Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.077
Filtrar
1.
Sci Rep ; 14(1): 23745, 2024 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-39390031

RESUMEN

The study of biological soft tissue structures at the micron scale details the function of healthy and pathological tissues, which is vital in the diagnosis and treatment of diseases. Speckle based X-ray phase contrast tomographic scans at a nanometer scale have the potential to thoroughly analyse such tissues in a quantitative and qualitative manner. Diamond light source, the UKs national synchrotron facility developed and refined a 1-D X-ray speckle-based imaging technique, referred to as Fly scan mode. This novel image acquisition technique was used to perform a rapid structural composition scan of rodent lung histology samples. The rodent samples were taken from healthy and Staphylococcus aureus induced acute respiratory distress syndrome models. The analysis and cross comparison of the fly scan method, absorption-based tomography and conventional histopathology H&E staining microscopy are discussed in this paper. This analysis and cross comparison outline the ways the speckle-based technique can be of benefit. These advantages include improved soft tissue contrast, 3-D volumetric rendering, segmentation of specific gross tissue structures, quantitative analysis of gross tissue volume. A further advantage is the analysis of cellular distribution throughout the volumetric rendering of the tissue sample. The study also details the current limitations of this technique and points to ways in which future work on this imaging modality may progress.


Asunto(s)
Modelos Animales de Enfermedad , Imagenología Tridimensional , Síndrome de Dificultad Respiratoria , Tomografía Computarizada por Rayos X , Animales , Síndrome de Dificultad Respiratoria/diagnóstico por imagen , Síndrome de Dificultad Respiratoria/patología , Imagenología Tridimensional/métodos , Tomografía Computarizada por Rayos X/métodos , Pulmón/diagnóstico por imagen , Pulmón/patología , Ratas , Staphylococcus aureus , Ratones
2.
Sci Rep ; 14(1): 22001, 2024 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-39322748

RESUMEN

The pulmonary vascular permeability index (PVPI) is a quotient of the extravascular lung water (EVLW) and the pulmonary blood volume (PBV). In acute respiratory distress syndrome (ARDS), the alveolar-capillary membrane integrity is disrupted. The result is a disproportionate increase of EVLW compared to the PBV and, hence, an increase in PVPI. Thus, PVPI has repetitively been discussed to extend the definition of ARDS. Besides sex, the influence of other anthropometric variables on PVPI has not been studied so far. However, since it is known that EVLW depends on body height and sex, we hypothesize that PVPI depends on anthropometric variables as well. This prospective single-center observational study included 1533 TPTD measurements of 251 non-critically ill patients (50.6% men) undergoing elective neuro-, thoracic, or abdominal surgery at the Munich Clinic Bogenhausen of the Technical University of Munich. Multivariate regressions were used to measure the influence of sex, age, and body height on PVPI. In all patients, PVPI was significantly higher in women (P < 0.001), with 34.4% having a PVPI > 2 compared to 15.9% of men. Mean PVPI significantly decreased with height (P < 0.001) and age (P < 0.001). Multivariate regressions allowed the calculation of mean reference surfaces. The 95th percentile surface for PVPI was > 3 for small and young women and well above 2 for all but tall and elderly men. In patients who underwent (lung reduction) thoracic surgery, the PVPI before and after surgery did not differ significantly (P = 0.531), and post-surgical PVPI did not correlate with the amount of lung resected (P = 0.536). Hence, we conclude that PVPI may be independent of the extent of lung volume reduction. However, PVPI is heavily dependent on sex, age, and body height. Anthropometric variables thus have a significant impact on the likelihood of misclassified abnormal PVPI. This warrants further studies since an increased PVPI, e.g. in the context of an ARDS, may be overlooked if anthropometric variables are not considered. We suggest reference surfaces based on the 95th-percentile corrected for sex, age, and height as a novel approach to normalize PVPI.


Asunto(s)
Estatura , Permeabilidad Capilar , Pulmón , Humanos , Masculino , Femenino , Persona de Mediana Edad , Estudios Prospectivos , Anciano , Factores Sexuales , Factores de Edad , Adulto , Síndrome de Dificultad Respiratoria/fisiopatología , Síndrome de Dificultad Respiratoria/patología , Agua Pulmonar Extravascular/metabolismo , Anciano de 80 o más Años
3.
Cells ; 13(18)2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39329762

RESUMEN

BACKGROUND: Despite promise in preclinical models of acute respiratory distress syndrome (ARDS), mesenchymal stem cells (MSC) have failed to translate to therapeutic benefit in clinical trials. The MSC is a live cell medicine and interacts with the patient's disease state. Here, we explored this interaction, seeking to devise strategies to enhance MSC therapeutic function. METHODS: Human bone-marrow-derived MSCs were exposed to lung homogenate from healthy and E. coli-induced ARDS rat models. Apoptosis and functional assays of the MSCs were performed. RESULTS: The ARDS model showed reduced arterial oxygenation, decreased lung compliance and an inflammatory microenvironment compared to controls. MSCs underwent more apoptosis after stimulation by lung homogenate from controls compared to E. coli, which may explain why MSCs persist longer in ARDS subjects after administration. Changes in expression of cell surface markers and cytokines were associated with lung homogenate from different groups. The anti-microbial effects of MSCs did not change with the stimulation. Moreover, the conditioned media from lung-homogenate-stimulated MSCs inhibited T-cell proliferation. CONCLUSIONS: These findings suggest that the ARDS microenvironment plays an important role in the MSC's therapeutic mechanism of action, and changes can inform strategies to modulate MSC-based cell therapy for ARDS.


Asunto(s)
Microambiente Celular , Pulmón , Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Animales , Humanos , Pulmón/patología , Ratas , Síndrome de Dificultad Respiratoria/terapia , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/metabolismo , Neumonía/patología , Neumonía/metabolismo , Neumonía/terapia , Masculino , Apoptosis , Proliferación Celular , Ratas Sprague-Dawley , Modelos Animales de Enfermedad , Escherichia coli , Citocinas/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Linfocitos T/inmunología , Linfocitos T/metabolismo
4.
ACS Appl Mater Interfaces ; 16(39): 51957-51969, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39305228

RESUMEN

Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) still faces great challenges due to uncontrollable inflammation disorders, complicated causes of occurrence, and high mortality. Small-activating RNA (saRNA) has emerged as a novel and powerful gene-activating tool that may be useful in the treatment of ALI/ARDS. However, effective saRNA therapy is still challenged by the lack of effective and safe gene delivery vehicles. In this study, we develop a type of artificial neutrophil that is used to deliver saRNAs for ALI/ARDS treatment. The saRNA targeting CCAAT-enhancer binding protein α (CEBPA-saRNA) is complexed with H1 histone and further camouflaged with neutrophil membranes (NHR). Interestingly, we are the first to find that the H1 histone possesses the most effective binding capability to saRNA, compared to other subtypes. The prepared NHR shows excellent physicochemical properties, effective cellular uptake by the inflammatory M1 macrophages, and efficient activation of CEBPA, leading to significant M2 polarization. NHR shows an extended circulation lifetime and high-level accumulation in the inflamed lungs. The in vivo experiments indicate that NHR ameliorates ALI in a mouse model. This type of artificial neutrophil shows powerful inflammatory inhibition both in vitro and in vivo, which opens a new avenue for the treatment of ALI/ARDS.


Asunto(s)
Lesión Pulmonar Aguda , Neutrófilos , Síndrome de Dificultad Respiratoria , Animales , Neutrófilos/metabolismo , Ratones , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/patología , Humanos , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Ratones Endogámicos C57BL , Histonas/metabolismo , Histonas/química
5.
Sci Rep ; 14(1): 20768, 2024 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-39237657

RESUMEN

Acute respiratory distress syndrome is a severe lung condition resulting from various causes, with life-threatening consequences that necessitate intensive care. The phenomenon can be modeled in preclinical models, notably through the use of lipopolysaccharide (LPS) instillation in mice. The phenotype induced closely recapitulates the human syndrome, including pulmonary edema, leukocyte infiltration, acute inflammation, impaired pulmonary function, and histological damage. However, the experimental designs using LPS instillations are extremely diverse in the literature. This highly complicates the interpretation of the induced phenotype chronology for future study design and hinders the proper identification of the optimal time frame to assess different readouts. Therefore, the definition of the treatment window in relation to the beginning of the disease onset also presents a significant challenge to address questions or test compound efficacy. In this context, the temporality of the different readouts usually measured in the model was evaluated in both normal and neutrophil-depleted male C57bl/6 mice using LPS-induction to assess the best window for proper readout evaluation with an optimal dynamic response range. Ventilation parameters were evaluated by whole-body plethysmography and neutrophil recruitment were evaluated in bronchoalveolar lavage fluids and in lung tissues directly. Imaging evaluation of myeloperoxidase along with activity in lung lysates and fluids were compared, along with inflammatory cytokines and lung extravasation by enzyme-linked immunoassays. Moreover, dexamethasone, the gold standard positive control in this model, was also administered at different times before and after phenotype induction to assess how kinetics affected each parameter. Overall, our data demonstrate that each readout evaluated in this study has a singular kinetic and highlights the key importance of the timing between ARDS phenotype and treatment administration and/or analysis. These findings also strongly suggest that analyzes, both in-life and post-mortem should be conducted at multiple time points to properly capture the dynamic phenotype of the LPS-ARDS model and response to treatment.


Asunto(s)
Modelos Animales de Enfermedad , Lipopolisacáridos , Ratones Endogámicos C57BL , Fenotipo , Síndrome de Dificultad Respiratoria , Animales , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/inducido químicamente , Síndrome de Dificultad Respiratoria/patología , Ratones , Masculino , Pulmón/patología , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Líquido del Lavado Bronquioalveolar/química , Factores de Tiempo , Citocinas/metabolismo , Neutrófilos/metabolismo
6.
Mol Med ; 30(1): 140, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39251905

RESUMEN

BACKGROUND: Sepsis-induced pulmonary injury (SPI) is a common complication of sepsis with a high rate of mortality. N4-acetylcytidine (ac4C) is mediated by the ac4C "writer", N-acetyltransferase (NAT)10, to regulate the stabilization of mRNA. This study aimed to investigate the role of NAT10 in SPI and the underlying mechanism. METHODS: Twenty-three acute respiratory distress syndrome (ARDS) patients and 27 non-ARDS volunteers were recruited. A sepsis rat model was established. Reverse transcription-quantitative polymerase chain reaction was used to detect the expression of NAT10 and transferrin receptor (TFRC). Cell viability was detected by cell counting kit-8. The levels of Fe2+, glutathione, and malondialdehyde were assessed by commercial kits. Lipid reactive oxygen species production was measured by flow cytometric analysis. Western blot was used to detect ferroptosis-related protein levels. Haematoxylin & eosin staining was performed to observe the pulmonary pathological symptoms. RESULTS: The results showed that NAT10 was increased in ARDS patients and lipopolysaccharide-treated human lung microvascular endothelial cell line-5a (HULEC-5a) cells. NAT10 inhibition increased cell viability and decreased ferroptosis in HULEC-5a cells. TFRC was a downstream regulatory target of NAT10-mediated ac4C acetylation. Overexpression of TFRC decreased cell viability and promoted ferroptosis. In in vivo study, NAT10 inhibition alleviated SPI. CONCLUSION: NAT10-mediated ac4C acetylation of TFRC aggravated SPI through promoting ferroptosis.


Asunto(s)
Ferroptosis , Receptores de Transferrina , Sepsis , Sepsis/metabolismo , Sepsis/complicaciones , Sepsis/etiología , Acetilación , Animales , Humanos , Ratas , Masculino , Receptores de Transferrina/metabolismo , Receptores de Transferrina/genética , Femenino , Lesión Pulmonar/metabolismo , Lesión Pulmonar/etiología , Lesión Pulmonar/patología , Modelos Animales de Enfermedad , Acetiltransferasas/metabolismo , Acetiltransferasas/genética , Persona de Mediana Edad , Antígenos CD/metabolismo , Antígenos CD/genética , Citidina/análogos & derivados , Citidina/farmacología , Línea Celular , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/patología , Ratas Sprague-Dawley , Supervivencia Celular
7.
Nat Commun ; 15(1): 7241, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39174557

RESUMEN

Type 2 alveolar epithelial (AT2) cells of the lung are fundamental in regulating alveolar inflammation in response to injury. Impaired mitochondrial long-chain fatty acid ß-oxidation (mtLCFAO) in AT2 cells is assumed to aggravate alveolar inflammation in acute lung injury (ALI), yet the importance of mtLCFAO to AT2 cell function needs to be defined. Here we show that expression of carnitine palmitoyltransferase 1a (CPT1a), a mtLCFAO rate limiting enzyme, in AT2 cells is significantly decreased in acute respiratory distress syndrome (ARDS). In mice, Cpt1a deletion in AT2 cells impairs mtLCFAO without reducing ATP production and alters surfactant phospholipid abundance in the alveoli. Impairing mtLCFAO in AT2 cells via deleting either Cpt1a or Acadl (acyl-CoA dehydrogenase long chain) restricts alveolar inflammation in ALI by hindering the production of the neutrophilic chemokine CXCL2 from AT2 cells. This study thus highlights mtLCFAO as immunometabolism to injury in AT2 cells and suggests impaired mtLCFAO in AT2 cells as an anti-inflammatory response in ARDS.


Asunto(s)
Lesión Pulmonar Aguda , Células Epiteliales Alveolares , Carnitina O-Palmitoiltransferasa , Ácidos Grasos , Mitocondrias , Oxidación-Reducción , Síndrome de Dificultad Respiratoria , Animales , Carnitina O-Palmitoiltransferasa/metabolismo , Carnitina O-Palmitoiltransferasa/genética , Mitocondrias/metabolismo , Células Epiteliales Alveolares/metabolismo , Ácidos Grasos/metabolismo , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/genética , Ratones , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/genética , Masculino , Humanos , Quimiocina CXCL2/metabolismo , Quimiocina CXCL2/genética , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Neutrófilos/metabolismo , Ratones Noqueados , Acil-CoA Deshidrogenasa de Cadena Larga/metabolismo , Acil-CoA Deshidrogenasa de Cadena Larga/genética , Inflamación/metabolismo , Inflamación/patología , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Alveolos Pulmonares/inmunología , Adenosina Trifosfato/metabolismo , Neumonía/metabolismo , Neumonía/inmunología , Neumonía/patología , Neumonía/genética
8.
Int J Mol Sci ; 25(15)2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39125585

RESUMEN

Acute respiratory distress syndrome (ARDS) occurs as an acute onset condition, and patients present with diffuse alveolar damage, refractory hypoxemia, and non-cardiac pulmonary edema. ARDS progresses through an initial exudative phase, an inflammatory phase, and a final fibrotic phase. Pirfenidone, a powerful anti-fibrotic agent, is known as an agent that inhibits the progression of fibrosis in idiopathic pulmonary fibrosis. In this study, we studied the treatment efficiency of pirfenidone on lipopolysaccharide (LPS) and bleomycin-induced ARDS using rats. The ARDS rat model was created by the intratracheal administration of 3 mg/kg LPS of and 3 mg/kg of bleomycin dissolved in 0.2 mL of normal saline. The pirfenidone treatment group was administered 100 or 200 mg/kg of pirfenidone dissolved in 0.5 mL distilled water orally 10 times every 2 days for 20 days. The administration of LPS and bleomycin intratracheally increased lung injury scores and significantly produced pro-inflammatory cytokines. ARDS induction increased the expressions of transforming growth factor (TGF)-ß1/Smad-2 signaling factors. Additionally, matrix metalloproteinase (MMP)-9/tissue inhibitor of metalloproteinase (TIMP)-1 imbalance occurred, resulting in enhanced fibrosis-related factors. Treatment with pirfenidone strongly suppressed the expressions of TGF-ß1/Smad-2 signaling factors and improved the imbalance of MMP-9/TIMP-1 compared to the untreated group. These effects led to a decrease in fibrosis factors and pro-inflammatory cytokines, promoting the recovery of damaged lung tissue. These results of this study showed that pirfenidone administration suppressed inflammation and fibrosis in the ARDS animal model. Therefore, pirfenidone can be considered a new early treatment for ARDS.


Asunto(s)
Bleomicina , Lipopolisacáridos , Piridonas , Síndrome de Dificultad Respiratoria , Transducción de Señal , Animales , Piridonas/farmacología , Piridonas/uso terapéutico , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/inducido químicamente , Transducción de Señal/efectos de los fármacos , Ratas , Masculino , Bleomicina/efectos adversos , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Proteína Smad2/metabolismo , Ratas Sprague-Dawley , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Modelos Animales de Enfermedad , Metaloproteinasa 9 de la Matriz/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Proteínas Smad/metabolismo
9.
Int J Mol Sci ; 25(15)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39125954

RESUMEN

In this study, we evaluated the ability of the synthetic amphipathic helical peptide (SAHP), L-37pA, which mediates pathogen recognition and innate immune responses, to treat acute respiratory distress syndrome (ARDS) accompanied by diffuse alveolar damage (DAD) and chronic pulmonary fibrosis (PF). For the modeling of ARDS/DAD, male ICR mice were used. Intrabronchial instillation (IB) of 200 µL of inflammatory agents was performed by an intravenous catheter 20 G into the left lung lobe only, leaving the right lobe unaffected. Intravenous injections (IVs) of L-37pA, dexamethasone (DEX) and physiological saline (saline) were used as therapies for ARDS/DAD. L37pA inhibited the circulating levels of inflammatory cytokines, such as IL-8, TNFα, IL1α, IL4, IL5, IL6, IL9 and IL10, by 75-95%. In all cases, the computed tomography (CT) data indicate that L-37pA reduced lung density faster to -335 ± 23 Hounsfield units (HU) on day 7 than with DEX and saline, to -105 ± 29 HU and -23 ± 11 HU, respectively. The results of functional tests showed that L-37pA treatment 6 h after ARDS/DAD initiation resulted in a more rapid improvement in the physiological respiratory lung by 30-45% functions compared with the comparison drugs. Our data suggest that synthetic amphipathic helical peptide L-37pA blocked a cytokine storm, inhibited acute and chronic pulmonary inflammation, prevented fibrosis development and improved physiological respiratory lung function in the ARDS/DAD mouse model. We concluded that a therapeutic strategy using SAHPs targeting SR-B receptors is a potential novel effective treatment for inflammation-induced ARDS, DAD and lung fibrosis of various etiologies.


Asunto(s)
Citocinas , Ratones Endogámicos ICR , Péptidos , Fibrosis Pulmonar , Síndrome de Dificultad Respiratoria , Animales , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/patología , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/patología , Ratones , Masculino , Citocinas/metabolismo , Péptidos/farmacología , Péptidos/química , Modelos Animales de Enfermedad , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo
10.
Int J Biol Macromol ; 279(Pt 1): 135077, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39191344

RESUMEN

The pathogenesis of this condition is intricate, characterized by the aberrant activation of numerous cytokines and signaling pathways. This study aimed to delve into the association between the expression of the MAPK14 protein and immune cell infiltration in patients suffering from Acute Respiratory Distress Syndrome (ARDS). Additionally, it sought to assess the viability of autophagy-related genes as potential diagnostic biomarkers. To achieve this, the researchers employed various techniques such as immunohistochemistry, real-time quantitative PCR, and western blotting to measure the MAPK14 protein levels in the lung tissues of ARDS patients. These measurements were then correlated with clinical data to provide a comprehensive analysis.In this study, the researchers conducted a gene expression profile analysis to identify genes associated with autophagy. The relationship between these genes, MAPK14 expression, and immune cell infiltration was thoroughly evaluated. The findings revealed a marked increase in the expression of MAPK14 protein in the lung tissues of ARDS patients. This increased expression was found to be positively correlated with the extent of immune cell infiltration. The study's further analysis highlighted that several genes associated with autophagy exhibited expression levels that were correlated with both MAPK14 expression and the degree of immune infiltration. This suggests a complex interplay between MAPK14 protein levels, autophagy-related genes, and immune responses in the pathogenesis of ARDS. The results underscore the potential of these molecular markers in understanding the disease mechanisms and possibly aiding in the diagnosis and treatment of ARDS.


Asunto(s)
Autofagia , Biomarcadores , Proteína Quinasa 14 Activada por Mitógenos , Síndrome de Dificultad Respiratoria , Síndrome de Dificultad Respiratoria/genética , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/metabolismo , Humanos , Autofagia/genética , Biomarcadores/metabolismo , Masculino , Femenino , Proteína Quinasa 14 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Persona de Mediana Edad , Pulmón/patología , Pulmón/metabolismo , Pulmón/inmunología , Adulto , Regulación de la Expresión Génica , Perfilación de la Expresión Génica , Anciano
11.
Sci Rep ; 14(1): 16317, 2024 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-39009819

RESUMEN

To study the effects of caspase inhibitors on hemodynamics and inflammatory factors in acute respiratory distress syndrome (ARDS) model rats. Sixty healthy male Wistar rats were randomly divided into three groups, namely, the control group, ARDS group and ARDS + Caspase inhibitor group, with 20 rats in each group. The control group was intraperitoneally injected with 2 mL/kg saline, and the ARDS model group was established by intraperitoneally injecting 4 mg/kg Lipopolysaccharide (LPS), ARDS + Caspase inhibitor group was adminstered 20 mg/kg caspase inhibitor after intraperitoneal LPS injection. Changes in pulmonary arterial pressure (PAP) and mean arterial pressure (MAP) at 6 and 12 h before and after administration were recorded. Moreover, arterial blood gas was evaluated with a blood gas analyzer and changes in the partial pressure of O2 (PaO2), partial pressure of CO2 (PaCO2), partial pressure of O2/fraction of inspired O2 (PaO2/FiO2) were evaluated. In addition, the lung wet/dry weight (W/D) ratio and inflammatory factor levels in lung tissue were determined. Finally, pathological sections were used to determine the pulmonary artery media thickness (MT), MT percentage (MT%), and the degree of muscle vascularization. The pulmonary arterial pressure of rats was determined at several time points. Compared with the control group, the model group had a significantly increased pulmonary arterial pressure at each time point (P < 0.01), and the mean arterial pressure significantly increased at 6 h (P < 0.05). Compared with that of rats in the model group, the pulmonary arterial pressure of rats in drug administration group was significantly reduced at each time point after administration (P < 0.01), and the mean arterial pressure was significantly reduced at 6 h (P < 0.05). The arterial blood gas analysis showed that compared with those in the control group, PaO2, PaCO2 and PaO2/FiO2 in the model group were significantly reduced (P < 0.01), and PaO2, PaCO2 and PaO2/FiO2 were significantly increased after caspase inhibitor treatment (P < 0.05 or 0.01). The levels of the inflammatory mediators tumor necrosis factor-alpha (TNF-α), interleukin-1ß (IL-1ß) and interleukin-6 (IL-6) in the model group were significantly higher than those in the control group (P < 0.01), and they were significantly decreased after caspase inhibitor treatment (P < 0.01). In the model group, pulmonary artery MT, MT% and the degree of muscle vascularization were significantly increased (P < 0.05 or 0.01), and pulmonary artery MT and the degree of muscle vascularization were significantly reduced after caspase inhibitor treatment (P < 0.05 or 0.01). Apoptosis Repressor with a Caspase Recuitment Domain (ARC) can alleviate the occurrence and development of pulmonary hypertension (PH) by affecting hemodynamics and reducing inflammation.


Asunto(s)
Inhibidores de Caspasas , Modelos Animales de Enfermedad , Hemodinámica , Ratas Wistar , Síndrome de Dificultad Respiratoria , Animales , Masculino , Hemodinámica/efectos de los fármacos , Ratas , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/patología , Inhibidores de Caspasas/farmacología , Pulmón/efectos de los fármacos , Pulmón/patología , Lipopolisacáridos , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/fisiopatología , Arteria Pulmonar/patología , Análisis de los Gases de la Sangre , Inflamación/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-6/metabolismo
12.
Am J Physiol Lung Cell Mol Physiol ; 327(3): L269-L281, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38887793

RESUMEN

Acute respiratory distress syndrome (ARDS) is a severe lung disease of high mortality (30-50%). Patients require lifesaving supplemental oxygen therapy; however, hyperoxia can induce pulmonary inflammation and cellular damage. Although alveolar macrophages (AMs) are essential for lung immune homeostasis, they become compromised during inflammatory lung injury. To combat this, stem cell-derived alveolar-like macrophages (ALMs) are a prospective therapeutic for lung diseases like ARDS. Using in vitro and in vivo approaches, we investigated the impact of hyperoxia on murine ALMs during acute inflammation. In vitro, ALMs retained their viability, growth, and antimicrobial abilities when cultured at 60% O2, whereas they die at 90% O2. In contrast, ALMs instilled in mouse lungs remained viable during exposure of mice to 90% O2. The ability of the delivered ALMs to phagocytose Pseudomonas aeruginosa was not impaired by exposure to 60 or 90% O2. Furthermore, ALMs remained immunologically stable in a murine model of LPS-induced lung inflammation when exposed to 60 and 90% O2 and effectively attenuated the accumulation of CD11b+ inflammatory cells in the airways. These results support the potential use of ALMs in patients with ARDS receiving supplemental oxygen therapy.NEW & NOTEWORTHY The current findings support the prospective use of stem cell-derived alveolar-like macrophages (ALMs) as a therapeutic for inflammatory lung disease such as acute respiratory distress syndrome (ARDS) during supplemental oxygen therapy where lungs are exposed to high levels of oxygen. Alveolar-like macrophages directly delivered to mouse lungs were found to remain viable, immunologically stable, phagocytic toward live Pseudomonas aeruginosa, and effective in reducing CD11b+ inflammatory cell numbers in LPS-challenged lungs during moderate and extreme hyperoxic exposure.


Asunto(s)
Modelos Animales de Enfermedad , Hiperoxia , Lipopolisacáridos , Macrófagos Alveolares , Neumonía , Pseudomonas aeruginosa , Animales , Hiperoxia/complicaciones , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Macrófagos Alveolares/metabolismo , Ratones , Neumonía/patología , Neumonía/inducido químicamente , Neumonía/inmunología , Neumonía/terapia , Ratones Endogámicos C57BL , Síndrome de Dificultad Respiratoria/terapia , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/inducido químicamente , Fagocitosis , Masculino , Pulmón/patología , Pulmón/inmunología
13.
Ann Med ; 56(1): 2362871, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38902986

RESUMEN

The lung is an important site of extramedullary platelet formation, and megakaryocytes in the lung participate in immune responses in addition to platelet production. In acute lung injury and chronic lung injury, megakaryocytes and platelets play a promoting or protective role through different mechanisms. The authors reviewed the role of megakaryocytes and platelets in common clinical lung injuries with different course of disease and different pathogenic factors in order to provide new thinking for the diagnosis and treatment of lung injuries.


What is the context?Platelets are specialized non-nucleated blood cells produced by cytoplasmic lysis of megakaryocytes.HSCs differentiate into granular mature megakaryocytes and produce platelets.Lung is a reservoir of megakaryocytes and a site where platelets are produced in addition to bone marrow and spleen.Lung injury can be divided into acute lung injury and chronic lung injury, and characterized by different pathogenesis.Platelets and megakaryocytes are involved in hemostasis and regulation of the body 's inflammatory response.The disease state of the lung affects the functions of megakaryocytes and platelets.The role of megakaryocytes and platelets in acute and chronic lung injury is poorly studied.What is new?Platelets in the lung are derived not only from the spleen and bone marrow, but also from megakaryocytes in the pulmonary circulation. In this study, we demonstrated that pulmonary megakaryocytes not only produce platelets to play a hemostatic role in lung injury, but also participate in inflammation and immune response with platelets to promote the process of lung injury or play a protective role.Therefore, it was suggested in our analysis that targeting lung megakaryocytes and platelets is currently a new direction for the treatment of a variety of lung injuries.What is the impact?This review intends to explain the relationship between megakaryocytes, platelets and many types of lung injury from the mechanism of platelet production in the lung, and make a prospect in the new progress in the diagnosis and treatment of lung injury.


Asunto(s)
Lesión Pulmonar Aguda , Plaquetas , Megacariocitos , Humanos , Lesión Pulmonar Aguda/patología , Lesión Pulmonar , Pulmón/patología , Animales , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/inmunología
14.
Sci Rep ; 14(1): 14545, 2024 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-38914619

RESUMEN

SARS-CoV-2 has become a global public health problem. Acute respiratory distress syndrome (ARDS) is the leading cause of death due to the SARS-CoV-2 infection. Pulmonary fibrosis (PF) is a severe and frequently reported COVID-19 sequela. In this study, an in vitro model of ARDS and PF caused by SARS-CoV-2 was established in MH-S, THP-1, and MRC-5 cells using pseudo-SARS-CoV-2 (PSCV). Expression of proinflammatory cytokines (IL-6, IL-1ß, and TNF-α) and HIF-1α was increased in PSCV-infected MH-S and THP-1 cells, ARDS model, consistent with other profiling data in SARS-CoV-2-infected patients have been reported. Hypoxia-inducible factor-1 alpha (HIF-1α) siRNA and cobalt chloride were tested using this in vitro model. HIF-1α knockdown reduces inflammation caused by PSCV infection in MH-S and THP-1 cells and lowers elevated levels of CTGF, COLA1, and α-SMA in MRC-5 cells exposed to CPMSCV. Furthermore, apigetrin, a glycoside bioactive dietary flavonoid derived from several plants, including Crataegus pinnatifida, which is reported to be a HIF-1α inhibitor, was tested in this in vitro model. Apigetrin significantly reduced the increased inflammatory cytokine (IL-6, IL-1ß, and TNF-α) expression and secretion by PSCV in MH-S and THP-1 cells. Apigetrin inhibited the binding of the SARS-CoV-2 spike protein RBD to the ACE2 protein. An in vitro model of PF induced by SARS-CoV-2 was produced using a conditioned medium of THP-1 and MH-S cells that were PSCV-infected (CMPSCV) into MRC-5 cells. In a PF model, CMPSCV treatment of THP-1 and MH-S cells increased cell growth, migration, and collagen synthesis in MRC-5 cells. In contrast, apigetrin suppressed the increase in cell growth, migration, and collagen synthesis induced by CMPSCV in THP-1 and MH-S MRC-5 cells. Also, compared to control, fibrosis-related proteins (CTGF, COLA1, α-SMA, and HIF-1α) levels were over two-fold higher in CMPSV-treated MRC-5 cells. Apigetrin decreased protein levels in CMPSCV-treated MRC-5 cells. Thus, our data suggest that hypoxia-inducible factor-1 alpha (HIF-1α) might be a novel target for SARS-CoV-2 sequela therapies and apigetrin, representative of HIF-1alpha inhibitor, exerts anti-inflammatory and PF effects in PSCV-treated MH-S, THP-1, and CMPVSC-treated MRC-5 cells. These findings indicate that HIF-1α inhibition and apigetrin would have a potential value in controlling SARS-CoV-2-related diseases.


Asunto(s)
COVID-19 , Citocinas , Subunidad alfa del Factor 1 Inducible por Hipoxia , Fibrosis Pulmonar , SARS-CoV-2 , Humanos , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/virología , Fibrosis Pulmonar/patología , SARS-CoV-2/fisiología , COVID-19/metabolismo , COVID-19/virología , COVID-19/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Citocinas/metabolismo , Inflamación/metabolismo , Inflamación/patología , Línea Celular , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/virología , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/etiología , Células THP-1
15.
Eur J Pharmacol ; 977: 176672, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38849041

RESUMEN

OBJECTIVE: Abscisic acid (ABA) is a phytohormone that inhibits airway inflammation in acute respiratory distress syndrome (ARDS) mouse models. However, the molecular mechanism underlying this phenomenon remains unclear. METHODS: Serum ABA level in patients and mice was measured via liquid chromatography-tandem mass spectrometry (LC-MS/MS). In-depth molecular mechanism was investigated through transmission electron microscopy, RNA-sequencing, and molecular docking in ARDS mice and cultured primary alveolar macrophages (AMs). RESULTS: We found that the serum ABA level was remarkably decreased in ARDS mice and patients. ABA inhibited lipopolysaccharide (LPS)-induced airway inflammation in mice; moreover, it downregulated genes associated with pyroptosis, as shown by RNA-sequencing and lung protein immunoblots. ABA inhibited the formation of membrane pores in AMs and suppressed the cleavage of gasdermin D (GSDMD) and the activation of caspase-11 and caspase-1 in vivo and in vitro; however, the overexpression of caspase-11 reversed the protective effect of ABA on LPS-induced pyroptosis of primary AMs. ABA inhibited intra-AM LPS accumulation while increasing the level of acyloxyacyl hydrolase (AOAH) in AMs, whereas AOAH deficiency abrogated the suppressive action of ABA on inflammation, pyroptosis, and intra-AM LPS accumulation in vivo and in vitro. Importantly, ABA promoted its intracellular receptor lanthionine C-like receptor 2 interacting with transcription factor peroxisome proliferator-activated receptor γ, which ultimately leading to increase AOAH expression to inactivate LPS and inhibit pyroptosis in AMs. CONCLUSIONS: ABA protected against LPS-induced lung injury by inhibiting pyroptosis in AMs via proliferator-activated receptor γ-mediated AOAH expression.


Asunto(s)
Ácido Abscísico , Macrófagos Alveolares , Piroptosis , Síndrome de Dificultad Respiratoria , Piroptosis/efectos de los fármacos , Animales , Ácido Abscísico/farmacología , Ratones , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/inducido químicamente , Masculino , Humanos , Lipopolisacáridos/farmacología , PPAR gamma/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Ratones Endogámicos C57BL , Femenino , Modelos Animales de Enfermedad
16.
Proc Natl Acad Sci U S A ; 121(26): e2319322121, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38900789

RESUMEN

Thymocyte selection-associated high-mobility group box (TOX) is a transcription factor that is crucial for T cell exhaustion during chronic antigenic stimulation, but its role in inflammation is poorly understood. Here, we report that TOX extracellularly mediates drastic inflammation upon severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection by binding to the cell surface receptor for advanced glycation end-products (RAGE). In various diseases, including COVID-19, TOX release was highly detectable in association with disease severity, contributing to lung fibroproliferative acute respiratory distress syndrome (ARDS). Recombinant TOX-induced blood vessel rupture, similar to a clinical signature in patients experiencing a cytokine storm, further exacerbating respiratory function impairment. In contrast, disruption of TOX function by a neutralizing antibody and genetic removal of RAGE diminished TOX-mediated deleterious effects. Altogether, our results suggest an insight into TOX function as an inflammatory mediator and propose the TOX-RAGE axis as a potential target for treating severe patients with pulmonary infection and mitigating lung fibroproliferative ARDS.


Asunto(s)
COVID-19 , Receptor para Productos Finales de Glicación Avanzada , SARS-CoV-2 , Humanos , Receptor para Productos Finales de Glicación Avanzada/metabolismo , COVID-19/inmunología , COVID-19/metabolismo , COVID-19/patología , COVID-19/complicaciones , COVID-19/virología , Animales , Ratones , Inflamación/metabolismo , Inflamación/patología , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/virología , Lesión Pulmonar/inmunología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas del Grupo de Alta Movilidad/genética , Masculino , Pulmón/patología , Pulmón/metabolismo , Pulmón/inmunología , Femenino
17.
J Transl Med ; 22(1): 535, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38840216

RESUMEN

BACKGROUND: Inflammation and endothelial barrier dysfunction are the major pathophysiological changes in acute respiratory distress syndrome (ARDS). Sphingosine-1-phosphate receptor 3 (S1PR3), a G protein-coupled receptor, has been found to mediate inflammation and endothelial cell (EC) integrity. However, the function of S1PR3 in ARDS has not been fully elucidated. METHODS: We used a murine lipopolysaccharide (LPS)-induced ARDS model and an LPS- stimulated ECs model to investigate the role of S1PR3 in anti-inflammatory effects and endothelial barrier protection during ARDS. RESULTS: We found that S1PR3 expression was increased in the lung tissues of mice with LPS-induced ARDS. TY-52156, a selective S1PR3 inhibitor, effectively attenuated LPS-induced inflammation by suppressing the expression of proinflammatory cytokines and restored the endothelial barrier by repairing adherens junctions and reducing vascular leakage. S1PR3 inhibition was achieved by an adeno-associated virus in vivo and a small interfering RNA in vitro. Both the in vivo and in vitro studies demonstrated that pharmacological or genetic inhibition of S1PR3 protected against ARDS by inhibiting the NF-κB pathway and improving mitochondrial oxidative phosphorylation. CONCLUSIONS: S1PR3 inhibition protects against LPS-induced ARDS via suppression of pulmonary inflammation and promotion of the endothelial barrier by inhibiting NF-κB and improving mitochondrial oxidative phosphorylation, indicating that S1PR3 is a potential therapeutic target for ARDS.


Asunto(s)
Lipopolisacáridos , Ratones Endogámicos C57BL , Mitocondrias , FN-kappa B , Fosforilación Oxidativa , Síndrome de Dificultad Respiratoria , Receptores de Esfingosina-1-Fosfato , Animales , Humanos , Masculino , Ratones , Citocinas/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/efectos de los fármacos , Inflamación/patología , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , FN-kappa B/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Sustancias Protectoras/farmacología , Receptores de Lisoesfingolípidos/metabolismo , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Síndrome de Dificultad Respiratoria/inducido químicamente , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/patología , Receptores de Esfingosina-1-Fosfato/metabolismo , Receptores de Esfingosina-1-Fosfato/antagonistas & inhibidores
18.
Am J Physiol Lung Cell Mol Physiol ; 327(2): L141-L149, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38772909

RESUMEN

Neutrophils are the first leukocytes to be recruited to sites of inflammation in response to chemotactic factors released by activated macrophages and pulmonary epithelial and endothelial cells in bacterial pneumonia, a common cause of acute respiratory distress syndrome (ARDS). Although neutrophilic inflammation facilitates the elimination of pathogens, neutrophils also may cause bystander tissue injury. Even though the presence of neutrophils in alveolar spaces is a key feature of acute lung injury and ARDS especially from pneumonia, their contribution to the pathogenesis of lung injury is uncertain. The goal of this study was to elucidate the role of neutrophils in a clinically relevant model of bacterial pneumonia. We investigated the effect of reducing neutrophils in a mouse model of pneumococcal pneumonia treated with antibiotics. Neutrophils were reduced with anti-lymphocyte antigen 6 complex locus G6D (Ly6G) monoclonal antibody 24 h before and immediately preceding infection. Mice were inoculated intranasally with Streptococcus pneumoniae and received ceftriaxone 12 h after bacterial inoculation. Neutrophil reduction in mice treated with ceftriaxone attenuated hypoxemia, alveolar permeability, epithelial injury, pulmonary edema, and inflammatory biomarker release induced by bacterial pneumonia, even though bacterial loads in the distal air spaces of the lung were modestly increased as compared with antibiotic treatment alone. Thus, when appropriate antibiotics are administered, lung injury in the early phase of bacterial pneumonia is mediated in part by neutrophils. In the early phase of bacterial pneumonia, neutrophils contribute to the severity of lung injury, although they also participate in host defense.NEW & NOTEWORTHY Neutrophil accumulation is a key feature of ARDS, but their contribution to the pathogenesis is still uncertain. We investigated the effect of reducing neutrophils in a clinically relevant mouse model of pneumococcal pneumonia treated with antibiotics. When appropriate antibiotics were administered, neutrophil reduction with Ly6G antibody markedly attenuated lung injury and improved oxygenation. In the early phase of bacterial pneumonia, neutrophils contribute to the severity of lung injury, although they also participate in host defense.


Asunto(s)
Ratones Endogámicos C57BL , Neutrófilos , Neumonía Neumocócica , Animales , Neumonía Neumocócica/inmunología , Neumonía Neumocócica/patología , Neumonía Neumocócica/tratamiento farmacológico , Neumonía Neumocócica/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Ratones , Streptococcus pneumoniae/patogenicidad , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/microbiología , Modelos Animales de Enfermedad , Pulmón/patología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Lesión Pulmonar/patología , Lesión Pulmonar/inmunología , Lesión Pulmonar/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/inmunología , Masculino
19.
Am J Pathol ; 194(8): 1443-1457, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38705380

RESUMEN

Acute respiratory distress syndrome (ARDS) is a heterogeneous clinical syndrome that is most commonly triggered by infection-related inflammation. Lung pericytes can respond to infection and act as immune and proangiogenic cells; moreover, these cells can differentiate into myofibroblasts in nonresolving ARDS and contribute to the development of pulmonary fibrosis. Here, we aimed to characterize the role of lung cells, which present characteristics of pericytes, such as peri-endothelial location and expression of a panel of specific markers. A murine model of lipopolysaccharide (LPS)-induced resolving ARDS was used to study their role in ARDS. The development of ARDS was confirmed after LPS instillation, which was resolved 14 days after onset. Immunofluorescence and flow cytometry showed early expansion of neural-glial antigen 2+ ß-type platelet-derived growth factor receptor+ pericytes in murine lungs with loss of CD31+ ß-type platelet-derived growth factor receptor+ endothelial cells. These changes were accompanied by specific changes in lung structure and loss of vascular integrity. On day 14 after ARDS onset, the composition of pericytes and endothelial cells returned to baseline values. LPS-induced ARDS activated NOTCH signaling in lung pericytes, the inhibition of which during LPS stimulation reduced the expression of its downstream target genes, pericyte markers, and angiogenic factors. Together, these data indicate that lung pericytes in response to inflammatory injury activate NOTCH signaling that supports their maintenance and in turn can contribute to recovery of the microvascular endothelium.


Asunto(s)
Lipopolisacáridos , Pericitos , Síndrome de Dificultad Respiratoria , Animales , Pericitos/patología , Pericitos/metabolismo , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/inducido químicamente , Síndrome de Dificultad Respiratoria/metabolismo , Lipopolisacáridos/farmacología , Ratones , Pulmón/patología , Pulmón/metabolismo , Ratones Endogámicos C57BL , Masculino , Modelos Animales de Enfermedad , Transducción de Señal , Células Endoteliales/metabolismo , Células Endoteliales/patología
20.
Biochem Biophys Res Commun ; 716: 150019, 2024 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-38703555

RESUMEN

- Acute respiratory distress syndrome (ARDS)/acute lung injury (ALI) is a life-threatening condition marked by severe lung inflammation and increased lung endothelial barrier permeability. Endothelial glycocalyx deterioration is the primary factor of vascular permeability changes in ARDS/ALI. Although previous studies have shown that phospholipase D2 (PLD2) is closely related to the onset and progression of ARDS/ALI, its role and mechanism in the damage of endothelial cell glycocalyx remains unclear. We used LPS-induced ARDS/ALI mice (in vivo) and LPS-stimulated injury models of EA.hy926 endothelial cells (in vitro). We employed C57BL/6 mice, including wild-type and PLD2 knockout (PLD2-/-) mice, to establish the ARDS/ALI model. We applied immunofluorescence and ELISA to examine changes in syndecan-1 (SDC-1), matrix metalloproteinase-9 (MMP9), inflammatory cytokines (TNF-α, IL-6, and IL-1ß) levels and the effect of external factors, such as phosphatidic acid (PA), 1-butanol (a PLD inhibitor), on SDC-1 and MMP9 expression levels. We found that PLD2 deficiency inhibits SDC-1 degradation and MMP9 expression in LPS-induced ARDS/ALI. Externally added PA decreases SDC-1 levels and increases MMP9 in endothelial cells, hence underlining PA's role in SDC-1 degradation. Additionally, PLD2 deficiency decreases the production of inflammatory cytokines (TNF-α, IL-6, and IL-1ß) in LPS-induced ARDS/ALI. In summary, these findings suggest that PLD2 deficiency plays a role in inhibiting the inflammatory process and protecting against endothelial glycocalyx injury in LPS-induced ARDS/ALI.


Asunto(s)
Lesión Pulmonar Aguda , Células Endoteliales , Glicocálix , Lipopolisacáridos , Fosfolipasa D , Síndrome de Dificultad Respiratoria , Animales , Humanos , Ratones , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/etiología , Línea Celular , Citocinas/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Glicocálix/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfolipasa D/metabolismo , Fosfolipasa D/genética , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/inducido químicamente , Sindecano-1/metabolismo , Sindecano-1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA