Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 751
Filtrar
1.
Funct Integr Genomics ; 24(6): 200, 2024 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-39453497

RESUMEN

Septic lung injury is an unmet clinical challenge due to its high mortality, and there is a lack of effective treatment. Accumulating evidence suggests that an uncontrolled pulmonary inflammatory response is important in the pathogenesis of lung injury in sepsis. Therefore, limiting excessive early inflammatory responses may be an effective strategy. We established a septic lung injury model using cecal ligation and puncture. Western blotting and immunofluorescence analyses were performed to assess the expression of PTP1B and endoplasmic reticulum (ER) stress and pyroptosis. Co-immunoprecipitation was used to analyze the binding of PTP1B and Src molecules. PTP1B is upregulated in both in vivo and in vitro models of septic lung injury. PTP1B directly binds to Src and aggravates inflammation by regulating the ER stress-pyroptosis axis. The inhibition of PTP1B alleviates inflammation and improves the prognosis of septic mice. Our study suggesting that PT1B inhibitors have clinical application value in the treatment of septic lung injury. This may provide a new strategy for the treatment of septic lung injury.


Asunto(s)
Lesión Pulmonar , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Piroptosis , Sepsis , Transducción de Señal , Animales , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Ratones , Sepsis/tratamiento farmacológico , Sepsis/metabolismo , Piroptosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Masculino , Familia-src Quinasas/metabolismo , Familia-src Quinasas/genética , Ratones Endogámicos C57BL , Humanos , Modelos Animales de Enfermedad
2.
Front Immunol ; 15: 1451577, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39430764

RESUMEN

Introduction: Pulmonary ischemia-reperfusion (IR) injury (IRI) plays a significant role in various lung disorders and is a key factor in the development of primary graft dysfunction following lung transplantation. Hemopexin (Hx) is the major serum scavenger protein for heme, which is a prooxidant and pro-inflammatory compound. In the current study, we hypothesized that Hx could confer beneficial effects in sterile inflammation induced by IR-mediated lung injury. Methods: To examine this hypothesis, we administered Hx in an experimental mouse model of unilateral lung IRI. Results: Our results demonstrate that treatment with Hx alleviated histopathological signs of inflammation in ischemic lungs, as evidenced by a reduction in the number of infiltrating neutrophils and decreased levels of perivascular edema. In addition, thrombotic vaso-occlusion in pulmonary blood vessels of IRI lungs was reduced by Hx. Immunohistochemical analysis revealed that Hx inhibited the up-regulation of heme oxygenase-1, an enzyme highly induced by heme, in ischemic lungs. Finally, Hx administration caused a decrease in the levels of circulating B- and CD8+ T-lymphocytes in the peripheral blood of mice with pulmonary IRI. Conclusion: These findings suggest that the serum heme scavenger protein Hx holds therapeutic promise in alleviating lung IRI-mediated sterile inflammation. Thus, Hx may represent a preemptive therapeutic approach in IR-related lung disorders such as primary graft dysfunction in lung transplantation.


Asunto(s)
Modelos Animales de Enfermedad , Hemopexina , Daño por Reperfusión , Animales , Hemopexina/metabolismo , Hemopexina/farmacología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Ratones , Masculino , Hemo-Oxigenasa 1/metabolismo , Pulmón/patología , Pulmón/inmunología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones Endogámicos C57BL , Inflamación , Lesión Pulmonar/etiología , Lesión Pulmonar/tratamiento farmacológico , Proteínas de la Membrana
3.
FASEB J ; 38(17): e70027, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39221615

RESUMEN

The complex pathogenesis of lung ischemia-reperfusion injury (LIRI) was examined in a murine model, focusing on the role of pyroptosis and its exacerbation of lung injury. We specifically examined the levels and cellular localization of pyroptosis within the lung, which revealed alveolar macrophages as the primary site. The inhibition of pyroptosis by VX-765 reduced the severity of lung injury, underscoring its significant role in LIRI. Furthermore, the therapeutic potential of ß-hydroxybutyrate (ß-OHB) in ameliorating LIRI was examined. Modulation of ß-OHB levels was evaluated by ketone ester supplementation and 3-hydroxybutyrate dehydrogenase 1 (BDH-1) gene knockout, along with the manipulation of the SIRT1-FOXO3 signaling pathway using EX-527 and pCMV-SIRT1 plasmid transfection. This revealed that ß-OHB exerts lung-protective and anti-pyroptotic effects, which were mediated through the upregulation of SIRT1 and the enhancement of FOXO3 deacetylation, leading to decreased pyroptosis markers and lung injury. In addition, ß-OHB treatment of MH-S cells in vitro showed a concentration-dependent improvement in pyroptosis, linking its therapeutic benefits to specific cell mechanisms. Overall, this study highlights the significance of alveolar macrophage pyroptosis in the exacerbation of LIRI and indicates the potential of ß-OHB in mitigating injury by modulating the SIRT1-FOXO3 signaling pathway.


Asunto(s)
Ácido 3-Hidroxibutírico , Proteína Forkhead Box O3 , Macrófagos Alveolares , Ratones Endogámicos C57BL , Piroptosis , Daño por Reperfusión , Transducción de Señal , Sirtuina 1 , Animales , Proteína Forkhead Box O3/metabolismo , Piroptosis/efectos de los fármacos , Sirtuina 1/metabolismo , Ratones , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Daño por Reperfusión/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Masculino , Ácido 3-Hidroxibutírico/farmacología , Pulmón/metabolismo , Pulmón/patología , Carbazoles/farmacología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/tratamiento farmacológico
4.
Appl Microbiol Biotechnol ; 108(1): 469, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39298023

RESUMEN

Prior research has indicated that the gut-lung-axis can be influenced by the intestinal microbiota, thereby impacting lung immunity. Rifaximin is a broad-spectrum antibacterial drug that can maintain the homeostasis of intestinal microflora. In this study, we established an influenza A virus (IAV)-infected mice model with or without rifaximin supplementation to investigate whether rifaximin could ameliorate lung injury induced by IAV and explore the molecular mechanism involved. Our results showed that IAV caused significant weight loss and disrupted the structure of the lung and intestine. The analysis results of 16S rRNA and metabolomics indicated a notable reduction in the levels of probiotics Lachnoclostridium, Ruminococcaceae_UCG-013, and tryptophan metabolites in the fecal samples of mice infected with IAV. In contrast, supplementation with 50 mg/kg rifaximin reversed these changes, including promoting the repair of the lung barrier and increasing the abundance of Muribaculum, Papillibacter and tryptophan-related metabolites content in the feces. Additionally, rifaximin treatment increased ILC3 cell numbers, IL-22 level, and the expression of RORγ and STAT-3 protein in the lung. Furthermore, our findings demonstrated that the administration of rifaximin can mitigate damage to the intestinal barrier while enhancing the expression of AHR, IDO-1, and tight junction proteins in the small intestine. Overall, our results provided that rifaximin alleviated the imbalance in gut microbiota homeostasis induced by IAV infection and promoted the production of tryptophan-related metabolites. Tryptophan functions as a signal to facilitate the activation and movement of ILC3 cells from the intestine to the lung through the AHR/STAT3/IL-22 pathway, thereby aiding in the restoration of the barrier. KEY POINTS: • Rifaximin ameliorated IAV infection-caused lung barrier injury and induced ILC3 cell activation. • Rifaximin alleviated IAV-induced gut dysbiosis and recovered tryptophan metabolism. • Tryptophan mediates rifaximin-induced ILC3 cell activation via the AHR/STAT3/IL-22 pathway.


Asunto(s)
Microbioma Gastrointestinal , Virus de la Influenza A , Pulmón , Infecciones por Orthomyxoviridae , Rifaximina , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Rifaximina/uso terapéutico , Ratones , Pulmón/microbiología , Pulmón/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Virus de la Influenza A/efectos de los fármacos , Modelos Animales de Enfermedad , ARN Ribosómico 16S/genética , Interleucinas/metabolismo , Interleucinas/genética , Interleucina-22 , Ratones Endogámicos C57BL , Antibacterianos/farmacología , Factor de Transcripción STAT3/metabolismo , Heces/microbiología , Triptófano/metabolismo , Lesión Pulmonar/tratamiento farmacológico , Probióticos/administración & dosificación , Probióticos/farmacología
5.
J Agric Food Chem ; 72(38): 20944-20958, 2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39259217

RESUMEN

Radiation-induced lung injury (RILI) is a prevalent complication of thoracic tumor radiotherapy and accidental radiation exposure. Pyrroloquinoline quinone (PQQ), a novel vitamin B, plays a crucial role in delaying aging, antioxidation, anti-inflammation, and antiapoptosis. This study aims to investigate the protective effect and mechanisms of PQQ against RILI. C57BL/6 mice were exposed to a 20 Gy dose of X-ray radiation on the entire thorax with or without daily oral administration of PQQ for 2 weeks. PQQ effectively mitigated radiation-induced lung tissue damage, inflammation, oxidative stress, and epithelial cell apoptosis. Additionally, PQQ significantly inhibited oxidative stress and mitochondrial damage in MLE-12 cells. Mechanistically, PQQ upregulated the mRNA and protein levels of MOTS-c in irradiated lung tissue and MLE-12 cells. Knockdown of MOTS-c by siRNA substantially attenuated the protective effects of PQQ on oxidative stress, inflammation, and apoptosis. In conclusion, PQQ alleviates RILI by preserving mitochondrial function through a MOTS-c-dependent mechanism, suggesting that PQQ may serve as a promising nutraceutical intervention against RILI.


Asunto(s)
Apoptosis , Lesión Pulmonar , Ratones Endogámicos C57BL , Mitocondrias , Estrés Oxidativo , Cofactor PQQ , Animales , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación , Cofactor PQQ/farmacología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Lesión Pulmonar/metabolismo , Lesión Pulmonar/etiología , Lesión Pulmonar/genética , Lesión Pulmonar/prevención & control , Lesión Pulmonar/tratamiento farmacológico , Humanos , Apoptosis/efectos de los fármacos , Masculino , Traumatismos por Radiación/metabolismo , Traumatismos por Radiación/genética , Traumatismos por Radiación/tratamiento farmacológico , Traumatismos por Radiación/prevención & control , Pulmón/efectos de la radiación , Pulmón/metabolismo , Pulmón/efectos de los fármacos
6.
In Vivo ; 38(5): 2179-2189, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39187362

RESUMEN

BACKGROUND/AIM: Silibinin, has been investigated for its potential benefits and mechanisms in addressing vanadium pentoxide (V2O5)-induced pulmonary inflammation. This study explored the anti-inflammatory activity of silibinin and elucidate the mechanisms by which it operates in a mouse model of vanadium-induced lung injury. MATERIALS AND METHODS: Eight-week-old male BALB/c mice were exposed to V2O5 to induce lung injury. Mice were pretreated with silibinin at doses of 50 mg/kg and 100 mg/kg. Histological analyses were performed to assess cell viability and infiltration of inflammatory cells. The expression of pro-inflammatory cytokines (TNF-α, IL-6, IL-1ß) and activation of the MAPK and NF-[Formula: see text]B signaling pathways, as well as the NLRP3 inflammasome, were evaluated using real-time PCR, western blot analysis, and immunohistochemistry. Whole blood analysis was conducted to measure white blood cell counts. RESULTS: Silibinin treatment significantly improved cell viability, reduced inflammatory cell infiltration, and decreased the expression of pro-inflammatory cytokines in V2O5-induced lung injury. It also notably suppressed the activation of the MAPK and NF-[Formula: see text]B signaling pathways, along with a marked reduction in NLRP3 inflammasome expression levels in lung tissues. Additionally, silibinin-treated groups exhibited a significant decrease in white blood cell counts, including neutrophils, lymphocytes, and eosinophils. CONCLUSION: These findings underscore the potent anti-inflammatory effects of silibinin in mice with V2O5-induced lung inflammation, highlighting its therapeutic potential. The study not only confirms the efficacy of silibinin in mitigating inflammatory responses but also provides a foundational understanding of its role in modulating key inflammatory pathways, paving the way for future therapeutic strategies against pulmonary inflammation induced by environmental pollutants.


Asunto(s)
Citocinas , Lesión Pulmonar , FN-kappa B , Transducción de Señal , Silibina , Receptor Toll-Like 4 , Animales , Silibina/farmacología , Ratones , FN-kappa B/metabolismo , Masculino , Transducción de Señal/efectos de los fármacos , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Lesión Pulmonar/etiología , Citocinas/metabolismo , Receptor Toll-Like 4/metabolismo , Modelos Animales de Enfermedad , Vanadio/farmacología , Ratones Endogámicos BALB C , Antiinflamatorios/farmacología , Silimarina/farmacología , Mediadores de Inflamación/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo
7.
Immun Inflamm Dis ; 12(8): e70001, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39172009

RESUMEN

BACKGROUND: The inhalation of paraquat (PQ), one of the most widely used herbicides in the world, can result in lung injury. Curcuma longa (Cl) has long history in traditional and folk medicine for the treatment of a wide range of disorders including respiratory diseases. AIM: The aim of the present work was to evaluate the preventive effect of Cl on inhaled PQ-induced lung injury in rats. METHODS: Male Wistar rats were divided into 8 groups (n = 7), one group exposed to saline (control) and other groups exposed to PQ aerosol. Saline (PQ), Cl extract, (two doses), curcumin (Cu), pioglitazone (Pio), and the combination of Cl-L + Pio and dexamethasone (Dex) were administered during the exposure period to PQ. Total and differential white blood cell (WBC) counts, oxidant and antioxidant indicators in the bronchoalveolar lavage (BALF), interleukin (IL)-10, and tumor necrosis alpha (TNF-α) levels in the lung tissues, lung histologic lesions score, and air way responsiveness to methacholine were evaluated. RESULTS: WBC counts (Total and differential), malondialdehyde level, tracheal responsiveness (TR), IL-10, TNF-α and histopathological changes of the lung were markedly elevated but total thiol content and the activities of catalase and superoxide dismutase were decreased in the BALF in the PQ group. Both doses of Cl, Cu, Pio, Cl-L + Pio, and Dex markedly improved all measured variables in comparison with the PQ group. CONCLUSION: CI, Pio, and Cl-L + Pio improved PQ-induced lung inflammation and oxidative damage comparable with the effects of Dex.


Asunto(s)
Curcuma , PPAR gamma , Paraquat , Pioglitazona , Extractos Vegetales , Ratas Wistar , Animales , Pioglitazona/farmacología , Pioglitazona/uso terapéutico , Paraquat/toxicidad , Masculino , Ratas , Curcuma/química , PPAR gamma/agonistas , PPAR gamma/metabolismo , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/prevención & control , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/patología , Lesión Pulmonar/metabolismo , Dexametasona/farmacología , Líquido del Lavado Bronquioalveolar/citología , Estrés Oxidativo/efectos de los fármacos , Tiazolidinedionas/farmacología , Tiazolidinedionas/uso terapéutico , Antioxidantes/farmacología , Curcumina/farmacología , Curcumina/uso terapéutico
8.
J Physiol Pharmacol ; 75(3)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-39042394

RESUMEN

Bronchopulmonary dysplasia (BPD) is a common serious complication of premature babies. No effective means control it. Hyperoxia damage is one of the important mechanisms of BPD. The reaserach confirmed pyroptosis existed in BPD. Dexmedetomidine is a new, high-specific α2 receptor agonist. Previous research foundation found that dexmedetomidine has a protective effect on BPD. To investigate how dexmedetomidine improves hyperoxic lung injury in neonatal mice by regulating pyroptosis. Neonatal rats were randomly divided into four groups: normal control group, hyperoxic injury group, air plus dexmedetomidine group, and hyperoxia plus dexmedetomidine group. After seven days the lungs of rats in each group were extracted, and the wet-to-dry weight ratio of the lung was measured. The lung injury in rats was observed using hematoxylin-eosin staining. Additionally, the expression and localization of nucleotide-binding oligomerization domain-like receptor thermal protein domain associated protein 3 (NLRP3), apoptosis-associated speck-like protein (ASC), and gasdermin D (GSDMD) proteins were examined in the lungs of rats using immunofluorescence staining. The mRNA levels of NLRP3, ASC, caspase-1, and interleukin 18 (IL-18) in the lungs of rats were determined using real-time PCR. Moreover, the protein levels of NLRP3, ASC, caspase-1/cleaved caspase-1, interleukin 1beta (IL-1ß), IL-18, and tunor necrosis factor alpha (TNF-α) were detected in lungs of rats using Western blot. The extent of mitochondrial damage in lung tissues of each group was observed by transmission electron microscopy. The lung tissue injury of the neonatal rats was significantly improved in the hyperoxia plus dexmedetomidine group compared to the hyperoxic injury group. Furthermore, the expressions of pyroptosis-related proteins such as NLRP3, ASC, cleaved-caspase-1, and GSDMD were significantly decreased, along with the expressions of inflammatory factors in lung tissues. By inhibiting the NLRP3/caspase-1/GSDMD pyroptosis pathway, dexmedetomidine reduces the activation and release of inflammatory factors and provides a protective effect against hyperoxic lung injury in neonatal mice.


Asunto(s)
Animales Recién Nacidos , Dexmedetomidina , Hiperoxia , Lesión Pulmonar , Pulmón , Proteína con Dominio Pirina 3 de la Familia NLR , Piroptosis , Ratas Sprague-Dawley , Animales , Dexmedetomidina/farmacología , Dexmedetomidina/uso terapéutico , Hiperoxia/metabolismo , Hiperoxia/complicaciones , Hiperoxia/tratamiento farmacológico , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo , Piroptosis/efectos de los fármacos , Lesión Pulmonar/metabolismo , Lesión Pulmonar/prevención & control , Lesión Pulmonar/patología , Lesión Pulmonar/tratamiento farmacológico , Ratas , Proteínas de Unión a Fosfato/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasa 1/metabolismo , Interleucina-18/metabolismo , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Agonistas de Receptores Adrenérgicos alfa 2/uso terapéutico , Masculino , Gasderminas
9.
Immunopharmacol Immunotoxicol ; 46(5): 594-603, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39048516

RESUMEN

BACKGROUND: Inflammation and oxidative stress are key players in lung injury stemming from cardiac ischemia (LISCI). Cannabidiol (CBD) demonstrates tissue-protective properties through its antioxidant, anti-inflammatory, and anti-apoptotic characteristics. This study aims to assess the preventive (p-CBD) and therapeutic (t-CBD) effects of CBD on LISCI. METHODS: Forty male Wistar Albino rats were divided into four groups: control (CON), LISCI, p-CBD, and t-CBD. The left anterior descending coronary artery was ligated for 30 min of ischemia followed by 30 min of reperfusion. Lung tissues were then extracted for histopathological, immunohistochemical, genetic, and biochemical analyses. RESULTS: Histopathologically, marked hyperemia, increased septal tissue thickness, and inflammatory cell infiltrations were observed in the lung tissues of the LISCI group. Spectrophotometrically, total oxidant status and oxidative stress index levels were elevated, while total antioxidant status levels were decreased. Immunohistochemically, expressions of cyclooxygenase-1 (COX1), granulocyte colony-stimulating factor (GCSF), interleukin-6 (IL6) were increased. In genetic analyses, PERK and CHOP expressions were increased, whereas Nuclear factor erythroid 2-related factor 2 (NRF2) and B-cell leukemia/lymphoma 2 protein (BCL2) expressions were decreased. These parameters were alleviated by both prophylactic and therapeutic CBD treatment protocols. CONCLUSION: In LISCI-induced damage, both endoplasmic reticulum and mitochondrial stress, along with oxidative and inflammatory markers, were triggered, resulting in lung cell damage. However, both p-CBD and t-CBD treatments effectively reversed these mechanisms, normalizing all histopathological, biochemical, and PCR parameters.


Asunto(s)
Cannabidiol , Lesión Pulmonar , Isquemia Miocárdica , Factor 2 Relacionado con NF-E2 , Proteínas Proto-Oncogénicas c-bcl-2 , Ratas Wistar , Factor de Transcripción CHOP , Animales , Cannabidiol/farmacología , Masculino , Ratas , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factor de Transcripción CHOP/metabolismo , Lesión Pulmonar/prevención & control , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Isquemia Miocárdica/tratamiento farmacológico , Isquemia Miocárdica/patología , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/prevención & control , eIF-2 Quinasa/metabolismo , Modelos Animales de Enfermedad , Transducción de Señal/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos
10.
J Ethnopharmacol ; 334: 118584, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39019418

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Fuzheng-Qushi decoction (FZQS) is a practical Chinese herbal formula for relieving cough and fever. Therefore, the action and specific molecular mechanism of FZQS in the treatment of lung injury with cough and fever as the main symptoms need to be further investigated. AIMS OF THE STUDY: To elucidate the protective effects of FZQS against lung injury in mice and reveal its potential targets and key biological pathways for the treatment of lung injury based on transcriptomics, microbiomics, and untargeted metabolomics analyses. MATERIALS AND METHODS: Lipopolysaccharide (LPS) was used to induce a mouse model of lung injury, followed by the administration of FZQS. ELISA was used to detect IL-1ß, IL-6, IL-17A, IL-4, IL-10, and TNF-α, in mouse lung tissues. Macrophage polarization and neutrophil activation were measured by flow cytometry. RNA sequencing (RNA-seq) was applied to screen for differentially expressed genes (DEGs) in lung tissues. RT-qPCR and Western blot assays were utilized to validate key DEGs and target proteins in lung tissues. 16S rRNA sequencing was employed to characterize the gut microbiota of mice. Metabolites in the gut were analyzed using untargeted metabolomics. RESULTS: FZQS treatment significantly ameliorated lung histopathological damage, decreased pro-inflammatory cytokine levels, and increased anti-inflammatory cytokine levels. M1 macrophage levels in the peripheral blood decreased, M2 macrophage levels increased, and activated neutrophils were inhibited in mice with LPS-induced lung injury. Importantly, transcriptomic analysis showed that FZQS downregulated macrophage and neutrophil activation and migration and adhesion pathways by reversing 51 DEGs, which was further confirmed by RT-qPCR and Western blot analysis. In addition, FZQS modulated the dysbiosis of the gut microbiota by reversing the abundance of Corynebacterium, Facklamia, Staphylococcus, Paenalcaligenes, Lachnoclostridium, norank_f_Muribaculaceae, and unclassified_f_Lachnospiraceae. Meanwhile, metabolomics analysis revealed that FZQS significantly regulated tryptophan metabolism by reducing the levels of 3-Indoleacetonitrile and 5-Hydroxykynurenine. CONCLUSION: FZQS effectively ameliorated LPS-induced lung injury by inhibiting the activation, migration, and adhesion of macrophages and neutrophils and modulating gut microbiota and its metabolites.


Asunto(s)
Medicamentos Herbarios Chinos , Lipopolisacáridos , Lesión Pulmonar , Metabolómica , Animales , Medicamentos Herbarios Chinos/farmacología , Ratones , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/metabolismo , Lesión Pulmonar/inducido químicamente , Masculino , Microbioma Gastrointestinal/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo , Ratones Endogámicos C57BL , Citocinas/metabolismo , Modelos Animales de Enfermedad , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo
11.
Tissue Cell ; 89: 102444, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38945090

RESUMEN

Injury to internal organs caused by myocardial infarction (MI), although often neglected, is a very serious condition which damages internal organs especially the lungs. Changes in microcirculation can begin with acute lung injury and result in severe respiratory failure. The aim of this study was to create new approaches that will explain the pathophysiology and treatment of the disease by examining the therapeutic effects of vitamin D (VITD) and Nerolidol (NRD) on the injuries of the lungs caused by MI, and their relationship with asprosin / spexin proteins. METHODS: Six groups of seven experimental animals each were constituted. Control, VITD (only 50 IU/day during the experiment), NRD (only 100 mg/kg/day during the experiment), MI (200 mg/kg isoproterenol was administered to rats as a single dose subcutaneously), MI+VITD (200 mg/kg isoproterenol +50 IU/day) and MI+NRD (200 mg/kg isoproterenol +100 mg/kg/day) were the six (6) groups constituted. Tissues were analyzed using histopathological and immunohistochemical methods, whereas serum samples were analyzed using ELISA method. RESULTS: The result of the histopathological study for the MI group showed an observed increase in inflammatory cells, congestion, interalveolar septal thickening, erythrocyteloaded macrophages and fibrosis in the lung tissues. The treatment groups however recorded significant differences with regards to these parameters. In the immunohistochemical analysis, expressions of asprosin and spexin were observed in the smooth muscle structures and interalveolar areas of the vessels and bronchioles of the lung, as well as the bronchiole epithelium. There was no significant difference between the groups in terms of asprosin and spexin expression in the bronchiol epithelium. When immunohistochemical and serum ELISA results were examined, it was observed that asprosin levels increased significantly in the lung tissues of the MI group compared to the control group, decreased significantly in the treatment groups treated with Vitamin D and NRD after MI. While spexin decreased significantly in the MI group compared to the control group, it increased significantly in the MI+VITD group, but did not change in the MI+NRD group. CONCLUSION: It was observed that serious injuries occurred in the lungs due to myocardial infarction and that, VITD and NRD treatments had a curative effect on those injuries. It was also observed that Asprosin and Speksin proteins can have effect on mechanisms of both injury and therapy of the lung. Furthermore, the curative effects of VITD are dependent on the expression of asprosin and spexin; whereas the observation indicated that nerolidol could be effective through asprosin-dependent mechanisms and specisin by independent mechanisms.


Asunto(s)
Infarto del Miocardio , Sesquiterpenos , Vitamina D , Animales , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Infarto del Miocardio/metabolismo , Sesquiterpenos/farmacología , Sesquiterpenos/uso terapéutico , Ratas , Vitamina D/farmacología , Masculino , Hormonas Peptídicas/metabolismo , Hormonas Peptídicas/farmacología , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/patología , Lesión Pulmonar/etiología , Lesión Pulmonar/metabolismo , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/metabolismo , Isoproterenol/farmacología , Ratas Wistar
12.
Int Immunopharmacol ; 137: 112450, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-38906007

RESUMEN

Inflammation, apoptosis and oxidative stress play crucial roles in the deterioration of severe acute pancreatitis-associated acute respiratory distress syndrome (SAP-ARDS). Unfortunately, despite a high mortality rate of 45 %[1], there are limited treatment options available for ARDS outside of last resort options such as mechanical ventilation and extracorporeal support strategies[2]. This study investigated the potential therapeutic role and mechanisms of AQP9 inhibitor RG100204 in two animal models of severe acute pancreatitis, inducing acute respiratory distress syndrome: 1) a sodium-taurocholate induced rat model, and 2) and Cerulein and lipopolysaccharide induced mouse model. RG100204 treatment led to a profound reduction in inflammatory cytokine expression in pancreatic, and lung tissue, in both models. In addition, infiltration of CD68 + and CD11b + cells into these tissues were reduced in RG100204 treated SAP animals, and edema and SAP associated tissue damage were improved. Moreover, we demonstrate that RG100204 reduced apoptosis in the lungs of rat SAP animals, and reduces NF-κB signaling, NLRP3, expression, while profoundly increasing the Nrf2-dependent anti oxidative stress response. We conclude that AQP9 inhibition is a promising strategy for the treatment of pancreatitis and its systemic complications, such as ARDS.


Asunto(s)
Factor 2 Relacionado con NF-E2 , Proteína con Dominio Pirina 3 de la Familia NLR , Pancreatitis , Síndrome de Dificultad Respiratoria , Transducción de Señal , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Pancreatitis/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/metabolismo , Masculino , Transducción de Señal/efectos de los fármacos , Ratones , Ratas , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/metabolismo , Acuaporinas/metabolismo , Acuaporinas/antagonistas & inhibidores , Modelos Animales de Enfermedad , Ratas Sprague-Dawley , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Lipopolisacáridos , Ratones Endogámicos C57BL , Ácido Taurocólico , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Páncreas/patología , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Apoptosis/efectos de los fármacos , Ceruletida , Humanos , Hemo Oxigenasa (Desciclizante)/metabolismo
13.
J Ethnopharmacol ; 333: 118404, 2024 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-38824977

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Sepsis presents complex pathophysiological challenges. Taohe Chengqi Decoction (THCQ), a traditional Chinese medicine, offers potential in managing sepsis-related complications, though its exact mechanisms are not fully understood. AIM OF THE STUDY: This research aimed to assess the therapeutic efficacy and underlying mechanisms of THCQ on sepsis-induced lung injury. MATERIALS AND METHODS: The study began with validating THCQ's anti-inflammatory effects through in vitro and in vivo experiments. Network pharmacology was employed for mechanistic exploration, incorporating GO, KEGG, and PPI analyses of targets. Hub gene-immune cell correlations were assessed using CIBERSORT, with further scrutiny at clinical and single-cell levels. Molecular docking explored THCQ's drug-gene interactions, culminating in qPCR and WB validations of hub gene expressions in sepsis and post-THCQ treatment scenarios. RESULTS: THCQ demonstrated efficacy in modulating inflammatory responses in sepsis, identified through network pharmacology. Key genes like MAPK14, MAPK3, MMP9, STAT3, LYN, AKT1, PTPN11, and HSP90AA1 emerged as central targets. Molecular docking revealed interactions between these genes and THCQ components. qPCR results showed significant modulation of these genes, indicating THCQ's potential in reducing inflammation and regulating immune responses in sepsis. CONCLUSION: This study sheds light on THCQ's anti-inflammatory and immune regulatory mechanisms in sepsis, providing a foundation for further research and potential clinical application.


Asunto(s)
Antiinflamatorios , Medicamentos Herbarios Chinos , Simulación del Acoplamiento Molecular , Sepsis , Sepsis/tratamiento farmacológico , Sepsis/complicaciones , Sepsis/inmunología , Animales , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Medicamentos Herbarios Chinos/química , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Humanos , Lesión Pulmonar/tratamiento farmacológico , Farmacología en Red , Modelos Animales de Enfermedad
14.
Biomed Pharmacother ; 177: 117026, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38936197

RESUMEN

Cyclophosphamide is an anti-neoplastic drug that has shown competence in the management of a broad range of malignant tumors. In addition, it represents a keystone agent for management of immunological conditions. Despite these unique properties, induction of lung toxicity may limit its clinical use. Omarigliptin is one of the dipeptidyl peptidase-4 inhibitors that has proven efficacy in management of diabetes mellitus. Rosinidin is an anthocyanidin flavonoid that exhibited promising results in management of diseases characterized by oxidative stress, inflammation, and apoptosis. The present work investigated the possible effects of omarigliptin with or without rosinidin on cyclophosphamide-induced lung toxicity with an exploration of the molecular mechanisms that contribute to these effects. In a rodent model of cyclophosphamide elicited lung toxicity, the potential efficacy of omarigliptin with or without rosinidin was investigated at both the biochemical and the histopathological levels. Both omarigliptin and rosinidin exhibited a synergistic ability to augment the tissue antioxidant defenses, mitigate the inflammatory pathways, restore glucagon-like peptide-1 levels, modulate high mobility group box 1 (HMGB1)/receptors of advanced glycation end products (RAGE)/nuclear factor kappa B (NF-κB) axis, downregulate the fibrogenic mediators, and create a balance between the pathways involved in apoptosis and the autophagy signals in the pulmonary tissues. In conclusion, omarigliptin/rosinidin combination may be introduced as a novel therapeutic modality that attenuates the different forms of lung toxicities induced by cyclophosphamide.


Asunto(s)
Ciclofosfamida , Péptido 1 Similar al Glucagón , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Piranos , Transducción de Señal , Animales , Ciclofosfamida/toxicidad , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Masculino , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Ratas , Fosfatidilinositol 3-Quinasas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Piranos/farmacología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Antocianinas/farmacología , Estrés Oxidativo/efectos de los fármacos , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Ratas Wistar , Pirimidinas/farmacología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Proteína Forkhead Box O1 , Compuestos Heterocíclicos con 2 Anillos
15.
Vet Res Commun ; 48(4): 2499-2511, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38865040

RESUMEN

Varicellovirus bovinealpha 1 (BoAHV-1) is a significant pathogen responsible for respiratory disease in cattle, capable of inducing lung damage independently or co-infection with bacteria. The widespread spread of BoAHV-1 in cattle herds has caused substantial economic losses to the cattle industry. The pathogenic mechanisms of BoAHV-1 are often relevant to robust inflammatory responses, increased oxidative burden, and the initiation of apoptosis. Glycyrrhizin (GLY) is a small-molecule triterpenoid saponin compound obtained from the herb liquorice, which has a broad spectrum of pharmacological properties such as antiviral, anti-inflammatory, and antioxidant effects. Furthermore, GLY regulates lung physiology by modulating oxidative stress, inflammatory response, and cell apoptosis through interference with the NF-κB/NLRP3 and Nrf2/HO-1 Signaling pathways. However, the potential of GLY to mitigate lung injury induced by BoAHV-1 and its underlying mechanism remains unclear. Therefore, in this study, we investigated the protective effect of GLY against pulmonary injury induced by BoAHV-1 in a guinea pig model by reducing viral load and suppressing the inflammatory response, oxidative stress, and apoptosis. The results of this study demonstrated that GLY exerted a protective effect against BoAHV-1-induced lung injury in guinea pigs. Specifically, GLY reduced the levels of pro-inflammatory cytokines interleukin (IL)-1ß, tumor necrosis factor (TNF)-α, and interleukin (IL)-8 in guinea pig tissues while suppressing the expression of Caspase-1. Additionally, GLY reduced BoAHV-1 load and the number of TUNEL-positive lung cells in guinea pig lungs while inhibiting Caspase 3 protein expression. Furthermore, GLY significantly enhanced lung antioxidant capacity by increasing superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px) activity while simultaneously reducing malondialdehyde (MDA) levels. Lung histological observation and score further validated the protective effect of GLY on BoAHV-1-induced lung injury. Furthermore, we observed that the expression of phosphorylated NF-κB p65 (p-NF-κB p65) and NLRP3 proteins in the lung tissue of BoAHV-1-infected guinea pigs decreased after GLY treatment while the expression of Nrf2 and HO-1 proteins increased. These results indicated that GLY inhibited the NF-κB/NLRP3 Signaling pathway and activated the Nrf2/HO-1 Signaling pathway during BoAHV-1 infection. Ultimately, our findings demonstrated that GLY alleviates BoAHV-1-induced inflammation response, oxidative stress, and cell apoptosis by inhibiting the NF-κB/NLRP3 Signaling pathway and activating the Nrf2/HO-1 Signaling pathway to protect guinea pigs from lung injury caused by BoAHV-1. Ultimately, our findings demonstrated that GLY alleviates BoAHV-1-induced inflammation response, oxidative stress, and cell apoptosis by inhibiting the NF-κB/NLRP3 Signaling pathway and activating the Nrf2/HO-1 Signaling pathway to protect guinea pigs from lung injury caused by BoAHV-1. Importantly, this study provides a compelling argument for the GLY in combating respiratory disease in cattle caused by BoAHV-1.


Asunto(s)
Ácido Glicirrínico , Factor 2 Relacionado con NF-E2 , FN-kappa B , Proteína con Dominio Pirina 3 de la Familia NLR , Transducción de Señal , Animales , Transducción de Señal/efectos de los fármacos , Cobayas , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ácido Glicirrínico/farmacología , Ácido Glicirrínico/uso terapéutico , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/veterinaria , Lesión Pulmonar/virología , Hemo-Oxigenasa 1/metabolismo , Hemo-Oxigenasa 1/genética , Masculino
16.
Drug Res (Stuttg) ; 74(5): 241-249, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38830372

RESUMEN

Pentoxifylline (PTX), a non-selective phosphodiesterase inhibitor, has demonstrated protective effects against lung injury in animal models. Given the significance of pulmonary toxicity resulting from paraquat (PQ) exposure, the present investigation was designed to explore the impact of PTX on PQ-induced pulmonary oxidative impairment in male mice.Following preliminary studies, thirty-six mice were divided into six groups. Group 1 received normal saline, group 2 received a single dose of PQ (20 mg/kg; i.p.), and group 3 received PTX (100 mg/kg/day; i.p.). Additionally, treatment groups 4-6 were received various doses of PTX (25, 50, and 100 mg/kg/day; respectively) one hour after a single dose of PQ. After 72 hours, the animals were sacrificed, and lung tissue was collected.PQ administration caused a significant decrease in hematocrit and an increase in blood potassium levels. Moreover, a notable increase was found in the lipid peroxidation (LPO), nitric oxide (NO), and myeloperoxidase (MPO) levels, along with a notable decrease in total thiol (TTM) and total antioxidant capacity (TAC) contents, catalase (CAT) and superoxide dismutase (SOD) enzymes activity in lung tissue. PTX demonstrated the ability to improve hematocrit levels; enhance SOD activity and TTM content; and decrease MPO activity, LPO and NO levels in PQ-induced pulmonary toxicity. Furthermore, these findings were well-correlated with the observed lung histopathological changes.In conclusion, our results suggest that the high dose of PTX may ameliorate lung injury by improving the oxidant/antioxidant balance in animals exposed to PQ.


Asunto(s)
Antioxidantes , Peroxidación de Lípido , Pulmón , Paraquat , Pentoxifilina , Superóxido Dismutasa , Animales , Pentoxifilina/farmacología , Pentoxifilina/uso terapéutico , Paraquat/toxicidad , Ratones , Masculino , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Antioxidantes/farmacología , Superóxido Dismutasa/metabolismo , Estrés Oxidativo/efectos de los fármacos , Catalasa/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Inhibidores de Fosfodiesterasa/uso terapéutico , Óxido Nítrico/metabolismo , Peroxidasa/metabolismo , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/tratamiento farmacológico , Hidrolasas Diéster Fosfóricas/metabolismo
19.
Redox Biol ; 75: 103252, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-38925040

RESUMEN

Pulmonary vascular remodeling and inflammation play an important role in the hypoxic-induced lung diseases. Our previous investigations showed that peptide from yak milk residues could alleviate inflammation. In this study, our results suggest that peptide (LV) from yak milk residues peptide had protective effect of lung in the animal models of hypoxic-induced lung injury. LV Gavage could improve pulmonary vascular remodeling in the lung tissues of hypoxic mice. A comprehensive analysis of metabolomics and transcriptomics revealed that 5-KETE, 8,9-EET, and 6-keto-prostaglandin F1a might be potential targets to prevent lung injury in the hypoxic mice. These metabolites can be regulated by MAPK/VEGF and inflammatory pathways. Our data indicated that LV treatment could inhibit apoptosis and inflammation via Nrf2/NF-κB/MAPK/PHD-2 pathway and protected hypoxic-induced lung epithelial cells injury. Taken together, our results suggest that LV provides a novel therapeutic clue for the prevention of hypoxia-induced lung injury and inflammation-related lung diseases.


Asunto(s)
Hipoxia , Lesión Pulmonar , Péptidos , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular , Animales , Ratones , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/metabolismo , Lesión Pulmonar/etiología , Lesión Pulmonar/patología , Bovinos , Hipoxia/metabolismo , Hipoxia/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Péptidos/farmacología , Péptidos/química , Péptidos/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Inflamación/metabolismo , Inflamación/tratamiento farmacológico , Modelos Animales de Enfermedad , Apoptosis/efectos de los fármacos , Humanos , Masculino
20.
Sci Rep ; 14(1): 14231, 2024 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-38902260

RESUMEN

Butorphanol is widely used as an anesthetic drug, whether butorphanol could reduce organ injury and protecting lung tissue is unknown. This study explored the effects of butorphanol on ALI and investigated its underlying mechanisms. We established a "two-hit" rat model and "two-hit" cell model to prove our hypothesis. Rats were divided into four groups [control, "two-hit" (OA + LPS), "two-hit" + butorphanol (4 mg/kg and 8 mg/kg) (OA + LPS + B1 and OA + LPS + B2)]. RPMVE cells were divided into four groups [control, "two-hit" (OA + LPS), "two-hit" + butorphanol (4 µM and 8 µM) (OA + LPS + 4 µM and OA + LPS + 8 µM)]. Inflammatory injury was assessed by the histopathology and W/D ratio, inflammatory cytokines, and arterial blood gas analysis. Apoptosis was assessed by Western blotting and flow cytometry. The effect of NF-κB p65 was detected by ELISA. Butorphanol could relieve the "two-hit" induced lung injury, the expression of TNF, IL-1ß, IL-6, and improve lung ventilation. In addition, butorphanol decreased Bax and cleaved caspase-3, increased an antiapoptotic protein (Bcl-2), and inhibited the "two-hit" cell apoptosis ratio. Moreover, butorphanol suppressed NF-κB p65 activity in rat lung injury. Our research showed that butorphanol may attenuate "two-hit"-induced lung injury by regulating the activity of NF-κB p65, which may supply more evidence for ALI treatment.


Asunto(s)
Lesión Pulmonar Aguda , Apoptosis , Butorfanol , Inflamación , Animales , Butorfanol/farmacología , Apoptosis/efectos de los fármacos , Ratas , Masculino , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/prevención & control , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Factor de Transcripción ReIA/metabolismo , Lipopolisacáridos , Ratas Sprague-Dawley , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Lesión Pulmonar/prevención & control , Modelos Animales de Enfermedad , Citocinas/metabolismo , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA