Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.070
Filtrar
1.
Allergol Immunopathol (Madr) ; 52(4): 53-59, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38970265

RESUMEN

BACKGROUND: Pulmonary fibrosis is a pathological hallmark of lung injury. It is an aggressive disease that replaces normal lung parenchyma by fibrotic tissue. The transforming growth factor-beta-mothers against decapentaplegic homolog 3 (TGF-ß1-Smad3) signaling pathway plays a key role in regulating lung fibrosis. Decorin (DCN), a small leucine-rich proteoglycan, has a modulatory effect on the immune system by reversibly binding with TGF-ß and reducing its bioavailability. Mesenchymal stem cell (MSC) therapy is a new strategy that has an immune-modulatory capacity. OBJECTIVE: The aim of this study was to introduce a new therapeutic approach to harness remodeling in injured lung. MATERIAL AND METHODS: Bone marrow MSCs were isolated and transduced by decorin gene. Lung injury was induced by bleomycin and mice were treated with MSCs, MSCs-decorin, and decorin. Then, oxidative stress biomarkers, remodeling biomarkers, bronchoalveolar lavage cells, and histopathology study were conducted. RESULTS: Reduced catalase and superoxide dismutase increased due to treatments. Elevated malondialdehyde, hydroxyproline, TGF-ß levels, and polymorphonuclear cells count decreased in the treated groups. Additionally, the histopathology of lung tissues showed controlled inflammation and fibrosis. CONCLUSION: Transfected decorin gene to MSCs and used cell therapy could control remodeling and bleomycin-induced lung injury.


Asunto(s)
Bleomicina , Decorina , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Fibrosis Pulmonar , Decorina/genética , Decorina/metabolismo , Animales , Ratones , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/terapia , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/terapia , Lesión Pulmonar/inmunología , Lesión Pulmonar/genética , Transducción Genética , Estrés Oxidativo , Células Cultivadas , Modelos Animales de Enfermedad , Masculino , Humanos
2.
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
3.
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
4.
Sci Total Environ ; 944: 173760, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-38857800

RESUMEN

Ferrate (Fe(VI)) is an environmentally friendly disinfectant that is widely used to eradicate microbes in reclaimed water. However, the potential health risks associated with inhalation of Fe(VI)-treated bacteria-laden reclaimed water remains uncertain. We aimed to explore the inhalation hazards and potential mechanisms of K2FeO4-treated Escherichia coli (E. coli, ATCC 25922). Our findings indicated that Fe(VI) disinfection induced a dose- and time-dependent E. coli inactivation, accompanied by a rapid release of the bacterial endotoxin, lipopolysaccharide (LPS). Scanning electron microscopy (SEM) observations indicate that Fe(VI)-induced endotoxin production consists of at least two stages: initial binding of endotoxin to bacteria and subsequent dissociation to release free endotoxin. Furthermore, Fe(VI) disinfection was not able to effectively eliminate pure or E. coli-derived endotoxins. The E. coli strain used in this study lacks lung infection capability, thus the inhalation of bacteria alone failed to induce severe lung injury. However, mice inhaled exposure to Fe(VI)-treated E. coli showed severe impairment of lung structure and function. Moreover, we observed an accumulation of neutrophil/macrophage recruitment, cell apoptosis, and ROS generation in the lung tissue of mice subjected to Fe(VI)-treated E. coli. RNA sequencing (RNA-seq) and PCR results revealed that genes involved with endotoxin stimuli, cell apoptosis, antioxidant defence, inflammation response, chemokines and their receptors were upregulated in response to Fe(VI)-treated E. coli. In conclusion, Fe(VI) is ineffective in eliminating endotoxins and can trigger secondary hazards owing to endotoxin release from inactivated bacteria. Aerosol exposure to Fe(VI)-treated E. coli causes considerable damage to lung tissue by inducing oxidative stress and inflammatory responses.


Asunto(s)
Endotoxinas , Escherichia coli , Inflamación , Lesión Pulmonar , Estrés Oxidativo , Escherichia coli/efectos de los fármacos , Ratones , Animales , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/microbiología , Hierro/metabolismo , Desinfección/métodos , Desinfectantes/toxicidad
5.
Anal Chem ; 96(26): 10488-10495, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38901019

RESUMEN

Hydrogen peroxide (H2O2) overexpressed in mitochondria has been regarded as a key biomarker in the pathological processes of various diseases. However, there is currently a lack of suitable mitochondria-targetable near-infrared (NIR) probes for the visualization of H2O2 in multiple diseases, such as PM2.5 exposure-induced lung injury, hepatic ischemia-reperfusion injury (HIRI), nonalcoholic fatty liver (NAFL), hepatic fibrosis (HF), and malignant tumor tissues containing clinical cancer patient samples. Herein, we conceived a novel NIR fluorescent probe (HCy-H2O2) by introducing pentafluorobenzenesulfonyl as a H2O2 sensing unit into the NIR hemicyanine platform. HCy-H2O2 exhibits good sensitivity and selectivity toward H2O2, accompanied by a remarkable "turn-on" fluorescence signal at 720 nm. Meanwhile, HCy-H2O2 has stable mitochondria-targetable ability and permits monitoring of the up-generated H2O2 level during mitophagy. Furthermore, using HCy-H2O2, we have successfully observed an overproduced mitochondrial H2O2 in ambient PM2.5 exposure-induced lung injury, HIRI, NAFL, and HF models through NIR fluorescence imaging. Significantly, the visualization of H2O2 has been achieved in both tumor-bear mice as well as surgical specimens of cancer patients, making HCy-H2O2 a promising tool for cancer diagnosis and imaging-guided surgery.


Asunto(s)
Colorantes Fluorescentes , Peróxido de Hidrógeno , Mitocondrias , Imagen Óptica , Colorantes Fluorescentes/química , Colorantes Fluorescentes/síntesis química , Peróxido de Hidrógeno/metabolismo , Animales , Mitocondrias/metabolismo , Mitocondrias/química , Ratones , Humanos , Lesión Pulmonar/diagnóstico por imagen , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Rayos Infrarrojos
7.
Disaster Med Public Health Prep ; 18: e86, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38706344

RESUMEN

Nuclear and chemical weapons of mass destruction share both a tragic and beneficial legacy in mankind's history and health. The horrific health effects of ionizing radiation and mustard gas exposures unleashed during disasters, wars, and conflicts have been harnessed to treat human health maladies. Both agents of destruction have been transformed into therapies to treat a wide range of cancers. The discovery of therapeutic uses of radiation and sulfur mustard was largely due to observations by clinicians treating victims of radiation and sulfur mustard gas exposures. Clinicians identified vulnerability of leukocytes to these agents and repurposed their use in the treatment of leukemias and lymphomas. Given the overlap in therapeutic modalities, it goes to reason that there may be common mechanisms to target as protective strategies against their damaging effects. This commentary will highlight oxidative stress as a common mechanism shared by both radiation and sulfur mustard gas exposures and discuss potential therapies targeting oxidative stress as medical countermeasures against the devastating lung diseases wrought by these agents.


Asunto(s)
Lesión Pulmonar , Gas Mostaza , Estrés Oxidativo , Humanos , Estrés Oxidativo/efectos de los fármacos , Lesión Pulmonar/inducido químicamente , Sustancias para la Guerra Química
8.
Int Immunopharmacol ; 134: 112165, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38692017

RESUMEN

Particulate matter (PM) is considered the fundamental component of atmospheric pollutants and is associated with the pathogenesis of many respiratory diseases. Fibroblast growth factor 10 (FGF10) mediates mesenchymal-epithelial signaling and has been linked with the repair process of PM-induced lung injury (PMLI). However, the pathogenic mechanism of PMLI and the specific FGF10 protective mechanism against this injury are still undetermined. PM was administered in vivo into murine airways or in vitro to human bronchial epithelial cells (HBECs), and the inflammatory response and ferroptosis-related proteins SLC7A11 and GPX4 were assessed. The present research investigates the FGF10-mediated regulation of ferroptosis in PMLI mice models in vivo and HBECs in vitro. The results showed that FGF10 pretreatment reduced PM-mediated oxidative damage and ferroptosis in vivo and in vitro. Furthermore, FGF10 pretreatment led to reduced oxidative stress, decreased secretion of inflammatory mediators, and activation of the Nrf2-dependent antioxidant signaling. Additionally, silencing of Nrf2 using siRNA in the context of FGF10 treatment attenuated the effect on ferroptosis. Altogether, both in vivo and in vitro assessments confirmed that FGF10 protects against PMLI by inhibiting ferroptosis via the Nrf2 signaling. Thus, FGF10 can be used as a novel ferroptosis suppressor and a potential treatment target in PMLI.


Asunto(s)
Ferroptosis , Factor 10 de Crecimiento de Fibroblastos , Lesión Pulmonar , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Material Particulado , Transducción de Señal , Ferroptosis/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Material Particulado/toxicidad , Humanos , Transducción de Señal/efectos de los fármacos , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factor 10 de Crecimiento de Fibroblastos/genética , Ratones , Estrés Oxidativo/efectos de los fármacos , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Lesión Pulmonar/prevención & control , Masculino , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Línea Celular , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Modelos Animales de Enfermedad , Sistema de Transporte de Aminoácidos y+
10.
Poult Sci ; 103(7): 103860, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38795514

RESUMEN

A large amount of hydrogen sulfide (H2S) is produced in the process of chicken breeding, which can cause serious inflammation and oxidative damage to the respiratory system of chickens. Tea tree oil (TTO) has antioxidant and anti-inflammatory properties. No studies have been reported on the use of TTO in H2S-induced lung injury in chickens. Therefore, in this study, 240 one-day-old Roman pink laying hens were randomly and equally divided into 3 groups: control group (CON), H2S exposure group (AVG, containing H2S), and TTO treatment group (TTG, containing H2S and 0.02 mL/L TTO) to establish an experimental model of TTO treatment with H2S exposure for a period of 42 d. Hematoxylin and eosin (H&E) staining was used to detect lung histopathology. Gene expression profiles were analyzed using transcriptomics. The underlying mechanism of the amelioration of lung injury by TTO was further revealed by antioxidant enzyme assays and qRT-PCR. The results showed that H2S exposure induced significant gene expression of CYP450s (CYP1B1 and CYP1C1) (P < 0.05), and caused intense oxidative stress, apoptosis and inflammation compared with CON. TTO could reduce ROS production and enhance antioxidant capacity (SOD, CAT, T-AOC, and GSH-PX) by regulating the CYP450s/ROS pathway (P < 0.05). Compared with the control group, the treatment group showed significantly decreased expression of apoptotic (Caspase-8, Caspase-3, Bid and Fas) (P < 0.05) and inflammatory (IL-4, IL-16, NF-κB, TNF-α and IFN-γ) (P < 0.05) factors in the lung. This study revealed that TTO regulated CYP450s/ROS pathway to alleviate H2S-induced lung injury in chickens. These results enrich the theory of the action mechanism of TTO on H2S-exposed chicken lungs and are of great value for the treatment of H2S-exposed animals.


Asunto(s)
Pollos , Sistema Enzimático del Citocromo P-450 , Sulfuro de Hidrógeno , Pulmón , Estrés Oxidativo , Aceite de Árbol de Té , Animales , Sulfuro de Hidrógeno/metabolismo , Estrés Oxidativo/efectos de los fármacos , Aceite de Árbol de Té/farmacología , Aceite de Árbol de Té/administración & dosificación , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Sistema Enzimático del Citocromo P-450/metabolismo , Sistema Enzimático del Citocromo P-450/genética , Femenino , Especies Reactivas de Oxígeno/metabolismo , Enfermedades de las Aves de Corral/inducido químicamente , Antioxidantes/metabolismo , Antioxidantes/farmacología , Proteínas Aviares/metabolismo , Proteínas Aviares/genética , Distribución Aleatoria , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/veterinaria , Lesión Pulmonar/tratamiento farmacológico
11.
Sci Total Environ ; 931: 172910, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38701926

RESUMEN

Significant impairment of pulmonary function has been demonstrated through long-term exposure to neonicotinoid insecticides, such as imidacloprid (IMI). However, the underlying mechanisms of lung injury induced by IMI remain unclear. In this study, a mouse model of IMI-induced pulmonary injury was established, and the toxicity and lung damage were assessed through mouse body weight, organ index, hematological parameters, and histopathological analysis of lung tissues. Furthermore, metabolomics and transcriptomics techniques were employed to explore the mechanistic aspects. Results from the toxicity assessments indicated that mouse body weight was significantly reduced by IMI, organ index was disturbed, and hematological parameters were disrupted, resulting in pulmonary injury. The mechanistic experimental results indicate that the differences in metabolites and gene expression in mouse lungs could be altered by IMI. Validation of the results through combined analysis of metabolomics and transcriptomics revealed that the mechanism by which IMI induces lung injury in mice might be associated with the activation of the TLR4 receptor, thereby activating the PI3K/AKT/NF-κB signaling pathway to induce inflammation in mouse lungs. This study provided valuable insights into the mechanisms underlying IMI-induced pulmonary damage, potentially contributing to the development of safer pest control strategies. The knowledge gained served as a robust scientific foundation for the prevention and treatment of IMI-related pulmonary injuries.


Asunto(s)
Insecticidas , Lesión Pulmonar , FN-kappa B , Neonicotinoides , Nitrocompuestos , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Receptor Toll-Like 4 , Animales , Neonicotinoides/toxicidad , Nitrocompuestos/toxicidad , Ratones , Lesión Pulmonar/inducido químicamente , Transducción de Señal/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Insecticidas/toxicidad , Receptor Toll-Like 4/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología
12.
Sci Rep ; 14(1): 11637, 2024 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773158

RESUMEN

Ricin, an extremely potent toxin produced from the seeds of castor plant, Ricinus communis, is ribosome-inactivating protein that blocks cell-protein synthesis. It is considered a biological threat due to worldwide availability of castor beans, massive quantities as a by-product of castor oil production, high stability and ease of production. The consequence of exposure to lethal dose of ricin was extensively described in various animal models. However, it is assumed that in case of aerosolized ricin bioterror attack, the majority of individuals would be exposed to sublethal doses rather than to lethal ones. Therefore, the purpose of current study was to assess short- and long-term effects on physiological parameters and function following sublethal pulmonary exposure. We show that in the short-term, sublethal exposure of mice to ricin resulted in acute lung injury, including interstitial pneumonia, cytokine storm, neutrophil influx, edema and cellular death. This damage was manifested in reduced lung performance and physiological function. Interestingly, although in the long-term, mice recovered from acute lung damage and restored pulmonary and physiological functionality, the reparative process was associated with lasting fibrotic lesions. Therefore, restriction of short-term acute phase of the disease and management of long-term pulmonary fibrosis by medical countermeasures is expected to facilitate the quality of life of exposed survivors.


Asunto(s)
Ricina , Animales , Ricina/toxicidad , Ratones , Pulmón/efectos de los fármacos , Pulmón/patología , Citocinas/metabolismo , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Femenino , Modelos Animales de Enfermedad
13.
Stem Cell Res Ther ; 15(1): 147, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38773627

RESUMEN

BACKGROUND: Bleomycin (BLM)-induced lung injury is characterized by mixed histopathologic changes with inflammation and fibrosis, such as observed in human patients with bronchopulmonary dysplasia, idiopathic pulmonary fibrosis, and chronic obstructive pulmonary disease. Although no curative therapies for these lung diseases exist, stem cell therapy has emerged as a potential therapeutic option. Multilineage-differentiating stress-enduring (Muse) cells are endogenous pluripotent- and macrophage-like stem cells distributed in various adult and fetal tissues as stage-specific embryonic antigen-3-positive cells. They selectively home to damaged tissue by sensing sphingosine-1-phosphate and replace the damaged/apoptotic cells by in vivo differentiation. Clinical trials for some human diseases suggest the safety and therapeutic efficacy of intravenously injected human leukocyte antigen-mismatched allogenic Muse cells from adult bone marrow (BM) without immunosuppressant. Here, we evaluated the therapeutic effects of human Muse cells from preterm and term umbilical cord (UC), and adult BM in a rat BLM-induced lung injury model. METHODS: Rats were endotracheally administered BLM to induce lung injury on day 0. On day 3, human preterm UC-Muse, term UC-Muse, or adult BM-Muse cells were administered intravenously without immunosuppressants, and rats were subjected to histopathologic analysis on day 21. Body weight, serum surfactant protein D (SP-D) levels, and oxygen saturation (SpO2) were monitored. Histopathologic lung injury scoring by the Ashcroft and modified American Thoracic Society document scales, quantitative characterization of engrafted Muse cells, RNA sequencing analysis, and in vitro migration assay of infused Muse cells were performed. RESULTS: Rats administered preterm- and term-UC-Muse cells exhibited a significantly better recovery based on weight loss, serum SP-D levels, SpO2, and histopathologic lung injury scores, and a significantly higher rate of both Muse cell homing to the lung and alveolar marker expression (podoplanin and prosurfactant protein-C) than rats administered BM-Muse cells. Rats receiving preterm-UC-Muse cells showed statistically superior results to those receiving term-UC-Muse cells in many of the measures. These findings are thought to be due to higher expression of genes related to cell migration, lung differentiation, and cell adhesion. CONCLUSION: Preterm UC-Muse cells deliver more efficient therapeutic effects than term UC- and BM-Muse cells for treating BLM-induced lung injury in a rat model.


Asunto(s)
Bleomicina , Modelos Animales de Enfermedad , Lesión Pulmonar , Cordón Umbilical , Animales , Humanos , Ratas , Lesión Pulmonar/terapia , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Cordón Umbilical/citología , Ratas Sprague-Dawley , Masculino , Diferenciación Celular , Femenino
14.
Am J Respir Cell Mol Biol ; 71(1): 23-29, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38593005

RESUMEN

Investigations into the mechanisms of injury and repair in fibroproliferative disease require consideration of the spatial heterogeneity inherent in the disease. Most scoring of fibrotic remodeling in preclinical animal models relies on the modified Ashcroft score, which is an ordinal rubric of macroscopic resolution. The obvious limitations of manual histopathologic scoring have generated an unmet need for unbiased, repeatable scoring of fibroproliferative burden in tissue. Using computer vision approaches on immunofluorescence imaging of the extracellular matrix component laminin, we generated a robust and repeatable quantitative remodeling scorer. In the bleomycin lung injury model, the quantitative remodeling scorer shows significant agreement with the modified Ashcroft scale. This antibody-based approach is easily integrated into larger multiplex immunofluorescence experiments, which we demonstrate by testing the spatial apposition of tertiary lymphoid structures to fibroproliferative tissue, a poorly characterized phenomenon observed in both human interstitial lung diseases and preclinical models of lung fibrosis. The tool reported in this article is available as a stand-alone application that is usable without programming knowledge.


Asunto(s)
Bleomicina , Laminina , Fibrosis Pulmonar , Laminina/metabolismo , Animales , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/inducido químicamente , Pulmón/patología , Pulmón/metabolismo , Ratones , Lesión Pulmonar/patología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/inducido químicamente , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Estructuras Linfoides Terciarias/patología , Estructuras Linfoides Terciarias/inmunología , Humanos , Técnica del Anticuerpo Fluorescente , Matriz Extracelular/metabolismo , Matriz Extracelular/patología
15.
Environ Sci Pollut Res Int ; 31(22): 33098-33106, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38676862

RESUMEN

A number of biocidal disinfectant chemicals are used as household products to prevent spread of pathogens. People are commonly exposed to multiple chemicals through those disinfectants. However, effects of interactions (e.g., synergism) between disinfectants on human health outcomes have been rarely studied. In this study, we aimed to investigate associations of a mixture of chloromethylisothiazolinone/methylisothiazolinone (CMIT/MIT) and polyhexamethylene guanidine (PHMG), which had been used as humidifier disinfectants (HDs) in South Korea, with HD-associated lung injury (HDLI) in a Korean population (n = 4058) with HD exposure through use of HD products. Exposure to HD was retrospectively assessed by an interview-based standardized survey, and HDLI was determined by clinical assessment. After adjusting for covariates, PHMG-specific exposure indices (e.g., amount of use, indoor air concentration, and weekly exposure level) were dose-dependently associated with HDLI (their odds ratios for the comparison of third tertile versus first tertile were 1.95, 1.77, and 2.16, respectively). CMIT/MIT exposure was not observed to have a significant association with HDLI in a single chemical exposure model; however, associations between PHMG exposure and HDLI were strengthened by co-exposure to CMIT/MIT in combined chemical exposure models, where synergistic interactions between CMIT/MIT use and PHMG indices (amount of use and weekly exposure level) were observed (p-interaction in additive scale: 0.02 and 0.03, respectively). Our findings imply that adverse effects of PHMG exposure on lung injury among HD users might be worsened by co-exposure to CMIT/MIT. Given that plenty of household products contain disinfectants on global markets, epidemiological and toxicological investigations are warranted on interaction effects of co-exposure to disinfectants.


Asunto(s)
Desinfectantes , Guanidinas , Humidificadores , Lesión Pulmonar , Humanos , Lesión Pulmonar/inducido químicamente , República de Corea , Masculino , Femenino , Tiazoles , Adulto , Persona de Mediana Edad
16.
Environ Int ; 187: 108700, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38678936

RESUMEN

The significant correlation between particulate matter with aerodynamic diameters of ≤ 2.5 µm (PM2.5) and the high morbidity and mortality of respiratory diseases has become the consensus of the research. Epidemiological studies have clearly pointed out that there is no safe concentration of PM2.5, and mechanism studies have also shown that exposure to PM2.5 will first cause pulmonary inflammation. Therefore, the purpose of this study is to explore the mechanism of early lung injury induced by low-level PM2.5 from the perspective of epigenetics. Based on the previous results of population samples, combined with an in vitro/vivo exposure model of PM2.5, it was found that low-level PM2.5 promoted the transport of circ_0092363 from intracellular to extracellular spaces. The decreased expression of intracellular circ_0092363 resulted in reduced absorption of miR-31-5p, leading to inhibition of Rho associated coiled-coil containing protein kinase 1 (ROCK1) and the subsequent abnormal expression of tight junction proteins such as Zonula occludens protein 1 (ZO-1) and Claudin-1, ultimately inducing the occurrence of early pulmonary injury. Furthermore, this study innovatively introduced organoid technology and conducted a preliminary exploration for a study of the relationship among environmental exposure genomics, epigenetics and disease genomics in organoids. The role of circ_0092363 in early pulmonary injury induced by low-level PM2.5 was elucidated, and its value as a potential diagnostic biomarker was confirmed.


Asunto(s)
Lesión Pulmonar , Material Particulado , Lesión Pulmonar/inducido químicamente , Humanos , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo , Animales , MicroARNs/genética , Contaminantes Atmosféricos/toxicidad , Exposición a Riesgos Ambientales/efectos adversos
17.
Eur J Pharmacol ; 974: 176612, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38677537

RESUMEN

One of the main pathological features of chronic obstructive pulmonary disease (COPD) is the loss of functional alveolar tissue as a consequence of impaired regenerative capacities (emphysema). Recent research suggests that the secretome from mesenchymal cells, particularly extracellular vesicles (EVs), may possess regenerative properties beneficial for lung repair. However, the regenerative potential of the soluble factors (SFs) within the secretome remains largely unexplored in COPD. To this extent, we purified EVs and SFs secreted by lung fibroblasts to generate EV-enriched and SF-enriched fractions, and evaluated their effects on elastase-induced lung injury in both precision-cut lung slices (PCLS) and a mouse model. EV- and SF-enriched fractions were concentrated and purified from the conditioned medium of cultured MRC-5 lung fibroblasts using a combination of ultrafiltration and size exclusion chromatography, and were subsequently characterized according to the MISEV guidelines. Treatment with EV- or SF-enriched concentrates prevented and improved elastase-induced emphysema in PCLS, leading to reduced lung injury and upregulated markers of alveolar epithelial cells (aquaporin 5 and surfactant protein C), indicating potential parenchymal regeneration. Accordingly, prophylactic intratracheal treatment with lung fibroblast-derived EV- and SF-enriched concentrates in vivo attenuated elastase-induced lung tissue destruction, improved lung function, and enhanced gene expression of alveolar epithelial cell markers. Here, alveolar repair not only serves the purpose of facilitating gas exchange, but also by reinstating the essential parenchymal tethering required for optimal airway mechanics. In conclusion, this study highlights the therapeutic potential of both lung fibroblast-derived EV- and SF-enriched concentrates for the treatment of lung injury and emphysema.


Asunto(s)
Vesículas Extracelulares , Fibroblastos , Pulmón , Elastasa Pancreática , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/trasplante , Animales , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Pulmón/patología , Pulmón/efectos de los fármacos , Ratones , Humanos , Lesión Pulmonar/patología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Línea Celular , Masculino , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Solubilidad
18.
Am J Physiol Cell Physiol ; 326(6): C1637-C1647, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38646782

RESUMEN

Bleomycin (BLM)-induced lung injury in mice is a valuable model for investigating the molecular mechanisms that drive inflammation and fibrosis and for evaluating potential therapeutic approaches to treat the disease. Given high variability in the BLM model, it is critical to accurately phenotype the animals in the course of an experiment. In the present study, we aimed to demonstrate the utility of microscopic computed tomography (µCT) imaging combined with an artificial intelligence (AI)-convolutional neural network (CNN)-powered lung segmentation for rapid phenotyping of BLM mice. µCT was performed in freely breathing C57BL/6J mice under isoflurane anesthesia on days 7 and 21 after BLM administration. Terminal invasive lung function measurement and histological assessment of the left lung collagen content were conducted as well. µCT image analysis demonstrated gradual and time-dependent development of lung injury as evident by alterations in the lung density, air-to-tissue volume ratio, and lung aeration in mice treated with BLM. The right and left lung were unequally affected. µCT-derived parameters such as lung density, air-to-tissue volume ratio, and nonaerated lung volume correlated well with the invasive lung function measurement and left lung collagen content. Our study demonstrates the utility of AI-CNN-powered µCT image analysis for rapid and accurate phenotyping of BLM mice in the course of disease development and progression.NEW & NOTEWORTHY Microscopic computed tomography (µCT) imaging combined with an artificial intelligence (AI)-convolutional neural network (CNN)-powered lung segmentation is a rapid and powerful tool for noninvasive phenotyping of bleomycin mice over the course of the disease. This, in turn, allows earlier and more reliable identification of therapeutic effects of new drug candidates, ultimately leading to the reduction of unnecessary procedures in animals in pharmacological research.


Asunto(s)
Bleomicina , Lesión Pulmonar , Pulmón , Ratones Endogámicos C57BL , Redes Neurales de la Computación , Fenotipo , Animales , Bleomicina/toxicidad , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/diagnóstico por imagen , Lesión Pulmonar/patología , Lesión Pulmonar/metabolismo , Pulmón/diagnóstico por imagen , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo , Ratones , Microtomografía por Rayos X/métodos , Modelos Animales de Enfermedad , Inteligencia Artificial , Masculino , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/diagnóstico por imagen , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/metabolismo , Colágeno/metabolismo
19.
Ecotoxicol Environ Saf ; 277: 116330, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38636406

RESUMEN

PIWI-interacting RNAs (piRNAs) is an emerging class of small non-coding RNAs that has been recently reported to have functions in infertility, tumorigenesis, and multiple diseases in humans. Previously, 5 toxicity pathways were proposed from hundreds of toxicological studies that underlie BaP-induced lung injuries, and a "Bottom-up" approach was established to identify small non-coding RNAs that drive BaP-induced pulmonary effects by investigating the activation of these pathways in vitro, and the expression of the candidate microRNAs were validated in tissues of patients with lung diseases from publications. Here in this study, we employed the "Bottom-up" approach to identifying the roles of piRNAs and further validated the mechanisms in vivo using mouse acute lung injury model. Specifically, by non-coding RNA profiling in in vitro BaP exposure, a total of 3 suppressed piRNAs that regulate 5 toxicity pathways were proposed, including piR-004153 targeting CYP1A1, FGFR1, ITGA5, IL6R, NGRF, and SDHA, piR-020326 targeting CDK6, and piR-020388 targeting RASD1. Animal experiments demonstrated that tail vein injection of respective formulated agomir-piRNAs prior to BaP exposure could all alleviate acute lung injury that was shown by histopathological and biochemical evidences. Immunohistochemical evaluation focusing on NF-kB and Bcl-2 levels showed that exogenous piRNAs protect against BaP-induced inflammation and apoptosis, which further support that the inhibition of the 3 piRNAs had an important impact on BaP-induced lung injuries. This mechanism-driven, endpoint-supported result once again confirmed the plausibility and efficiency of the approach integrating in silico, in vitro, and in vivo evidences for the purpose of identifying key molecules.


Asunto(s)
Benzo(a)pireno , ARN Interferente Pequeño , Animales , Ratones , Benzo(a)pireno/toxicidad , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/patología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Masculino , Ratones Endogámicos C57BL , Humanos , ARN de Interacción con Piwi
20.
Ecotoxicol Environ Saf ; 277: 116364, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38657461

RESUMEN

The purpose of this study was to investigate the effect of Treg/Th1 imbalance in cadmium-induced lung injury and the potential protective effect of astilbin against cadmium-induced lung injury in chicken. Cadmium exposure significantly decreased T-AOC and GSH-Px levels and SOD activity in the chicken lung tissues. In contrast, it significantly increased the MDA and NO levels. These results indicate that cadmium triggers oxidative stress in lungs. Histopathological analysis revealed that cadmium exposure further induced infiltration of lymphocytes in the chicken lungs, indicating that cadmium causes pulmonary damage. Further analysis revealed that cadmium decreased the expression of IL-4 and IL-10 but increased those of IL-17, Foxp3, TNF-α, and TGF-ß, indicating that the exposure of cadmium induced the imbalance of Treg/Th1. Moreover, cadmium adversely affected chicken lung function by activating the NF-kB pathway and inducing expression of genes downstream to these pathways (COX-2, iNOS), associated with inflammatory injury in the lung tissue. Astilbin reduced cadmium-induced oxidative stress and inflammation in the lungs by increasing antioxidant enzyme activities and restoring Treg/Th1 balance. In conclusion, our results suggest that astilbin treatment alleviated the effects of cadmium-mediated lung injury in chickens by restoring the Treg/Th1 balance.


Asunto(s)
Cadmio , Pollos , Flavonoles , Lesión Pulmonar , Pulmón , Estrés Oxidativo , Transducción de Señal , Linfocitos T Reguladores , Animales , Cadmio/toxicidad , Estrés Oxidativo/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/patología , Transducción de Señal/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Flavonoles/farmacología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA