Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.006
Filtrar
1.
J Control Release ; 365: 301-316, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38007195

RESUMEN

Changes in bodily fluid pressures, such as pulmonary artery pressure, play key roles in high-altitude pulmonary edema (HAPE) and other disorders. Smart delivery systems releasing a drug in response to these pressures might facilitate early medical interventions. However, pressure-responsive delivery systems are unavailable. We here constructed hydrostatic pressure-sensitive multivesicular liposomes (PSMVLs) based on the incomplete filling of the internal vesicle space with neutral lipids. These liposomes were loaded with amlodipine besylate (AB), a next-generation calcium channel inhibitor, to treat HAPE on time. AB-loaded PSMVLs (AB-PSMVLs) were destroyed, and AB was released through treatment under hydrostatic pressure of at least 25 mmHg. At 25 mmHg, which is the minimum pulmonary artery pressure value in HAPE, 38.8% of AB was released within 1 h. In a mouse HAPE model, AB-PSMVLs concentrated in the lung and released AB to diffuse into the vascular wall. Intravenously injected AB-PSMVLs before HAPE modeling resulted in a stronger protection of lung tissues and respiratory function and lower occurrence of pulmonary edema than treatment with free drug or non-pressure-sensitive AB-loaded liposomes. This study offers a new strategy for developing smart drug delivery systems that respond to changes in bodily fluid pressures.


Asunto(s)
Mal de Altura , Hipertensión Pulmonar , Edema Pulmonar , Ratones , Animales , Edema Pulmonar/tratamiento farmacológico , Edema Pulmonar/prevención & control , Liposomas , Altitud , Sistemas de Liberación de Medicamentos
2.
Biochem Pharmacol ; 218: 115905, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37949322

RESUMEN

BACKGROUND AND PURPOSE: Neurogenic pulmonary edema (NPE) frequently arises as a complication subsequent to subarachnoid hemorrhage (SAH). Heterodimers of S100A8 and S100A9 are commonly formed, thereby initiating an inflammatory reaction through receptor binding on the cell surface. Paquinimod serves as a specific inhibitor of S100A9. The objective of this investigation is to assess the impact of Paquinimod administration and S100A9 knockout on NPE following SAH. METHODS: In this study, SAH models of C57BL/6J wild-type (WT) and S100A9 knockout mice were established through intravascular perforation. These models were then divided into several groups, including the WT-sham group, S100A9-KO-sham group, WT-SAH group, WT-SAH + Paquinimod group, and S100A9-KO-SAH group. After 24 h of SAH induction, pulmonary edema was assessed using the lung wet-dry weight method and Hematoxylin and eosin (HE) staining. Additionally, the expression levels of various proteins, such as interleukin-1ß (IL-1ß), tumor necrosis factor α (TNF-α), occludin, claudin-3, Bax, Bcl-2, TLR4, MYD88, and pNF-κB, in lung tissue were analyzed using western blot and immunofluorescence staining. Lung tissue apoptosis was detected by TUNEL staining. RESULTS: Firstly, our findings indicate that the knockout of S100A9 has a protective effect on early brain injury following subarachnoid hemorrhage (SAH). Additionally, the reduction of brain injury after SAH can also alleviate neurogenic pulmonary edema (NPE). Immunofluorescence staining and western blot analysis revealed that compared to SAH mice with wild-type S100A9 expression (WT-SAH), the lungs of S100A9 knockout SAH mice (S100A9-KO-SAH) and mice treated with Paquinimod exhibited decreased levels of inflammatory molecules (IL-1ß and TNF-α) and increased levels of tight junction proteins. Furthermore, the knockout of S100A9 resulted in upregulated expression of the apoptotic-associated protein Bax and down-regulated expression of Bcl-2. Furthermore, a decrease in TLR4, MYD88, and phosphorylated pNF-κB was noted in S100A9-KO-SAH and Paquinimod treated mice, indicating the potential involvement of the TLR4/MYD88/NF-κB signaling pathway in the inhibition of the protective effect of S100A9 on NPE following SAH. CONCLUSION: The knockout of S100A9 not only ameliorated initial cerebral injury following subarachnoid hemorrhage (SAH), but also mitigated SAH-associated neurogenic pulmonary edema (NPE). Additionally, Paquinimod was found to diminish NPE. These findings imply a correlation between the central nervous system and peripheral organs, highlighting the potential of safeguarding the brain to mitigate harm to peripheral organs.


Asunto(s)
Lesiones Encefálicas , Edema Pulmonar , Hemorragia Subaracnoidea , Animales , Ratones , Proteína X Asociada a bcl-2/metabolismo , Lesiones Encefálicas/patología , Calgranulina B , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Edema Pulmonar/tratamiento farmacológico , Edema Pulmonar/etiología , Edema Pulmonar/prevención & control , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/tratamiento farmacológico , Hemorragia Subaracnoidea/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
3.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 39(11): 996-1002, 2023.
Artículo en Chino | MEDLINE | ID: mdl-37980551

RESUMEN

Objective To investigate the effect of dexamethasone (DEX) combined with glutamine (Gln) on lung inflammation and pulmonary edema in rats with acute lung injury induced by lipopolysaccharide (LPS) and its related mechanisms. Methods Fifty Wistar rats were randomly divided into control group, model group, dexamethasone group (DEX) and DEX combined with Gln group. Except for the control group, rats in other groups were injected with 6 mg/kg LPS intraperitoneally to induce an acute lung injury. The mRNA expression of p38 MAPK, NLRP3, and NF-κB in lung tissue were detected by real-time quantitative PCR. The protein expressions of p-p38 MAPK, NLRP3, phosphorylated inhibitor of nuclear factor κB (p-IκB), NF-κB p65, aquaporin 1 (AQP1) and AQP5 in lung tissue were detected by Western blot analysis. ELISA was used to detect the content of serum tumor necrosis factor-α (TNF-α), interleukin 6 (IL-6), interleukin 1ß (IL-1ß). Spectrophotometer was employed to detect the content of superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione peroxidase (GSH-Px) in lung tissue. Results Compared with the control group, the lung index of the model group decreased, the content of the serum inflammatory factors TNF-α, IL-6 and IL-1ß significantly increased, and the protein expression of p38 MAPK, NLRP3, NF-κB mRNA, p-p38 MAPK, NLRP3, p-IκB and NF-κB p65 in the lung tissue significantly increased, while that of AQP1, AQP5 decreased, and the content of SOD and GSH-Px in lung tissue decreased, while that of MDA increased; Compared with the model group, the above mentioned symptoms and indicators in each treatment group were significantly improved, among which the DEX combined with Gln group was the most significant. Conclusion DEX combined with Gln can inhibit inflammation, resist oxidative damage, relieve pulmonary edema, and prevent acute lung injury. Its mechanism is related to inhibiting the activation of p38 MAPK, NLRP3, and NF-κB signaling pathways, promoting the expression of AQP1 and AQP5, and promoting the activity of antioxidant products.


Asunto(s)
Lesión Pulmonar Aguda , Neumonía , Edema Pulmonar , Ratas , Animales , Edema Pulmonar/tratamiento farmacológico , Edema Pulmonar/prevención & control , Edema Pulmonar/metabolismo , FN-kappa B/metabolismo , Glutamina , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Lipopolisacáridos , Ratas Sprague-Dawley , Ratas Wistar , Lesión Pulmonar Aguda/inducido químicamente , Proteínas I-kappa B , Dexametasona/farmacología , ARN Mensajero , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Superóxido Dismutasa
4.
Toxicol Appl Pharmacol ; 480: 116742, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37923178

RESUMEN

Acute hypobaric hypoxia at high altitude can cause fatal non-cardiogenic high altitude pulmonary edema. Anti-inflammatory and anti-oxidant treatments appear to be a prospective way to alleviate acute hypoxia lung injury. Kaempferol (KA) and ginsenoside Rg1 (GRg1) can be isolated and purified from ginseng with anti-inflammatory, antioxidant, anti-carcinogenic, neuroprotective, and antiaging effects. However, their effects and pharmacological mechanisms on lung injury remains unclear. Network pharmacology analyses were used to explore potential targets of KA and GRg1 against acute hypobaric hypoxia induced lung injury. Rat lung tissues were further used for animal experiment verification. Among the putative targets of KA and GRg1 for inhibition of acute hypobaric hypoxia induced lung injury, AKT1, PIK3R1, PTK2, STAT3, HSP90AA1 and AKT2 were recognized as higher interrelated targets. And PI3K-AKT signaling pathway is considered to be the most important and relevant pathway. The rat experimental results showed that KA and GRg1 significantly improved histopathological changes and decreased pulmonary edema in rats with lung injury caused by acute hypobaric hypoxia. The concentrations of IL-6, TNF-α, MDA, SOD and CAT in rats treated with KA and GRg1 were significantly ameliorated. Protein and mRNA levels of PI3K and AKTI were significantly inhibited after KA administration. KA and GRg1 can lower lung water content, improve lung tissue damage, reduce the production of pro-inflammatory cytokines and the oxidative stress level.


Asunto(s)
Lesión Pulmonar Aguda , Edema Pulmonar , Ratas , Animales , Edema Pulmonar/tratamiento farmacológico , Edema Pulmonar/prevención & control , Fosfatidilinositol 3-Quinasas/metabolismo , Quempferoles/farmacología , Quempferoles/uso terapéutico , Farmacología en Red , Hipoxia/complicaciones , Hipoxia/tratamiento farmacológico , Antioxidantes , Lesión Pulmonar Aguda/tratamiento farmacológico , Antiinflamatorios
5.
Eur J Pharmacol ; 959: 176065, 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37775017

RESUMEN

High-altitude pulmonary edema (HAPE) is a potentially fatal disease. Notoginsenoside R1 is a novel phytoestrogen with anti-inflammatory, antioxidant and anti-apoptosis properties. However, its effects and underlying mechanisms in the protection of hypobaric hypoxia-induced HAPE rats remains unclear. This study aimed to explore the protective effects and underlying mechanisms of Notoginsenoside R1 in hypobaric hypoxia-induced HAPE. We found that Notoginsenoside R1 alleviated the lung tissue injury, decreased lung wet/dry ratio, and reduced inflammation and oxidative stress. Additionally, Notoginsenoside R1 ameliorated the changes in arterial blood gas, decreased the total protein concentration in bronchoalveolar lavage fluid, and inhibited the occurrence of apoptosis caused by HAPE. In the process of further exploration of the mechanism, it was found that Notoginsenoside R1 could promote the activation of ERK1/2-P90rsk-BAD signaling pathway, and the effect of Notoginsenoside R1 was attenuated after the use of ERK1/2 inhibitor U0126. Our study indicated that the protective effects of Notoginsenoside R1 against HAPE were mainly related to the inhibition of inflammation, oxidative stress, and apoptosis. Notoginsenoside R1 may be a potential candidate for preventing HAPE.


Asunto(s)
Altitud , Edema Pulmonar , Ratas , Animales , Sistema de Señalización de MAP Quinasas , Edema Pulmonar/prevención & control , Hipoxia/complicaciones , Hipoxia/metabolismo , Inflamación
6.
Rev Esp Cardiol (Engl Ed) ; 76(10): 759-766, 2023 Oct.
Artículo en Inglés, Español | MEDLINE | ID: mdl-36801376

RESUMEN

INTRODUCTION AND OBJECTIVES: Contrast-associated acute kidney injury (CA-AKI) is a potential complication of procedures requiring administration of iodinated contrast medium. RenalGuard, which provides real-time matching of intravenous hydration with furosemide-induced diuresis, is an alternative to standard periprocedural hydration strategies. The evidence on RenalGuard in patients undergoing percutaneous cardiovascular procedures is sparse. We used a Bayesian framework to perform a meta-analysis of RenalGuard as a CA-AKI preventive strategy. METHODS: We searched Medline, Cochrane Library and Web of Science for randomized trials of RenalGuard vs standard periprocedural hydration strategies. The primary outcome was CA-AKI. Secondary outcomes were all-cause death, cardiogenic shock, acute pulmonary edema, and renal failure requiring renal replacement therapy. A Bayesian random-effect risk ratio (RR) with corresponding 95% credibility interval (95%CrI) was calculated for each outcome. PROSPERO database number CRD42022378489. RESULTS: Six studies were included. RenalGuard was associated with a significant relative reduction in CA-AKI (median RR, 0.54; 95%CrI, 0.31-0.86) and acute pulmonary edema (median RR, 0.35; 95%CrI, 0.12-0.87). No significant differences were observed for the other secondary endpoints [all-cause death (RR, 0.49; 95%CrI, 0.13-1.08), cardiogenic shock (RR, 0.06; 95%CrI, 0.00-1.91), and renal replacement therapy (RR, 0.52; 95%CrI, 0.18-1.18)]. The Bayesian analysis also showed that RenalGuard had a high probability of ranking first for all the secondary outcomes. These results were consistent in multiple sensitivity analyses. CONCLUSIONS: In patients undergoing percutaneous cardiovascular procedures, RenalGuard was associated with a reduced risk of CA-AKI and acute pulmonary edema compared with standard periprocedural hydration strategies.


Asunto(s)
Lesión Renal Aguda , Edema Pulmonar , Humanos , Diuréticos , Edema Pulmonar/etiología , Edema Pulmonar/prevención & control , Edema Pulmonar/tratamiento farmacológico , Choque Cardiogénico , Teorema de Bayes , Ensayos Clínicos Controlados Aleatorios como Asunto , Diuresis , Medios de Contraste/efectos adversos , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/epidemiología , Lesión Renal Aguda/prevención & control , Factores de Riesgo
7.
Drug Chem Toxicol ; 46(5): 1024-1034, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36069203

RESUMEN

We aimed to observe the possible effects of melatonin (MLT) deprivation (pinealectomy) and exogenous MLT administration on pulmonary edema induced by alpha-naphthylthiourea (ANTU), a toxic chemical agent, in rats. Seventy animals were assigned to seven groups: control, sham pinealectomy (PINX), PINX, ANTU (10 mg/kg intraperitoneal on day 30), ANTU + MLT (10 mg/kg/day i.p. for 30 days), ANTU + PINX, and ANTU + PINX + MLT.In this study, pleural effusion (PE) formation, lung weight/body weight (LW/BW) and PE/BW ratios (fluid accumulation and weight values in the lungs) increase detected. Pre-ANTU MLT administration led to significant decreases in PE, LW/BW, and PE/BW levels. The inhibited glutathione (GSH) and superoxide dismutase (SOD) levels and high malondialdehyde (MDA) levels that ANTU increase lipid peroxidation in the study. MLT administration eliminated oxidative stress by reducing MDA and ameliorating GSH and SOD levels.Pre-ANTU MLT administration led to a significant decrease in interleukin-1 beta (IL-1ß) and tumor necrosis factor-alpha (TNF-α) levels in the lung when compared to the ANTU group without MLT administration. Post-pinealectomy ANTU administration significantly increased IL-1ß and TNF-α levels when compared to ANTU and MLT administration without pinealectomy. Diffused inflammatory cell infiltration, interstitial pulmonary edema, and histopathological congestion were observed after the administration of ANTU. Severity of the damage was elevated in the ANTU + PINX group. MLT treatment regressed pulmonary effusion and edema and improves lung structure. In brief, the findings suggested that MLT inhibited proinflammatory mediators and could serve as a therapeutic agent to prevent inflammatory disorders.


Asunto(s)
Melatonina , Edema Pulmonar , Ratas , Animales , Edema Pulmonar/inducido químicamente , Edema Pulmonar/prevención & control , Edema Pulmonar/patología , Melatonina/farmacología , Pinealectomía , Factor de Necrosis Tumoral alfa , Tiourea/toxicidad
8.
Korean J Anesthesiol ; 75(6): 518-529, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35912428

RESUMEN

BACKGROUND: Acute lung injury (ALI) is the most serious complication of subarachnoid hemorrhage (SAH). We investigated role of autophagy and inflammatory signaling pathways in lung damage and therapeutic effects of dexmedetomidine (DEX). METHODS: Fifty male Wistar rats were randomly divided into five groups: sham, SAH, SAH+ DEX5, SAH+DEX25, and SAH+DEX50. SAH was induced using endovascular perforation technique. All rats received mechanical ventilation for 60 minutes. At 2 and 24 h of SAH induction, SAH+DEX groups were treated with 5, 25, and 50 µg/kg of DEX, respectively. Histological ALI score and pulmonary edema were assessed after 48 h. Lung expression of LC3B, ATG3, p62, TLR4, TLR9, and NFκB was assessed using western blotting and quantitative PCR. Blood levels of IL-6, IL-1ß, IFN-γ, and TNFα were also assessed. RESULTS: SAH induced ALI and pulmonary edema, which were attenuated in SAH+DEX5 (P < 0.001 for both) and SAH+DEX25 groups (P = 0.001 and P < 0.001 for ALI and edema, respectively). Lung expressions of LC3B and ATG3 were upregulated in SAH group, which was attenuated in SAH+DEX5 and SAH+DEX25 groups. Lung expressions of TLR4, TLR9, and NFκB were increased in SAH group, which was attenuated in SAH+DEX5 group. Blood IL-6 level was increased in SAH group and attenuated in SAH+DEX5 and SAH+DEX25 groups. Blood IFN-γ level was lower in SAH group than in sham group, and it was increased in SAH+DEX25 group. CONCLUSIONS: Low-dose DEX treatment after SAH may protect against ALI by disrupting pathological brain-lung crosstalk and alleviating autophagy flux and TLR-dependent inflammatory pathways.


Asunto(s)
Lesión Pulmonar Aguda , Dexmedetomidina , Edema Pulmonar , Hemorragia Subaracnoidea , Animales , Masculino , Ratas , Lesión Pulmonar Aguda/prevención & control , Lesión Pulmonar Aguda/complicaciones , Autofagia , Dexmedetomidina/farmacología , Interleucina-6/uso terapéutico , FN-kappa B/metabolismo , Edema Pulmonar/prevención & control , Edema Pulmonar/complicaciones , Ratas Wistar , Transducción de Señal , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/tratamiento farmacológico , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 9/metabolismo
9.
Pflugers Arch ; 474(10): 1053-1067, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35778581

RESUMEN

Acute hypoxia impairs left ventricular (LV) inotropic function and induces development of pulmonary edema (PE). Enhanced and uneven hypoxic pulmonary vasoconstriction is an important pathogenic factor of hypoxic PE. We hypothesized that the potent vasodilator relaxin might reduce hypoxic pulmonary vasoconstriction and prevent PE formation. Furthermore, as relaxin has shown beneficial effects in acute heart failure, we expected that relaxin might also improve LV inotropic function in hypoxia. Forty-two rats were exposed over 24 h to normoxia or hypoxia (10% N2 in O2). They were infused with either 0.9% NaCl solution (normoxic/hypoxic controls) or relaxin at two doses (15 and 75 µg kg-1 day-1). After 24 h, hemodynamic measurements and bronchoalveolar lavage were performed. Lung tissue was obtained for histological and immunohistochemical analyses. Hypoxic control rats presented significant depression of LV systolic pressure by 19% and of left and right ventricular contractility by about 40%. Relaxin did not prevent the hypoxic decrease in LV inotropic function, but re-increased right ventricular contractility. Moreover, hypoxia induced moderate interstitial PE and inflammation in the lung. Contrasting to our hypothesis, relaxin did not prevent hypoxia-induced pulmonary edema and inflammation. In hypoxic control rats, PE was similarly distributed in the apical and basal lung lobes. In relaxin-treated rats, PE index was 35-40% higher in the apical than in the basal lobe, which is probably due to gravity effects. We suggest that relaxin induced exaggerated vasodilation, and hence pulmonary overperfusion. In conclusion, the results show that relaxin does not prevent but rather may aggravate PE formation.


Asunto(s)
Edema Pulmonar , Relaxina , Animales , Hipoxia/complicaciones , Neumonía/terapia , Arteria Pulmonar , Edema Pulmonar/etiología , Edema Pulmonar/prevención & control , Ratas , Relaxina/farmacología , Relaxina/uso terapéutico , Solución Salina/farmacología , Vasodilatadores/farmacología
10.
Biomed Pharmacother ; 153: 113307, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35753262

RESUMEN

Acute lung injury (ALI), hallmarked with alveolar epithelial barrier impairment and pulmonary edema induced by acute inflammation, presents a severe health burden to the public, due to the limited available interventions. Oxyberberine (OBB), having improved anti-inflammatory activity and safety, is a representative component with various activities derived from berberine, whereas its role against ALI with alveolar epithelial barrier injury remains uncertain. To investigate the influence and underlying mechanisms of OBB on ALI, we induced acute inflammation in mice and A549 cells by using lipopolysaccharide (LPS). Changes in alveolar permeability were assessed by analyzing lung histopathology, measuring the dry/wet weight ratio of the lungs, and altering proinflammatory cytokines and neutrophils levels in the bronchoalveolar lavage fluid (BALF). Parameters of pulmonary permeability were assessed through ELISA, western blotting, quantitative real-time PCR, and immunofluorescence analysis. U46619, the agonist of RhoA/ROCK, was employed to further investigate the mechanism of OBB on ALI. Unexpectedly, we found OBB mitigated lung impairment, pulmonary edema, inflammatory reactions in BALF and lung tissue, reduction in ZO-1, and addition of connexin-43. Besides, OBB markedly reduced the expression of RhoA in association with its downstream factors, which are linked to the intercellular junctions and permeability both in vivo and in vitro. Nevertheless, U46619 abolished the benefits obtained from OBB in A549 cells. In conclusion, these outcomes indicated that OBB exerted RhoA/ROCK inhibitor-like effect to moderate alveolar epithelial barrier impairment and permeability, ultimately preventing ALI progression.


Asunto(s)
Lesión Pulmonar Aguda , Edema Pulmonar , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/prevención & control , Animales , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Pulmón , Ratones , Edema Pulmonar/tratamiento farmacológico , Edema Pulmonar/prevención & control , Transducción de Señal
11.
J Appl Physiol (1985) ; 132(6): 1361-1369, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35511718

RESUMEN

Acetazolamide prevents acute mountain sickness (AMS) by inhibition of carbonic anhydrase. Since it also reduces acute hypoxic pulmonary vasoconstriction (HPV), it may also prevent high-altitude pulmonary edema (HAPE) by lowering pulmonary artery pressure. We tested this hypothesis in a randomized, placebo-controlled, double-blind study. Thirteen healthy, nonacclimatized lowlanders with a history of HAPE ascended (<22 h) from 1,130 to 4,559 m with one overnight stay at 3,611 m. Medications were started 48 h before ascent (acetazolamide: n = 7, 250 mg 3 times/day; placebo: n = 6, 3 times/day). HAPE was diagnosed by chest radiography and pulmonary artery pressure by measurement of right ventricular to atrial pressure gradient (RVPG) by transthoracic echocardiography. AMS was evaluated with the Lake Louise Score (LLS) and AMS-C score. The incidence of HAPE was 43% versus 67% (acetazolamide vs. placebo, P = 0.39). Ascent to altitude increased RVPG from 20 ± 5 to 43 ± 10 mmHg (P < 0.001) without a group difference (P = 0.68). Arterial Po2 fell to 36 ± 9 mmHg (P < 0.001) and was 8.5 mmHg higher with acetazolamide at high altitude (P = 0.025). At high altitude, the LLS and AMS-C score remained lower in those taking acetazolamide (both P < 0.05). Although acetazolamide reduced HAPE incidence by 35%, this effect was not statistically significant, and was considerably less than reductions of about 70%-100% with prophylactic dexamethasone, tadalafil, and nifedipine performed with the same ascent profile at the same location. We could not demonstrate a reduction in RVPG compared with placebo treatment despite reductions in AMS severity and better arterial oxygenation. Limited by small sample size, our data do not support recommending acetazolamide for the prevention of HAPE in mountaineers ascending rapidly to over 4,500 m.NEW & NOTEWORTHY This randomized, placebo-controlled, double-blind study is the first to investigate whether acetazolamide, which reduces acute mountain sickness (AMS), inhibits short-term hypoxic pulmonary vasoconstriction, and also prevents high-altitude pulmonary edema (HAPE) in a fast-climbing ascent to 4,559 m. We found no statistically significant reduction in HAPE incidence or differences in hypoxic pulmonary artery pressures compared with placebo despite reductions in AMS and greater ventilation-induced arterial oxygenation. Our data do not support recommending acetazolamide for HAPE prevention.


Asunto(s)
Mal de Altura , Edema Pulmonar , Acetazolamida/uso terapéutico , Enfermedad Aguda , Altitud , Mal de Altura/diagnóstico , Mal de Altura/tratamiento farmacológico , Mal de Altura/prevención & control , Humanos , Hipertensión Pulmonar , Hipoxia/tratamiento farmacológico , Arteria Pulmonar , Edema Pulmonar/prevención & control
12.
Inflamm Res ; 71(2): 183-185, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34993559

RESUMEN

Growth Hormone-Releasing Hormone (GHRH) is a neuropeptide regulating the release of Growth Hormone (GH) from the anterior pituitary gland, and acts as a growth factor in a diverse variety of tissues. GHRH antagonists (GHRHAnt) have been developed to counteract those events, and the beneficial effects of those peptides toward homeostasis have been associated with anti-inflammatory activities. Our lab is interested in delineating the mechanisms governing endothelial barrier function. Our goal is to establish new grounds on the development of efficient countermeasures against Acute Respiratory Distress Syndrome (ARDS), which has been associated with thousands of deaths worldwide due to COVID-19. Herein we demonstrate in vivo that GHRHAnt suppresses LPS-induced increase in bronchoalveolar lavage fluid (BALF) protein concentration, thus protecting the lungs against edema and inflammation.


Asunto(s)
Líquido del Lavado Bronquioalveolar/química , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Lipopolisacáridos , Animales , COVID-19/complicaciones , Hormona Liberadora de Hormona del Crecimiento , Inflamación/etiología , Inflamación/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas/química , Edema Pulmonar/etiología , Edema Pulmonar/prevención & control , Especies Reactivas de Oxígeno , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/etiología , SARS-CoV-2
13.
Nat Commun ; 12(1): 6791, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34815389

RESUMEN

Angiotensin-converting enzyme 2 (ACE2) is a receptor for cell entry of SARS-CoV-2, and recombinant soluble ACE2 protein inhibits SARS-CoV-2 infection as a decoy. ACE2 is a carboxypeptidase that degrades angiotensin II, thereby improving the pathologies of cardiovascular disease or acute lung injury. Here we show that B38-CAP, an ACE2-like enzyme, is protective against SARS-CoV-2-induced lung injury. Endogenous ACE2 expression is downregulated in the lungs of SARS-CoV-2-infected hamsters, leading to elevation of angiotensin II levels. Recombinant Spike also downregulates ACE2 expression and worsens the symptoms of acid-induced lung injury. B38-CAP does not neutralize cell entry of SARS-CoV-2. However, B38-CAP treatment improves the pathologies of Spike-augmented acid-induced lung injury. In SARS-CoV-2-infected hamsters or human ACE2 transgenic mice, B38-CAP significantly improves lung edema and pathologies of lung injury. These results provide the first in vivo evidence that increasing ACE2-like enzymatic activity is a potential therapeutic strategy to alleviate lung pathologies in COVID-19 patients.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Tratamiento Farmacológico de COVID-19 , COVID-19/prevención & control , Lesión Pulmonar/prevención & control , SARS-CoV-2/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Lesión Pulmonar Aguda , Angiotensina II , Animales , COVID-19/patología , Carboxipeptidasas , Chlorocebus aethiops , Cricetinae , Modelos Animales de Enfermedad , Femenino , Humanos , Pulmón/patología , Masculino , Ratones , Ratones Transgénicos , Edema Pulmonar/patología , Edema Pulmonar/prevención & control , Glicoproteína de la Espiga del Coronavirus/efectos de los fármacos , Células Vero
14.
Anal Cell Pathol (Amst) ; 2021: 6653681, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34426780

RESUMEN

Acute lung injury (ALI) is a common clinical syndrome in the cardiac intensive care unit with a high mortality rate. Inflammation and oxidative stress have been reported to play a crucial role in the development of ALI. Previous studies have shown that human umbilical cord mesenchymal stem cells (hucMSCs) have anti-inflammatory and antioxidative effects in various diseases. However, the anti-inflammatory and antioxidative effects of the hucMSC conditioned medium (CM) on LPS-induced ALI remain unclear. Therefore, in this study, we assessed whether the hucMSC conditioned medium could attenuate LPS-induced ALI and the underlying mechanisms. Mice were randomly divided into four groups: the control group, PBS group, LPS+PBS group, and LPS+CM group. The lung histopathology and bronchoalveolar lavage fluid (BALF) were analyzed after intervention. The Nrf2/NF-κB signaling pathway and its downstream target genes were tested, and the cytokines and growth factors in CM were also measured. The results showed that CM significantly attenuated the histological alterations; decreased the wet/dry weight ratio; reduced the levels of MPO, MDA and ROS; increased SOD and GSH activity; and downregulated the level of proinflammatory cytokines such as IL-1ß, IL-6, and TNF-α. Furthermore, CM promoted the expression of Nrf2 and its target genes NQ01, HO-1, and GCLC and inhibited the expression of NF-κB and its target genes IL-6, IL-1ß, and TNF-α. These effects may be closely related to the large amounts of cytokines and growth factors in the CM. In conclusion, our results demonstrated that CM could attenuate LPS-induced ALI, probably due to inhibition of inflammation and oxidative stress via the Nrf2/NF-κB signaling pathway.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Antioxidantes/farmacología , Medios de Cultivo Condicionados/farmacología , Pulmón/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Neumonía/prevención & control , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Antiinflamatorios/metabolismo , Antiinflamatorios/farmacología , Antioxidantes/metabolismo , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Lipopolisacáridos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Neumonía/inducido químicamente , Neumonía/metabolismo , Neumonía/patología , Edema Pulmonar/inducido químicamente , Edema Pulmonar/metabolismo , Edema Pulmonar/patología , Edema Pulmonar/prevención & control , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
15.
Int Immunopharmacol ; 99: 107972, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34298401

RESUMEN

We sought to assess the protective effect of different doses of Fingolimod (FTY720) in a rat model of acute lung injury (ALI) induced by intratracheal instillation of lipopolysaccharide (LPS) and explored the underlying mechanisms. The ALI model was established in rats and different doses of FTY720 (0.1 mg/kg, 0.2 mg/kg, 0.5 mg/kg, 1 mg/kg, or 2 mg/kg) were injected intraperitoneally. Lung computed tomography and blood gas analyses were performed at 6 h, 24 h, and 48 h after intraperitoneal injection, and the lung tissues were extracted to prepare paraffin sections for histopathological examination. The levels of inflammatory cytokines (TNF-α, IL-6, and IL-1ß) were detected by ELISA, and the expressions of inflammatory pathway proteins in each group were measured by Western blot analysis. A single intraperitoneal injection of FTY720 inhibited LPS-induced NF-κB activation, reduced the level of inflammatory cytokines, and decreased the infiltration of inflammatory cells. Moreover, it alleviated lung tissue injury, as shown by marked attenuation of pulmonary oedema and improved arterial partial pressure of oxygen (PaO2) and the general condition of ALI rats. In conclusion, our results demonstrate the protective effect of FTY720 against LPS-induced ALI. The underlying mechanism of the protective effect may involve inhibition of LPS-induced activation of NF-κB and regulation of the inflammatory pathway to alleviate barrier dysfunction of alveolar capillaries.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Clorhidrato de Fingolimod/uso terapéutico , Inmunosupresores/uso terapéutico , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/patología , Animales , Análisis de los Gases de la Sangre , Barrera Alveolocapilar/efectos de los fármacos , Citocinas/sangre , Relación Dosis-Respuesta a Droga , Clorhidrato de Fingolimod/administración & dosificación , Inmunosupresores/administración & dosificación , Inyecciones Intraperitoneales , Lipopolisacáridos , Pulmón/patología , Masculino , FN-kappa B/efectos de los fármacos , Oxígeno/sangre , Edema Pulmonar/prevención & control , Ratas , Ratas Sprague-Dawley , Tomografía Computarizada por Rayos X
16.
Inflammation ; 44(6): 2193-2202, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34085161

RESUMEN

Vanillin, the main constituents of vanillin beans, has been reported to exhibit anti-inflammatory effects. However, the effects of vanillin on the cadmium-induced lung injury are still unclear. Therefore, we assay whether vanillin has potential preventive activity on cadmium-induced lung injury in mice. Mice were given vanillin (5, 10, 20 mg/kg) and treated with cadmium for 7 days. The detection data of vanillin on lung tissue changes were analyzed after the cadmium treatment. The results displayed that vanillin obviously decreased the lung histological alterations and myeloperoxidase (MPO) activity. Vanillin also suppressed the levels of TNF-α, IL-1ß, and IL-6 in BALF. Furthermore, vanillin prevented cadmium-induced NF-κB activation and upregulation the expression of tight junction protein ZO-1 and occludin. In addition, vanillin significantly increased the expression of aryl hydrocarbon receptor (AhR), and inhibition of AhR by its agonist could reverse the protective effects of vanillin on cadmium-induced lung injury. To sum up, vanillin could be a potential drug for the treatment of cadmium-induced lung injury.


Asunto(s)
Antiinflamatorios/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/agonistas , Benzaldehídos/farmacología , Lesión Pulmonar/prevención & control , Pulmón/efectos de los fármacos , Neumonía/prevención & control , Receptores de Hidrocarburo de Aril/agonistas , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cloruro de Cadmio , Citocinas/metabolismo , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Pulmón/metabolismo , Pulmón/patología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Masculino , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Ocludina/metabolismo , Peroxidasa/metabolismo , Neumonía/inducido químicamente , Neumonía/metabolismo , Neumonía/patología , Edema Pulmonar/inducido químicamente , Edema Pulmonar/metabolismo , Edema Pulmonar/patología , Edema Pulmonar/prevención & control , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Proteína de la Zonula Occludens-1/metabolismo
17.
Inflammation ; 44(6): 2170-2179, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34109517

RESUMEN

Acute ethanol intoxication increases the risk of sepsis and aggravates the symptoms of sepsis and lung injury. Therefore, this study aimed to explore whether sphingosine kinase 1 (SphK1)/sphingosine-1-phosphate (S1P)/S1P receptor 1 (S1PR1) signaling pathway functions in lung injury caused by acute ethanol intoxication-enhanced sepsis, as well as its underlying mechanism. The acute ethanol intoxication model was simulated by intraperitoneally administering mice with 32% ethanol solution, and cecal ligation and puncture (CLP) was used to construct the sepsis model. The lung tissue damage was observed by hematoxylin-eosin (H&E) staining, and the wet-to-dry (W/D) ratio was used to evaluate the degree of pulmonary edema. Inflammatory cell counting and protein concentration in bronchoalveolar lavage fluid (BALF) were, respectively, detected by hemocytometer and bicinchoninic acid (BCA) method. The levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1ß, and IL-18 in BALF were detected by their commercial enzyme-linked immunosorbent assay (ELISA) kits. The myeloperoxidase (MPO) activity and expression of apoptosis-related proteins and SphK1/S1P/S1PR1 pathway-related proteins were, respectively, analyzed by MPO ELISA kit and Western blot analysis. The cell apoptosis in lung tissues was observed by TUNEL assay. Acute ethanol intoxication (EtOH) decreased the survival rate of mice and exacerbated the lung injury caused by sepsis through increasing pulmonary vascular permeability, neutrophil infiltration, release of inflammatory factors, and cell apoptosis. In addition, EtOH could activate the SphK1/S1P/S1PR1 pathway in CLP mice. However, PF-543, as a specific inhibitor of SphK1, could partially reverse the deleterious effects on lung injury of CLP mice. PF-543 alleviated lung injury caused by sepsis in acute ethanol intoxication rats by suppressing the SphK1/S1P/S1PR1 signaling pathway.


Asunto(s)
Intoxicación Alcohólica/complicaciones , Inhibidores Enzimáticos/farmacología , Lesión Pulmonar/prevención & control , Pulmón/efectos de los fármacos , Lisofosfolípidos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Pirrolidinas/farmacología , Sepsis/tratamiento farmacológico , Receptores de Esfingosina-1-Fosfato/metabolismo , Esfingosina/análogos & derivados , Sulfonas/farmacología , Animales , Apoptosis/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Pulmón/enzimología , Pulmón/patología , Lesión Pulmonar/enzimología , Lesión Pulmonar/etiología , Lesión Pulmonar/patología , Masculino , Metanol/farmacología , Ratones Endogámicos C57BL , Infiltración Neutrófila/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Neumonía/enzimología , Neumonía/etiología , Neumonía/patología , Neumonía/prevención & control , Edema Pulmonar/enzimología , Edema Pulmonar/etiología , Edema Pulmonar/patología , Edema Pulmonar/prevención & control , Sepsis/complicaciones , Sepsis/enzimología , Transducción de Señal , Esfingosina/metabolismo
18.
Front Immunol ; 12: 625957, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33767697

RESUMEN

Endotoxin-induced lung injury is one of the major causes of death induced by endotoxemia, however, few effective therapeutic options exist. Hydrogen inhalation has recently been shown to be an effective treatment for inflammatory lung injury, but the underlying mechanism is unknown. In the current study we aim to investigate how hydrogen attenuates endotoxin-induced lung injury and provide reference values for the clinical application of hydrogen. LPS was used to establish an endotoxin-induced lung injury mouse model. The survival rate and pulmonary pathologic changes were evaluated. THP-1 and HUVECC cells were cultured in vitro. The thioredoxin 1 (Trx1) inhibitor was used to evaluate the anti-inflammatory effects of hydrogen. Hydrogen significantly improved the survival rate of mice, reduced pulmonary edema and hemorrhage, infiltration of neutrophils, and IL-6 secretion. Inhalation of hydrogen decreased tissue factor (TF) expression and MMP-9 activity, while Trx1 expression was increased in the lungs and serum of endotoxemia mice. LPS-stimulated THP-1 and HUVEC-C cells in vitro and showed that hydrogen decreases TF expression and MMP-9 activity, which were abolished by the Trx1 inhibitor, PX12. Hydrogen attenuates endotoxin-induced lung injury by decreasing TF expression and MMP-9 activity via activating Trx1. Targeting Trx1 by hydrogen may be a potential treatment for endotoxin-induced lung injury.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Antiinflamatorios/farmacología , Hidrógeno/farmacología , Pulmón/efectos de los fármacos , Tiorredoxinas/metabolismo , Tromboplastina/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Interleucina-6/metabolismo , Lipopolisacáridos , Pulmón/metabolismo , Pulmón/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos ICR , Infiltración Neutrófila/efectos de los fármacos , Edema Pulmonar/inducido químicamente , Edema Pulmonar/metabolismo , Edema Pulmonar/patología , Edema Pulmonar/prevención & control , Transducción de Señal , Células THP-1
20.
Int J Mol Med ; 47(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33760150

RESUMEN

Paraquat (PQ) causes serious oxidative stress and inflammatory responses, particularly to the lungs. Since lipoxin A4 (LXA4) functions as an anti­inflammatory mediator, the present study aimed to explore its effects on PQ­induced acute lung injury (ALI) and to elucidate the possible underlying mechanisms. PQ was administered to male SD rats and RAW264.7 cells to establish a model of poisoning, and LXA4 was used as an intervention drug. LXA4 treatment attenuated PQ­induced lung injury, and this was accompanied by decreased tumor necrosis factor (TNF)­α and interleukin (IL)­1ß secretion levels, and reduced oxidative stress damage. Additionally, LXA4 treatment inhibited the activation of the inflammation­related signaling molecules, Toll­like receptor 4 (TLR4), myeloid differentiation primary response 88 (MyD88), nuclear factor (NF)­κB p65, p­phosphoinositide 3­kinase (PI3K) and p­AKT. Furthermore, the in vitro experiments further confirmed that the beneficial effects of LXA4 on PQ­induced damage were TLR4­dependent. Hence, the present study demonstrated that LXA4 attenuated PQ­induced toxicity in lung tissue and RAW264.7 macrophages, and that this protective effect may be closely related to the mitigation of inflammatory responses, oxidative stress damage and the TLR4/MyD88­mediated activation of the PI3K/AKT/NF­κB pathway.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/prevención & control , Lipoxinas/farmacología , Edema Pulmonar/patología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/patología , Animales , Citocinas/metabolismo , Masculino , Malondialdehído/metabolismo , Ratones , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Paraquat/toxicidad , Fosfatidilinositol 3-Quinasas/metabolismo , Sustancias Protectoras/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Edema Pulmonar/inducido químicamente , Edema Pulmonar/prevención & control , Células RAW 264.7 , Ratas Sprague-Dawley , Superóxido Dismutasa/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...