Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.554
Filtrar
Más filtros

Intervalo de año de publicación
1.
Mem Inst Oswaldo Cruz ; 119: e240100, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39442103

RESUMEN

BACKGROUND: The Golden Syrian hamster (Mesocricetus auratus), Ferrets (Mustela putorius furo), and macaques have been described as useful laboratory animals naturally susceptible to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. OBJECTIVES: To study the mechanism of lung injury, we describe the histopathological features of SARS-CoV-2 infection in Golden Syrian hamsters inoculated intranasally with the A.2 Brazilian strain. METHODS: Hamsters were intranasally inoculated with the A.2 variant and euthanised at 3-, 5-, 10- and 15-days post-inoculation. The physical examination and body weight were recorded daily. Neutralising antibodies and viral RNA load of the respiratory tract were assessed during necropsies. FINDINGS: The coronavirus disease 2019 (COVID-19) model presented body weight loss, high levels of respiratory viral RNA load, severe segmentary pneumonitis, and bronchial fistula besides lymphatic trapping and infiltration, like the human SARS-COV-2 pathogenesis. The presence of subepithelial lymphoeosinophilic infiltrate was highlighted in our results; it contributed to the detachment of SARS-CoV-2 nucleocapsid-positive epithelial cells resulting in the infectious cell plugs. MAIN CONCLUSIONS: The SARS-CoV-2 caused segmentary pneumonia and vascular damage. In our comprehension, the infectious cell plugs, as being aspirated from the upper respiratory tract into the terminal bronchial lumen, work as a "Trojan horse", thus contributing to the dissemination of the SARS-CoV-2 infection into specific regions of the deep lung parenchyma.


Asunto(s)
COVID-19 , Modelos Animales de Enfermedad , Células Epiteliales , Mesocricetus , SARS-CoV-2 , Animales , COVID-19/patología , Células Epiteliales/virología , Lesión Pulmonar/virología , Lesión Pulmonar/patología , Carga Viral , Cricetinae , ARN Viral/análisis , Pulmón/patología , Pulmón/virología , Masculino , Brasil
2.
Nat Commun ; 15(1): 8672, 2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39375377

RESUMEN

Aberrant repair underlies the pathogenesis of pulmonary fibrosis while effective strategies to convert fibrosis to normal regeneration are scarce. Here, we found that thyroid hormone is decreased in multiple models of lung injury but is essential for lung regeneration. Moreover, thyroid hormone receptor α (TRα) promotes cell proliferation, while TRß fuels cell maturation in lung regeneration. Using a specific TRß agonist, sobetirome, we demonstrate that the anti-fibrotic effects of thyroid hormone mainly rely on TRß in mice. Cellularly, TRß activation enhances alveolar type-2 (AT2) cell differentiation into AT1 cell and constrains AT2 cell hyperplasia. Molecularly, TRß activation directly regulates the expression of KLF2 and CEBPA, both of which further synergistically drive the differentiation program of AT1 cells and benefit regeneration and anti-fibrosis. Our findings elucidate the modulation function of the TRß-KLF2/CEBPA axis on AT2 cell fate and provide a potential treatment strategy to facilitate lung regeneration and anti-fibrosis.


Asunto(s)
Diferenciación Celular , Factores de Transcripción de Tipo Kruppel , Pulmón , Fibrosis Pulmonar , Animales , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Diferenciación Celular/efectos de los fármacos , Ratones , Pulmón/patología , Pulmón/metabolismo , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/genética , Ratones Endogámicos C57BL , Regeneración , Masculino , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Modelos Animales de Enfermedad , Proteínas Potenciadoras de Unión a CCAAT
3.
Stem Cell Res Ther ; 15(1): 344, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39380037

RESUMEN

BACKGROUND: Lung injury and pulmonary fibrosis (PF), frequently arising as sequelae of severe and acute lung disease, currently face a dearth of effective therapeutic potions. Mesenchymal stem cells (MSCs) with immunomodulatory and tissue repair functions have immense potential to treat lung injury and PF. However, the optimal route of administration, timing, and frequency of dosing remain elusive. Human embryonic stem cell-derived immunity-and-matrix-regulatory cells (IMRCs) have shown therapeutic potential for lung injury and PF. METHODS: To ascertain the optimal therapeutic regimen for IMRCs in PF, we conducted an experimental study. Utilizing a mouse model of PF induced by bleomycin (BLM), IMRCs were administered via either a single or double intravenous (IV) or intratracheal (IT) injection on the first and seventh days post-BLM induction. RESULTS: Our findings revealed that IV infusion of IMRCs surpassed IT infusion in enhancing survival rates, facilitating body weight recovery, and optimizing Ashcroft and Szapiel scores among the model mice. Notably, IV administration exhibited a more profound ability to mitigate lung inflammation and fibrosis. Moreover, earlier and more frequent administrations of IMRCs were found to be advantageous in enhancing their therapeutic effects. Specifically, early administration with two IV infusions significantly improved body weight, lung organ coefficient, pulmonary ventilation and diffusion functions, and PF. This was accompanied by an increase in alveolar type I and II epithelial cells and a suppression of macrophage infiltration via CD24. CONCLUSION: Collectively, these results suggested that IMRCs infusion ameliorated lung injury by promoting lung regeneration and inhibiting macrophage infiltration in a route, time, and frequency-dependent manner.


Asunto(s)
Bleomicina , Células Madre Embrionarias Humanas , Lesión Pulmonar , Fibrosis Pulmonar , Animales , Ratones , Humanos , Células Madre Embrionarias Humanas/citología , Fibrosis Pulmonar/terapia , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/inducido químicamente , Lesión Pulmonar/terapia , Lesión Pulmonar/patología , Modelos Animales de Enfermedad , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL
4.
Physiol Rep ; 12(20): e70018, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39450926

RESUMEN

Parenchymal distribution of nebulized drug in healthy and diseased lungs has not, as evident from a literature review, been well characterized. We use a vibrating mesh nebulizer to deliver fluorescein solution in vivo to healthy or intratracheal-lipopolysaccharide (LPS)-instilled anesthetized rats in dorsal recumbency, or ex vivo to the lungs of LPS-instilled rats. Following in vivo nebulization (healthy/LPS-instilled), we quantify fluorescein intensity distribution by confocal microscopy in standard locations on the surface of freshly isolated lungs. Following LPS instillation (in vivo/ex vivo nebulization), we quantify fluorescein intensity in visibly injured locations. In standard locations, there is uniform, low-intensity basal fluorescein deposition. Focal regions receive high deposition that is, in upper (cranial), middle, and lower (caudal) locations, 6.4 ± 4.9, 3.3 ± 3.0, and 2.3 ± 2.8 times greater, respectively, than average basal intensity. Following LPS instillation, deposition in moderately injured regions can be high or low; deposition in severely injured regions is low. Further, actively phagocytic cells are observed in healthy and LPS-instilled lungs. And LPS particularly impairs mechanics and activates phagocytic cells in the male sex. We conclude that a low level of nebulized drug can be distributed across the parenchyma excepting to severely injured regions.


Asunto(s)
Lesión Pulmonar , Microscopía Confocal , Nebulizadores y Vaporizadores , Alveolos Pulmonares , Ratas Sprague-Dawley , Animales , Masculino , Microscopía Confocal/métodos , Ratas , Lesión Pulmonar/metabolismo , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Femenino , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/toxicidad , Fluoresceína/farmacocinética , Fluoresceína/administración & dosificación , Administración por Inhalación
5.
Nat Commun ; 15(1): 9169, 2024 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-39448571

RESUMEN

Cell plasticity, changes in cell fate, is crucial for tissue regeneration. In the lung, failure of regeneration leads to diseases, including fibrosis. However, the mechanisms governing alveolar cell plasticity during lung repair remain elusive. We previously showed that PCLAF remodels the DREAM complex, shifting the balance from cell quiescence towards cell proliferation. Here, we find that PCLAF expression is specific to proliferating lung progenitor cells, along with the DREAM target genes transactivated by lung injury. Genetic ablation of Pclaf impairs AT1 cell repopulation from AT2 cells, leading to lung fibrosis. Mechanistically, the PCLAF-DREAM complex transactivates CLIC4, triggering TGF-ß signaling activation, which promotes AT1 cell generation from AT2 cells. Furthermore, phenelzine that mimics the PCLAF-DREAM transcriptional signature increases AT2 cell plasticity, preventing lung fibrosis in organoids and mice. Our study reveals the unexpected role of the PCLAF-DREAM axis in promoting alveolar cell plasticity, beyond cell proliferation control, proposing a potential therapeutic avenue for lung fibrosis prevention.


Asunto(s)
Células Epiteliales Alveolares , Plasticidad de la Célula , Proliferación Celular , Pulmón , Regeneración , Animales , Ratones , Plasticidad de la Célula/genética , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/citología , Pulmón/patología , Pulmón/metabolismo , Canales de Cloruro/metabolismo , Canales de Cloruro/genética , Ratones Endogámicos C57BL , Factor de Crecimiento Transformador beta/metabolismo , Lesión Pulmonar/patología , Lesión Pulmonar/genética , Lesión Pulmonar/metabolismo , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/metabolismo , Transducción de Señal , Humanos , Ratones Noqueados , Masculino
6.
Stem Cell Res Ther ; 15(1): 295, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39256862

RESUMEN

BACKGROUND: Premature infants requiring mechanical ventilation and supplemental oxygen for respiratory support are at increased risk for bronchopulmonary dysplasia (BPD), wherein inflammation have been proposed as a driver of hyperoxia-induced injuries, including persistent loss of endothelial progenitor cells (EPCs), impaired vascularization and eventual alveolar simplification in BPD lungs. However, the underlying mechanisms linking these phenomena remain poorly defined. METHODS: We used clodronate liposomes to deplete macrophages in a mouse model of neonatal hyperoxia-induced lung injury to evaluate if EPC loss in BPD lungs could be an effect of macrophage infiltration. We further generated in vitro culture systems initiated with cord blood (CB)-derived CD34+ EPCs and neonatal macrophages either polarized from CB-derived monocytes or isolated from tracheal aspirates of human preterm infants requiring mechanical ventilation and oxygen supplementation, to identify EV-transmitted molecular mechanism that is critical for inhibitory actions of hyperoxic macrophages on EPCs. RESULTS: Initial experiments using mouse model identified the crucial role of macrophage infiltration in eliciting significant reduction of c-Kit+ EPCs in BPD lungs. Further examination of this concept in human system, we found that hyperoxia-exposed neonatal macrophages hamper human CD34+ EPC maintenance and impair endothelial function in the differentiated progeny via the EV transmission of miR-23a-3p. Notably, treatment with antagomiR-23a-3p to silence miR-23a-3p in vivo enhances c-Kit+ EPC maintenance, and increases capillary density, and consequently mitigates simplified alveolarization in BPD lungs. CONCLUSION: Our findings highlight the importance of pulmonary intercellular communication in the pathophysiology of BPD, by identifying a linkage through vesicle transfer of miR-23a-3p from hyperoxic macrophages to EPCs, and thus demonstrating potential for novel therapeutic target in BPD.


Asunto(s)
Células Progenitoras Endoteliales , Vesículas Extracelulares , Hiperoxia , Lesión Pulmonar , Macrófagos , MicroARNs , MicroARNs/genética , MicroARNs/metabolismo , Humanos , Animales , Células Progenitoras Endoteliales/metabolismo , Hiperoxia/metabolismo , Vesículas Extracelulares/metabolismo , Ratones , Macrófagos/metabolismo , Lesión Pulmonar/patología , Lesión Pulmonar/metabolismo , Recién Nacido , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patología , Displasia Broncopulmonar/genética , Animales Recién Nacidos , Modelos Animales de Enfermedad
7.
Mol Med ; 30(1): 140, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39251905

RESUMEN

BACKGROUND: Sepsis-induced pulmonary injury (SPI) is a common complication of sepsis with a high rate of mortality. N4-acetylcytidine (ac4C) is mediated by the ac4C "writer", N-acetyltransferase (NAT)10, to regulate the stabilization of mRNA. This study aimed to investigate the role of NAT10 in SPI and the underlying mechanism. METHODS: Twenty-three acute respiratory distress syndrome (ARDS) patients and 27 non-ARDS volunteers were recruited. A sepsis rat model was established. Reverse transcription-quantitative polymerase chain reaction was used to detect the expression of NAT10 and transferrin receptor (TFRC). Cell viability was detected by cell counting kit-8. The levels of Fe2+, glutathione, and malondialdehyde were assessed by commercial kits. Lipid reactive oxygen species production was measured by flow cytometric analysis. Western blot was used to detect ferroptosis-related protein levels. Haematoxylin & eosin staining was performed to observe the pulmonary pathological symptoms. RESULTS: The results showed that NAT10 was increased in ARDS patients and lipopolysaccharide-treated human lung microvascular endothelial cell line-5a (HULEC-5a) cells. NAT10 inhibition increased cell viability and decreased ferroptosis in HULEC-5a cells. TFRC was a downstream regulatory target of NAT10-mediated ac4C acetylation. Overexpression of TFRC decreased cell viability and promoted ferroptosis. In in vivo study, NAT10 inhibition alleviated SPI. CONCLUSION: NAT10-mediated ac4C acetylation of TFRC aggravated SPI through promoting ferroptosis.


Asunto(s)
Ferroptosis , Receptores de Transferrina , Sepsis , Sepsis/metabolismo , Sepsis/complicaciones , Sepsis/etiología , Acetilación , Animales , Humanos , Ratas , Masculino , Receptores de Transferrina/metabolismo , Receptores de Transferrina/genética , Femenino , Lesión Pulmonar/metabolismo , Lesión Pulmonar/etiología , Lesión Pulmonar/patología , Modelos Animales de Enfermedad , Acetiltransferasas/metabolismo , Acetiltransferasas/genética , Persona de Mediana Edad , Antígenos CD/metabolismo , Antígenos CD/genética , Citidina/análogos & derivados , Citidina/farmacología , Línea Celular , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/patología , Ratas Sprague-Dawley , Supervivencia Celular
8.
Tomography ; 10(9): 1342-1353, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39330747

RESUMEN

BACKGROUND: Radiation-induced lung injury (RILI), a serious side effect of thoracic radiotherapy, can lead to acute radiation pneumonitis (RP) and chronic pulmonary fibrosis (PF). Despite various interventions, no effective protocol exists to prevent pneumonitis. Oxytocin (OT), known for its anti-inflammatory, antiapoptotic, and antioxidant properties, has not been explored for its potential in mitigating RILI. MATERIALS AND METHODS: This study involved 24 female Wistar albino rats, divided into three groups: control group, radiation (RAD) + saline, and RAD + OT. The RAD groups received 18 Gy of whole-thorax irradiation. The RAD + OT group was treated with OT (0.1 mg/kg/day) intraperitoneally for 16 weeks. Computerizing tomography (CT) imaging and histopathological, biochemical, and blood gas analyses were performed to assess lung tissue damage and inflammation. RESULTS: Histopathological examination showed significant reduction in alveolar wall thickening, inflammation, and vascular changes in the RAD + OT group compared to the RAD + saline group. Biochemical analysis revealed decreased levels of TGF-beta, VEGF, and PDGF, and increased BMP-7 and prostacyclin in the RAD + oxytocin group (p < 0.05). Morphometric analysis indicated significant reductions in fibrosis, edema, and immune cell infiltration. CT imaging demonstrated near-normal lung parenchyma density in the RAD + oxytocin group (p < 0.001). CONCLUSION: Oxytocin administration significantly mitigates radiation-induced pneumonitis in rats, implying that is has potential as a therapeutic agent for preventing and treating RILI.


Asunto(s)
Oxitocina , Ratas Wistar , Animales , Oxitocina/farmacología , Oxitocina/uso terapéutico , Femenino , Ratas , Tomografía Computarizada por Rayos X/métodos , Pulmón/efectos de la radiación , Pulmón/patología , Pulmón/diagnóstico por imagen , Neumonitis por Radiación/patología , Neumonitis por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/diagnóstico por imagen , Lesión Pulmonar/etiología , Lesión Pulmonar/diagnóstico por imagen , Lesión Pulmonar/patología , Lesión Pulmonar/prevención & control , Protectores contra Radiación/farmacología , Protectores contra Radiación/uso terapéutico
9.
Artículo en Inglés | MEDLINE | ID: mdl-39326935

RESUMEN

Long-term exposure to fine particulate matter (PM2.5) can lead to chronic lung injury, including inflammation, idiopathic pulmonary fibrosis, and cancer. Mesenchymal cells, such as fibroblasts, myeloid-derived suppressor cells (MDSCs), and interstitial macrophages (IMs), contribute to immune regulation in lung, yet their diversity and functions upon long-term exposure to particulate matter (PM) remain inadequately characterized. In this study, we conducted a 16-week real-ambient PM exposure experiment on C57BL/6 J male mice in Shijiazhuang, China. We used single-cell RNA sequencing to analyze the cellular and molecular changes in lung tissues. Notably, we revealed a significant increase in specific fibroblast (ATX+, Col5a1+Meg3+, universal fibroblasts) and monocyte-derived cell subpopulations (monocytic-MDSCs (M-MDSCs), Lyve1loMHC-Ⅱhi IMs, Lyve1hiMHC-Ⅱlo IMs) that exhibited pro-inflammatory and pro-fibrotic functions. These cell subpopulations engaged in immunosuppressive signaling pathways and interactions with various cytokines, shaping a pulmonary microenvironment similar to those associated with cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs). This altered immune environment may promote the development of pulmonary fibrosis caused by PM exposure, underscoring the intricate roles of mesenchymal cells in chronic lung injury and highlighting the cancer-causing potential of PM2.5 exposure.


Asunto(s)
Fibroblastos , Lesión Pulmonar , Ratones Endogámicos C57BL , Monocitos , Material Particulado , Animales , Material Particulado/toxicidad , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Ratones , Masculino , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/metabolismo , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo
10.
Nat Commun ; 15(1): 8226, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39300060

RESUMEN

Hemolysis drives susceptibility to lung injury and predicts poor outcomes in diseases, such as malaria and sickle cell disease (SCD). However, the underlying pathological mechanism remains elusive. Here, we report that major facilitator superfamily domain containing 7 C (MFSD7C) protects the lung from hemolytic-induced damage by preventing ferroptosis. Mechanistically, MFSD7C deficiency in HuLEC-5A cells leads to mitochondrial dysfunction, lipid remodeling and dysregulation of ACSL4 and GPX4, thereby enhancing lipid peroxidation and promoting ferroptosis. Furthermore, systemic administration of MFSD7C mRNA-loaded nanoparticles effectively prevents lung injury in hemolytic mice, such as HbSS-Townes mice and PHZ-challenged 7 C-/- mice. These findings present the detailed link between hemolytic complications and ferroptosis, providing potential therapeutic targets for patients with hemolytic disorders.


Asunto(s)
Ferroptosis , Hemólisis , Ratones Noqueados , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Animales , Femenino , Humanos , Masculino , Ratones , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/genética , Coenzima A Ligasas/metabolismo , Coenzima A Ligasas/genética , Modelos Animales de Enfermedad , Ferroptosis/efectos de los fármacos , Ferroptosis/genética , Hemólisis/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Lesión Pulmonar/prevención & control , Lesión Pulmonar/genética , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Nanopartículas/química , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética
11.
Biomolecules ; 14(9)2024 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-39334901

RESUMEN

Fine particulate matter (PM2.5) is a significant cause of respiratory diseases and associated cellular damage. The mechanisms behind this damage have not been fully explained. This study investigated two types of cellular damage (inflammation and pyroptosis) induced by PM2.5, focusing on their relationship with two organelles (the endoplasmic reticulum and mitochondria). Animal models have demonstrated that PM2.5 induces excessive endoplasmic reticulum stress (ER stress), which is a significant cause of lung damage in rats. This was confirmed by pretreatment with an ER stress inhibitor (4-Phenylbutyric acid, 4-PBA). We found that, in vitro, the intracellular Ca2+ ([Ca2+]i) dysregulation induced by PM2.5 in rat alveolar macrophages was associated with ER stress. Changes in mitochondria-associated membranes (MAMs) result in abnormal mitochondrial function. This further induced the massive expression of NLRP3 and GSDMD-N, which was detrimental to cell survival. In conclusion, our findings provide valuable insights into the relationship between [Ca2+]i dysregulation, mitochondrial damage, inflammation and pyroptosis under PM2.5-induced ER stress conditions. Their interactions ultimately have an impact on respiratory health.


Asunto(s)
Calcio , Estrés del Retículo Endoplásmico , Retículo Endoplásmico , Lesión Pulmonar , Mitocondrias , Material Particulado , Fenilbutiratos , Animales , Fenilbutiratos/farmacología , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Calcio/metabolismo , Ratas , Estrés del Retículo Endoplásmico/efectos de los fármacos , Material Particulado/toxicidad , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Lesión Pulmonar/metabolismo , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Masculino , Piroptosis/efectos de los fármacos , Ratas Sprague-Dawley , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
12.
J Biochem Mol Toxicol ; 38(8): e23790, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39108137

RESUMEN

Pulmonary injury is one of the key restricting factors for the therapy of malignancies with chemotherapy or following radiotherapy for chest cancers. The lung is a sensitive organ to some severely toxic antitumor drugs, consisting of bleomycin and alkylating agents. Furthermore, treatment with radiotherapy may drive acute and late adverse impacts on the lung. The major consequences of radiotherapy and chemotherapy in the lung are pneumonitis and fibrosis. Pneumonitis may arise some months to a few years behind cancer therapy. However, fibrosis is a long-term effect that appears years after chemo/or radiotherapy. Several mechanisms such as oxidative stress and severe immune reactions are implicated in the progression of pulmonary fibrosis. Epithelial-mesenchymal transition (EMT) is offered as a pivotal mechanism for lung fibrosis behind chemotherapy and radiotherapy. It seems that pulmonary fibrosis is the main consequence of EMT after chemo/radiotherapy. Several biological processes, consisting of the liberation of pro-inflammatory and pro-fibrosis molecules, oxidative stress, upregulation of nuclear factor of κB and Akt, epigenetic changes, and some others, may participate in EMT and pulmonary fibrosis behind cancer therapy. In this review, we aim to discuss how chemotherapy or radiotherapy may promote EMT and lung fibrosis. Furthermore, we review potential targets and effective agents to suppress EMT and lung fibrosis after cancer therapy.


Asunto(s)
Quimioradioterapia , Transición Epitelial-Mesenquimal , Fibrosis Pulmonar , Humanos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/etiología , Quimioradioterapia/efectos adversos , Animales , Estrés Oxidativo/efectos de los fármacos , Lesión Pulmonar/etiología , Lesión Pulmonar/patología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo
13.
In Vivo ; 38(5): 2179-2189, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39187362

RESUMEN

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


Asunto(s)
Citocinas , Lesión Pulmonar , FN-kappa B , Transducción de Señal , Silibina , Receptor Toll-Like 4 , Animales , Silibina/farmacología , Ratones , FN-kappa B/metabolismo , Masculino , Transducción de Señal/efectos de los fármacos , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Lesión Pulmonar/etiología , Citocinas/metabolismo , Receptor Toll-Like 4/metabolismo , Modelos Animales de Enfermedad , Vanadio/farmacología , Ratones Endogámicos BALB C , Antiinflamatorios/farmacología , Silimarina/farmacología , Mediadores de Inflamación/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo
14.
Life Sci ; 355: 123000, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39168238

RESUMEN

BACKGROUND: Lung ischemia-reperfusion injury (LIRI) poses a significant challenge in various clinical scenarios. Despite extensive research on the pathogenesis and potential treatments of LIRI, there is a notable absence of bibliometric analysis. MATERIALS AND METHODS: We summarized the results of LIRI research through two searches on the Web of Science, covering data from 1979 to 2023 with topic words "lung" and "reperfusion injury". The collected data were analyzed and visualized based on country, author(s), and keywords by bibliometric software. The keyword "programmed cell death" was further added to explore the hotspot of the LIRI research field. RESULTS: The initial analysis of 1648 research articles showed a total of 40 countries and 7031 researchers were involved in the publications, with America being the most productive country in the research field of LIRI. Keyword analysis revealed that the evolving focus of LIRI research has progressively transitioned from, lung transplantation, primary graft dysfunction, inflammation, oxidative stress, and ex vivo lung perfusion to cell death. Subsequently, 212 publications specifically addressing programmed cell death (PCD) in LIRI were identified, which clarified the recent hotspot of the LIRI field. CONCLUSION: With closer international cooperation and increasing research scale, the LIRI research focused mainly on the pathogenesis and potential therapeutic interventions for LIRI. PCD in LIRI is becoming a trending topic and will continue to be a hotspot in this field. Our study may offer valuable guidance for future research endeavors concerning LIRI.


Asunto(s)
Bibliometría , Daño por Reperfusión , Daño por Reperfusión/patología , Humanos , Animales , Pulmón/patología , Investigación Biomédica/tendencias , Investigación Biomédica/métodos , Apoptosis , Lesión Pulmonar/patología
15.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167472, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39154794

RESUMEN

COVID-19, caused by SARS-CoV-2 infection, results in irreversible or fatal lung injury. We assumed that necroptosis of virus-infected alveolar epithelial cells (AEC) could promote local inflammation and further lung injury in COVID-19. Since CD8+ lymphocytes induced AEC cell death via cytotoxic molecules such as FAS ligands, we examined the involvement of FAS-mediated cell death in COVID-19 patients and murine COVID-19 model. We identified the occurrence of necroptosis and subsequent release of HMGB1 in the admitted patients with COVID-19. In the mouse model of COVID-19, lung inflammation and injury were attenuated in Fas-deficient mice compared to Fas-intact mice. The infection enhanced Type I interferon-inducible genes in both groups, while inflammasome-associated genes were specifically upregulated in Fas-intact mice. The treatment with necroptosis inhibitor, Nec1s, improved survival rate, lung injury, and systemic inflammation. SARS-CoV-2 induced necroptosis causes cytokine induction and lung damage, and its inhibition could be a novel therapeutic strategy for COVID-19.


Asunto(s)
Células Epiteliales Alveolares , COVID-19 , Necroptosis , SARS-CoV-2 , COVID-19/patología , COVID-19/inmunología , COVID-19/metabolismo , COVID-19/virología , COVID-19/complicaciones , Animales , Humanos , Ratones , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Células Epiteliales Alveolares/virología , Proteína HMGB1/metabolismo , Proteína HMGB1/genética , Lesión Pulmonar/patología , Lesión Pulmonar/virología , Lesión Pulmonar/inmunología , Lesión Pulmonar/metabolismo , Masculino , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos C57BL , Receptor fas/metabolismo , Receptor fas/genética , Ratones Noqueados , Neumonía/patología , Neumonía/virología , Neumonía/metabolismo , Neumonía/inmunología , Persona de Mediana Edad , Imidazoles , Indoles
16.
Redox Biol ; 75: 103296, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-39098263

RESUMEN

The lung macrophages play a crucial role in health and disease. Sexual dimorphism significantly impacts the phenotype and function of tissue-resident macrophages. The primary mechanisms responsible for sexually dimorphic outcomes in bronchopulmonary dysplasia (BPD) remain unidentified. We tested the hypothesis that biological sex plays a crucial role in the transcriptional state of alveolar macrophages, using neonatal murine hyperoxia-induced lung injury as a relevant model for human BPD. The effects of neonatal hyperoxia exposure (95 % FiO2, PND1-5: saccular stage) on the lung myeloid cells acutely after injury and during normoxic recovery were measured. Alveolar macrophages (AM) from room air- and hyperoxia exposed from male and female neonatal murine lungs were subjected to bulk-RNA Sequencing. AMs are significantly depleted in the hyperoxia-exposed lung acutely after injury, with subsequent recovery in both sexes. The transcriptome of the alveolar macrophages is impacted by neonatal hyperoxia exposure and by sex as a biological variable. Pathways related to DNA damage and interferon-signaling were positively enriched in female AMs. Metabolic pathways related to glucose and carbohydrate metabolism were positively enriched in the male AMs, while oxidative phosphorylation was negatively enriched. These pathways were shared with monocytes and airway macrophages from intubated male and female human premature neonates.


Asunto(s)
Animales Recién Nacidos , Hiperoxia , Macrófagos Alveolares , Femenino , Animales , Masculino , Macrófagos Alveolares/metabolismo , Ratones , Hiperoxia/metabolismo , Humanos , Transcriptoma , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patología , Displasia Broncopulmonar/etiología , Caracteres Sexuales , Factores Sexuales , Modelos Animales de Enfermedad , Recién Nacido , Pulmón/metabolismo , Pulmón/patología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Lesión Pulmonar/etiología
17.
Immun Inflamm Dis ; 12(8): e1343, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39092750

RESUMEN

The involvement of neutrophils in the lungs during the recovery phase of coronavirus disease 2019 (COVID-19) is not well defined mainly due to the limited accessibility of lung tissues from COVID-19 survivors. The lack of an appropriate small animal model has affected the development of effective therapeutic strategies. We here developed a long COVID mouse model to study changes in neutrophil phenotype and association with lung injury. Our data shows persistent neutrophil recruitment and neutrophil extracellular trap formation in the lungs for up to 30 days post-infection which correlates with lung fibrosis and inflammation.


Asunto(s)
COVID-19 , Modelos Animales de Enfermedad , Trampas Extracelulares , Pulmón , Neutrófilos , SARS-CoV-2 , Animales , Trampas Extracelulares/inmunología , COVID-19/inmunología , COVID-19/complicaciones , Ratones , Neutrófilos/inmunología , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Pulmón/patología , Pulmón/inmunología , Pulmón/virología , Lesión Pulmonar/inmunología , Lesión Pulmonar/virología , Lesión Pulmonar/patología , Lesión Pulmonar/etiología , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Infiltración Neutrófila/inmunología , Humanos , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/etiología
18.
JCI Insight ; 9(18)2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39106107

RESUMEN

Gas flow is fundamental for driving tidal ventilation and, thus, the speed of lung motion, but current bias flow settings to support the preterm lung after birth do not have an evidence base. We aimed to determine the role of gas bias flow rates to generate positive pressure ventilation in initiating early lung injury pathways in the preterm lamb. Using slower speeds to inflate the lung during tidal ventilation (gas flow rates 4-6 L/min) did not affect lung mechanics, mechanical power, or gas exchange compared with those currently used in clinical practice (8-10 L/min). Speed of pressure and volume change during inflation were faster with higher flow rates. Lower flow rates resulted in less bronchoalveolar fluid protein, better lung morphology, and fewer detached epithelial cells. Overall, relative to unventilated fetal controls, there was greater protein change using 8-10 L/min, which was associated with enrichment of acute inflammatory and innate responses. Slowing the speed of lung motion by supporting the preterm lung from birth with lower flow rates than in current clinical use resulted in less lung injury without compromising tidal ventilation or gas exchange.


Asunto(s)
Animales Recién Nacidos , Lesión Pulmonar , Pulmón , Animales , Ovinos , Pulmón/patología , Pulmón/fisiopatología , Lesión Pulmonar/fisiopatología , Lesión Pulmonar/patología , Femenino , Respiración con Presión Positiva/métodos , Respiración con Presión Positiva/efectos adversos , Nacimiento Prematuro/fisiopatología , Modelos Animales de Enfermedad , Intercambio Gaseoso Pulmonar/fisiología , Embarazo , Volumen de Ventilación Pulmonar
19.
Respir Res ; 25(1): 299, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39113018

RESUMEN

BACKGROUND: Although recent studies provide mechanistic understanding to the pathogenesis of radiation induced lung injury (RILI), rare therapeutics show definitive promise for treating this disease. Type II alveolar epithelial cells (AECII) injury in various manner results in an inflammation response to initiate RILI. RESULTS: Here, we reported that radiation (IR) up-regulated the TNKS1BP1, causing progressive accumulation of the cellular senescence by up-regulating EEF2 in AECII and lung tissue of RILI mice. Senescent AECII induced Senescence-Associated Secretory Phenotype (SASP), consequently activating fibroblasts and macrophages to promote RILI development. In response to IR, elevated TNKS1BP1 interacted with and decreased CNOT4 to suppress EEF2 degradation. Ectopic expression of EEF2 accelerated AECII senescence. Using a model system of TNKS1BP1 knockout (KO) mice, we demonstrated that TNKS1BP1 KO prevents IR-induced lung tissue senescence and RILI. CONCLUSIONS: Notably, this study suggested that a regulatory mechanism of the TNKS1BP1/CNOT4/EEF2 axis in AECII senescence may be a potential strategy for RILI.


Asunto(s)
Células Epiteliales Alveolares , Senescencia Celular , Ratones Endogámicos C57BL , Ratones Noqueados , Animales , Humanos , Masculino , Ratones , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/efectos de la radiación , Células Epiteliales Alveolares/patología , Células Cultivadas , Senescencia Celular/efectos de la radiación , Senescencia Celular/fisiología , Quinasa del Factor 2 de Elongación/metabolismo , Quinasa del Factor 2 de Elongación/genética , Lesión Pulmonar/metabolismo , Lesión Pulmonar/genética , Lesión Pulmonar/patología , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/genética , Proteína 1 de Unión a Repeticiones Teloméricas/genética , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo
20.
Immun Inflamm Dis ; 12(8): e70001, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39172009

RESUMEN

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


Asunto(s)
Curcuma , PPAR gamma , Paraquat , Pioglitazona , Extractos Vegetales , Ratas Wistar , Animales , Pioglitazona/farmacología , Pioglitazona/uso terapéutico , Paraquat/toxicidad , Masculino , Ratas , Curcuma/química , PPAR gamma/agonistas , PPAR gamma/metabolismo , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/prevención & control , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/patología , Lesión Pulmonar/metabolismo , Dexametasona/farmacología , Líquido del Lavado Bronquioalveolar/citología , Estrés Oxidativo/efectos de los fármacos , Tiazolidinedionas/farmacología , Tiazolidinedionas/uso terapéutico , Antioxidantes/farmacología , Curcumina/farmacología , Curcumina/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA