Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 380
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 326(5): L524-L538, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38375572

RESUMEN

Lung surfactant collectins, surfactant protein A (SP-A) and D (SP-D), are oligomeric C-type lectins involved in lung immunity. Through their carbohydrate recognition domain, they recognize carbohydrates at pathogen surfaces and initiate lung innate immune response. Here, we propose that they may also be able to bind to other carbohydrates present in typical cell surfaces, such as the alveolar epithelial glycocalyx. To test this hypothesis, we analyzed and quantified the binding affinity of SP-A and SP-D to different sugars and glycosaminoglycans (GAGs) by microscale thermophoresis (MST). In addition, by changing the calcium concentration, we aimed to characterize any consequences on the binding behavior. Our results show that both oligomeric proteins bind with high affinity (in nanomolar range) to GAGs, such as hyaluronan (HA), heparan sulfate (HS) and chondroitin sulfate (CS). Binding to HS and CS was calcium-independent, as it was not affected by changing calcium concentration in the buffer. Quantification of GAGs in bronchoalveolar lavage (BAL) fluid from animals deficient in either SP-A or SP-D showed changes in GAG composition, and electron micrographs showed differences in alveolar glycocalyx ultrastructure in vivo. Taken together, SP-A and SP-D bind to model sulfated glycosaminoglycans of the alveolar epithelial glycocalyx in a multivalent and calcium-independent way. These findings provide a potential mechanism for SP-A and SP-D as an integral part of the alveolar epithelial glycocalyx binding and interconnecting free GAGs, proteoglycans, and other glycans in glycoproteins, which may influence glycocalyx composition and structure.NEW & NOTEWORTHY SP-A and SP-D function has been related to innate immunity of the lung based on their binding to sugar residues at pathogen surfaces. However, their function in the healthy alveolus was considered as limited to interaction with surfactant lipids. Here, we demonstrated that these proteins bind to glycosaminoglycans present at typical cell surfaces like the alveolar epithelial glycocalyx. We propose a model where these proteins play an important role in interconnecting alveolar epithelial glycocalyx components.


Asunto(s)
Calcio , Glicocálix , Glicosaminoglicanos , Alveolos Pulmonares , Proteína A Asociada a Surfactante Pulmonar , Proteína D Asociada a Surfactante Pulmonar , Glicocálix/metabolismo , Animales , Proteína A Asociada a Surfactante Pulmonar/metabolismo , Glicosaminoglicanos/metabolismo , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Calcio/metabolismo , Alveolos Pulmonares/metabolismo , Unión Proteica , Líquido del Lavado Bronquioalveolar , Ratones , Células Epiteliales Alveolares/metabolismo , Heparitina Sulfato/metabolismo , Ratones Endogámicos C57BL
2.
Eur J Pharmacol ; 963: 176219, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38040079

RESUMEN

Sepsis-associated lung injury often coexists with intestinal dysfunction. Butyrate, an essential gut microbiota metabolite, participates in gut-lung crosstalk and has immunoregulatory effects. This study aims to investigate the effect and mechanism of sodium butyrate (NaB) on lung injury. Sepsis-associated lung injury was established in mice by cecal ligation and puncture (CLP). Mice in treatment groups received NaB gavage after surgery. The survival rate, the oxygenation index and the lung wet-to-dry weight (W/D) ratio were calculated respectively. Pulmonary and intestinal histologic changes were observed. The total protein concentration in bronchoalveolar lavage fluid (BALF) was measured, and inflammatory factors in serum and BALF were examined. Diamine oxidase (DAO), lipopolysaccharide (LPS), and surfactant-associated protein D (SP-D) levels in serum and amphiregulin in lung tissue were assessed. Intercellular junction protein expression in the lung and intestinal tissues were examined. Changes in immune cells were analyzed. NaB treatment improved the survival rate, the oxygenation index and the histologic changes. NaB decreased the W/D ratio, total protein concentration, and the levels of proinflammatory cytokines, as well as SP-D, DAO and LPS, while increased the levels of anti-inflammatory cytokines and amphiregulin. The intercellular junction protein expression were improved by NaB. Furthermore, the CD4+/CD8+ T-cell ratio and the proportion of CD4+Foxp3+ regulatory T cells (Tregs) were increased by NaB. Our data suggested that NaB gavage effectively improved the survival rate and mitigated lung injury in CLP mice. The possible mechanism was that NaB augmented CD4+Foxp3+ Tregs and enhanced the barrier function of the gut and the lung.


Asunto(s)
Lesión Pulmonar Aguda , Sepsis , Ratones , Animales , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/complicaciones , Ácido Butírico/farmacología , Ácido Butírico/uso terapéutico , Ácido Butírico/metabolismo , Anfirregulina/metabolismo , Linfocitos T Reguladores/metabolismo , Lipopolisacáridos/metabolismo , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Pulmón/patología , Citocinas/metabolismo , Factores de Transcripción/metabolismo , Sepsis/complicaciones , Sepsis/tratamiento farmacológico , Sepsis/metabolismo , Factores de Transcripción Forkhead/metabolismo
3.
Protein Pept Lett ; 30(9): 743-753, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37622713

RESUMEN

BACKGROUND: Apelin-13 is an endogenous adipocytokine known for its antioxidant, antiinflammatory, and antiapoptotic properties. OBJECTIVE: We aimed to investigate the possible protective effects of exogenous Apelin-13 administration on oxidative stress, inflammation, and apoptosis induced by the cytotoxic agent cyclophosphamide (CP) in the lungs. METHODS: Twenty-four male Wistar albino rats were divided into four groups: Control (saline), CP (200 mg/kg), Apelin-13 (10 µg/kg/day), and CP+Apelin-13. CP was administered as a single dose on the fifth day, and apelin-13 was administered intraperitoneally for five days. Total oxidant status (TOS), total antioxidant status (TAS), and lipid peroxidation were determined with spectrophotometry, TNFα and IL1ß were determined with ELISA, APJ, Sirt1, NF-κB, and p53 mRNA expressions were determined with qRT-PCR, cytochrome (Cyt) C and caspase-3 protein expressions were studied with western blotting in lung tissues. The oxidative stress index (OSI) was also calculated. Furthermore, serum surfactant protein-D (SP-D) and Krebs von den Lungen-6 (KL-6) levels were measured with ELISA. RESULTS: Compared to the control group, TOS, OSI, lipid peroxidation, TNFα, IL1ß, cyt C, caspase-3, APJ, NF-κB, and p53 were higher, and Sirt1 was lower in the lung tissue of rats in the CP group. Serum KL-6 and SP-D levels were higher in the CP group. Co-administration of CP with Apelin-13 completely reversed the changes induced by CP administration. CONCLUSION: Exogenous Apelin-13 treatment protected lung tissue against injury by inhibiting cyclophosphamide-induced oxidative stress, inflammation, and apoptosis. This protective effect of apelin-13 was accompanied by upregulation of the Sirt1 and downregulation of NF-κB/p53 in the lungs.


Asunto(s)
Antioxidantes , FN-kappa B , Ratas , Masculino , Animales , FN-kappa B/metabolismo , Ratas Wistar , Antioxidantes/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Sirtuina 1/metabolismo , Sirtuina 1/farmacología , Caspasa 3/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Proteína D Asociada a Surfactante Pulmonar/farmacología , Estrés Oxidativo , Ciclofosfamida/efectos adversos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Pulmón , Apoptosis , Apelina/efectos adversos , Apelina/metabolismo
4.
In Vivo ; 37(4): 1721-1728, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37369511

RESUMEN

BACKGROUND/AIM: The lung-specific soluble lectins, SP-A and SP-D have been clinically used to diagnose interstitial lung disease, but their clinical significance in COVID-19 remains controversial. This study was undertaken to determine their association with other lectins (MBL and FCN1), disease severity, and radiographs in COVID-19 patients. PATIENTS AND METHODS: A total of 131 patients with COVID-19 admitted in the Sapporo Medical University Hospital between May 22 and September 19, 2021, were enrolled in the study. Data including demographics, medical history, symptoms, signs, laboratory findings, and radiological images were collected from the patients' medical records. Chest computed tomography (CT) scanning was performed at admission. Serum levels of surfactant protein A and D (SP-A and SP-D), mannose-binding lectin (MBL) and ficolin1 (FCN1) were measured using enzyme-linked immunosorbent assay (ELISA) kits. RESULTS: Compared to the control group, the COVID-19 group had significantly higher serum SP-A and FCN1 levels on admission (SP-A: 59.60±38.89 vs. 35.61±11.22 ng/ml; p<0.01, FCN1: 542.45±506.04 vs. 250.6±161.1 ng/ml; p<0.01). The severe group in COVID-19 had significantly higher serum SP-D and lower MBL levels than the non-severe group (SP-D: 141.7±155.7 vs. 61.41±54.54 ng/ml; p<0.01, MBL: 1,670±1,240 vs. 2,170±1,140 ng/ml; p<0.05). SP-D strongly reflected the degree of imaging findings, whereas SP-A showed a significant correlation, albeit slightly weaker than SP-D. Conversely, MBL and FNC1 were not significantly correlated with imaging findings. CONCLUSION: Among soluble serum lectins, SP-A and SP-D may be more sensitive to CT findings than reported disease biomarkers such as IL-6, LDH, and CRP due to their lung-specific characteristics.


Asunto(s)
COVID-19 , Lectinas , Humanos , Proteína D Asociada a Surfactante Pulmonar/metabolismo , COVID-19/diagnóstico , Biomarcadores , Pulmón/diagnóstico por imagen , Pulmón/metabolismo
5.
Am J Physiol Lung Cell Mol Physiol ; 325(3): L288-L298, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37366541

RESUMEN

Pompe disease is an autosomal recessive glycogen storage disease caused by mutations in the gene that encodes acid alpha-glucosidase (GAA)-an enzyme responsible for hydrolyzing lysosomal glycogen. GAA deficiency results in systemic lysosomal glycogen accumulation and cellular disruption. Glycogen accumulation in skeletal muscles, motor neurons, and airway smooth muscle cells is known to contribute to respiratory insufficiency in Pompe disease. However, the impact of GAA deficiency on the distal alveolar type 1 and type 2 cells (AT1 and AT2) has not been evaluated. AT1 cells rely on lysosomes for cellular homeostasis so that they can maintain a thin barrier for gas exchange, whereas AT2 cells depend on lysosome-like structures (lamellar bodies) for surfactant production. Using a mouse model of Pompe disease, the Gaa-/- mouse, we investigated the consequences of GAA deficiency on AT1 and AT2 cells using histology, pulmonary function and mechanics, and transcriptional analysis. Histological analysis revealed increased accumulation of lysosomal-associated membrane protein 1 (LAMP1) in the Gaa-/- mice lungs. Furthermore, ultrastructural examination showed extensive intracytoplasmic vacuoles enlargement and lamellar body engorgement. Respiratory dysfunction was confirmed using whole body plethysmography and forced oscillometry. Finally, transcriptomic analysis demonstrated dysregulation of surfactant proteins in AT2 cells, specifically reduced levels of surfactant protein D in the Gaa-/- mice. We conclude that GAA enzyme deficiency leads to glycogen accumulation in the distal airway cells that disrupts surfactant homeostasis and contributes to respiratory impairments in Pompe disease.NEW & NOTEWORTHY This research highlights the impact of Pompe disease on distal airway cells. Prior to this work, respiratory insufficiency in Pompe disease was classically attributed to pathology in respiratory muscles and motor neurons. Using the Pompe mouse model, we note significant pathology in alveolar type 1 and 2 cells with reductions in surfactant protein D and disrupted surfactant homeostasis. These novel findings highlight the potential contributions of alveolar pathology to respiratory insufficiency in Pompe disease.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II , Insuficiencia Respiratoria , Humanos , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Proteína D Asociada a Surfactante Pulmonar/metabolismo , alfa-Glucosidasas/genética , alfa-Glucosidasas/metabolismo , Músculo Esquelético/metabolismo , Glucógeno/metabolismo
6.
Environ Health Perspect ; 131(5): 57002, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37141245

RESUMEN

BACKGROUND: Exposure to traffic-related air pollution (TRAP) has been associated with increased risks of respiratory diseases, but the biological mechanisms are not yet fully elucidated. OBJECTIVES: Our aim was to evaluate the respiratory responses and explore potential biological mechanisms of TRAP exposure in a randomized crossover trial. METHODS: We conducted a randomized crossover trial in 56 healthy adults. Each participant was exposed to high- and low-TRAP exposure sessions by walking in a park and down a road with high traffic volume for 4 h in random order. Respiratory symptoms and lung function, including forced expiratory volume in the first second (FEV1), forced vital capacity (FVC), the ratio of FEV1 to FVC, and maximal mid-expiratory flow (MMEF), were measured before and after each exposure session. Markers of 8-isoprostane, tumor necrosis factor-α (TNF-α), and ezrin in exhaled breath condensate (EBC), and surfactant proteins D (SP-D) in serum were also measured. We used linear mixed-effects models to estimate the associations, adjusted for age, sex, body mass index, meteorological condition, and batch (only for biomarkers). Liquid chromatography-mass spectrometry was used to profile the EBC metabolome. Untargeted metabolome-wide association study (MWAS) analysis and pathway enrichment analysis using mummichog were performed to identify critical metabolomic features and pathways associated with TRAP exposure. RESULTS: Participants had two to three times higher exposure to traffic-related air pollutants except for fine particulate matter while walking along the road compared with in the park. Compared with the low-TRAP exposure at the park, high-TRAP exposure at the road was associated with a higher score of respiratory symptoms [2.615 (95% CI: 0.605, 4.626), p=1.2×10-2] and relatively lower lung function indicators [-0.075L (95% CI: -0.138, -0.012), p=2.1×10-2] for FEV1 and -0.190L/s (95% CI: -0.351, -0.029; p=2.4×10-2) for MMEF]. Exposure to TRAP was significantly associated with changes in some, but not all, biomarkers, particularly with a 0.494-ng/mL (95% CI: 0.297, 0.691; p=9.5×10-6) increase for serum SP-D and a 0.123-ng/mL (95% CI: -0.208, -0.037; p=7.2×10-3) decrease for EBC ezrin. Untargeted MWAS analysis revealed that elevated TRAP exposure was significantly associated with perturbations in 23 and 32 metabolic pathways under positive- and negative-ion modes, respectively. These pathways were most related to inflammatory response, oxidative stress, and energy use metabolism. CONCLUSIONS: This study suggests that TRAP exposure might lead to lung function impairment and respiratory symptoms. Possible underlying mechanisms include lung epithelial injury, inflammation, oxidative stress, and energy metabolism disorders. https://doi.org/10.1289/EHP11139.


Asunto(s)
Contaminantes Atmosféricos , Contaminación del Aire , Adulto , Humanos , Contaminantes Atmosféricos/toxicidad , Contaminantes Atmosféricos/análisis , Contaminación del Aire/efectos adversos , Contaminación del Aire/análisis , Exposición a Riesgos Ambientales/análisis , Proteína D Asociada a Surfactante Pulmonar/análisis , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Material Particulado/toxicidad , Material Particulado/análisis , Emisiones de Vehículos/toxicidad , Emisiones de Vehículos/análisis , Biomarcadores/análisis , Metaboloma , Pulmón
7.
Ann Anat ; 247: 152048, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36690045

RESUMEN

BACKGROUND: Surfactant protein D (SP-D) is an innate host defense protein that clears infectious pathogens from the lung and regulates pulmonary host defense cells. SP-D is also detected in lower concentrations in plasma and many other non-pulmonary tissues. Plasma levels of SP-D increase during infection and other proinflammatory states; however, the source and functions of SP-D in the systemic circulation are largely unknown. We hypothesized that systemic SP-D may clear infectious pathogens and regulate host defense cells in extrapulmonary systems. METHODS: To determine if SP-D inhibited inflammation induced by systemic lipopolysaccharide (LPS), E.coli LPS was administered to mice via tail vein injection with and without SP-D and the inflammatory response was measured. RESULTS: Systemic SP-D has a circulating half-life of 6 h. Systemic IL-6 levels in mice lacking the SP-D gene were similar to wild type mice at baseline but were significantly higher than wild type mice following LPS treatment (38,000 vs 29,900 ng/ml for 20 mg/kg LPS and 100,700 vs 73,700 ng/ml for 40 mg/kg LPS). In addition, treating wild type mice with purified intravenous SP-D inhibited LPS induced secretion of IL-6 and TNFα in a concentration dependent manner. Inhibition of LPS induced inflammation by SP-D correlated with SP-D LPS binding suggesting SP-D mediated inhibition of systemic LPS requires direct SP-D LPS interactions. CONCLUSIONS: Taken together, the above results suggest that circulating SP-D decreases systemic inflammation and raise the possibility that a physiological purpose of increasing systemic SP-D levels during infection is to scavenge systemic infectious pathogens and limit inflammation-induced tissue injury.


Asunto(s)
Lipopolisacáridos , Proteína D Asociada a Surfactante Pulmonar , Ratones , Animales , Proteína D Asociada a Surfactante Pulmonar/genética , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Proteína D Asociada a Surfactante Pulmonar/farmacología , Lipopolisacáridos/farmacología , Interleucina-6 , Inflamación , Pulmón
8.
Alcohol Clin Exp Res (Hoboken) ; 47(1): 95-103, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36352814

RESUMEN

BACKGROUND: Over 43% of the world's population regularly consumes alcohol. Although not commonly known, alcohol can have a significant impact on the respiratory environment. Living in the time of the COVID-19 pandemic, alcohol misuse can have a particularly deleterious effect on SARS-CoV-2-infected individuals and, in turn, the overall healthcare system. Patients with alcohol use disorders have higher odds of COVID-19-associated hospitalization and mortality. Even though the detrimental role of alcohol on COVID-19 outcomes has been established, the underlying mechanisms are yet to be fully understood. Alcohol misuse has been shown to induce oxidative damage in the lungs through the production of reactive aldehydes such as malondialdehyde and acetaldehyde (MAA). MAA can then form adducts with proteins, altering their structure and function. One such protein is surfactant protein D (SPD), which plays an important role in innate immunity against pathogens. METHODS AND RESULTS: In this study, we examined whether MAA adduction of SPD (SPD-MAA) attenuates the ability of SPD to bind SARS-CoV-2 spike protein, reversing SPD-mediated virus neutralization. Using ELISA, we show that SPD-MAA is unable to competitively bind spike protein and prevent ACE2 receptor binding. Similarly, SPD-MAA fails to inhibit entry of wild-type SARS-CoV-2 virus into Calu-3 cells, a lung epithelial cell line, as well as ciliated primary human bronchial epithelial cells isolated from healthy individuals. CONCLUSIONS: Overall, MAA adduction of SPD, a consequence of alcohol overconsumption, represents one mechanism of compromised lung innate defense against SARS-CoV-2, highlighting a possible mechanism underlying COVID-19 severity and related mortality in patients who misuse alcohol.


Asunto(s)
Alcoholismo , COVID-19 , Humanos , Acetaldehído/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Malondialdehído/metabolismo , Pandemias , SARS-CoV-2/metabolismo , Etanol , Proteínas/metabolismo , Unión Proteica
9.
Cell Mol Immunol ; 20(1): 38-50, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36376488

RESUMEN

Increased levels of surfactant protein D (SP-D) and lipid-laden foamy macrophages (FMs) are frequently found under oxidative stress conditions and/or in patients with chronic obstructive pulmonary disease (COPD) who are also chronically exposed to cigarette smoke (CS). However, the roles and molecular mechanisms of SP-D and FMs in COPD have not yet been determined. In this study, increased levels of SP-D were found in the bronchoalveolar lavage fluid (BALF) and sera of ozone- and CS-exposed mice. Furthermore, SP-D-knockout mice showed increased lipid-laden FMs and airway inflammation caused by ozone and CS exposure, similar to that exhibited by our study cohort of chronic smokers and COPD patients. We also showed that an exogenous recombinant fragment of human SP-D (rfhSP-D) prevented the formation of oxidized low-density lipoprotein (oxLDL)-induced FMs in vitro and reversed the airway inflammation and emphysematous changes caused by oxidative stress and CS exposure in vivo. SP-D upregulated bone marrow-derived macrophage (BMDM) expression of genes involved in countering the oxidative stress and lipid metabolism perturbations induced by CS and oxLDL. Our study demonstrates the crucial roles of SP-D in the lipid homeostasis of dysfunctional alveolar macrophages caused by ozone and CS exposure in experimental mouse emphysema, which may provide a novel opportunity for the clinical application of SP-D in patients with COPD.


Asunto(s)
Ozono , Neumonía , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Ratones , Animales , Pulmón/metabolismo , Proteína D Asociada a Surfactante Pulmonar/genética , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Macrófagos/metabolismo , Líquido del Lavado Bronquioalveolar , Inflamación/metabolismo , Ozono/farmacología , Ozono/metabolismo , Lípidos , Ratones Endogámicos C57BL
10.
Acta Biochim Pol ; 69(4): 697-702, 2022 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-36515569

RESUMEN

This study was conducted to investigate the changes of Club cell protein 16 (CC16) and surfactant protein D (SP-D) levels in serum and bronchoalveolar lavage fluid (BALF) in silicotic rats and to explore their potential as early biomarkers for silicosis. Pulmonary fibrosis models of rats were constructed by exposing them to silica particles. BALF and serum were collected to determine CC16 and SP-D levels using enzyme-linked immunosorbent assay (ELISA) at different times after the exposure. Hydroxyproline (HYP) level in BALF and CC16 level in the lung tissues were also measured immunohistochemistrially. The BALF levels of CC16 decreased from 49.65 to 38.02 ng/mg after the rats were exposed to silica for 3 and 28 days, which were all significantly lower as compared with the controls (P<0.05), where the levels remained barely changed during the same period (61.27 to 56.76 ng/mg). The serum CC16 also showed a similar decrease from 9.8 ng/ml to 8.78 ng/ml during the period, while in the controls, the serum CC16 levels remained constantly between 11.04 and 10.96 ng/ml. The levels of SP-D in the serum of silica-exposed rats did not decrease as compared with the controls and BALF SP-D presented a parabolic curve change with silica exposure. Immunohistochemical examinations showed that the lung Club cells were severely damaged and CC16 expression was obviously decreased after silica exposure. BALF HYP level was higher in silica-exposed rats than in control only when the exposure was at 50 mg/ml. Our work demonstrates that expressions of CC16 and SP-D are pulmonary tissue-specific and CC16 expression is down-regulated as a result of silica-exposure. The significant relationship between CC16 and silica dose indicates that CC16 may be exploited as an early biomarker to assess silica-induced pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar , Ratas , Animales , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Uteroglobina/metabolismo , Dióxido de Silicio/efectos adversos , Dióxido de Silicio/metabolismo , Pulmón/metabolismo , Biomarcadores/metabolismo
11.
Allergol Immunopathol (Madr) ; 50(6): 176-186, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36335462

RESUMEN

BACKGROUND: Injury to the lung is a common, clinically serious inflammatory disease. However, its pathogenesis remains unclear, and the existing treatments, including cytokine therapy, stem cell therapy, and hormone therapy, are not completely effective in treating this disease. Dimethyl itaconate (DMI) is a surfactant with important anti-inflammatory effects. OBJECTIVE: The present study used alveolar type II (AT II) and bronchial epithelial cells as models to determine the role of DMI in lung injury. MATERIAL AND METHODS: First, the effects of DMI were established on the survival, inflammatory release, and apoptosis in lipopolysaccharide (LPS)-induced AT II and bronchial epithelial cells. The association between DMI and Sirtuin1 (SIRT1) was assessed using molecular docking. Next, by constructing interference plasmids to inhibit surfactant protein (SP)-A and SP-D expressions, the effect of DMI was observed on inflammatory release and apoptosis. RESULTS: The results revealed that DMI increased the survival rate and expression levels of SP-A, SP-D, and SIRT1, and inhibited inflammatory factors as well as apoptosis in LPS-induced cells. Furthermore, DMI could bind to SIRT1 to regulate SP-A and SP-D expressions. After SP-A and SP-D expressions were inhibited, the inhibitory effect of DMI was reversed on inflammatory release and apoptosis. CONCLUSION: The findings of the present study revealed that DMI inhibited LPS-induced inflammatory release and apoptosis in cells by targeting SIRT1 and then activating SP-A and SP-D. This novel insight into the pharmacological mechanism of DMI lays the foundation for its later use for alleviating lung injury.


Asunto(s)
Lesión Pulmonar , Surfactantes Pulmonares , Humanos , Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Surfactantes Pulmonares/metabolismo , Surfactantes Pulmonares/farmacología , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Proteína D Asociada a Surfactante Pulmonar/farmacología , Sirtuina 1/metabolismo , Sirtuina 1/farmacología , Lesión Pulmonar/metabolismo , Simulación del Acoplamiento Molecular , Células Epiteliales/metabolismo , Proteína A Asociada a Surfactante Pulmonar/metabolismo , Proteína A Asociada a Surfactante Pulmonar/farmacología , Apoptosis , Tensoactivos/metabolismo , Tensoactivos/farmacología
12.
Allergol. immunopatol ; 50(6): 176-186, 01 nov. 2022. graf, ilus
Artículo en Inglés | IBECS | ID: ibc-211519

RESUMEN

Background Injury to the lung is a common, clinically serious inflammatory disease. However, its pathogenesis remains unclear, and the existing treatments, including cytokine therapy, stem cell therapy, and hormone therapy, are not completely effective in treating this disease. Dimethyl itaconate (DMI) is a surfactant with important anti-inflammatory effects. Objective The present study used alveolar type II (AT II) and bronchial epithelial cells as models to determine the role of DMI in lung injury. Material and Methods First, the effects of DMI were established on the survival, inflammatory release, and apoptosis in lipopolysaccharide (LPS)-induced AT II and bronchial epithelial cells. The association between DMI and Sirtuin1 (SIRT1) was assessed using molecular docking. Next, by constructing interference plasmids to inhibit surfactant protein (SP)-A and SP-D expressions, the effect of DMI was observed on inflammatory release and apoptosis. Results The results revealed that DMI increased the survival rate and expression levels of SP-A, SP-D, and SIRT1, and inhibited inflammatory factors as well as apoptosis in LPS-induced cells. Furthermore, DMI could bind to SIRT1 to regulate SP-A and SP-D expressions. After SP-A and SP-D expressions were inhibited, the inhibitory effect of DMI was reversed on inflammatory release and apoptosis. Conclusion The findings of the present study revealed that DMI inhibited LPS-induced inflammatory release and apoptosis in cells by targeting SIRT1 and then activating SP-A and SP-D. This novel insight into the pharmacological mechanism of DMI lays the foundation for its later use for alleviating lung injury (AU)


Asunto(s)
Humanos , Surfactantes Pulmonares/metabolismo , Surfactantes Pulmonares/farmacología , Células Epiteliales/metabolismo , Apoptosis , Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Lesión Pulmonar/metabolismo , Simulación del Acoplamiento Molecular , Proteína A Asociada a Surfactante Pulmonar/metabolismo , Proteína A Asociada a Surfactante Pulmonar/farmacología , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Proteína D Asociada a Surfactante Pulmonar/farmacocinética , Sirtuina 1/metabolismo , Sirtuina 1/farmacología
13.
Front Immunol ; 13: 913901, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35865531

RESUMEN

Osteoarthritis (OA) is a deteriorating disease of cartilage tissues mainly characterized as low-grade inflammation of the joint. Innate immune molecule surfactant protein D (SP-D) is a member of collectin family of collagenous Ca2+-dependent defense lectins and plays a vital role in the inflammatory and innate immune responses. The present study investigated the SP-D-mediated innate/inflammatory bioregulation in OA and explored the underlying molecular mechanism. Transcriptome analysis revealed that SP-D regulated genes were strongly enriched in the inflammatory response, immune response, cellular response to lipopolysaccharide (LPS), PI3K-Akt signaling, Toll-like receptor (TLR) signaling, and extracellular matrix (ECM)-receptor interaction pathways. Knockdown of the SP-D gene by the recombinant adeno-associated virus promoted the macrophage specific markers of CD68, F4/80 and TLR4 in the articular cartilage in vivo. SP-D alleviated the infiltration of synovial macrophages and neutrophils, and inhibited TLR4, TNF-α and the phosphorylation of PI3K, Akt and NF-κB p65 in cartilage. SP-D suppressed cartilage degeneration, inflammatory and immune responses in the rat OA model, whilst TAK-242 strengthened this improvement. In in vitro conditions, SP-D pre-treatment inhibited LPS-induced overproduction of inflammation-correlated cytokines such as IL-1ß and TNF-α, and suppressed the overexpression of TLR4, MD-2 and NLRP3. SP-D prevented the LPS-induced degradation of ECM by down-regulating MMP-13 and up-regulating collagen II. Blocking of TLR4 by TAK-242 further enhanced these manifestations. We also demonstrated that SP-D binds to the TLR4/MD-2 complex to suppress TLR4-mediated PI3K/Akt and NF-κB signaling activation in chondrocytes. Taken together, these findings indicate that SP-D has chondroprotective properties dependent on TLR4-mediated PI3K/Akt and NF-κB signaling and that SP-D has an optimal bioregulatory effect on the inflammatory and innate responses in OA.


Asunto(s)
Osteoartritis , Proteína D Asociada a Surfactante Pulmonar , Receptor Toll-Like 4 , Animales , Inflamación , Lipopolisacáridos/efectos adversos , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Ratas , Factor de Necrosis Tumoral alfa
14.
Front Immunol ; 13: 866795, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35669781

RESUMEN

Alcohol consumption with concurrent cigarette smoking produces malondialdehyde acetaldehyde (MAA)-adducted lung proteins. Lung surfactant protein D (SPD) supports innate immunity via bacterial aggregation and lysis, as well as by enhancing macrophage-binding and phagocytosis. MAA-adducted SPD (SPD-MAA) has negative effects on lung cilia beating, macrophage function, and epithelial cell injury repair. Because changes in SPD multimer structure are known to impact SPD function, we hypothesized that MAA-adduction changes both SPD structure and function. Purified human SPD and SPD-MAA (1 mg/mL) were resolved by gel filtration using Sephadex G-200 and protein concentration of each fraction determined by Bradford assay. Fractions were immobilized onto nitrocellulose by slot blot and assayed by Western blot using antibodies to SPD and to MAA. Binding of SPD and SPD-MAA was determined fluorometrically using GFP-labeled Streptococcus pneumoniae (GFP-SP). Anti-bacterial aggregation of GFP-SP and macrophage bacterial phagocytosis were assayed by microscopy and permeability determined by bacterial phosphatase release. Viral injury was measured as LDH release in RSV-treated airway epithelial cells. Three sizes of SPD were resolved by gel chromatography as monomeric, trimeric, and multimeric forms. SPD multimer was the most prevalent, while the majority of SPD-MAA eluted as trimer and monomer. SPD dose-dependently bound to GFP-SP, but SPD-MAA binding to bacteria was significantly reduced. SPD enhanced, but MAA adduction of SPD prevented, both aggregation and macrophage phagocytosis of GFP-SP. Likewise, SPD increased bacterial permeability while SPD-MAA did not. In the presence of RSV, BEAS-2B cell viability was enhanced by SPD, but not protected by SPD-MAA. Our results demonstrate that MAA adduction changes the quaternary structure of SPD from multimer to trimer and monomer leading to a decrease in the native anti-microbial function of SPD. These findings suggest one mechanism for increased pneumonia observed in alcohol use disorders.


Asunto(s)
Acetaldehído , Alcoholismo , Acetaldehído/química , Acetaldehído/metabolismo , Alcoholismo/metabolismo , Humanos , Pulmón/metabolismo , Malondialdehído , Proteína D Asociada a Surfactante Pulmonar/metabolismo
15.
Int Ophthalmol ; 42(11): 3611-3623, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35639299

RESUMEN

PURPOSE: Our review explains the role of surfactant protein D (SP-D) in different kinds of bacterial infection based on its presence in different ocular surface tissues. We discuss the potential role of SP-D against invasion by pathogens, with the aim of identifying new prospects for the possible mechanism of SP-D-mediated immune processes, and the diagnosis, prognosis, or treatment of ocular bacterial infection. METHODS: We reviewed articles about the role of SP-D in various ocular bacterial infections or infection-related ocular diseases through PubMed, Google Scholar, and the Web of Science databases. RESULTS: SP-D acts as an important immune factor that can resemble molecules in different polymerization states and that defends against pathogen invasion. The increased SP-D production and secretion in tear fluid and the cornea after ocular bacterial infections such as Staphylococcus aureus, Pseudomonas aeruginosa keratitis, and infection-related ocular diseases, was shown to have potential anti-inflammatory effects. The mechanisms of SP-D's action against ocular bacterial infections include presenting, aggregating, opsonizing, and phagocytizing antigens, as well as regulating anti-bacterial immunity processes, including toll-like receptor-5 (TLR-5) pathway and IL-8 effect, TLR-4 and TLR-2 pathways and other possible ways remained to be elucidated in more detail. The findings demonstrate the potential of SP-D as an important clinical diagnostic biomarker prognosis predictor, and target for ocular immunotherapy. CONCLUSION: SP-D participates in invasion by different ocular bacteria and infection-related ocular diseases through multiple immune mechanisms. This finding provides new prospects for the diagnosis, prognosis and treatment of ocular bacterial infection.


Asunto(s)
Infecciones Bacterianas del Ojo , Proteína D Asociada a Surfactante Pulmonar , Humanos , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Interleucina-8/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 5/metabolismo , Córnea/metabolismo , Infecciones Bacterianas del Ojo/diagnóstico , Infecciones Bacterianas del Ojo/microbiología , Antiinflamatorios , Tensoactivos/metabolismo
16.
Comput Intell Neurosci ; 2022: 7205016, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35463266

RESUMEN

Objective: To study the mechanism of chronic obstructive pulmonary disease (COPD) in diagnosing alveolar factors and analyze the effect of miR-149-3p on alveolar inflammatory factors and the expression of surfactant protein D (SP-D) and SP-A on the lung surface mediated by Wnt pathway. Methods: Patients with stable COPD were taken as the research subjects, and healthy volunteers as the control group. Cardiac color Doppler ultrasound was adopted to measure the ventricular structure of patients. The ultrasound simulation method was introduced in the ultrasound imaging. The ultrasound image was processed based on the intelligent ultrasound simulation algorithm. The changes in the structure of the left and right ventricles were analyzed and compared in the two groups. The expression changes of miR-149-3p, Wnt1, ß-catenin, RhoA, and Wnt5a in lung tissues of mice in three groups were detected, as well as the content of tumor necrosis factor- (TNF-) α, IL-1ß, interleukin (IL-6), nuclear factor kB (NF-kB), and other inflammatory factors in bronchoalveolar tissues of mice in three groups. Results: The position where the attenuation ratio was less than 0.92 in the experiment under the ultrasonic simulation algorithm had a gray value of 50. Compared with the control group, the right ventricular mass index of patients with stable COPD was statistically considerable (P < 0.05). In patients with stable COPD, the overall right ventricular longitudinal strain, right ventricular diastolic longitudinal strain rate (RV DLSR), right ventricular diastolic circumferential strain rate, and right ventricular longitudinal displacement were significantly impaired (P < 0.05). The content of miR-149-3p in the lung tissue of the model group was dramatically inferior to that of the control group and the interference group (P < 0.05). The contents of Wnt1, ß-catenin, RhoA, and Wnt5a in the lung tissue of the model group were dramatically superior to those of the control group (P < 0.05). In addition, the expressions of TNF-α, IL-1ß, IL-6, and NF-kB in the alveolar lavage fluid of the model group were statistically different from those of control group (P < 0.05). The expression levels of SP-D and surfactant protein A (SP-A) in the COPD group were also statistically different from those of control group (P < 0.05). Conclusion: miR-149-3p regulated the expression of Wnt1, ß-catenin, RhoA, and Wnt5a, which also affected the signal transmission of the Wnt pathway, causing changes in the expression of alveolar inflammatory factors. Eventually, it affected the development of COPD.


Asunto(s)
MicroARNs , Enfermedad Pulmonar Obstructiva Crónica , Animales , Humanos , Interleucina-6/metabolismo , Interleucina-6/farmacología , Pulmón , Ratones , MicroARNs/genética , MicroARNs/metabolismo , MicroARNs/farmacología , FN-kappa B/metabolismo , FN-kappa B/farmacología , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico por imagen , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Proteína A Asociada a Surfactante Pulmonar/metabolismo , Proteína A Asociada a Surfactante Pulmonar/farmacología , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Proteína D Asociada a Surfactante Pulmonar/farmacología , Tensoactivos/metabolismo , Tensoactivos/farmacología , Vía de Señalización Wnt , beta Catenina/genética , beta Catenina/metabolismo , beta Catenina/farmacología
17.
Inhal Toxicol ; 34(5-6): 145-158, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35452355

RESUMEN

OBJECTIVE: Ammonia (NH3) is a corrosive alkaline gas that can cause life-threatening injuries by inhalation. The aim was to establish a disease model for NH3-induced injuries similar to acute lung injury (ALI) described in exposed humans and investigate the progression of lung damage, respiratory dysfunction and evaluate biomarkers for ALI and inflammation over time. METHODS: Female BALB/c mice were exposed to an NH3 dose of 91.0 mg/kg·bw using intratracheal instillation and the pathological changes were followed for up to 7 days. RESULTS: NH3 instillation resulted in the loss of body weight along with a significant increase in pro-inflammatory mediators in both bronchoalveolar lavage fluid (e.g. IL-1ß, IL-6, KC, MMP-9, SP-D) and blood (e.g. IL-6, Fibrinogen, PAI-1, PF4/CXCL4, SP-D), neutrophilic lung inflammation, alveolar damage, increased peripheral airway resistance and methacholine-induced airway hyperresponsiveness compared to controls at 20 h. On day 7 after exposure, deteriorating pathological changes such as increased macrophage lung infiltration, heart weights, lung hemorrhages and coagulation abnormalities (elevated plasma levels of PAI-1, fibrinogen, endothelin and thrombomodulin) were observed but no increase in lung collagen. Some of the analyzed blood biomarkers (e.g. RAGE, IL-1ß) were unaffected despite severe ALI and may not be significant for NH3-induced damages. CONCLUSIONS: NH3 induces severe acute lung injuries that deteriorate over time and biomarkers in lungs and blood that are similar to those found in humans. Therefore, this model has potential use for developing diagnostic tools for NH3-induced ALI and for finding new therapeutic treatments, since no specific antidote has been identified yet.


Asunto(s)
Lesión Pulmonar Aguda , Amoníaco , Lesión Pulmonar Aguda/patología , Amoníaco/toxicidad , Animales , Líquido del Lavado Bronquioalveolar , Modelos Animales de Enfermedad , Femenino , Fibrinógeno/metabolismo , Interleucina-6/metabolismo , Pulmón , Ratones , Ratones Endogámicos BALB C , Inhibidor 1 de Activador Plasminogénico/metabolismo , Proteína D Asociada a Surfactante Pulmonar/metabolismo
18.
Front Immunol ; 12: 687506, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34484184

RESUMEN

Surfactant protein D (SP-D) plays an important role in innate and adaptive immune responses. In this study, we found that the expression of total and de-oligomerized SP-D was significantly elevated in mice with lipopolysaccharide (LPS)-induced acute lung injury (ALI). To investigate the role of the lower oligomeric form of SP-D in the pathogenesis of ALI, we treated bone marrow-derived macrophages (BMDMs) with ALI-derived bronchoalveolar lavage (BAL) and found that SP-D in ALI BAL predominantly bound to calreticulin (CALR) on macrophages, subsequently increasing the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and expression of interleukin (IL)-6, tumor necrosis factor (TNF)-alpha, IL-10, and CD80. However, anti-SP-D (aSP-D) and anti-calreticulin (aCALR) pretreatment reversed the SP-D binding and activation of macrophages induced by ALI BAL or de-oligomerized recombinant murine SP-D (rSP-D). Lack of signal transducer and activator of transcription (STAT)6 in STAT6-/- macrophages resulted in resistance to suppression by aCALR. Further studies in an ALI mouse model showed that blockade of pulmonary SP-D by intratracheal (i.t.), but not intraperitoneal (i.p.), administration of aSP-D attenuated the severity of ALI, accompanied by lower neutrophil infiltrates and expression of IL-1beta and IL-6. Furthermore, i.t. administration of de-oligomerized rSP-D exacerbated the severity of ALI in association with more pro-inflammatory CD45+Siglec-F(-) M1 subtype macrophages and production of IL-6, TNF-alpha, IL-1beta, and IL-18. The results indicated that SP-D in the lungs of murine ALI was de-oligomerized and participated in the pathogenesis of ALI by predominantly binding to CALR on macrophages and subsequently activating the pro-inflammatory downstream signaling pathway. Targeting de-oligomerized SP-D is a promising therapeutic strategy for the treatment of ALI and acute respiratory distress syndrome (ARDS).


Asunto(s)
Lesión Pulmonar Aguda/enzimología , Calbindina 2/metabolismo , Pulmón/enzimología , Activación de Macrófagos , Macrófagos/enzimología , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/inmunología , Animales , Anticuerpos/farmacología , Calbindina 2/antagonistas & inhibidores , Citocinas/metabolismo , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Lipopolisacáridos , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Infiltración Neutrófila , Fenotipo , Fosforilación , Proteína D Asociada a Surfactante Pulmonar/antagonistas & inhibidores , Células RAW 264.7 , Transducción de Señal
19.
Biomolecules ; 11(8)2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34439781

RESUMEN

SARS-CoV-2 infection of host cells is driven by binding of the SARS-CoV-2 spike-(S)-protein to lung type II pneumocytes, followed by virus replication. Surfactant protein SP-D, member of the front-line immune defense of the lungs, binds glycosylated structures on invading pathogens such as viruses to induce their clearance from the lungs. The objective of this study is to measure the pulmonary SP-D levels in COVID-19 patients and demonstrate the activity of SP-D against SARS-CoV-2, opening the possibility of using SP-D as potential therapy for COVID-19 patients. Pulmonary SP-D concentrations were measured in bronchoalveolar lavage samples from patients with corona virus disease 2019 (COVID-19) by anti-SP-D ELISA. Binding assays were performed by ELISAs. Protein bridge and aggregation assays were performed by gel electrophoresis followed by silver staining and band densitometry. Viral replication was evaluated in vitro using epithelial Caco-2 cells. Results indicate that COVID-19 patients (n = 12) show decreased pulmonary levels of SP-D (median = 68.9 ng/mL) when compared to levels reported for healthy controls in literature. Binding assays demonstrate that SP-D binds the SARS-CoV-2 glycosylated spike-(S)-protein of different emerging clinical variants. Binding induces the formation of protein bridges, the critical step of viral aggregation to facilitate its clearance. SP-D inhibits SARS-CoV-2 replication in Caco-2 cells (EC90 = 3.7 µg/mL). Therefore, SP-D recognizes and binds to the spike-(S)-protein of SARS-CoV-2 in vitro, initiates the aggregation, and inhibits viral replication in cells. Combined with the low levels of SP-D observed in COVID-19 patients, these results suggest that SP-D is important in the immune response to SARS-CoV-2 and that rhSP-D supplementation has the potential to be a novel class of anti-viral that will target SARS-CoV-2 infection.


Asunto(s)
COVID-19/metabolismo , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Adulto , Anciano , COVID-19/virología , Células CACO-2 , Femenino , Humanos , Masculino , Persona de Mediana Edad , Unión Proteica , Proteína D Asociada a Surfactante Pulmonar/genética , Proteína D Asociada a Surfactante Pulmonar/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad , SARS-CoV-2/fisiología , Replicación Viral
20.
Can J Vet Res ; 85(3): 170-176, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34248260

RESUMEN

The lung is a complex organ, and its physiology and immunology are regulated by various immune molecules and cells. Lung surfactant, a mixture of phospholipids and proteins produced by the bronchiolar and type II alveolar epithelial cells, is one such important player in lung physiology. Compared to knowledge about the biology of the surfactant in rodents and humans, only limited data are available on the surfactant in the horse. Although there are data linking levels of surfactant proteins with respiratory disease in the horse, there are no data on the cellular localization of surfactant protein A (SP-A) and surfactant protein D (SP-D). A member of the tetraspanin family of proteins, CD9 is a cell-signaling and adhesion protein and its expression has been detected in both normal and cancer cells, including those in the lung. Because there are no immunolocalization data on SP-A, SP-D, and CD9 in the normal lungs of the horse, our objective was to conduct a light and electron microscopic immunocytochemical study on normal lungs of the horse. The data showed SP-A and SP-D in bronchiolar epithelial and type II alveolar epithelial cells. These proteins were also localized in type I alveolar epithelial cells, pulmonary intravascular macrophages, and neutrophils, which is likely an outcome of endocytosis of the proteins by these cells. CD9 was present in the airway and vascular smooth muscle cells, endothelium, and blood cells, but not in the airway epithelium. These new data provide a baseline to further examine the expression and functions of SP-A, SP-D, and CD9 proteins in inflammation associated with respiratory diseases in the horse.


Le poumon est un organe complexe, et sa physiologie et son immunologie sont régulées par diverses molécules et cellules immunitaires. Le surfactant pulmonaire, un mélange de phospholipides et de protéines produits par les cellules épithéliales bronchiolaires et alvéolaires de type II, est un acteur important de la physiologie pulmonaire. Par rapport aux connaissances sur la biologie du surfactant chez les rongeurs et les humains, seules des données limitées sont disponibles sur le surfactant chez le cheval. Bien qu'il existe des données reliant les niveaux de protéines du surfactant à une maladie respiratoire chez le cheval, il n'y a pas de données sur la localisation cellulaire de la protéine de surfactant A (SP-A) et de la protéine de surfactant D (SP-D). Membre de la famille des protéines tétraspanines, CD9 est une protéine de signalisation et d'adhésion cellulaire et son expression a été détectée dans les cellules normales et cancéreuses, y compris celles du poumon. Comme il n'y a pas de données d'immunolocalisation pour SP-A, SP-D et CD9 dans les poumons normaux du cheval, notre objectif était de mener une étude immunocytochimique au microscope optique et électronique sur les poumons normaux du cheval. Les données ont montré la présence de SP-A et SP-D dans les cellules épithéliales bronchiolaires et alvéolaires de type II. Ces protéines étaient également localisées dans les cellules épithéliales alvéolaires de type I, les macrophages intravasculaires pulmonaires et les neutrophiles, ce qui est probablement le résultat de l'endocytose des protéines par ces cellules. Le CD9 était présent dans les cellules des voies respiratoires et des muscles lisses vasculaires, l'endothélium et les cellules sanguines, mais pas dans l'épithélium des voies respiratoires. Ces nouvelles données fournissent une base de référence pour examiner plus à fond l'expression et les fonctions des protéines SP-A, SP-D et CD9 dans l'inflammation associée aux maladies respiratoires chez le cheval.(Traduit par Docteur Serge Messier).


Asunto(s)
Caballos , Pulmón/metabolismo , Proteína A Asociada a Surfactante Pulmonar/metabolismo , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Tetraspanina 29/metabolismo , Animales , Regulación de la Expresión Génica , Inmunohistoquímica/métodos , Inmunohistoquímica/veterinaria , Pulmón/ultraestructura , Microscopía Electrónica , Proteína A Asociada a Surfactante Pulmonar/genética , Proteína D Asociada a Surfactante Pulmonar/genética , Tetraspanina 29/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...