Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.082
Filtrar
1.
Syst Rev ; 13(1): 243, 2024 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-39342304

RESUMEN

BACKGROUND: Postoperative pneumonia is a common but serious complication in patients with lung cancer. This meta-analysis aims to evaluate the effect of respiratory exercise on reducing postoperative pneumonia in patients with lung cancer and to provide a reliable basis for clinical treatment and nursing of patients with lung cancer. METHODS: Two reviewers searched PubMed, Embase, Web of Science, Cochrane Library, China Knowledge Network, Wanfang, and Weipu databases. We searched for the randomized controlled trials (RCTs) published in Chinese or English on the breathing exercises in patients with lung cancer up to January 30, 2024. The quality of the literature was evaluated with the Cochrane Risk of Bias Tool 2 (ROB 2). RevMan 5.3 software was used for meta-analysis. RESULTS: Eleven RCTs with 1429 patients with lung cancer were included, and 710 patients received breathing exercises. The meta-analysis results showed that breathing exercises could significantly reduce the incidence of postoperative pneumonia [RR = 0.35, 95%CI (0.25, 0.51)], improve the FEV1 [MD = - 0.49, 95%CI (- 0.73, - 0.24)], FVC [MD = - 0.59, 95%CI (- 0.83, - 0.35)] in patients with lung cancer (all P < 0.05). There were significant differences in the incidence of pneumonia for patients undergoing breathing exercises with single exercise time ≥ 15 min (RR = 0.37, 95%CI 0.24 ~ 0.62), breathing exercises for 1 week (RR = 0.29, 95%CI 0.16 ~ 0.55) or for 2 weeks (RR = 0.48, 95%CI 0.28 ~ 0.85) and breathing exercises > 4 times (RR = 0.36, 95%CI 0.23 ~ 0.57) per day (all P < 0.05). CONCLUSION: Breathing exercises have shown the capacity to augment pulmonary function in patients with lung cancer, concurrently mitigating the risk of postoperative pneumonia.


Asunto(s)
Ejercicios Respiratorios , Neoplasias Pulmonares , Neumonía , Complicaciones Posoperatorias , Humanos , Ejercicios Respiratorios/métodos , Neoplasias Pulmonares/cirugía , Complicaciones Posoperatorias/prevención & control , Neumonía/prevención & control , Ensayos Clínicos Controlados Aleatorios como Asunto
2.
Antiviral Res ; 229: 105968, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-39004311

RESUMEN

Since human angiotensin-converting enzyme 2 (ACE2) serves as a primary receptor for SARS-CoV-2, characterizing ACE2 regions that allow SARS-CoV-2 to enter human cells is essential for designing peptide-based antiviral blockers and elucidating the pathogenesis of the virus. We identified and synthesized a 25-mer mimetic peptide (encompassing positions 22-46 of the ACE2 alpha-helix α1) implicated in the S1 receptor-binding domain (RBD)-ACE2 interface. The mimetic (wild-type, WT) ACE2 peptide significantly inhibited SARS-CoV-2 infection of human pulmonary Calu-3 cells in vitro. In silico protein modeling predicted that residues F28, K31, F32, F40, and Y41 of the ACE2 alpha-helix α1 are critical for the original, Delta, and Omicron strains of SARS-CoV-2 to establish the Spike RBD-ACE2 interface. Substituting these residues with alanine (A) or aspartic acid (D) abrogated the antiviral protective effect of the peptides, indicating that these positions are critical for viral entry into pulmonary cells. WT ACE2 peptide, but not the A or D mutated peptides, exhibited significant interaction with the SARS-CoV-2 S1 RBD, as shown through molecular dynamics simulations. Through identifying the critical amino acid residues of the ACE2 alpha-helix α1, which is necessary for the Spike RBD-ACE2 interface and mobilized during the in vitro viral infection of cells, we demonstrated that the WT ACE2 peptide protects susceptible K18-hACE2 mice against in vivo SARS-CoV-2 infection and is effective for the treatment of COVID-19.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Péptidos , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/química , Humanos , Animales , SARS-CoV-2/efectos de los fármacos , COVID-19/virología , Ratones , Glicoproteína de la Espiga del Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Péptidos/farmacología , Péptidos/química , Péptidos/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Antivirales/farmacología , Antivirales/química , Línea Celular , Neumonía/tratamiento farmacológico , Neumonía/virología , Neumonía/prevención & control , Pulmón/virología , Pulmón/patología , Femenino
3.
In Vivo ; 38(4): 1790-1798, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38936906

RESUMEN

BACKGROUND/AIM: We evaluated the usefulness of prophylactic mini-tracheostomy (PMT) and perioperative administration of tazobactam/piperacillin (TAZ/PIPC) in high-risk patients after esophagectomy. PATIENTS AND METHODS: We retrospectively studied 89 consecutive high-risk patients who underwent esophagectomy for esophageal cancer between January 2013 and December 2021. We defined patients with two or more of the following factors as high risk: age ≥70 years, performance status ≥1, respiratory dysfunction, liver dysfunction, cardiac dysfunction, renal dysfunction, diabetes mellitus, albumin <3.5 g/dl, and Brinkman index >600. Standard management was administered to the first 50 patients (standard group). PMT and TAZ/PIPC were administered to the next 39 patients (combination group). Patient characteristics and short-term outcomes were compared before and after propensity-score matching. RESULTS: Before propensity-score matching, 24-hour urine creatinine clearance, retrosternal route, 3-field lymph node dissection, and open abdominal approach were more common, postoperative pneumonia (13% vs. 36%, p=0.045) and complications of grade ≥3b (2.6% vs. 22%, p=0.01) were less frequent, and the postoperative hospital stay was shorter (median: 23 vs. 28 days, p=0.022) in the combination group than in the standard group. In propensity-score matching, patient characteristics, except for 24-h creatinine clearance and reconstructive route, were matched for 23 paired patients. Postoperative pneumonia (8.7% vs. 39%, p=0.035) and complications of grade ≥3b (0% vs. 26%, p=0.022) were less frequent and postoperative hospital stay was shorter (median: 22 vs. 25 days, p=0.021) in the combination group than in the standard group. CONCLUSION: PMT with TAZ/PIPC can potentially prevent postoperative pneumonia in high-risk patients after esophagectomy.


Asunto(s)
Neoplasias Esofágicas , Esofagectomía , Combinación Piperacilina y Tazobactam , Neumonía , Complicaciones Posoperatorias , Humanos , Masculino , Femenino , Anciano , Esofagectomía/efectos adversos , Esofagectomía/métodos , Neoplasias Esofágicas/cirugía , Neumonía/prevención & control , Neumonía/etiología , Neumonía/epidemiología , Combinación Piperacilina y Tazobactam/uso terapéutico , Combinación Piperacilina y Tazobactam/administración & dosificación , Persona de Mediana Edad , Complicaciones Posoperatorias/prevención & control , Complicaciones Posoperatorias/etiología , Antibacterianos/uso terapéutico , Antibacterianos/administración & dosificación , Estudios Retrospectivos , Factores de Riesgo
4.
Br J Anaesth ; 133(3): 647-657, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38937217

RESUMEN

BACKGROUND: Prior studies have reported inconsistent results regarding the association between driving pressure-guided ventilation and postoperative pulmonary complications (PPCs). We aimed to investigate whether driving pressure-guided ventilation is associated with a lower risk of PPCs. METHODS: We systematically searched electronic databases for RCTs comparing driving pressure-guided ventilation with conventional protective ventilation in adult surgical patients. The primary outcome was a composite of PPCs. Secondary outcomes were pneumonia, atelectasis, and acute respiratory distress syndrome (ARDS). Meta-analysis and subgroup analysis were conducted to calculate risk ratios (RRs) with 95% confidence intervals (CI). Trial sequential analysis (TSA) was used to assess the conclusiveness of evidence. RESULTS: Thirteen RCTs with 3401 subjects were included. Driving pressure-guided ventilation was associated with a lower risk of PPCs (RR 0.70, 95% CI 0.56-0.87, P=0.001), as indicated by TSA. Subgroup analysis (P for interaction=0.04) found that the association was observed in non-cardiothoracic surgery (nine RCTs, 1038 subjects, RR 0.61, 95% CI 0.48-0.77, P< 0.0001), with TSA suggesting sufficient evidence and conclusive result; however, it did not reach significance in cardiothoracic surgery (four RCTs, 2363 subjects, RR 0.86, 95% CI 0.67-1.10, P=0.23), with TSA indicating insufficient evidence and inconclusive result. Similarly, a lower risk of pneumonia was found in non-cardiothoracic surgery but not in cardiothoracic surgery (P for interaction=0.046). No significant differences were found in atelectasis and ARDS between the two ventilation strategies. CONCLUSIONS: Driving pressure-guided ventilation was associated with a lower risk of postoperative pulmonary complications in non-cardiothoracic surgery but not in cardiothoracic surgery. SYSTEMATIC REVIEW PROTOCOL: INPLASY 202410068.


Asunto(s)
Complicaciones Posoperatorias , Humanos , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/etiología , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/epidemiología , Respiración con Presión Positiva/métodos , Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/epidemiología , Enfermedades Pulmonares/prevención & control , Respiración Artificial/métodos , Atelectasia Pulmonar/prevención & control , Atelectasia Pulmonar/etiología , Atelectasia Pulmonar/epidemiología , Ensayos Clínicos Controlados Aleatorios como Asunto , Neumonía/epidemiología , Neumonía/etiología , Neumonía/prevención & control
6.
Sci Total Environ ; 933: 173222, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38750750

RESUMEN

Ozone (O3) is a major air pollutant that directly threatens the respiratory system, lung fatty acid metabolism disorder is an important molecular event in pulmonary inflammatory diseases. Liver kinase B1 (LKB1) and nucleotide-binding domain leucine-rich repeat-containing protein 3 (NLRP3) inflammasome not only regulate inflammation, but also have close relationship with fatty acid metabolism. However, the role and mechanism of LKB1 and NLRP3 inflammasome in lung fatty acid metabolism, which may contribute to ozone-induced lung inflammation, remain unclear, and effective strategy for preventing O3-induced pulmonary inflammatory injury is lacking. To explore these, mice were exposed to 1.00 ppm O3 (3 h/d, 5 days), and pulmonary inflammation was determined by airway hyperresponsiveness, histopathological examination, total cells and cytokines in bronchoalveolar lavage fluid (BALF). Targeted fatty acids metabolomics was used to detect medium and long fatty acid in lung tissue. Then, using LKB1-overexpressing adenovirus and NLRP3 knockout (NLRP3-/-) mice to explore the mechanism of O3-induced lung fatty acid metabolism disorder. Results demonstrated that O3 exposure caused pulmonary inflammatory injury and lung medium and long chain fatty acids metabolism disorder, especially decreased dihomo-γ-linolenic acid (DGLA). Meanwhile, LKB1 expression was decreased, and NLRP3 inflammasome was activated in lung of mice after O3 exposure. Additionally, LKB1 overexpression alleviated O3-induced lung inflammation and inhibited the activation of NLRP3 inflammasome. And we found that pulmonary fatty acid metabolism disorder was ameliorated of NLRP3 -/- mice compared with those in wide type mice after O3 exposure. Furthermore, administrating DGLA intratracheally prior to O3 exposure significantly attenuated O3-induced pulmonary inflammatory injury. Taken together, these findings suggest that fatty acids metabolism disorder is involved in O3-induced pulmonary inflammation, which is regulated by LKB1-mediated NLRP3 pathway, DGLA supplement could be a useful preventive strategy to ameliorate ozone-associated lung inflammatory injury.


Asunto(s)
Ácidos Grasos , Proteína con Dominio Pirina 3 de la Familia NLR , Ozono , Animales , Ratones , Ácidos Grasos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neumonía/metabolismo , Neumonía/prevención & control , Contaminantes Atmosféricos/toxicidad , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Inflamasomas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo
7.
BMC Pulm Med ; 24(1): 224, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38720270

RESUMEN

BACKGROUND: Simvastatin (Sim), a hydroxy-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor, has been widely used in prevention and treatment of cardiovascular diseases. Studies have suggested that Sim exerts anti-fibrotic effects by interfering fibroblast proliferation and collagen synthesis. This study was to determine whether Sim could alleviate silica-induced pulmonary fibrosis and explore the underlying mechanisms. METHODS: The rat model of silicosis was established by the tracheal perfusion method and treated with Sim (5 or 10 mg/kg), AICAR (an AMPK agonist), and apocynin (a NOX inhibitor) for 28 days. Lung tissues were collected for further analyses including pathological histology, inflammatory response, oxidative stress, epithelial mesenchymal transformation (EMT), and the AMPK-NOX pathway. RESULTS: Sim significantly reduced silica-induced pulmonary inflammation and fibrosis at 28 days after administration. Sim could reduce the levels of interleukin (IL)-1ß, IL-6, tumor necrosis factor-α and transforming growth factor-ß1 in lung tissues. The expressions of hydroxyproline, α-SMA and vimentin were down-regulated, while E-cad was increased in Sim-treated rats. In addition, NOX4, p22pox, p40phox, p-p47phox/p47phox expressions and ROS levels were all increased, whereas p-AMPK/AMPK was decreased in silica-induced rats. Sim or AICAR treatment could notably reverse the decrease of AMPK activity and increase of NOX activity induced by silica. Apocynin treatment exhibited similar protective effects to Sim, including down-regulating of oxidative stress and inhibition of the EMT process and inflammatory reactions. CONCLUSIONS: Sim attenuates silica-induced pulmonary inflammation and fibrosis by downregulating EMT and oxidative stress through the AMPK-NOX pathway.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Fibrosis Pulmonar , Dióxido de Silicio , Simvastatina , Animales , Masculino , Ratas , Acetofenonas/farmacología , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , NADPH Oxidasa 4/metabolismo , NADPH Oxidasas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Neumonía/inducido químicamente , Neumonía/prevención & control , Neumonía/tratamiento farmacológico , Neumonía/metabolismo , Neumonía/patología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Ribonucleótidos/farmacología , Transducción de Señal/efectos de los fármacos , Silicosis/tratamiento farmacológico , Silicosis/patología , Silicosis/metabolismo , Simvastatina/farmacología , Factor de Crecimiento Transformador beta1/metabolismo
8.
Physiol Res ; 73(2): 239-251, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38710061

RESUMEN

Oxygen therapy provides an important treatment for preterm and low-birth-weight neonates, however, it has been shown that prolonged exposure to high levels of oxygen (hyperoxia) is one of the factors contributing to the development of bronchopulmonary dysplasia (BPD) by inducing lung injury and airway hyperreactivity. There is no effective therapy against the adverse effects of hyperoxia. Therefore, this study was undertaken to test the hypothesis that natural phytoalexin resveratrol will overcome hyperoxia-induced airway hyperreactivity, oxidative stress, and lung inflammation. Newborn rats were exposed to hyperoxia (fraction of inspired oxygen - FiO2>95 % O2) or ambient air (AA) for seven days. Resveratrol was supplemented either in vivo (30 mg·kg-1·day-1) by intraperitoneal administration or in vitro to the tracheal preparations in an organ bath (100 mikroM). Contractile and relaxant responses were studied in tracheal smooth muscle (TSM) using the in vitro organ bath system. To explain the involvement of nitric oxide in the mechanisms of the protective effect of resveratrol against hyperoxia, a nitric oxide synthase inhibitor - Nomega-nitro-L-arginine methyl ester (L-NAME), was administered in some sets of experiments. The superoxide dismutase (SOD) and glutathione peroxidase (GPx) activities and the tumor necrosis factor-alpha (TNF-alpha) and interleukin-1beta (IL-1beta) levels in the lungs were determined. Resveratrol significantly reduced contraction and restored the impaired relaxation of hyperoxia-exposed TSM (p<0.001). L-NAME reduced the inhibitory effect of resveratrol on TSM contractility, as well as its promotion relaxant effect (p<0.01). Resveratrol preserved the SOD and GPx activities and decreased the expression of TNF-alpha and IL-1beta in hyperoxic animals. The findings of this study demonstrate the protective effect of resveratrol against hyperoxia-induced airway hyperreactivity and lung damage and suggest that resveratrol might serve as a therapy to prevent the adverse effects of neonatal hyperoxia. Keywords: Bronchopulmonary dysplasia, Hyperoxia, Airway hyperreactivity, Resveratrol, Pro-inflammatory cytokines.


Asunto(s)
Animales Recién Nacidos , Displasia Broncopulmonar , Modelos Animales de Enfermedad , Estrés Oxidativo , Neumonía , Resveratrol , Animales , Resveratrol/farmacología , Estrés Oxidativo/efectos de los fármacos , Displasia Broncopulmonar/prevención & control , Displasia Broncopulmonar/metabolismo , Neumonía/prevención & control , Neumonía/metabolismo , Neumonía/inducido químicamente , Ratas , Hiperoxia/complicaciones , Hiperoxia/metabolismo , Estilbenos/farmacología , Estilbenos/uso terapéutico , Antioxidantes/farmacología , Hiperreactividad Bronquial/prevención & control , Hiperreactividad Bronquial/metabolismo , Hiperreactividad Bronquial/fisiopatología , Hiperreactividad Bronquial/inducido químicamente , Ratas Sprague-Dawley , Masculino
9.
Cells ; 13(10)2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38786103

RESUMEN

Cigarette smoke is one of the main factors in Chronic Obstructive Pulmonary Disease (COPD), a respiratory syndrome marked by persistent respiratory symptoms and increasing airway obstruction. Perturbed NAD+/NADH levels may play a role in various diseases, including lung disorders like COPD. In our study, we investigated the preventive effect of NADH supplementation in an experimental model of COPD induced by cigarette smoke extract (CSE). N = 64 mice randomly distributed in eight groups were injected with NADH (two doses of 100 mg/kg or 200 mg/kg) or dexamethasone (2 mg/kg) before being exposed to CSE for up to 9 weeks. Additionally, NADH supplementation preserved lung antioxidant defenses by preventing the functional loss of key enzymes such as superoxide dismutase (SOD), glutathione peroxidase (GPX), catalase, and the expression levels of glutathione (GSH) (n = 4, p < 0.001). It also reduced oxidative damage markers, such as malondialdehyde (MDA) and nitrites (n = 4, p < 0.001). A marked increase in tissue myeloperoxidase activity was assessed (MPO), confirming neutrophils implication in the inflammatory process. The latter was significantly ameliorated in the NADH-treated groups (p < 0.001). Finally, NADH prevented the CSE-induced secretion of cytokines such as Tumor Necrosis Factor alpha (TNF-α), IL-17, and IFN-y (n = 4, p < 0.001). Our study shows, for the first time, the clinical potential of NADH supplementation in preventing key features of COPD via its unique anti-inflammatory and antioxidant properties.


Asunto(s)
Modelos Animales de Enfermedad , Ratones Endogámicos BALB C , NAD , Neumonía , Enfermedad Pulmonar Obstructiva Crónica , Animales , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfermedad Pulmonar Obstructiva Crónica/prevención & control , Enfermedad Pulmonar Obstructiva Crónica/etiología , NAD/metabolismo , Ratones , Neumonía/prevención & control , Neumonía/metabolismo , Neumonía/patología , Inyecciones Intraperitoneales , Humo/efectos adversos , Estrés Oxidativo/efectos de los fármacos , Masculino , Antioxidantes/metabolismo , Antioxidantes/farmacología , Citocinas/metabolismo , Pulmón/patología , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Peroxidasa/metabolismo
10.
J Agric Food Chem ; 72(17): 9782-9794, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38597360

RESUMEN

Uncontrolled inflammation contributes significantly to the mortality in acute respiratory infections. Our previous research has demonstrated that maize bran feruloylated oligosaccharides (FOs) possess notable anti-inflammatory properties linked to the NF-kB pathway regulation. In this study, we clarified that the oral administration of FOs moderately inhibited H1N1 virus infection and reduced lung inflammation in influenza-infected mice by decreasing a wide spectrum of cytokines (IFN-α, IFN-ß, IL-6, IL-10, and IL-23) in the lungs. The mechanism involves FOs suppressing the transduction of the RIG-I/MAVS/TRAF3 signaling pathway, subsequently lowering the expression of NF-κB. In silico analysis suggests that FOs have a greater binding affinity for the RIG-I/MAVS signaling complex. This indicates that FOs have potential as promising targets for immune modulation. Moreover, in MAVS knockout mice, we confirmed that the anti-inflammatory function of FOs against influenza depends on MAVS. Comprehensive analysis using 16S rRNA gene sequencing and metabolite profiling techniques showed that FOs have the potential to restore immunity by modulating the gut microbiota. In conclusion, our study demonstrates that FOs are effective anti-inflammatory phytochemicals in inhibiting lung inflammation caused by influenza. This suggests that FOs could serve as a potential nutritional strategy for preventing the H1N1 virus infection and associated lung inflammation.


Asunto(s)
Proteína 58 DEAD Box , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana , Ratones Noqueados , Oligosacáridos , Infecciones por Orthomyxoviridae , Transducción de Señal , Factor 3 Asociado a Receptor de TNF , Animales , Ratones , Oligosacáridos/administración & dosificación , Oligosacáridos/química , Oligosacáridos/farmacología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/metabolismo , Subtipo H1N1 del Virus de la Influenza A/inmunología , Humanos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Gripe Humana/inmunología , Gripe Humana/prevención & control , Gripe Humana/metabolismo , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/metabolismo , Factor 3 Asociado a Receptor de TNF/inmunología , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/metabolismo , Proteína 58 DEAD Box/inmunología , Neumonía/inmunología , Neumonía/prevención & control , Neumonía/metabolismo , Neumonía/virología , Ratones Endogámicos C57BL , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Pulmón/virología , Citocinas/metabolismo , Citocinas/inmunología , Citocinas/genética , Femenino , FN-kappa B/inmunología , FN-kappa B/genética , FN-kappa B/metabolismo , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología
11.
Cell Biochem Biophys ; 82(2): 1145-1158, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38609738

RESUMEN

Recently, we have shown that guggulsterone is the principal constituent responsible for protective effects of Commiphora wightii against elastase-induced chronic obstructive pulmonary disease (COPD)-linked inflammation/emphysema. Given that cigarette smoke (CS) exposure is a primary risk factor for COPD and beneficial effects of guggulsterone have not been investigated in CS-induced COPD-linked lung inflammation. The present work was designed to validate the potential of guggulsterone in amelioration of COPD-linked lung inflammation by using a CS-based mouse model of the condition. Male BALB/c mice were exposed to 9 cigarettes/day with 1 h interval for 4 days daily. Guggulsterone was administered daily at a dose of 10 mg/kg orally for 4 consecutive days, 1 h before initiation of CS exposure. Mice were subjected to measurement of lung function followed by procurement of bronchoalveolar lavage fluid (BALF)/lung tissue. BALF was analyzed for inflammatory cells and pro-inflammatory cytokines. Lung tissue was subjected to RT-PCR for gene expression analysis. Data showed that CS exposure resulted in a significant increase in total BALF cells, predominantly neutrophils, and macrophages. Interestingly, guggulsterone administration significantly blunted CS-induced inflammation as reflected by reduced neutrophil and macrophage count. Further, the compound inhibited CS-induced gene expression of pro-inflammatory mediators TNF-α/ IL-1ß/ G-CSF/and KC in lungs along with the production of pro-inflammatory mediators TNF-α/ IL-1ß/ IL-6/ G-CSF/ KC/and MCP-1 in BALF. Further, guggulsterone improved the lung function parameters upon CS exposure. Analysis of mRNA expression of matrix metalloproteinase (MMP)-9 and tissue inhibitor of matrix metalloproteinase (TIMP)-1 suggests that guggulsterone may restore the fine balance between matrix-degrading proteases and its inhibitor in lung tissue upon CS exposure, which may contribute in the development of emphysema at later stages. Overall, our data show that guggulsterone protects against CS-induced COPD-linked lung inflammation by modulating relevant molecular players. Based on the potential effects of guggulsterone in the amelioration of CS-induced lung inflammation, we speculate that guggulsterone might alter chronic CS-induced emphysema.


Asunto(s)
Líquido del Lavado Bronquioalveolar , Ratones Endogámicos BALB C , Neumonía , Pregnenodionas , Enfermedad Pulmonar Obstructiva Crónica , Animales , Pregnenodionas/farmacología , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Enfermedad Pulmonar Obstructiva Crónica/prevención & control , Enfermedad Pulmonar Obstructiva Crónica/etiología , Masculino , Ratones , Neumonía/prevención & control , Neumonía/tratamiento farmacológico , Neumonía/metabolismo , Líquido del Lavado Bronquioalveolar/citología , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo , Humo/efectos adversos , Modelos Animales de Enfermedad , Citocinas/metabolismo , Commiphora/química , Nicotiana/efectos adversos
12.
Sci Rep ; 14(1): 9442, 2024 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658777

RESUMEN

Lung isolation usually refers to the isolation of the operative from the non-operative lung without isolating the non-operative lobe(s) of the operative lung. We aimed to evaluate whether protecting the non-operative lobe of the operative lung using a double-bronchial blocker (DBB) with continuous positive airway pressure (CPAP) could reduce the incidence of postoperative pneumonia. Eighty patients were randomly divided into two groups (n = 40 each): the DBB with CPAP (Group DBB) and routine bronchial blocker (Group BB) groups. In Group DBB, a 7-Fr BB was placed in the middle bronchus of the right lung for right lung surgery and in the inferior lobar bronchus of the left lung for left lung surgery. Further, a 9-Fr BB was placed in the main bronchus of the operative lung. In Group BB, routine BB placement was performed on the main bronchus on the surgical side. The primary endpoint was the postoperative pneumonia incidence. Compared with Group BB, Group DBB had a significantly lower postoperative pneumonia incidence in the operative (27.5% vs 5%, P = 0.013) and non-operative lung (40% vs 15%) on postoperative day 1. Compared with routine BB use for thoracoscopic lobectomy, using the DBB technique to isolate the operative lobe from the non-operative lobe(s) of the operative lung and providing CPAP to the non-operative lobe(s) through a BB can reduce the incidence of postoperative pneumonia in the operative and non-operative lungs.


Asunto(s)
Neumonectomía , Neumonía , Complicaciones Posoperatorias , Humanos , Masculino , Femenino , Persona de Mediana Edad , Neumonía/prevención & control , Neumonía/epidemiología , Neumonía/etiología , Incidencia , Neumonectomía/efectos adversos , Neumonectomía/métodos , Complicaciones Posoperatorias/prevención & control , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/etiología , Anciano , Pulmón/cirugía , Presión de las Vías Aéreas Positiva Contínua/métodos , Toracoscopía/métodos , Toracoscopía/efectos adversos , Bronquios/cirugía
13.
Med Sci Monit ; 30: e942585, 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38384124

RESUMEN

BACKGROUND Hospital-acquired infections negatively impact the health of inpatients and are highly costly to treat. Oral care reduces the microorganism number in the mouth and lungs and is essential in preventing postoperative oral inflammation, lung infection, and other complications. This study was designed to determine the effects of oral care with glutamine on oral health, oral flora, and incidence of pneumonia in patients after neurosurgery. MATERIAL AND METHODS This was a parallel, double-blind, randomized trial. Patients admitted to the Neurosurgery Department of the hospital from July to October 2021 were selected. Three hundred patients who met the inclusion criteria were randomized into 3 groups. The control group (n=100) received oral care with routine oral nursing methods with saline, whereas the experimental group (n=100) received oral care with 5% glutamine. A compound chlorhexidine group (n=100) was set as a positive control. All patients, care providers, and investigators were blinded to the group assignment. The incidence of local debris, oral mucositis, halitosis, dryness, oral mucositis disorders, and oral flora types were collected and analyzed in all groups. RESULTS The incidence of local debris, oral mucositis, halitosis, dryness, and other oral mucositis disorders in the glutamine oral care group was significantly decreased, compared with that of the control group. Oral flora types in the glutamine and chlorhexidine groups were significantly reduced. CONCLUSIONS Oral care with 5% glutamine after neurosurgery is associated with a lower incidence of oral disorders and pneumonia, and a significant reduction in oral flora.


Asunto(s)
Halitosis , Mucositis , Neurocirugia , Neumonía , Estomatitis , Humanos , Clorhexidina/farmacología , Salud Bucal , Glutamina/farmacología , Glutamina/uso terapéutico , Mucosa Bucal , Halitosis/complicaciones , Halitosis/tratamiento farmacológico , Estomatitis/tratamiento farmacológico , Mucositis/tratamiento farmacológico , Neumonía/prevención & control , Neumonía/complicaciones
14.
Respir Physiol Neurobiol ; 323: 104240, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38417564

RESUMEN

In this study, we hypothesized that long-term administration of hesperidin can modulate the inflammatory response and oxidative stress in animals submitted to mechanical ventilation (MV). Twenty-five C57BL/6 male mice were divided into 5 groups: control, MV, animals receiving hesperidin in three doses 10, 25 and 50 mg/kg. The animals received the doses of hesperidin for 30 days via orogastric gavage, and at the end of the period the animals were submitted to MV. In animals submitted to MV, increased lymphocyte, neutrophil and monocyte/macrophage cell counts were observed in the blood and airways. Associated to this, MV promoted an increase in inflammatory cytokine levels such as CCL2, IL-12 and TNFα. The daily administration of hesperidin in the three doses prevented the effects caused by MV, which was observed by a lower influx of inflammatory cells into the airways, a reduction in inflammatory markers and less oxidative damage.


Asunto(s)
Hesperidina , Neumonía , Ratones , Animales , Masculino , Hesperidina/farmacología , Hesperidina/uso terapéutico , Ratones Endogámicos C57BL , Citocinas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Estrés Oxidativo , Neumonía/prevención & control , Inflamación/prevención & control
15.
JCO Oncol Pract ; 20(4): 503-508, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38261984

RESUMEN

PURPOSE: Prophylactic anti-infective strategies are used in patients with cancer to decrease the risk for infection. Dietary restrictions do not allow raw vegetables and fresh fruits to limit the introduction of potentially harmful pathogens in the gastrointestinal tract, but the efficacy is unclear. PATIENTS AND METHODS: In this study analyzing the impact of the dietary restrictions on infectious complications, all children treated between April 2014 and March 2018 for ALL and AML or non-Hodgkin lymphoma (NHL) were included. Dietary restrictions were standard until March 2016, but were stopped in April 2016. Patients with dietary restrictions (treated April 2014-March 2016) and patients not advised for dietary restrictions (treated April 2016-March 2018) were compared regarding infectious complications, including bloodstream infection, pneumonia, diarrhea, and fever of unknown origin (FUO). RESULTS: Eighty-six patients (25 female; 62 ALL; nine AML, 15 NHL) experienced 223 infections. The 46 patients with dietary restrictions and the 40 patients without food restrictions did not significantly differ regarding the number of infections per patient, bloodstream infections, pneumonia, diarrhea, FUO, admission to intensive care, and death. CONCLUSION: Our data suggest that dietary restrictions do not affect the risk for infectious complications. Therefore, the indication of dietary restrictions should be reconsidered in pediatric patients with cancer.


Asunto(s)
Fiebre de Origen Desconocido , Leucemia Mieloide Aguda , Neumonía , Sepsis , Humanos , Niño , Femenino , Fiebre de Origen Desconocido/etiología , Fiebre de Origen Desconocido/prevención & control , Neumonía/epidemiología , Neumonía/prevención & control , Neumonía/complicaciones , Leucemia Mieloide Aguda/complicaciones , Diarrea/epidemiología , Diarrea/complicaciones
16.
Inflammation ; 47(1): 323-332, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37819455

RESUMEN

Inflammatory response in the pulmonary endothelium drives the pathogenesis of acute lung injury and sepsis. Sirtuin 6 (SIRT6), a member of class III NAD+-dependent deacetylases belonging to the sirtuin family, regulates senescence, metabolism, and inflammation and extends lifespan in mice and model organisms. However, the role of SIRT6 in pulmonary endothelial inflammation is unknown. Thus, we hypothesized that SIRT6 suppresses inflammatory response in human lung microvascular cells (HLMEC) and ensues monocyte adhesion to endothelial cells. Primary HLMECs were treated with control or SIRT6 adenovirus or SIRT6 agonist, with or without lipopolysaccharide (LPS) treatment. We observed that treatment with LPS did not affect the protein expression of SIRT6 in HLMECs. However, adenovirus-mediated SIRT6 overexpression attenuated LPS-induced VCAM1 gene and protein expression, followed by decreased monocyte adhesion to endothelial cells. Similarly, activation of SIRT6 by a recently reported SIRT6 activator UBCS039, but not the regioisomer negative control compound UBCS060, ameliorated LPS-induced VCAM1 mRNA and protein expression as well as monocyte adhesion. Moreover, luciferase assay revealed that SIRT6 adenovirus decreased the activity of NF-κB, the master regulator of vascular inflammation. Taken together, these results indicate that molecular and pharmacological activation of SIRT6 protects against lung microvascular inflammation via suppressing NF-κB activation, implicating the therapeutic potential of the SIRT6 activators for lung disorders associated with microvascular inflammation.


Asunto(s)
Neumonía , Sirtuinas , Humanos , Ratones , Animales , FN-kappa B/metabolismo , Lipopolisacáridos/farmacología , Células Endoteliales/metabolismo , Inflamación/inducido químicamente , Inflamación/prevención & control , Inflamación/metabolismo , Neumonía/inducido químicamente , Neumonía/prevención & control , Neumonía/metabolismo , Pulmón/metabolismo , Endotelio Vascular/metabolismo
17.
J Clin Anesth ; 93: 111345, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-37988813

RESUMEN

INTRODUCTION: Dexmedetomidine improves intrapulmonary shunt in thoracic surgery and minimizes inflammatory response during one-lung ventilation (OLV). However, it is unclear whether such benefits translate into less postoperative pulmonary complications (PPCs). Our objective was to determine the impact of dexmedetomidine on the incidence of PPCs after thoracic surgery. METHODS: Major databases were used to identify randomized trials that compared dexmedetomidine versus placebo during thoracic surgery in terms of PPCs. Our primary outcome was atelectasis within 7 days after surgery. Other specific PPCs included hypoxemia, pneumonia, and acute respiratory distress syndrome (ARDS). Secondary outcome included intraoperative respiratory mechanics (respiratory compliance [Cdyn]) and postoperative lung function (forced expiratory volume [FEV1]). Random effects models were used to estimate odds ratios (OR). RESULTS: Twelve randomized trials, including 365 patients in the dexmedetomidine group and 359 in the placebo group, were analyzed in this meta-analysis. Patients in the dexmedetomidine group were less likely to develop postoperative atelectasis (2.3% vs 6.8%, OR 0.42, 95%CI 0.18-0.95, P = 0.04; low certainty) and hypoxemia (3.4% vs 11.7%, OR 0.26, 95%CI 0.10-0.68, P = 0.01; moderate certainty) compared to the placebo group. The incidence of postoperative pneumonia (3.2% vs 5.8%, OR 0.57, 95%CI 0.25-1.26, P = 0.17; moderate certainty) or ARDS (0.9% vs 3.5%, OR 0.39, 95%CI 0.07-2.08, P = 0.27; moderate certainty) was comparable between groups. Both intraoperative Cdyn and postoperative FEV1 were higher among patients that received dexmedetomidine with a mean difference of 4.42 mL/cmH2O (95%CI 3.13-5.72) and 0.27 L (95%CI 0.12-0.41), respectively. CONCLUSION: Dexmedetomidine administration during thoracic surgery may potentially reduce the risk of postoperative atelectasis and hypoxemia. However, current evidence is insufficient to demonstrate an effect on pneumonia or ARDS.


Asunto(s)
Dexmedetomidina , Ventilación Unipulmonar , Neumonía , Atelectasia Pulmonar , Síndrome de Dificultad Respiratoria , Cirugía Torácica , Humanos , Dexmedetomidina/efectos adversos , Ventilación Unipulmonar/efectos adversos , Pulmón , Atelectasia Pulmonar/epidemiología , Atelectasia Pulmonar/etiología , Atelectasia Pulmonar/prevención & control , Neumonía/epidemiología , Neumonía/etiología , Neumonía/prevención & control , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/prevención & control , Hipoxia/epidemiología , Hipoxia/etiología , Hipoxia/prevención & control
18.
J Physiother ; 70(1): 16-24, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38036402

RESUMEN

QUESTIONS: What is the effect of preoperative respiratory muscle training (RMT) on the incidence of postoperative pulmonary complications (PPCs) after open cardiac surgery? What is the effect of RMT on the duration of mechanical ventilation, postoperative length of stay and respiratory muscle strength? DESIGN: Systematic review of randomised trials with meta-analysis. PARTICIPANTS: Adults undergoing elective open cardiac surgery. INTERVENTION: The experimental groups received preoperative RMT and the comparison groups received no intervention. OUTCOME MEASURES: The primary outcomes were PPCs, length of hospital stay, respiratory muscle strength, oxygenation and duration of mechanical ventilation. The methodological quality of studies was assessed using the PEDro scale and the overall certainty of the evidence was assessed using the GRADE approach. RESULTS: Eight trials involving 696 participants were included. Compared with the control group, the respiratory training group had fewer PPCs (RR 0.51, 95% CI 0.38 to 0.70), less pneumonia (RR 0.44, 95% CI 0.25 to 0.78), shorter hospital stay (MD -1.7 days, 95% CI -2.4 to -1.1) and higher maximal inspiratory pressure values at the end of the training protocol (MD 12 cmH2O, 95% CI 8 to 16). The mechanical ventilation time was similar in both groups. The quality of evidence was high for pneumonia, length of hospital stay and maximal inspiratory pressure. CONCLUSION: Preoperative RMT reduced the risk of PPCs and pneumonia after cardiac surgery. The training also improved the maximal inspiratory pressure and reduced hospital stay. The effects on PPCs were large enough to warrant use of RMT in this population. REGISTRATION: CRD42021227779.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos , Neumonía , Adulto , Humanos , Tiempo de Internación , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Ejercicios Respiratorios/métodos , Neumonía/prevención & control , Músculos Respiratorios/fisiología , Complicaciones Posoperatorias/prevención & control
19.
Dis Esophagus ; 37(3)2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38018252

RESUMEN

Esophagectomy for esophageal cancer is associated with high morbidity. It remains unclear whether prehabilitation, a strategy aimed at optimizing patients' physical and mental functioning prior to surgery, improves postoperative outcomes. A systematic review and meta-analysis was conducted to evaluate the effect of prehabilitation on post-operative outcomes after esophagectomy. Data sources included Cochrane Central Register of Controlled Trials, MEDLINE, EMBASE, CINAHL, and PEDro, with information from 1 January 2000 to 5 August 2023. The analysis included randomized controlled trials and observational studies that compared prehabilitation interventions to standard care prior to esophagectomy. A random effects model was used to generate a pooled estimate for pairwise meta-analysis, meta-analysis of proportions, and meta-analysis of means. A total of 1803 patients were included with 584 in randomized controlled trials (RCTs) and 1219 in observational studies. In the randomized evidence, there were no significant differences between prehabilitation and control in the odds of postoperative pneumonia (15.0 vs. 18.9%, odds ratio (OR) 1.06 [95% confidence interval (CI): 0.66;1.72]) or pulmonary complications (14 vs. 25.6%, OR 0.68 [95% CI: 0.32;1.45]). In the observational data, there was a reduction in both postoperative pneumonia (22.5 vs. 32.9%, OR 0.48 [95% CI: 0.28;0.83]) and pulmonary complications (26.1 vs. 52.3%, OR 0.35 [95% CI: 0.17;0.75]) with prehabilitation. Hospital and intensive care unit length of stay (days), operative mortality, and severe complications (Clavien-Dindo ≥ 3) did not differ between groups in both the randomized data and observational data. Prehabilitation demonstrated reductions in postoperative pneumonia and pulmonary complications in observational studies, but not RCTs. The overall certainty of these findings is limited by the low quality of the available evidence.


Asunto(s)
Neoplasias Esofágicas , Neumonía , Humanos , Neoplasias Esofágicas/cirugía , Esofagectomía/efectos adversos , Unidades de Cuidados Intensivos , Neumonía/epidemiología , Neumonía/etiología , Neumonía/prevención & control , Ejercicio Preoperatorio , Ensayos Clínicos Controlados Aleatorios como Asunto , Estudios Observacionales como Asunto
20.
Nutrients ; 15(22)2023 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-38004123

RESUMEN

Multiple studies have confirmed that Lactiplantibacillus plantarum has beneficial effects in respiratory diseases, including respiratory tract infections, asthma, and chronic obstructive pulmonary disease. However, the role of L. plantarum lysates in respiratory diseases is unclear. Staphylococcus aureus infects the lungs of mice, recruits immune cells, and induces structural changes in alveoli. Lung diseases can be further aggravated by inflammatory cytokines such as CCL2 and interleukin (IL)-6. In in vivo studies, L. plantarum K8 nanoparticles (K8NPs) restored lung function and prevented lung damage caused by S. aureus infection. They inhibited the S. aureus infection and the infiltration of immune cells and prevented the increase in goblet cell numbers in the lungs of S. aureus-infected mice. K8NPs suppressed the expression of CCL2 and IL-6, which were increased by the combination treatment of tumor necrosis factor alpha and interferon gamma (TI), in a dose-dependent manner. In in vitro studies, the anti-inflammatory effect of K8NPs in TI-treated A549 cells and TI-injected mice occurred through the reduction in activated mitogen-activated protein kinases and nuclear factor kappa-B. These findings suggest that the efficacy of K8NPs in controlling respiratory inflammation and infection can be used to develop functional materials that can prevent or alleviate respiratory diseases.


Asunto(s)
Lactobacillus plantarum , Neumonía , Infecciones del Sistema Respiratorio , Infecciones Estafilocócicas , Animales , Ratones , Factor de Necrosis Tumoral alfa/metabolismo , Interferón gamma/metabolismo , Staphylococcus aureus , Citocinas/metabolismo , Neumonía/prevención & control , Interleucina-6/metabolismo , Infecciones del Sistema Respiratorio/prevención & control , Lactobacillus plantarum/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA