , Diferenciación Celular/inmunología , Modelos Animales de Enfermedad , Humanos , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/cirugía , Hipoxia/inmunología , Hipoxia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Pulmón/irrigación sanguínea , Pulmón/inmunología , Pulmón/patología , Trasplante de Pulmón , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Ratones Transgénicos , Monocitos/metabolismo , Arteria Pulmonar/citología , Arteria Pulmonar/inmunología , Arteria Pulmonar/patología , Quimera por Trasplante/inmunología , Remodelación Vascular/genética
13.
Proc Natl Acad Sci U S A ; 116(26): 13122-13130, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31152133

RESUMEN

Iron deficiency augments hypoxic pulmonary arterial pressure in healthy individuals and exacerbates pulmonary arterial hypertension (PAH) in patients, even without anemia. Conversely, iron supplementation has been shown to be beneficial in both settings. The mechanisms underlying the effects of iron availability are not known, due to lack of understanding of how cells of the pulmonary vasculature respond to changes in iron levels. The iron export protein ferroportin (FPN) and its antagonist peptide hepcidin control systemic iron levels by regulating release from the gut and spleen, the sites of absorption and recycling, respectively. We found FPN to be present in pulmonary arterial smooth muscle cells (PASMCs) and regulated by hepcidin cell autonomously. To interrogate the importance of this regulation, we generated mice with smooth muscle-specific knock in of the hepcidin-resistant isoform fpn C326Y. While retaining normal systemic iron levels, this model developed PAH and right heart failure as a consequence of intracellular iron deficiency and increased expression of the vasoconstrictor endothelin-1 (ET-1) within PASMCs. PAH was prevented and reversed by i.v. iron and by the ET receptor antagonist BQ-123. The regulation of ET-1 by iron was also demonstrated in healthy humans exposed to hypoxia and in PASMCs from PAH patients with mutations in bone morphogenetic protein receptor type II. Such mutations were further associated with dysregulation of the HAMP/FPN axis in PASMCs. This study presents evidence that intracellular iron deficiency specifically within PASMCs alters pulmonary vascular function. It offers a mechanistic underpinning for the known effects of iron availability in humans.


Asunto(s)
Deficiencias de Hierro , Miocitos del Músculo Liso/patología , Hipertensión Arterial Pulmonar/etiología , Arteria Pulmonar/patología , Administración Intravenosa , Animales , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Modelos Animales de Enfermedad , Antagonistas de los Receptores de la Endotelina A/administración & dosificación , Endotelina-1/metabolismo , Técnicas de Sustitución del Gen , Hepcidinas/metabolismo , Humanos , Hierro/administración & dosificación , Masculino , Ratones , Miocitos del Músculo Liso/metabolismo , Hipertensión Arterial Pulmonar/patología , Hipertensión Arterial Pulmonar/prevención & control , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Receptor de Endotelina A/metabolismo , Regulación hacia Arriba
14.
Int J Mol Sci ; 23(19)2022 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-36233060

RESUMEN

Idiopathic pulmonary arterial hypertension (IPAH) is a disease with complex etiology. Currently, IPAH treatment is limited, and patients' prognosis is poor. This study aimed to explore new therapeutic targets in IPAH through bioinformatics. Two data sets (GSE113439 and GSE130391) meeting the requirements were obtained from the Gene Expression Omnibus (GEO) database. Then, differentially expressed genes (DEGs) were identified and analyzed by NetworkAnalyst platform. By enriching Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG), we examined the function of DEGs. A protein-protein interaction (PPI) network was constructed to identify central genes using the CytoNCA plug-in. Finally, four central genes, ASPM, CENPE, NCAPG, and TOP2A, were screened out. We selected NCAPG for protein-level verification. We established an animal model of PAH and found that the expression of NCAPG was significantly increased in the lung tissue of PAH rats. In vitro experiments showed that the expression of NCAPG was significantly increased in proliferative pulmonary arterial smooth muscle cells (PASMCs). When NCAPG of PASMCs was knocked down, the cell proliferation was inhibited, which suggested that NCAPG was related to the proliferation of PASMCs. Therefore, these results may provide new therapeutic targets for IPAH.


Asunto(s)
Proteínas de Ciclo Celular , Miocitos del Músculo Liso , Hipertensión Arterial Pulmonar , Animales , Proteínas de Ciclo Celular/genética , Proliferación Celular , Biología Computacional , Hipertensión Pulmonar Primaria Familiar/metabolismo , Humanos , Miocitos del Músculo Liso/citología , Proteínas del Tejido Nervioso/metabolismo , Hipertensión Arterial Pulmonar/genética , Arteria Pulmonar/citología , Ratas
15.
J Mol Cell Cardiol ; 161: 9-22, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34339758

RESUMEN

OBJECTIVE: Pulmonary vascular remodeling due to excessive growth factor production and pulmonary artery smooth muscle cells (PASMCs) proliferation is the hallmark feature of pulmonary arterial hypertension (PAH). Recent studies suggest that miR-663 is a potent modulator for tumorigenesis and atherosclerosis. However, whether miR-663 involves in pulmonary vascular remodeling is still unclear. METHODS AND RESULTS: By using quantitative RT-PCR, we found that miR-663 was highly expressed in normal human PASMCs. In contrast, circulating level of miR-663 dramatically reduced in PAH patients. In addition, in situ hybridization showed that expression of miR-663 was decreased in pulmonary vasculature of PAH patients. Furthermore, MTT and cell scratch-wound assay showed that transfection of miR-663 mimics significantly inhibited platelet derived growth factor (PDGF)-induced PASMCs proliferation and migration, while knockdown of miR-663 expression enhanced these effects. Mechanistically, dual-luciferase reporter assay revealed that miR-663 directly targets the 3'UTR of TGF-ß1. Moreover, western blots and ELISA results showed that miR-663 decreased PDGF-induced TGF-ß1 expression and secretion, which in turn suppressed the downstream smad2/3 phosphorylation and collagen I expression. Finally, intratracheal instillation of adeno-miR-663 efficiently inhibited the development of pulmonary vascular remodeling and right ventricular hypertrophy in monocrotaline (MCT)-induced PAH rat models. CONCLUSION: These results indicate that miR-663 is a potential biomarker for PAH. MiR-663 decreases PDGF-BB-induced PASMCs proliferation and prevents pulmonary vascular remodeling and right ventricular hypertrophy in MCT-PAH by targeting TGF-ß1/smad2/3 signaling. These findings suggest that miR-663 may represent as an attractive approach for the diagnosis and treatment for PAH.


Asunto(s)
MicroARNs/sangre , Hipertensión Arterial Pulmonar/genética , Factor de Crecimiento Transformador beta1/metabolismo , Remodelación Vascular/genética , Anciano , Animales , Becaplermina/farmacología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Monocrotalina/toxicidad , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Hipertensión Arterial Pulmonar/inducido químicamente , Hipertensión Arterial Pulmonar/metabolismo , Arteria Pulmonar/citología , Ratas Sprague-Dawley , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/genética , Remodelación Vascular/efectos de los fármacos
16.
Am J Respir Cell Mol Biol ; 65(5): 555-567, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34185619

RESUMEN

Impaired angiogenesis function in pulmonary artery endothelial cells (PAEC) contributes to persistent pulmonary hypertension of the newborn (PPHN). Decreased nitric oxide (NO) amounts in PPHN lead to impaired mitochondrial biogenesis and angiogenesis in the lung; the mechanisms remain unclear. We hypothesized that decreased cyclic guanosine monophosphate (cGMP)-PKG (protein kinase G) signaling downstream of NO leads to decreased mitochondrial biogenesis and angiogenesis in PPHN. PPHN was induced by ductus arteriosus constriction from 128-136 days' gestation in fetal lambs. Control animals were gestation-matched lambs that did not undergo ductal constriction. PAEC isolated from PPHN lambs were treated with the sGC (soluble guanylate cyclase) activator cinaciguat, the PKG activator 8-bromo-cGMP, or the PDE-V (PDE type V) inhibitor sildenafil. Lysates were immunoblotted for mitochondrial transcription factors and electron transport chain C-I (complex I), C-II, C-III, C-IV, and C-V proteins. The in vitro angiogenesis of PAEC was evaluated by using tube-formation and scratch-recovery assays. cGMP concentrations were measured by using an enzyme immunoassay. Fetal lambs with ductal constriction were given sildenafil or control saline through continuous infusion in utero, and the lung histology, capillary counts, vessel density, and right ventricular pressure were assessed at birth. PPHN PAEC showed decreased mitochondrial transcription factor levels, electron transport chain protein levels, and in vitro tube formation and cell migration; these were restored by cinaciguat, 8-bromo-cGMP, and sildenafil. Cinaciguat and sildenafil increased cGMP concentrations in PPHN PAEC. Radial alveolar and capillary counts and vessel density were lower in PPHN lungs, and the right ventricular pressure and Fulton Index were higher in PPHN lungs; these were improved by in utero sildenafil infusion. cGMP-PKG signaling is a potential therapeutic target to restore decreased mitochondrial biogenesis and angiogenesis in PPHN.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Guanosina Monofosfato/metabolismo , Hipertensión Pulmonar/metabolismo , Neovascularización Patológica/metabolismo , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Femenino , Humanos , Hipertensión Pulmonar/sangre , Hipertensión Pulmonar/fisiopatología , Recién Nacido , Mitocondrias/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Óxido Nítrico Sintasa de Tipo III/metabolismo , Embarazo , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Ovinos , Transducción de Señal , Citrato de Sildenafil/farmacología
17.
Mol Pharmacol ; 100(2): 53-60, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34031187

RESUMEN

Pulmonary arterial hypertension (PAH) is characterized by elevated pulmonary arterial pressure and carries a very poor prognosis. Understanding of PAH pathogenesis is needed to support the development of new therapeutic strategies. Transforming growth factor ß (TGF-ß) drives vascular remodeling and increases vascular resistance by regulating differentiation and proliferation of smooth muscle cells (SMCs). Also, sphingosine-1-phosphate (S1P) has been implicated in PAH, but the relation between these two signaling mechanisms is not well understood. Here, we characterize the signaling networks downstream of TGF-ß in human pulmonary arterial smooth muscle cells (HPASMCs), which involves mothers against decapentaplegic homolog (SMAD) signaling as well as Rho GTPases. Activation of Rho GTPases regulates myocardin-related transcription factor (MRTF) and serum response factor (SRF) transcription activity and results in upregulation of contractile gene expression. Our genetic and pharmacologic data show that in HPASMCs upregulation of α smooth muscle actin (αSMA) and calponin by TGF-ß is dependent on both SMAD and Rho/MRTF-A/SRF transcriptional mechanisms.The kinetics of TGF-ß-induced myosin light chain (MLC) 2 phosphorylation, a measure of RhoA activation, are slow, as is regulation of the Rho/MRTF/SRF-induced αSMA expression. These results suggest that TGF-ß1 activates Rho/phosphorylated MLC2 through an indirect mechanism, which was confirmed by sensitivity to cycloheximide treatment. As a potential mechanism for this indirect action, TGF-ß1 upregulates mRNA for sphingosine kinase (SphK1), the enzyme that produces S1P, an upstream Rho activator, as well as mRNA levels of the S1P receptor (S1PR) 3. SphK1 inhibitor and S1PR3 inhibitors (PF543 and TY52156/VPC23019) reduce TGF-ß1-induced αSMA upregulation. Overall, we propose a model in which TGF-ß1 activates Rho/MRTF-A/SRF by potentiating an autocrine/paracrine S1P signaling mechanism through SphK1 and S1PR3. SIGNIFICANCE STATEMENT: In human pulmonary arterial smooth muscle cells, transforming growth factor ß depends on sphingosine-1-phosphate signaling to bridge the interaction between mothers against decapentaplegic homolog and Rho/myocardin-related transcription factor (MRTF) signaling in regulating α smooth muscle actin (αSMA) expression. The Rho/MRTF pathway is a signaling node in the αSMA regulatory network and is a potential therapeutic target for the treatment of pulmonary arterial hypertension.


Asunto(s)
Lisofosfolípidos/metabolismo , Arteria Pulmonar/citología , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Factor de Crecimiento Transformador beta1/farmacología , Actinas/genética , Proteínas de Unión al Calcio/genética , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas de Microfilamentos/genética , Contracción Muscular/efectos de los fármacos , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Factor de Respuesta Sérica/genética , Proteínas Smad/genética , Proteínas Smad/metabolismo , Esfingosina/metabolismo , Calponinas
18.
J Cell Mol Med ; 25(22): 10663-10673, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34698450

RESUMEN

The proliferation of pulmonary artery smooth muscle cells (PASMCs) is an important cause of pulmonary vascular remodelling in hypoxia-induced pulmonary hypertension (HPH). However, its underlying mechanism has not been well elucidated. Connexin 43 (Cx43) plays crucial roles in vascular smooth muscle cell proliferation in various cardiovascular diseases. Here, the male Sprague-Dawley (SD) rats were exposed to hypoxia (10% O2 ) for 21 days to induce rat HPH model. PASMCs were treated with CoCl2 (200 µM) for 24 h to establish the HPH cell model. It was found that hypoxia up-regulated the expression of Cx43 and phosphorylation of Cx43 at Ser 368 in rat pulmonary arteries and PASMCs, and stimulated the proliferation and migration of PASMCs. HIF-1α inhibitor echinomycin attenuated the CoCl2 -induced Cx43 expression and phosphorylation of Cx43 at Ser 368 in PASMCs. The interaction between HIF-1α and Cx43 promotor was also identified using chromatin immunoprecipitation assay. Moreover, Cx43 specific blocker (37,43 Gap27) or knockdown of Cx43 efficiently alleviated the proliferation and migration of PASMCs under chemically induced hypoxia. Therefore, the results above suggest that HIF-1α, as an upstream regulator, promotes the expression of Cx43, and the HIF-1α/Cx43 axis regulates the proliferation and migration of PASMCs in HPH.


Asunto(s)
Conexina 43/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Conexina 43/agonistas , Conexina 43/genética , Hipoxia/genética , Hipoxia/metabolismo , Inmunohistoquímica , Modelos Biológicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Ratas
19.
Am J Physiol Lung Cell Mol Physiol ; 320(5): L688-L704, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33502939

RESUMEN

Early therapeutic effect of intratracheally (IT)-administered extracellular vesicles secreted by mesenchymal stem cells (MSC-EVs) has been demonstrated in a rat model of bronchopulmonary dysplasia (BPD) involving hyperoxia exposure in the first 2 postnatal weeks. The aim of this study was to evaluate the protective effects of IT-administered MSC-EVs in the long term. EVs were produced from MSCs following GMP standards. At birth, rats were distributed in three groups: (a) animals raised in ambient air for 6 weeks (n = 10); and animals exposed to 60% hyperoxia for 2 weeks and to room air for additional 4 weeks and treated with (b) IT-administered saline solution (n = 10), or (c) MSC-EVs (n = 10) on postnatal days 3, 7, 10, and 21. Hyperoxia exposure produced significant decreases in total number of alveoli, total surface area of alveolar air spaces, and proliferation index, together with increases in mean alveolar volume, mean linear intercept and fibrosis percentage; all these morphometric changes were prevented by MSC-EVs treatment. The medial thickness index for <100 µm vessels was higher for hyperoxia-exposed/sham-treated than for normoxia-exposed rats; MSC-EV treatment significantly reduced this index. There were no significant differences in interstitial/alveolar and perivascular F4/8-positive and CD86-positive macrophages. Conversely, hyperoxia exposure reduced CD163-positive macrophages both in interstitial/alveolar and perivascular populations and MSC-EV prevented these hyperoxia-induced reductions. These findings further support that IT-administered EVs could be an effective approach to prevent/treat BPD, ameliorating the impaired alveolarization and pulmonary artery remodeling also in a long-term model. M2 macrophage polarization could play a role through anti-inflammatory and proliferative mechanisms.


Asunto(s)
Displasia Broncopulmonar/complicaciones , Modelos Animales de Enfermedad , Vesículas Extracelulares/fisiología , Lesión Pulmonar/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Administración por Inhalación , Animales , Animales Recién Nacidos , Femenino , Hiperoxia/fisiopatología , Lesión Pulmonar/etiología , Lesión Pulmonar/patología , Masculino , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Ratas , Ratas Sprague-Dawley , Tráquea
20.
Am J Physiol Heart Circ Physiol ; 321(3): H542-H557, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34296965

RESUMEN

Pulmonary arterial hypertension (PAH) is a fatal cardiopulmonary disease characterized by increased vascular cell proliferation with apoptosis resistance and occlusive remodeling of the small pulmonary arteries. The Notch family of proteins subserves proximal signaling of an evolutionarily conserved pathway that effects cell proliferation, fate determination, and development. In endothelial cells (ECs), Notch receptor 2 (Notch2) was shown to promote endothelial apoptosis. However, a pro- or antiproliferative role for Notch2 in pulmonary endothelial proliferation and ensuing PAH is unknown. We postulated that suppressed Notch2 signaling drives pulmonary endothelial proliferation in the context of PAH. We observed that levels of Notch2 are ablated in lungs from PAH subjects compared with non-PAH controls. Notch2 expression was attenuated in human pulmonary artery endothelial cells (hPAECs) exposed to vasoactive stimuli including hypoxia, TGF-ß, ET-1, and IGF-1. Notch2-deficient hPAECs activated Akt, Erk1/2, and antiapoptotic protein Bcl-2 and reduced levels of p21cip and Bax associated with increased EC proliferation and reduced apoptosis. In addition, Notch2 suppression elicited a paradoxical activation of Notch1 and canonical Notch target gene Hes1, Hey1, and Hey2 transcription. Furthermore, reduction in Rb and increased E2F1 binding to the Notch1 promoter appear to explain the Notch1 upregulation. Yet, when Notch1 was decreased in Notch2-suppressed cells, the wound injury response was augmented. In aggregate, our results demonstrate that loss of Notch2 in hPAECs derepresses Notch1 and elicits EC hallmarks of PAH. Augmented EC proliferation upon Notch1 knockdown points to a context-dependent role for Notch1 and 2 in endothelial cell homeostasis.NEW & NOTEWORTHY This study demonstrates a previously unidentified role for Notch2 in the maintenance of lung vascular endothelial cell quiescence and pulmonary artery hypertension (PAH). A key novel finding is that Notch2 suppression activates Notch1 via Rb-E2F1-mediated signaling and induces proliferation and apoptosis resistance in human pulmonary artery endothelial cells. Notably, PAH patients show reduced levels of endothelial Notch2 in their pulmonary arteries, supporting Notch2 as a fundamental driver of PAH pathogenesis.


Asunto(s)
Proliferación Celular , Células Endoteliales/metabolismo , Hipertensión Pulmonar/metabolismo , Receptor Notch2/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Células Endoteliales/fisiología , Endotelio Vascular/citología , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Receptor Notch1/metabolismo , Receptor Notch2/genética , Proteínas Represoras/metabolismo , Factor de Transcripción HES-1/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda