Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
1.
Int J Mol Sci ; 24(7)2023 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-37047587

RESUMEN

Mast cells are responsible for IgE-dependent allergic responses, but they also produce various bioactive mediators and contribute to the pathogenesis of various cardiovascular diseases, including pulmonary hypertension (PH). The importance of lipid mediators in the pathogenesis of PH has become evident in recent years, as exemplified by prostaglandin I2, the most central therapeutic target in pulmonary arterial hypertension. New bioactive lipids other than eicosanoids have also been identified that are associated with the pathogenesis of PH. However, it remains largely unknown how mast cell-derived lipid mediators are involved in pulmonary vascular remodeling. Recently, it has been demonstrated that mast cells produce epoxidized n-3 fatty acid (n-3 epoxides) in a degranulation-independent manner, and that n-3 epoxides produced by mast cells regulate the abnormal activation of pulmonary fibroblasts and suppress the progression of pulmonary vascular remodeling. This review summarizes the role of mast cells and bioactive lipids in the pathogenesis of PH. In addition, we introduce the pathophysiological role and therapeutic potential of n-3 epoxides, a mast cell-derived novel lipid mediator, in the pulmonary vascular remodeling in PH. Further knowledge of mast cells and lipid mediators is expected to lead to the development of innovative therapies targeting pulmonary vascular remodeling.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Ácidos Grasos Insaturados , Hipertensión Pulmonar , Lisofosfolípidos , Mastocitos , Arteria Pulmonar , Mastocitos/metabolismo , Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Hipertensión Pulmonar/inmunología , Hipertensión Pulmonar/patología , Arteria Pulmonar/inmunología , Arteria Pulmonar/patología , Lisofosfolípidos/biosíntesis , Ácidos Grasos Insaturados/biosíntesis , Humanos , Animales
2.
Int J Mol Sci ; 23(5)2022 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-35269553

RESUMEN

Pulmonary arterial hypertension (PAH) is a progressive disease characterized by (mal)adaptive remodeling of the pulmonary vasculature, which is associated with inflammation, fibrosis, thrombosis, and neovascularization. Vascular remodeling in PAH is associated with cellular metabolic and inflammatory reprogramming that induce profound endothelial and smooth muscle cell phenotypic changes. Multiple signaling pathways and regulatory loops act on metabolic and inflammatory mediators which influence cellular behavior and trigger pulmonary vascular remodeling in vivo. This review discusses the role of bioenergetic and inflammatory impairments in PAH development.


Asunto(s)
Músculo Liso Vascular/patología , Hipertensión Arterial Pulmonar/patología , Arteria Pulmonar/patología , Animales , Metabolismo Energético , Humanos , Músculo Liso Vascular/inmunología , Hipertensión Arterial Pulmonar/inmunología , Arteria Pulmonar/inmunología , Transducción de Señal , Remodelación Vascular
3.
JCI Insight ; 6(15)2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34185707

RESUMEN

We previously reported heightened expression of the human endogenous retroviral protein HERV-K deoxyuridine triphosphate nucleotidohydrolase (dUTPase) in circulating monocytes and pulmonary arterial (PA) adventitial macrophages of patients with PA hypertension (PAH). Furthermore, recombinant HERV-K dUTPase increased IL-6 in PA endothelial cells (PAECs) and caused pulmonary hypertension in rats. Here we show that monocytes overexpressing HERV-K dUTPase, as opposed to GFP, can release HERV-K dUTPase in extracellular vesicles (EVs) that cause pulmonary hypertension in mice in association with endothelial mesenchymal transition (EndMT) related to induction of SNAIL/SLUG and proinflammatory molecules IL-6 as well as VCAM1. In PAECs, HERV-K dUTPase requires TLR4-myeloid differentiation primary response-88 to increase IL-6 and SNAIL/SLUG, and HERV-K dUTPase interaction with melanoma cell adhesion molecule (MCAM) is necessary to upregulate VCAM1. TLR4 engagement induces p-p38 activation of NF-κB in addition to p-pSMAD3 required for SNAIL and pSTAT1 for IL-6. HERV-K dUTPase interaction with MCAM also induces p-p38 activation of NF-κB in addition to pERK1/2-activating transcription factor-2 (ATF2) to increase VCAM1. Thus in PAH, monocytes or macrophages can release HERV-K dUTPase in EVs, and HERV-K dUTPase can engage dual receptors and signaling pathways to subvert PAEC transcriptional machinery to induce EndMT and associated proinflammatory molecules.


Asunto(s)
Retrovirus Endógenos , Transición Epitelial-Mesenquimal/inmunología , Hipertensión Pulmonar , Macrófagos/inmunología , Monocitos/inmunología , Arteria Pulmonar , Pirofosfatasas/metabolismo , Animales , Antígeno CD146/metabolismo , Retrovirus Endógenos/metabolismo , Retrovirus Endógenos/patogenicidad , Células Endoteliales/metabolismo , Hipertensión Pulmonar/inmunología , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/virología , Inflamación/metabolismo , Inflamación/virología , Ratones , Arteria Pulmonar/inmunología , Arteria Pulmonar/patología , Transducción de Señal , Factores de Transcripción de la Familia Snail/metabolismo
5.
Pathol Res Pract ; 220: 153382, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33647866

RESUMEN

OBJECTIVE: The pulmonary vascular remodeling in systemic sclerosis (SSc) is poorly understood and animal models are lacking. Type V collagen (COLV) is elevated in SSc and is implicated in the pathogenesis, and immunization with human COLV induces SSc-like skin and lung changes in rabbits and mice. Here we tested the hypothesis that COLV immunization will induce pathological and functional changes that phenocopy SSc-associated pulmonary vascular disease. METHODS: Pulmonary vascular changes in rabbits immunized with human COLV were extensively characterized by a combination of histology, electron microscopy and immunohistochemistry. Physiologic changes induced by COLV in explanted pulmonary artery rings were evaluated. The pattern of histopathologic alterations and gene expression induced in immunized rabbits were compared to those in SSc patients. RESULTS: COLV immunization was accompanied by striking pulmonary vascular abnormalities, characterized by reduced capillary density, perivascular inflammation, endothelial cell injury and collagen accumulation, that closely phenocopy changes seen in SSc patients. Moreover, pulmonary arteries from immunized rabbits showed impaired ex vivo vascular relaxation. Expression of COL5A2 was significantly increased in the lungs from immunized rabbits (p = 0.02), as well as in patients with SSc (P = 0.02). CONCLUSION: COLV immunity in rabbits is associated with marked vascular remodeling in the lung that phenocopies early-stage human SSc-associated pulmonary vascular disease. COLV immunization therefore represents a novel approach to model SSc pulmonary vascular pathology. Moreover, our findings suggest that COLV might represent a novel pathogenic autoantigen in SSc and future studies with the present model should be developed for possible association with PAH.


Asunto(s)
Colágeno Tipo V/inmunología , Pulmón/irrigación sanguínea , Arteria Pulmonar/patología , Esclerodermia Sistémica/patología , Remodelación Vascular , Adulto , Animales , Estudios de Casos y Controles , Colágeno Tipo V/metabolismo , Modelos Animales de Enfermedad , Femenino , Hemodinámica , Humanos , Persona de Mediana Edad , Arteria Pulmonar/inmunología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , Conejos , Esclerodermia Sistémica/inmunología , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/fisiopatología
6.
Physiol Rep ; 9(3): e14613, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33512067

RESUMEN

The Transient Receptor Potential Vanilloid 4 (TRPV4) of endothelial cells contributes to many important functions including the regulation of Ca2+ homeostasis, cell volume, endothelial barrier permeability, and smooth muscle tone. However, its role in the transition of endothelial cells toward a pro-inflammatory phenotype has not been studied so far. Using both arterial and venous endothelial cells, we first show that the pharmacological activation of TRPV4 channels with GSK1016790A, a potent TRPV4 agonist, triggers robust and sustained Ca2+ increases, which are blocked by both TRPV4 antagonists HC067047 and RN9893. TRPV4 activation also triggers the actin cytoskeleton and adherens junction (VE-Cadherin) rearrangement in both arterial and venous endothelial cells and leads to rapid decreases of trans-endothelial electrical resistance. In addition to its effect on endothelial barrier integrity, TRPV4 activation selectively increases ICAM-1 surface expression in arterial and venous endothelial cells, due to the stimulation of ICAM-1 gene expression through the NF-κB transcription factor. TRPV4 channel activation also induced apoptosis of venous and arterial endothelial cells, while TRPV4 blockade reduced apoptosis, even in the absence of TRPV4 activation. As altered barrier integrity, increased adhesion molecule expression and apoptosis are hallmarks of the pro-inflammatory state of endothelial cells, our results indicate that TRPV4 channel activity can induce the transition of both venous and arterial endothelial cells toward a pro-inflammatory phenotype.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Leucina/análogos & derivados , Arteria Pulmonar/efectos de los fármacos , Sulfonamidas/farmacología , Canales Catiónicos TRPV/agonistas , Antígenos CD/metabolismo , Apoptosis , Cadherinas/metabolismo , Señalización del Calcio , Células Cultivadas , Impedancia Eléctrica , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Leucina/farmacología , FN-kappa B/metabolismo , Permeabilidad , Fenotipo , Arteria Pulmonar/inmunología , Arteria Pulmonar/metabolismo , Canales Catiónicos TRPV/metabolismo
7.
Sci Rep ; 10(1): 13708, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32792588

RESUMEN

Endothelial cell (EC) inflammation and permeability are critical pathogenic mechanisms in many inflammatory conditions including acute lung injury. In this study, we investigated the role of ATG7, an essential autophagy regulator with no autophagy-unrelated functions, in the mechanism of EC inflammation and permeability. Knockdown of ATG7 using si-RNA significantly attenuated thrombin-induced expression of proinflammatory molecules such as IL-6, MCP-1, ICAM-1 and VCAM-1. Mechanistic study implicated reduced NF-κB activity in the inhibition of EC inflammation in ATG7-silenced cells. Moreover, depletion of ATG7 markedly reduced the binding of RelA/p65 to DNA in the nucleus. Surprisingly, the thrombin-induced degradation of IκBα in the cytosol was not affected in ATG7-depleted cells, suggesting a defect in the translocation of released RelA/p65 to the nucleus in these cells. This is likely due to suppression of thrombin-induced phosphorylation and thereby inactivation of Cofilin1, an actin-depolymerizing protein, in ATG7-depleted cells. Actin stress fiber dynamics are required for thrombin-induced translocation of RelA/p65 to the nucleus, and indeed our results showed that ATG7 silencing inhibited this response via inactivation of Cofilin1. ATG7 silencing also reduced thrombin-mediated EC permeability by inhibiting the disassembly of VE-cadherin at adherens junctions. Together, these data uncover a novel function of ATG7 in mediating EC inflammation and permeability, and provide a mechanistic basis for the linkage between autophagy and EC dysfunction.


Asunto(s)
Proteína 7 Relacionada con la Autofagia/metabolismo , Autofagia , Permeabilidad de la Membrana Celular , Endotelio Vascular/inmunología , Inflamación/inmunología , FN-kappa B/metabolismo , Arteria Pulmonar/inmunología , Proteína 7 Relacionada con la Autofagia/genética , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Regulación de la Expresión Génica , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , FN-kappa B/genética , Fosforilación , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Transducción de Señal , Trombina/farmacología
8.
Int J Mol Sci ; 21(14)2020 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-32708482

RESUMEN

According to a widespread theory, thrombotic masses are not formed in the pulmonary artery (PA) but result from migration of blood clots from the venous system. This concept has prevailed in clinical practice for more than a century. However, a new technologic era has brought forth more diagnostic possibilities, and it has been shown that thrombotic masses in the PA could, in many cases, be found without any obvious source of emboli. Chronic obstructive pulmonary disease, asthma, sickle cell anemia, emergency and elective surgery, viral pneumonia, and other conditions could be complicated by PA thrombosis development without concomitant deep vein thrombosis (DVT). Different pathologies have different causes for local PA thrombotic process. As evidenced by experimental results and clinical observations, endothelial and platelet activation are the crucial mechanisms of this process. Endothelial dysfunction can impair antithrombotic function of the arterial wall through downregulation of endothelial nitric oxide synthase (eNOS) or via stimulation of adhesion receptor expression. Hypoxia, proinflammatory cytokines, or genetic mutations may underlie the procoagulant phenotype of the PA endothelium. Both endotheliocytes and platelets could be activated by protease mediated receptor (PAR)- and receptors for advanced glycation end (RAGE)-dependent mechanisms. Hypoxia, in particular induced by high altitudes, could play a role in thrombotic complications as a trigger of platelet activity. In this review, we discuss potential mechanisms of PA thrombosis in situ.


Asunto(s)
Plaquetas/metabolismo , Micropartículas Derivadas de Células/metabolismo , Endotelio Vascular/metabolismo , Activación Plaquetaria/inmunología , Arteria Pulmonar/metabolismo , Embolia Pulmonar/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Trombosis/metabolismo , Plaquetas/enzimología , Plaquetas/inmunología , Hipoxia de la Célula , Micropartículas Derivadas de Células/patología , Citocinas/metabolismo , Endotelio Vascular/enzimología , Endotelio Vascular/inmunología , Proteína HMGB1/metabolismo , Humanos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Arteria Pulmonar/enzimología , Arteria Pulmonar/inmunología , Arteria Pulmonar/patología , Embolia Pulmonar/genética , Embolia Pulmonar/fisiopatología , Embolia Pulmonar/virología , Receptor PAR-1/metabolismo , Factores de Riesgo
9.
Biomed Pharmacother ; 129: 110355, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32559622

RESUMEN

Pulmonary arterial hypertension (PAH) is a progressive disease with limited effective therapies that can be lethal in the terminal stage. Since the universal use of traditional PAH-specific drugs affecting the nitric oxide pathway, endothelin pathway and prostacyclin pathway, major advances to treat this devastating condition have been made. For example, it has been recently demonstrated that PAH is associated with various dysfunctional genes and molecular signaling pathways, abnormal metabolism and endocrine systems, inflammation and immune dysfunction. Numerous descriptions of novel mechanisms in PAH have been presented, and some new drugs targeted to ease, reverse or cure PAH have been developed at the laboratory-level and/or tested in clinical trials. There are also some investigated drugs that have shown promising efficacy and safety in animal models but have failed in human beings. Inspiringly, immunotherapies also seem to be valuable therapies for PAH patients. In this review, we update the advances in investigational pharmacotherapy and immunotherapy of PAH. Hopefully this work can help promote more non-invasive treatment for PAH patients.


Asunto(s)
Antihipertensivos/uso terapéutico , Presión Arterial/efectos de los fármacos , Inmunoterapia , Hipertensión Arterial Pulmonar/terapia , Arteria Pulmonar/efectos de los fármacos , Animales , Antihipertensivos/efectos adversos , Humanos , Inmunosupresores/uso terapéutico , Inmunoterapia/efectos adversos , Terapia Molecular Dirigida , Hipertensión Arterial Pulmonar/inmunología , Hipertensión Arterial Pulmonar/metabolismo , Hipertensión Arterial Pulmonar/fisiopatología , Arteria Pulmonar/inmunología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , Transducción de Señal , Vacunas/uso terapéutico
10.
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
11.
Immunol Lett ; 218: 22-29, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31866401

RESUMEN

OBJECTIVE: Pulmonary arterial hypertension (PAH) is an intractable complication in connective tissue diseases, but the pathological mechanisms responsible for progression remain obscure. This study aims to test whether patient IgG possesses biological activity promoting the migration of pulmonary artery smooth muscle cells (PASMCs). METHODS: Cell migration was estimated by lamellipodia formation and by utilizing a Boyden chamber method. The specificity of autoantibodies was established by western blotting, ELISA, and immunocytochemistry. The target antigen was investigated by mass spectrometry. RESULTS: IgG obtained from a patient with systemic lupus erythematosus (SLE) accompanied by PAH was found to promote lamellipodia formation and migration of PASMCs. The IgG bound to a ∼50 kDa protein expressed on the cell membrane, and in the cytoplasm and nucleus. This molecule was identified as enolase 1. Removal of enolase 1-binding antibodies from the IgG fraction, or treatment of the cells with an enolase inhibitor, significantly suppressed the migration of PASMCs. CONCLUSION: Patients with SLE may possess autoantibodies to enolase 1 which stimulate the migration of PASMCs and are likely to play a role in the progression of PAH.


Asunto(s)
Autoanticuerpos/inmunología , Biomarcadores de Tumor/inmunología , Proteínas de Unión al ADN/inmunología , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/inmunología , Miocitos del Músculo Liso/metabolismo , Fosfopiruvato Hidratasa/inmunología , Hipertensión Arterial Pulmonar/complicaciones , Hipertensión Arterial Pulmonar/metabolismo , Proteínas Supresoras de Tumor/inmunología , Secuencia de Aminoácidos , Autoanticuerpos/sangre , Autoantígenos/inmunología , Biomarcadores de Tumor/química , Biomarcadores de Tumor/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Ligandos , Miocitos del Músculo Liso/inmunología , Fosfopiruvato Hidratasa/química , Fosfopiruvato Hidratasa/metabolismo , Unión Proteica , Hipertensión Arterial Pulmonar/diagnóstico , Arteria Pulmonar/citología , Arteria Pulmonar/inmunología , Arteria Pulmonar/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/metabolismo
12.
Eur J Pharmacol ; 867: 172823, 2020 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-31770525

RESUMEN

Leukotriene B4 (LTB4) has been found to contribute to pulmonary arterial smooth muscle cells (PASMCs) proliferation and pulmonary arterial remodeling therefore the development of pulmonary arterial hypertension (PAH). Yet, the underlying molecular mechanisms remain poorly understood. The present study aims to address this issue. Our results demonstrate that LTB4 dose- and time-dependently induced proliferation of primary cultured rat PASMCs, this was accompanied with the activation of phosphatidylinositol-3-kinase/Akt (PI3K/Akt) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathways, and consequent inactivation of glycogen synthase kinase-3ß (GSK-3ß), up-regulation of ß-catenin and induction of cyclin D1 expression. The presence of PI3K inhibitor (LY294002) or MEK inhibitor (U0126) or prior silencing of ß-catenin with siRNA suppressed LTB4-induced cyclin D1 up-regulation and PASMCs proliferation. In addition, inactivation or lack of GSK-3ß up-regulated ß-catenin and cyclin D1 in PASMCs. Taken together, our study indicates that activation of PI3K/Akt and ERK1/2 pathways mediates LTB4-induced PASMCs proliferation by modulating GSK-3ß/ß-catenin/cyclin D1 axis and suggests that targeting this pathway might have potential value in alleviating vascular remodeling and benefit PAH.


Asunto(s)
Hipertensión Pulmonar/inmunología , Leucotrieno B4/inmunología , Miocitos del Músculo Liso/patología , Arteria Pulmonar/patología , Remodelación Vascular/inmunología , Animales , Butadienos/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cromonas/farmacología , Ciclina D1/inmunología , Ciclina D1/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Hipertensión Pulmonar/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Morfolinas/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/inmunología , Nitrilos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/inmunología , Cultivo Primario de Células , Arteria Pulmonar/inmunología , Arteria Pulmonar/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Remodelación Vascular/efectos de los fármacos , beta Catenina/genética , beta Catenina/metabolismo
13.
J Immunol ; 203(11): 2862-2871, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31611261

RESUMEN

Pulmonary hypertension (PH) is a debilitating disease characterized by remodeling of the lung vasculature. In rodents, resistin-like molecule-α (RELMα, also known as HIMF or FIZZ1) can induce PH, but the signaling mechanisms are still unclear. In this study, we used human lung samples and a hypoxia-induced mouse model of PH. We found that the human homolog of RELMα, human (h) resistin, is upregulated in macrophage-like inflammatory cells from lung tissues of patients with idiopathic PH. Additionally, at PH onset in the mouse model, we observed RELMα-dependent lung accumulation of macrophages that expressed high levels of the key damage-associated molecular pattern (DAMP) molecule high-mobility group box 1 (HMGB1) and its receptor for advanced glycation end products (RAGE). In vitro, RELMα/hresistin-induced macrophage-specific HMGB1/RAGE expression and facilitated HMGB1 nucleus-to-cytoplasm translocation and extracellular secretion. Mechanistically, hresistin promoted HMGB1 posttranslational lysine acetylation by preserving the NAD+-dependent deacetylase sirtuin (Sirt) 1 in human macrophages. Notably, the hresistin-stimulated macrophages promoted apoptosis-resistant proliferation of human pulmonary artery smooth muscle cells in an HMGB1/RAGE-dependent manner. In the mouse model, RELMα also suppressed the Sirt1 signal in pulmonary macrophages in the early posthypoxic period. Notably, recruited macrophages in the lungs of these mice carried the RELMα binding partner Bruton tyrosine kinase (BTK). hResistin also mediated the migration of human macrophages by activating BTK in vitro. Collectively, these data reveal a vascular-immune cellular interaction in the early PH stage and suggest that targeting RELMα/DAMP-driven macrophages may offer a promising strategy to treat PH and other related vascular inflammatory diseases.


Asunto(s)
Hipertensión Pulmonar/inmunología , Péptidos y Proteínas de Señalización Intercelular/inmunología , Macrófagos Alveolares/inmunología , Arteria Pulmonar/inmunología , Remodelación Vascular/inmunología , Adolescente , Animales , Células Cultivadas , Humanos , Hipertensión Pulmonar/patología , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Macrófagos Alveolares/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Arteria Pulmonar/patología
14.
Am J Physiol Heart Circ Physiol ; 317(5): H1093-H1101, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31490732

RESUMEN

Pulmonary arterial hypertension (PAH) is a fatal disease with a median survival of only 5-7 yr. PAH is characterized by remodeling of the pulmonary vasculature causing reduced pulmonary arterial compliance (PAC) and increased pulmonary vascular resistance (PVR), ultimately resulting in right ventricular failure and death. Better therapies for PAH will require a paradigm shift in our understanding of the early pathophysiology. PAC decreases before there is an increase in the PVR. Unfortunately, present treatment has little effect on PAC. The loss of compliance correlates with extracellular matrix remodeling and fibrosis in the pulmonary vessels, which have been linked to chronic perivascular inflammation and immune dysregulation. However, what initiates the perivascular inflammation and immune dysregulation in PAH is unclear. Alteration of the gut microbiota composition and function underlies the level of immunopathogenic involvement in several diseases, including atherosclerosis, obesity, diabetes mellitus, and depression, among others. In this review, we discuss evidence that raises the possibility of an etiologic role for changes in the gut and circulating microbiome in the initiation of perivascular inflammation in the early pathogenesis of PAH.


Asunto(s)
Presión Arterial , Bacterias/metabolismo , Microbioma Gastrointestinal , Mediadores de Inflamación/sangre , Intestinos/microbiología , Hipertensión Arterial Pulmonar/microbiología , Arteria Pulmonar/microbiología , Animales , Bacterias/inmunología , Disbiosis , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/inmunología , Hipertensión Arterial Pulmonar/sangre , Hipertensión Arterial Pulmonar/inmunología , Hipertensión Arterial Pulmonar/fisiopatología , Arteria Pulmonar/inmunología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , Factores de Riesgo , Transducción de Señal
15.
Int J Chron Obstruct Pulmon Dis ; 14: 1323-1332, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31417249

RESUMEN

Background: Previous studies have shown that the arterial wall is a potential source of inflammatory markers in COPD. Here, we sought to compare the expression of acute phase reactants (APRs) in COPD patients and controls both at the local (pulmonary arteries and lung parenchyma) and systemic (peripheral blood leukocytes and plasma) compartments. Methods: Consecutive patients undergoing elective surgery for suspected primary lung cancer were eligible for the study. Patients were categorized either as COPD or control group based on the spirometry results. Pulmonary arteries and lung parenchyma sections, peripheral blood leukocytes, and plasma samples were obtained from all participants. Gene expression levels of C-reactive protein (CRP) and serum amyloid A (SAA1, SAA2, and SAA4) were evaluated in tissue samples and peripheral blood leukocytes by reverse transciption-PCR. Plasma CRP and SAA protein levels were measured by enzyme-linked immunosorbent assays. Proteins were evaluated in paraffin-embedded lung tissues by immunohistochemistry. Results: A total of 40 patients with COPD and 62 controls were enrolled. We did not find significant differences in the gene expression between COPD and control group. Both CRP and SAA were overexpressed in the lung parenchyma compared with pulmonary arteries and peripheral blood leukocytes. The expression of SAA was significantly higher in the lung parenchyma than in the pulmonary artery (2-fold higher for SAA1 and SAA4, P=0.015 and P<0.001, respectively; 8-fold higher for SAA2, P<0.001) and peripheral blood leukocytes (16-fold higher for SAA1, 439-fold higher for SAA2, and 5-fold higher for SAA4; P<0.001). No correlation between plasma levels of inflammatory markers and their expression in the lung and peripheral blood leukocytes was observed. Conclusions: The expression of SAA in lung parenchyma is higher than in pulmonary artery and peripheral blood leukocytes. Notably, no associations were noted between lung expression of APRs and their circulating plasma levels, making the leakage of inflammatory proteins from the lung to the bloodstream unlikely. Based on these results, other potential sources of systemic inflammation in COPD (eg, the liver) need further scrutiny.


Asunto(s)
Reacción de Fase Aguda , Pulmón , Linfocitos/inmunología , Arteria Pulmonar , Enfermedad Pulmonar Obstructiva Crónica , Proteína Amiloide A Sérica/análisis , Proteínas de Fase Aguda/análisis , Proteínas de Fase Aguda/inmunología , Reacción de Fase Aguda/sangre , Reacción de Fase Aguda/inmunología , Correlación de Datos , Femenino , Humanos , Pulmón/inmunología , Pulmón/patología , Masculino , Persona de Mediana Edad , Arteria Pulmonar/inmunología , Arteria Pulmonar/patología , Enfermedad Pulmonar Obstructiva Crónica/sangre , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/patología , Espirometría/métodos
16.
Mod Rheumatol ; 29(2): 388-392, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27535710

RESUMEN

A 20-year-old man presented with recurrent hemoptysis for seven months. A small subpleural nodule in his right lower lobe was found and excised surgically. Based on the presence of antiphospholipid antibodies (aPL) and vascular wall hypertrophy without vasculitis or an intraluminal thrombus, nonthrombotic proliferative vasculopathy (NTPV) affecting pulmonary arteries was diagnosed. Recently, aPL have been postulated to directly induce the proliferation of vascular cells in the intima and media, leading to NTPV. We review 5 cases of NTPV-associated aPL with critical ischemia in the lower extremities and gastrointestinal infarction. NTPV-associated aPL might be distinct from classic antiphospholipid syndrome and should be considered in aPL-positive patients who present with vascular occlusions of medium-sized vessels in the absence of atherosclerotic risk factors and systemic or local inflammation.


Asunto(s)
Anticuerpos Antifosfolípidos/análisis , Síndrome Antifosfolípido , Hemoptisis , Arteria Pulmonar , Túnica Íntima , Túnica Media , Vasculitis , Síndrome Antifosfolípido/complicaciones , Síndrome Antifosfolípido/diagnóstico , Síndrome Antifosfolípido/inmunología , Proliferación Celular , Diagnóstico Diferencial , Hemoptisis/diagnóstico , Hemoptisis/etiología , Humanos , Masculino , Arteria Pulmonar/inmunología , Arteria Pulmonar/patología , Túnica Íntima/patología , Túnica Íntima/fisiopatología , Túnica Media/patología , Túnica Media/fisiopatología , Vasculitis/diagnóstico , Vasculitis/etiología , Vasculitis/inmunología , Vasculitis/fisiopatología , Adulto Joven
18.
Int J Mol Sci ; 19(9)2018 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-30150544

RESUMEN

Pulmonary arterial hypertension (PAH) is a progressive disease characterized by remodeling of the pulmonary arteries, increased pulmonary infiltrates, loss of vascular cross-sectional area, and elevated pulmonary vascular resistance. Despite recent advances in the management of PAH, there is a pressing need for the development of new tools to effectively treat and reduce the risk of further complications. Dysregulated immunity underlies the development of PAH, and macrophages orchestrate both the initiation and resolution of pulmonary inflammation, thus, manipulation of lung macrophage function represents an attractive target for emerging immunomodulatory therapies, including cell-based approaches. Indeed, mesenchymal stem cell (MSC)-based therapies have shown promise, effectively modulating the macrophage fulcrum to favor an anti-inflammatory, pro-resolving phenotype, which is associated with both histological and functional benefits in preclinical models of pulmonary hypertension (PH). The complex interplay between immune system homeostasis and MSCs remains incompletely understood. Here, we highlight the importance of macrophage function in models of PH and summarize the development of MSC-based therapies, focusing on the significance of MSC exosomes (MEx) and the immunomodulatory and homeostatic mechanisms by which such therapies may afford their beneficial effects.


Asunto(s)
Micropartículas Derivadas de Células/inmunología , Exosomas/inmunología , Hipertensión Pulmonar/inmunología , Inmunomodulación/inmunología , Macrófagos/inmunología , Células Madre Mesenquimatosas/inmunología , Animales , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/trasplante , Exosomas/metabolismo , Exosomas/trasplante , Humanos , Hipertensión Pulmonar/fisiopatología , Hipertensión Pulmonar/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Arteria Pulmonar/inmunología , Arteria Pulmonar/fisiopatología
19.
PLoS One ; 13(7): e0199206, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29990318

RESUMEN

Although the accumulation of amyloidogenic proteins in neuroinflammatory conditions is generally considered pathologic, in a murine model of multiple sclerosis, amyloid-forming fibrils, comprised of hexapeptides, are anti-inflammatory. Whether these molecules modulate systemic inflammatory conditions remains unknown. We hypothesized that an amylin hexapeptide that forms fibrils can attenuate the systemic inflammatory response in a murine model of sepsis. To test this hypothesis, mice were pre-treated with either vehicle or amylin hexapeptide (20 µg) at 12 hours and 6 hours prior to intraperitoneal (i.p.) lipopolysaccharide (LPS, 20 mg/kg) administration. Illness severity and survival were monitored every 6 hours for 3 days. Levels of pro- (IL-6, TNF-α, IFN-γ) and anti-inflammatory (IL-10) cytokines were measured via ELISA at 1, 3, 6, 12, and 24 hours after LPS (i.p.). As a metric of lung injury, pulmonary artery endothelial cell (PAEC) barrier function was tested 24 hours after LPS administration by comparing lung wet-to-dry ratios, Evan's blue dye (EBD) extravasation, lung histology and caspase-3 activity. Compared to controls, pretreatment with amylin hexapeptide significantly reduced mortality (p<0.05 at 72 h), illness severity (p<0.05), and pro-inflammatory cytokine levels, while IL-10 levels were elevated (p<0.05). Amylin pretreatment attenuated LPS-induced lung injury, as demonstrated by decreased lung water and caspase-3 activity (p<0.05, versus PBS). Hence, in a murine model of systemic inflammation, pretreatment with amylin hexapeptide reduced mortality, disease severity, and preserved lung barrier function. Amylin hexapeptide may represent a novel therapeutic tool to mitigate sepsis severity and lung injury.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Proteínas Amiloidogénicas/farmacología , Polipéptido Amiloide de los Islotes Pancreáticos/química , Pulmón/efectos de los fármacos , Oligopéptidos/farmacología , Sepsis/tratamiento farmacológico , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/mortalidad , Proteínas Amiloidogénicas/síntesis química , Animales , Caspasa 3/genética , Caspasa 3/inmunología , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales/patología , Femenino , Regulación de la Expresión Génica , Inflamación , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Lipopolisacáridos/administración & dosificación , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Oligopéptidos/síntesis química , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/inmunología , Arteria Pulmonar/patología , Sepsis/inducido químicamente , Sepsis/inmunología , Sepsis/mortalidad , Análisis de Supervivencia , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
20.
Physiol Rep ; 6(11): e13721, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29894584

RESUMEN

The importance of myeloid cells in promoting neovascularization has been shown in a number of pathological settings in several organs. However, the specific role of macrophages in promoting systemic angiogenesis during pulmonary ischemia is not fully determined. Our past work suggested that cells of monocytic lineage contributed to systemic angiogenesis in the lung since clodronate-induced depletion of all macrophages resulted in attenuated neovascularization. Our current goals were to define the population of macrophages important for systemic vessel growth into the lung after the onset of pulmonary ischemia in mice. Interstitial macrophages (CD64+ MerTK+ CD11b+ ) increased significantly as did the percent of CD45+ Ly6G+ neutrophils 1 day after the induction of left lung ischemia, despite the fact there was limited cell recruitment due to complete obstruction of the left pulmonary artery in this ischemia model. Since both interstitial macrophages and neutrophils express CD11b, we used CD11b+ DTR mice and showed the critical role for these cells since CD11b+ depleted mice showed no systemic angiogenesis 7 days after the onset of ischemia when compared to control mice. Coculture of mouse aortic endothelial cells with macrophages showed increased proliferation relative to endothelial cells in culture without inflammatory cells, or pulmonary artery endothelial cells. We conclude that CD11b+ leukocytes, trapped within the lung at the onset of ischemia, contribute to growth factor release, and are critical for new blood vessel proliferation.


Asunto(s)
Isquemia/complicaciones , Pulmón/irrigación sanguínea , Pulmón/inmunología , Macrófagos/inmunología , Neovascularización Patológica , Animales , Antígeno CD11b/inmunología , Células Cultivadas , Citocinas/inmunología , Células Endoteliales/inmunología , Inflamación/inmunología , Leucocitos/inmunología , Masculino , Ratones Endogámicos C57BL , Arteria Pulmonar/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA