Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 914
Filtrer
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167358, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39025374

RÉSUMÉ

Radiation-induced pulmonary fibrosis (RIPF) is a frequently encountered late complication in patients undergoing radiation therapy, presenting a substantial risk to patient mortality and quality of life. The pathogenesis of RIPF remains unclear, and current treatment options are limited in efficacy. High-dose vitamin C has demonstrated potential when used in conjunction with other adjuvant therapies due to potent anticancer properties. However, the potential relationship between high-dose vitamin C and RIPF has not yet been explored in existing literature. In our study, the RIPF model and the LLC tumor model were used as two animal models to explore how high-dose vitamin C can improve RIPF without hampering the antitumour efficacy of radiotherapy. The impact of high-dose vitamin C on RIPF was assessed through various assays, including micro-CT, HE staining, Masson staining, and immunohistochemistry. Our results indicated that administering high-dose vitamin C 2 days before radiation and continuing for a duration of 6 weeks significantly inhibited the progression of RIPF. In order to explore the mechanism by which high-dose vitamin C attenuates RIPF, we utilized RNA-seq analysis of mouse lung tissue in conjunction with publicly available databases. Our findings indicated that high-dose vitamin C inhibits the differentiation of fibroblasts into myofibroblasts by targeting S100A8 and S100A9 derived from neutrophils. Additionally, the combination of high-dose vitamin C and radiation demonstrated enhanced inhibition of tumor growth in a murine LLC tumor model. These results revealed that the combination of radiotherapy and high-dose vitamin C may offer a promising therapeutic approach for the clinical management of thoracic tumors and the prevention of RIPF.


Sujet(s)
Acide ascorbique , Calgranuline A , Calgranuline B , Fibrose pulmonaire , Animaux , Acide ascorbique/pharmacologie , Acide ascorbique/usage thérapeutique , Acide ascorbique/administration et posologie , Souris , Calgranuline A/métabolisme , Calgranuline A/génétique , Fibrose pulmonaire/prévention et contrôle , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/étiologie , Fibrose pulmonaire/traitement médicamenteux , Calgranuline B/métabolisme , Calgranuline B/génétique , Souris de lignée C57BL , Modèles animaux de maladie humaine , Humains , Mâle
2.
Eur J Pharmacol ; 980: 176843, 2024 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-39068977

RÉSUMÉ

PURPOSE: Idiopathic pulmonary fibrosis (IPF) is a fatal progressive condition often requiring lung transplantation. Accelerated senescence of type II alveolar epithelial cells (AECII) plays a crucial role in pulmonary fibrosis progression through the secretion of the senescence-associated secretory phenotype (SASP). Low-dose carbon monoxide (CO) possesses anti-inflammatory, anti-oxidative, and anti-aging properties. This study aims to explore the preventive effects of CO-releasing molecule 2 (CORM2) in a bleomycin-induced pulmonary fibrosis model. METHODS: We established an pulmonary fibrosis model in C57BL/6J mice and evaluated the impact of CORM2 on fibrosis pathology using Masson's trichrome staining, fluorescence staining, and pulmonary function tests. Fibrogenic marker expression and SASP secretion in tissues and AECII cells were analyzed using qRT-PCR, Western blot, and ELISA assays both in vivo and in vitro. Additionally, we investigated DNA damage and cellular senescence through immunofluorescence and SA-ß-gal staining. RESULTS: CORM2 showed a preventive effect on bleomycin-induced lung fibrosis by improving pulmonary function and reducing the expression of fibrosis-related genes, such as TGF-ß, α-SMA, Collagen I/III. CORM2 decreased the DNA damage response by inhibiting γ-H2AX, p53, and p21. We identified PAI-1 as a new target gene that was downregulated by CORM2, and which was associated with cellular senescence and fibrosis. CORM2 effectively inhibited cellular senescence and delayed EMT occurrence in AECII cells. CONCLUSION: Our study highlights the potential of CORM2 in preventing DNA damage-induced cellular senescence in bleomycin-induced pulmonary fibrosis through modulation of the p53/PAI-1 signaling pathway. These findings underscore the promising prospects of CORM2 in targeting cellular senescence and the p53/PAI-1 pathway as a potential preventive strategy for IPF.


Sujet(s)
Bléomycine , Monoxyde de carbone , Vieillissement de la cellule , Souris de lignée C57BL , Inhibiteur-1 d'activateur du plasminogène , Transduction du signal , Protéine p53 suppresseur de tumeur , Animaux , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/métabolisme , Monoxyde de carbone/pharmacologie , Monoxyde de carbone/métabolisme , Souris , Bléomycine/toxicité , Transduction du signal/effets des médicaments et des substances chimiques , Inhibiteur-1 d'activateur du plasminogène/métabolisme , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/prévention et contrôle , Mâle , Composés organométalliques/pharmacologie , Altération de l'ADN/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine
3.
Respir Res ; 25(1): 242, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38877465

RÉSUMÉ

BACKGROUND: Silicosis represents a paramount occupational health hazard globally, with its incidence, morbidity, and mortality on an upward trajectory, posing substantial clinical dilemmas due to limited effective treatment options available. Trigonelline (Trig), a plant alkaloid extracted mainly from coffee and fenugreek, have diverse biological properties such as protecting dermal fibroblasts against ultraviolet radiation and has the potential to inhibit collagen synthesis. However, it's unclear whether Trig inhibits fibroblast activation to attenuate silicosis-induced pulmonary fibrosis is unclear. METHODS: To evaluate the therapeutic efficacy of Trig in the context of silicosis-related pulmonary fibrosis, a mouse model of silicosis was utilized. The investigation seeks to elucidated Trig's impact on the progression of silica-induced pulmonary fibrosis by evaluating protein expression, mRNA levels and employing Hematoxylin and Eosin (H&E), Masson's trichrome, and Sirius Red staining. Subsequently, we explored the mechanism underlying of its functions. RESULTS: In vivo experiment, Trig has been demonstrated the significant efficacy in mitigating SiO2-induced silicosis and BLM-induced pulmonary fibrosis, as evidenced by improved histochemical staining and reduced fibrotic marker expressions. Additionally, we showed that the differentiation of fibroblast to myofibroblast was imped in Trig + SiO2 group. In terms of mechanism, we obtained in vitro evidence that Trig inhibited fibroblast-to-myofibroblast differentiation by repressing TGF-ß/Smad signaling according to the in vitro evidence. Notably, our finding indicated that Trig seemed to be safe in mice and fibroblasts. CONCLUSION: In summary, Trig attenuated the severity of silicosis-related pulmonary fibrosis by alleviating the differentiation of myofibroblasts, indicating the development of novel therapeutic approaches for silicosis fibrosis.


Sujet(s)
Alcaloïdes , Différenciation cellulaire , Fibroblastes , Souris de lignée C57BL , Myofibroblastes , Fibrose pulmonaire , Silice , Silicose , Animaux , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/prévention et contrôle , Alcaloïdes/pharmacologie , Silice/toxicité , Souris , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Myofibroblastes/effets des médicaments et des substances chimiques , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Silicose/anatomopathologie , Silicose/métabolisme , Silicose/traitement médicamenteux , Mâle
4.
Biomed Pharmacother ; 177: 117016, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38943992

RÉSUMÉ

Idiopathic pulmonary fibrosis is an aging-related, chronic lung disease, with unclear pathogenesis and no effective treatment. One of the triggering factors in cell aging is oxidative stress and it is known to have a role in idiopathic pulmonary fibrosis. In this paper, the protective effect of the E-CG-01 (3,4-lacto-cycloastragenol) molecule in terms of its antioxidant properties was evaluated in the bleomycin induced mice lung fibrosis model. Bleomycin sulfate was administered as a single dose (2.5 U/kg body weight) intratracheally to induce lung fibrosis. E-CG-01 was administered intraperitoneally in three different doses (2 mg/kg/day, 6 mg/kg/day, and 10 mg/kg/day) for 14 days, starting three days before the bleomycin administration. Fibrosis was examined by Hematoxylin-Eosin, Masson Trichrome, and immunohistochemical staining for TGF-beta1, Type I collagen Ki-67, and gama-H2AX markers. Activity analysis of catalase and Superoxide dismutase enzymes, measurement of total oxidant, total glutathione, and Malondialdehyde levels. In histological analysis, it was determined that all three different doses of the molecule provided a prophylactic effect against the progression of fibrosis compared to the bleomycin control group. However, it was observed that only the molecule applied in the high dose decreased the total oxidant stress level. Lung weight ratio increased in the BLM group but significantly reduced with high-dose E-CG-01. E-CG-01 at all doses reduced collagen deposition, TGF-ß expression, and Ki-67 expression compared to the BLM group. Intermediate and high doses of E-CG-01 also significantly reduced alveolar wall thickness and edema formation. These findings suggest that E-CG-01 has potential therapeutic effects in mitigating lung fibrosis through its antioxidant properties.


Sujet(s)
Antioxydants , Bléomycine , Souris de lignée C57BL , Stress oxydatif , Fibrose pulmonaire , Animaux , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/prévention et contrôle , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/métabolisme , Antioxydants/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Souris , Mâle , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Superoxide dismutase/métabolisme
5.
Arthritis Res Ther ; 26(1): 94, 2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38702742

RÉSUMÉ

BACKGROUND: Systemic sclerosis (SSc) is an autoimmune disease characterized by vascular injury and inflammation, followed by excessive fibrosis of the skin and other internal organs, including the lungs. CX3CL1 (fractalkine), a chemokine expressed on endothelial cells, supports the migration of macrophages and T cells that express its specific receptor CX3CR1 into targeted tissues. We previously reported that anti-CX3CL1 monoclonal antibody (mAb) treatment significantly inhibited transforming growth factor (TGF)-ß1-induced expression of type I collagen and fibronectin 1 in human dermal fibroblasts. Additionally, anti-mouse CX3CL1 mAb efficiently suppressed skin inflammation and fibrosis in bleomycin- and growth factor-induced SSc mouse models. However, further studies using different mouse models of the complex immunopathology of SSc are required before the initiation of a clinical trial of CX3CL1 inhibitors for human SSc. METHODS: To assess the preclinical utility and functional mechanism of anti-CX3CL1 mAb therapy in skin and lung fibrosis, a sclerodermatous chronic graft-versus-host disease (Scl-cGVHD) mouse model was analyzed with immunohistochemical staining for characteristic infiltrating cells and RNA sequencing assays. RESULTS: On day 42 after bone marrow transplantation, Scl-cGVHD mice showed increased serum CX3CL1 level. Intraperitoneal administration of anti-CX3CL1 mAb inhibited the development of fibrosis in the skin and lungs of Scl-cGVHD model, and did not result in any apparent adverse events. The therapeutic effects were correlated with the number of tissue-infiltrating inflammatory cells and α-smooth muscle actin (α-SMA)-positive myofibroblasts. RNA sequencing analysis of the fibrotic skin demonstrated that cGVHD-dependent induction of gene sets associated with macrophage-related inflammation and fibrosis was significantly downregulated by mAb treatment. In the process of fibrosis, mAb treatment reduced cGVHD-induced infiltration of macrophages and T cells in the skin and lungs, especially those expressing CX3CR1. CONCLUSIONS: Together with our previous findings in other SSc mouse models, the current results indicated that anti-CX3CL1 mAb therapy could be a rational therapeutic approach for fibrotic disorders, such as human SSc and Scl-cGVHD.


Sujet(s)
Anticorps monoclonaux , Chimiokine CX3CL1 , Modèles animaux de maladie humaine , Maladie du greffon contre l'hôte , Fibrose pulmonaire , Sclérodermie systémique , Peau , Animaux , Maladie du greffon contre l'hôte/traitement médicamenteux , Maladie du greffon contre l'hôte/immunologie , Maladie du greffon contre l'hôte/anatomopathologie , Sclérodermie systémique/traitement médicamenteux , Sclérodermie systémique/anatomopathologie , Sclérodermie systémique/immunologie , Souris , Chimiokine CX3CL1/métabolisme , Chimiokine CX3CL1/antagonistes et inhibiteurs , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/usage thérapeutique , Fibrose pulmonaire/immunologie , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/prévention et contrôle , Peau/anatomopathologie , Peau/effets des médicaments et des substances chimiques , Peau/métabolisme , Peau/immunologie , Fibrose , Femelle , Souris de lignée C57BL , Humains , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Poumon/immunologie
6.
Respir Res ; 25(1): 213, 2024 May 18.
Article de Anglais | MEDLINE | ID: mdl-38762465

RÉSUMÉ

BACKGROUND: Obesity is associated with airway hyperresponsiveness and lung fibrosis, which may reduce the effectiveness of standard asthma treatment in individuals suffering from both conditions. Statins and proprotein convertase subtilisin/kexin-9 inhibitors not only reduce serum cholesterol, free fatty acids but also diminish renin-angiotensin system activity and exhibit anti-inflammatory effects. These mechanisms may play a role in mitigating lung pathologies associated with obesity. METHODS: Male C57BL/6 mice were induced to develop obesity through high-fat diet for 16 weeks. Conditional TGF-ß1 transgenic mice were fed a normal diet. These mice were given either atorvastatin or proprotein convertase subtilisin/kexin-9 inhibitor (alirocumab), and the impact on airway hyperresponsiveness and lung pathologies was assessed. RESULTS: High-fat diet-induced obesity enhanced airway hyperresponsiveness, lung fibrosis, macrophages in bronchoalveolar lavage fluid, and pro-inflammatory mediators in the lung. These lipid-lowering agents attenuated airway hyperresponsiveness, macrophages in BALF, lung fibrosis, serum leptin, free fatty acids, TGF-ß1, IL-1ß, IL-6, and IL-17a in the lung. Furthermore, the increased RAS, NLRP3 inflammasome, and cholecystokinin in lung tissue of obese mice were reduced with statin or alirocumab. These agents also suppressed the pro-inflammatory immune responses and lung fibrosis in TGF-ß1 over-expressed transgenic mice with normal diet. CONCLUSIONS: Lipid-lowering treatment has the potential to alleviate obesity-induced airway hyperresponsiveness and lung fibrosis by inhibiting the NLRP3 inflammasome, RAS and cholecystokinin activity.


Sujet(s)
Alimentation riche en graisse , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase , Souris de lignée C57BL , Souris transgéniques , Obésité , Fibrose pulmonaire , Animaux , Mâle , Alimentation riche en graisse/effets indésirables , Obésité/traitement médicamenteux , Obésité/métabolisme , Souris , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/pharmacologie , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/usage thérapeutique , Fibrose pulmonaire/prévention et contrôle , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/traitement médicamenteux , Inhibiteurs de PCSK9 , Atorvastatine/pharmacologie , Atorvastatine/usage thérapeutique , Souris obèse , Proprotéine convertase 9/métabolisme , Proprotéine convertase 9/génétique , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/usage thérapeutique , Hyperréactivité bronchique/prévention et contrôle , Hyperréactivité bronchique/traitement médicamenteux , Hyperréactivité bronchique/métabolisme , Hyperréactivité bronchique/physiopathologie , Anticorps monoclonaux humanisés
7.
Biochem Pharmacol ; 225: 116282, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38762147

RÉSUMÉ

IPF is a chronic, progressive, interstitial lung disease with high mortality. Current drugs have limited efficacy in curbing disease progression and improving quality of life. Selpercatinib, a highly selective inhibitor of receptor tyrosine kinase RET (rearranged during transfection), was approved in 2020 for the treatment of a variety of solid tumors with RET mutations. In this study, the action and mechanism of Selpercatinib in pulmonary fibrosis were evaluated in vivo and in vitro. In vivo experiments demonstrated that Selpercatinib significantly ameliorated bleomycin (BLM)-induced pulmonary fibrosis in mice. In vitro, Selpercatinib inhibited the proliferation, migration, activation and extracellular matrix deposition of fibroblasts by inhibiting TGF-ß1/Smad and TGF-ß1/non-Smad pathway, and suppressed epithelial-mesenchymal transition (EMT) like process of lung epithelial cells via inhibiting TGF-ß1/Smad pathway. The results of in vivo pharmacological tests corroborated the results obtained from the in vitro experiments. Further studies revealed that Selpercatinib inhibited abnormal phenotypes of lung fibroblasts and epithelial cells in part by regulating its target RET. In short, Selpercatinib inhibited the activation of fibroblasts and EMT-like process of lung epithelial cells by inhibiting TGF-ß1/Smad and TGF-ß1/non-Smad pathways, thus alleviating BLM-induced pulmonary fibrosis in mice.


Sujet(s)
Bléomycine , Souris de lignée C57BL , Fibrose pulmonaire , Transduction du signal , Facteur de croissance transformant bêta-1 , Animaux , Bléomycine/toxicité , Facteur de croissance transformant bêta-1/métabolisme , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/prévention et contrôle , Mâle , Pyrazoles/pharmacologie , Pyrazoles/usage thérapeutique , Pyridines/pharmacologie , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Humains , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme
8.
Vaccine ; 42(18): 3774-3788, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38714443

RÉSUMÉ

Interleukin-1ß (IL-1ß) contributes to interstitial lung disease (ILD) and pulmonary fibrosis (PF), thus representing a potential therapeutic target for PF. In this study, we first verified the increased expression of IL-1ß in human fibrotic lung specimens and mouse lung tissues after intratracheal (i.t.) instillation of bleomycin (BLM), after which the pro-inflammatory and pro-fibrotic effects of recombinant IL-1ß were tested in mice. The results above suggested that vaccination against IL-1ß could be an effective strategy for managing PF. An anti-IL-1ß vaccine (PfTrx-IL-1ß) was designed by incorporating two IL-1ß-derived polypeptides, which have been verified as the key domains that mediate the binding of IL-1ß to its type I receptor, into Pyrococcus furiosus thioredoxin (PfTrx). The fusion protein PfTrx-IL-1ß was prepared by using E. coli expression system. The vaccine was well tolerated; it induced robust and long-lasting antibody responses in mice and neutralized the biological activity of IL-1ß, as shown in cellular assays. Pre-immunization with PfTrx-IL-1ß effectively protected mice from BLM-induced lung injury, inflammation, and fibrosis. In vitro experiments further showed that anti-PfTrx-IL-1ß antibodies counteracted the effects of IL-1ß concerning pro-inflammatory and pro-fibrotic cytokine production by primary mouse lung fibroblast, macrophages (RAW264.7), and type II alveolar epithelial cell (A549), primary mouse lung fibroblast activation and epithelial-mesenchymal transition (EMT) of alveolar epithelial cells. In addition, the vaccination did not compromise the anti-infection immunity in mice, as validated by a sepsis model. Our preliminary study suggests that the anti-IL-1ß vaccine we prepared has the potential to be developed as a therapeutic measure for PF. Further experiments are warranted to evaluate whether IL-1ß vaccination has the capacity of inhibiting chronic progressive PF and reversing established PF.


Sujet(s)
Bléomycine , Interleukine-1 bêta , Fibrose pulmonaire , Animaux , Fibrose pulmonaire/prévention et contrôle , Fibrose pulmonaire/immunologie , Fibrose pulmonaire/induit chimiquement , Interleukine-1 bêta/immunologie , Souris , Humains , Vaccins synthétiques/immunologie , Vaccins synthétiques/administration et posologie , Poumon/anatomopathologie , Poumon/immunologie , Modèles animaux de maladie humaine , Femelle , Souris de lignée C57BL , Protéines de fusion recombinantes/immunologie , Thiorédoxines/immunologie
9.
Ecotoxicol Environ Saf ; 279: 116483, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38788565

RÉSUMÉ

Oxidative stress and inflammation play a fundamental role in the beginning and advancement of silicosis. Hence, questing active phytocompounds (APCs) with anti-oxidative and anti-inflammatory properties such as diosgenin (DG) and emodin (ED) can be a therapeutic intervention targeting silica-induced pulmonary inflammation and fibrosis. Hydrophobicity and low bioavailability are the barriers that restrict the therapeutic efficacy of DG and ED against pulmonary defects. Encapsulating these APCs in polymeric nanoparticles can overcome this limitation. The present study has thus explored the anti-inflammatory and anti-fibrotic effects of polylactic-co-glycolic acid (PLGA) nanoparticles (NPs) individually loaded with DG (DGn) or ED (EDn) and in combine DG+ED [(DG+ED)n] in respirable silica dust (RSD)-induced pulmonary fibrosis silicosis rat model. Our study found that individual and combined NPs revealed physiochemical characteristics appropriate for IV administration with sustained-drug release purposes. Physiological evaluations of RSD-induced silicosis rats suggested that no treatment could improve the body weight. Still, they reduced the lung coefficient by maintaining lung moisture. Only (DG+ED)n significantly cleared free lung silica. All interventions were found to attribute the increased per cent cell viability in BALF, reduce cytotoxicity via minimizing LDH levels, and balance the oxidant-antioxidant status in silicotic rats. The expression of inflammatory cytokines (TNF-α, IL-1ß, IL-6, MCP-1, and TGF-ß1) were efficiently down-regulated with NPs interventions compared to pure (DG+ED) treatment. All drug treatments significantly declined, the 8-HdG and HYP productions indicate that RSD-induced oxidative DNA damage and collagen deposition were successfully repaired. Moreover, histopathological investigations proposed that individual or combined drugs NPs interventions could decrease the fibrosis and alveolitis grades in RSD-induced silicosis rats. However, (DG+ED)n intervention significantly inhibited pulmonary fibrosis and alveolitis compared to pure (DG+ED) treatment. In conclusion, the RSD can induce oxidative stress and inflammation in rats, producing reactive oxygen species (ROS)-mediated cytotoxicity to pulmonary cells and leading to silicosis development. The IV administration of combined NP suppressed lung inflammation and collagen formation by maintaining oxidant-antioxidant status and effectively interrupting the fibrosis-silicosis progression. These results may be attributed to the improved bioavailability of DG and ED through their combined nano-encapsulation-mediated targeted drug delivery.


Sujet(s)
Diosgénine , Émodine , Nanoparticules , Fibrose pulmonaire , Silice , Silicose , Animaux , Diosgénine/pharmacologie , Silicose/traitement médicamenteux , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/prévention et contrôle , Rats , Émodine/pharmacologie , Mâle , Poussière , Stress oxydatif/effets des médicaments et des substances chimiques , Anti-inflammatoires , Rat Wistar , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Copolymère d'acide poly(lactique-co-glycolique)/composition chimique
10.
Basic Clin Pharmacol Toxicol ; 135(1): 23-42, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38745367

RÉSUMÉ

This study investigated the therapeutic benefits of para-hydroxycinnamic acid in mice with bleomycin-induced lung fibrosis. Forty male BALB/c mice were randomly assigned to four groups: normal, which received 0.9% normal saline; induced, which received a single dose of bleomycin (5 mg/kg) by oropharyngeal challenge; pirfenidone-treated; and para-hydroxycinnamic acid-treated, which challenged with bleomycin and received a daily oral dose of 300 and 50 mg/kg, respectively, from day 7 to day 21. Tissue pro-fibrotic and inflammatory cytokines, oxidative indicators, pulmonary histopathology, immunohistochemistry of fibrotic proteins and the assessment of gene expression by RT-qPCR were evaluated on day 22 after euthanizing animals. Pirfenidone and para-hydroxycinnamic acid managed to alleviate the fibrotic endpoints by statistically improving the weight index, histopathological score and reduced expression of fibrotic-related proteins in immune-stained lung sections, as well as fibrotic markers measured in serum samples. They also managed to alleviate tissue levels of oxidative stress and inflammatory and pro-fibrotic mediators. para-Hydroxycinnamic acid enhanced the expression of crucial genes associated with oxidative stress, inflammation and fibrosis in vivo. para-Hydroxycinnamic acid has demonstrated similar effectiveness to pirfenidone, suggesting it could be a promising treatment for fibrotic lung conditions by inhibiting the TGF-ß1/Smad3 pathway or through its anti-inflammatory and antioxidant properties.


Sujet(s)
Bléomycine , Acides coumariques , Poumon , Souris de lignée BALB C , Stress oxydatif , Fibrose pulmonaire , Animaux , Bléomycine/toxicité , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/prévention et contrôle , Mâle , Stress oxydatif/effets des médicaments et des substances chimiques , Souris , Acides coumariques/pharmacologie , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Pyridones/pharmacologie , Inflammation/traitement médicamenteux , Inflammation/induit chimiquement , Inflammation/métabolisme , Cytokines/métabolisme , Modèles animaux de maladie humaine , Antioxydants/pharmacologie , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/génétique
11.
Toxicol Appl Pharmacol ; 486: 116939, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38643951

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is an irreversible disease which considered the most fatal pulmonary fibrosis. Pulmonary toxicity including IPF is the most severe adverse effect of bleomycin, the chemotherapeutic agent. Based on the fact that, exogenous surfactants could induce alveolar stabilization in many lung diseases, the aim of this study was to explore the effects of low cost biosurfactants, surfactin (SUR) and sophorolipids (SLs), against bleomycin-induced pulmonary fibrosis in mice due to their antioxidant, and anti-inflammatory properties. Surfactin and sophorolipids were produced by microbial conversion of frying oil and potato peel wastes using Bacillus halotolerans and Candida parapsilosis respectively. These biosurfactants were identified by FTIR, 1H NMR, and LC-MS/MS spectra. C57BL/6 mice were administered the produced biosurfactants daily at oral dose of 200 mg kg-1 one day after the first bleomycin dose (35 U/kg). We evaluated four study groups: Control, Bleomycin, Bleomycin+SUR, Bleomycin+SLs. After 30 days, lungs from each mouse were sampled for oxidative stress, ELISA, Western blot, histopathological, immunohistochemical analyses. Our results showed that the produced SUR and SLs reduced pulmonary oxidative stress and inflammatory response in the lungs of bleomycin induced mice as they suppressed SOD, CAT, and GST activities also reduced NF-κß, TNF-α, and CD68 levels. Furthermore, biosurfactants suppressed the expression of TGF-ß1, Smad-3, and p-JNK fibrotic signaling pathway in pulmonary tissues. Histologically, SUR and SLs protected against lung ECM deposition caused by bleomycin administration. Biosurfactants produced from microbial sources can inhibit the induced inflammatory and fibrotic responses in bleomycin-induced pulmonary fibrosis.


Sujet(s)
Anti-inflammatoires , Antioxydants , Bléomycine , Candida parapsilosis , Souris de lignée C57BL , microARN , Fibrose pulmonaire , Protéine Smad-3 , Tensioactifs , Facteur de croissance transformant bêta-1 , Animaux , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/prévention et contrôle , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/métabolisme , Bléomycine/toxicité , Antioxydants/pharmacologie , Facteur de croissance transformant bêta-1/métabolisme , Anti-inflammatoires/pharmacologie , Protéine Smad-3/métabolisme , Souris , Candida parapsilosis/effets des médicaments et des substances chimiques , Tensioactifs/pharmacologie , microARN/métabolisme , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Bacillus , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Acides oléiques
12.
Toxicol Appl Pharmacol ; 487: 116949, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38688425

RÉSUMÉ

Pulmonary fibrosis is a lung disorder characterized by the accumulation of abnormal extracellular matrix, scar tissue formation, and tissue stiffness. Type II alveolar epithelial cells (AEII) play a critical role in repairing lung tissue after injury, and repeated injury to these cells is a key factor in the development of pulmonary fibrosis. Chronic exposure to PM2.5, a type of air pollution, has been shown to increase the incidence and severity of pulmonary fibrosis by enhancing the activation of EMT in lung epithelial cells. Melatonin, a hormone with antioxidant properties, has been shown to prevent EMT and reduce fibrosis in previous studies. However, the mechanism through which melatonin targets EMT to prevent pulmonary fibrosis caused by PM2.5 exposure has not been extensively discussed before. In this current study, we found that melatonin effectively prevented pulmonary fibrosis caused by prolonged exposure to PM2.5 by targeting EMT. The study demonstrated changes in cellular morphology and expression of EMT markers. Furthermore, the cell migratory potential induced by prolonged exposure to PM2.5 was greatly reduced by melatonin treatment. Finally, in vivo animal studies showed reduced EMT markers and improved pulmonary function. These findings suggest that melatonin has potential clinical use for the prevention of pulmonary fibrosis.


Sujet(s)
Transition épithélio-mésenchymateuse , Mélatonine , Matière particulaire , Fibrose pulmonaire , Mélatonine/pharmacologie , Mélatonine/usage thérapeutique , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Animaux , Fibrose pulmonaire/prévention et contrôle , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/anatomopathologie , Matière particulaire/toxicité , Mâle , Souris , Souris de lignée C57BL , Mouvement cellulaire/effets des médicaments et des substances chimiques , Humains , Pneumocytes/effets des médicaments et des substances chimiques , Pneumocytes/anatomopathologie , Pneumocytes/métabolisme , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Antioxydants/pharmacologie , Antioxydants/usage thérapeutique
13.
Biomed Pharmacother ; 174: 116431, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38522238

RÉSUMÉ

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a progressive and life-threatening lung disease with high mortality rates. The limited availability of effective drugs for IPF treatment, coupled with concerns regarding adverse effects and restricted responsiveness, underscores the need for alternative approaches. Kefir peptides (KPs) have demonstrated antioxidative, anti-inflammatory, and antifibrotic properties, along with the capability to modulate gut microbiota. This study aims to investigate the impact of KPs on bleomycin-induced pulmonary fibrosis. METHODS: Mice were treated with KPs for four days, followed by intratracheal injection of bleomycin for 21 days. Comprehensive assessments included pulmonary functional tests, micro-computed tomography (µ-CT), in vivo image analysis using MMPsense750, evaluation of inflammation- and fibrosis-related gene expression in lung tissue, and histopathological examinations. Furthermore, a detailed investigation of the gut microbiota community was performed using full-length 16 S rRNA sequencing in control mice, bleomycin-induced fibrotic mice, and KPs-pretreated fibrotic mice. RESULTS: In KPs-pretreated bleomycin-induced lung fibrotic mice, notable outcomes included the absence of significant bodyweight loss, enhanced pulmonary functions, restored lung tissue architecture, and diminished thickening of inter-alveolar septa, as elucidated by morphological and histopathological analyses. Concurrently, a reduction in the expression levels of oxidative biomarkers, inflammatory factors, and fibrotic indicators was observed. Moreover, 16 S rRNA sequencing demonstrated that KPs pretreatment induced alterations in the relative abundances of gut microbiota, notably affecting Barnesiella_intestinihominis, Kineothrix_alysoides, and Clostridium_viride. CONCLUSIONS: Kefir peptides exerted preventive effects, protecting mice against bleomycin-induced lung oxidative stress, inflammation, and fibrosis. These effects are likely linked to modifications in the gut microbiota community. The findings highlight the therapeutic potential of KPs in mitigating pulmonary fibrosis and advocate for additional exploration in clinical settings.


Sujet(s)
Bléomycine , Microbiome gastro-intestinal , Kéfir , Souris de lignée C57BL , Stress oxydatif , Fibrose pulmonaire , Animaux , Stress oxydatif/effets des médicaments et des substances chimiques , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Souris , Kéfir/microbiologie , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/prévention et contrôle , Fibrose pulmonaire/traitement médicamenteux , Inflammation/anatomopathologie , Mâle , Peptides/pharmacologie , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Anti-inflammatoires/pharmacologie , Modèles animaux de maladie humaine
14.
Hum Vaccin Immunother ; 20(1): 2319965, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38408907

RÉSUMÉ

Mimotope, a kind of peptide vaccine, is developed to bind natural receptor and inhibit the downstream signaling. We have demonstrated that the vaccination of Tocilizumab mimotopes could alleviate the renal fibrosis by interfering with both IL-6 and ferroptosis signaling. However, the effect of the vaccination of Tocilizumab mimotopes on the fibroblast was not investigated in previous study. Thus, we sought to explore the changes in the fibroblast induced by the Tocilizumab mimotopes vaccination. Bleomycin instillation was performed to construct the pulmonary fibrosis model after the immunization of Tocilizumab mimotopes. Lung histological analysis showed that the Tocilizumab mimotopes could significantly reduce the maladaptive repairment and abnormal remodeling. Immunoblotting assay and fluorescence staining showed that Immunization with the Tocilizumab mimotopes reduces the accumulation of fibrosis-related proteins. High level of lipid peroxidation product was observed in the animal model, while the Tocilizumab mimotopes vaccination could reduce the generation of lipid peroxidation product. Mechanism analysis further showed that Nrf-2 signaling, but not GPX-4 and FSP-1 signaling, was upregulated, and reduced the lipid peroxidation. Our results revealed that in the BLM-induced pulmonary fibrosis, high level of lipid peroxidation product was significantly accumulation in the lung tissues, which might lead to the occurrence of ferroptosis. The IL-6 pathway block therapy could inhibit lipid peroxidation product generation in the lung tissues by upregulating the Nrf-2 signaling, and further alleviate the pulmonary fibrosis.


Sujet(s)
Anticorps monoclonaux humanisés , Fibrose pulmonaire , Animaux , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/prévention et contrôle , Interleukine-6 , Bléomycine/effets indésirables , Bléomycine/métabolisme , Poumon/anatomopathologie , Vaccination
15.
Immunopharmacol Immunotoxicol ; 46(2): 183-191, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38224264

RÉSUMÉ

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a pulmonary fibrotic disease characterized by a poor prognosis, which its pathogenesis involves the accumulation of abnormal fibrous tissue, inflammation, and oxidative stress. Ivermectin, a positive allosteric modulator of GABAA receptor, exerts anti-inflammatory and antioxidant properties in preclinical studies. The present study investigates the potential protective effects of ivermectin treatment in rats against bleomycin-induced IPF. MATERIALS AND METHODS: The present study involved 42 male Wistar rats, which were divided into five groups: control (without induction of IPF), bleomycin (IPF-induced by bleomycin 2.5 mg/kg, by intratracheal administration), and three fibrosis groups receiving ivermectin (0.5, 1, and 3 mg/kg). lung tissues were harvested for measurement of oxidative stress [via myeloperoxidase (MPO), superoxide dismutase (SOD), glutathione (GSH)] and inflammatory markers (tumor necrosis factor-α [TNF-α], interleukin-1ß [IL-1ß], and transforming growth factor-ß [TGF-ß]). Histological assessments of tissue damage were performed using hematoxylin-eosin (H&E) and Masson's trichrome staining methods. RESULTS: The induction of fibrosis via bleomycin was found to increase levels of MPO as well as TNF-α, IL-1ß, and TGF-ß while decrease SOD activity and GSH level. Treatment with ivermectin at a dosage of 3 mg/kg was able to reverse the effects of bleomycin-induced fibrosis on these markers. In addition, results from H&E and Masson's trichrome staining showed that ivermectin treatment at this same dose reduced tissue damage and pulmonary fibrosis. CONCLUSION: The data obtained from this study indicate that ivermectin may have therapeutic benefits for IPF, likely due to its ability to reduce inflammation and mitigate oxidative stress-induced toxicity.


Sujet(s)
Fibrose pulmonaire , Rats , Mâle , Animaux , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/prévention et contrôle , Bléomycine/effets indésirables , Ivermectine/effets indésirables , Facteur de nécrose tumorale alpha/métabolisme , Rat Wistar , Inflammation/induit chimiquement , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Poumon/métabolisme , Stress oxydatif , Facteur de croissance transformant bêta , Glutathion/métabolisme , Superoxide dismutase/métabolisme
16.
Biol Pharm Bull ; 47(1): 303-310, 2024.
Article de Anglais | MEDLINE | ID: mdl-38281774

RÉSUMÉ

Methotrexate (MTX) is an indispensable drug used for the treatment of many autoimmune and cancerous diseases. However, its clinical use is associated with serious side effects, such as lung fibrosis. The main objective of this study is to test the hypothesis that hydroxytyrosol (HT) can mitigate MTX-induced lung fibrosis in rats while synergizing MTX anticancer effects. Pulmonary fibrosis was induced in the rats using MTX (14 mg/kg/week, per os (p.o.)). The rats were treated with or without HT (10, 20, and 40 mg/kg/d p.o.) or dexamethasone (DEX; 0.5 mg/kg/d, intraperitoneally (i.p.)) for two weeks concomitantly with MTX. Transforming growth factor beta 1 (TGF-ß1), interleukin-4 (IL-4), thromboxane A2 (TXA2), vascular endothelial growth factor (VEGF), 8-hydroxy-2-deoxy-guanosine (8-OHdG), tissue factor (TF) and fibrin were assessed using enzyme-linked immunosorbent assay (ELISA), immunofluorescence, and RT-PCR. Pulmonary fibrosis was manifested by an excessive extracellular matrix (ECM) deposition and a marked increase in TGF-ß1 and IL-4 in lung tissues. Furthermore, cotreatment with HT or dexamethasone (DEX) significantly attenuated MTX-induced ECM deposition, TGF-ß1, and IL-4 expression. Similarly, HT or DEX notably reduced hydroxyproline contents, TXA2, fibrin, and TF expression in lung tissues. Moreover, using HT or DEX downregulated the gene expression of TF. A significant decrease in lung contents of VEGF, IL-8, and 8-OHdG was also observed in HT + MTX- or DEX + MTX -treated animals in a dose-dependent manner. Collectively, the results of our study suggest that HT might represent a potential protective agent against MTX-induced pulmonary fibrosis.


Sujet(s)
Méthotrexate , Alcool phénéthylique , Fibrose pulmonaire , Animaux , Rats , Dexaméthasone/pharmacologie , Fibrine/métabolisme , Interleukine-4/métabolisme , Poumon/anatomopathologie , Méthotrexate/effets indésirables , Alcool phénéthylique/analogues et dérivés , Alcool phénéthylique/pharmacologie , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/prévention et contrôle , Thromboplastine/métabolisme , Facteur de croissance transformant bêta-1/génétique , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme
17.
Life Sci ; 336: 122272, 2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-37981228

RÉSUMÉ

AIMS: Pulmonary fibrosis (PF) is a chronic interstitial lung disease with an increasing incidence following the COVID-19 outbreak. Pirfenidone (Pirf), an FDA-approved pulmonary anti-fibrotic drug, is poorly tolerated and exhibits limited efficacy. Trigonelline (Trig) is a natural plant alkaloid with diverse pharmacological actions. We investigated the underlying prophylactic and therapeutic mechanisms of Trig in ameliorating bleomycin (BLM)-induced PF and the possible synergistic antifibrotic activity of Pirf via its combination with Trig. MATERIALS AND METHODS: A single dose of BLM was administered intratracheally to male Sprague-Dawley rats for PF induction. In the prophylactic study, Trig was given orally 3 days before BLM and then for 28 days. In the therapeutic study, Trig and/or Pirf were given orally from day 8 after BLM until the 28th day. Biochemical assay, histopathology, qRT-PCR, ELISA, and immunohistochemistry were performed on lung tissues. KEY FINDINGS: Trig prophylactically and therapeutically mitigated the inflammatory process via targeting NF-κB/NLRP3/IL-1ß signaling. Trig activated the autophagy process which in turn attenuated alveolar epithelial cells apoptosis and senescence. Remarkably, Trig attenuated lung SPHK1/S1P axis and its downstream Hippo targets, YAP-1, and TAZ, with a parallel decrease in YAP/TAZ profibrotic genes. Interestingly, Trig upregulated lung miR-375 and miR-27a expression. Consequently, epithelial-mesenchymal transition in lung tissues was reversed upon Trig administration. These results were simultaneously associated with profound improvement in lung histological alterations. SIGNIFICANCE: The current study verifies Trig's prophylactic and antifibrotic effects against BLM-induced PF via targeting multiple signaling. Trig and Pirf combination may be a promising approach to synergize Pirf antifibrotic effect.


Sujet(s)
Alcaloïdes , microARN , Pneumopathie infectieuse , Fibrose pulmonaire , Rats , Animaux , Bléomycine/pharmacologie , Inflammasomes/métabolisme , Voie de signalisation Hippo , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Rat Sprague-Dawley , Poumon/métabolisme , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/prévention et contrôle , Pneumopathie infectieuse/anatomopathologie , Alcaloïdes/usage thérapeutique , microARN/métabolisme
18.
Naunyn Schmiedebergs Arch Pharmacol ; 397(2): 1071-1079, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-37581637

RÉSUMÉ

PURPOSE: Lung fibrosis is a heterogeneous lung condition characterized by excessive accumulation of scarred tissue, leading to lung architecture destruction and restricted ventilation. The current work was conducted to examine the probable shielding influence of cinnamic acid against lung fibrosis induced by methotrexate. METHODS: Rats were pre-treated with oral administration of cinnamic acid (50 mg/kg/day) for 14 days, whereas methotrexate (14 mg/kg) was orally given on the 5th and 12th days of the experiment. Pirfenidone (50 mg/kg/day) was used as a standard drug. At the end of the experiment, oxidative parameters (malondialdehyde, myeloperoxidase, nitric oxide, and total glutathione) and inflammatory mediators (tumor necrosis factor-α and interleukin-8), as well as transforming growth factor-ß and collagen content, as fibrosis indicators, were measured in lung tissue. RESULTS: Our results revealed that cinnamic acid, as pirfenidone, effectively prevented the methotrexate-induced overt histopathological damage. This was associated with parallel improvements in oxidative, inflammatory, and fibrotic parameters measured. The outcomes of cinnamic acid administration were more or less the same as those of pirfenidone. In conclusion, pre-treatment with cinnamic acid protects against methotrexate-induced fibrosis, making it a promising prophylactic adjuvant therapy to methotrexate and protecting against its possible induction of lung fibrosis.


Sujet(s)
Cinnamates , Fibrose pulmonaire , Pyridones , Rats , Animaux , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/prévention et contrôle , Méthotrexate/toxicité , Poumon , Fibrose
19.
Naunyn Schmiedebergs Arch Pharmacol ; 397(7): 4809-4822, 2024 07.
Article de Anglais | MEDLINE | ID: mdl-38153514

RÉSUMÉ

Pulmonary fibrosis is a chronic and progressive lung disease with high mortality. This study aims to explore the protective mechanism of quercetin against pulmonary fibrosis regarding cell senescence and gut microbiota. Rats were intratracheally injected with bleomycin (BLM) to establish a pulmonary fibrosis rat model. RLE-6TN cells were stimulated with BLM to build the model of alveolar epithelial cell senescence, and RLE-6TN-derived conditional medium (CM) was harvested to further culture fibroblasts. Histopathological changes were assessed by H&E and Masson staining. α-SMA expression was assessed by immunofluorescence assay. Senescence-associated ß-galactosidase (SA-ß-gal) staining and senescence-associated secretory phenotype (SASP) cytokine assay were conducted to assess cellular senescence. Gut microbiota was analyzed by 16S rRNA gene sequencing. The fibrosis-, senescence-, and PTEN/PI3K/AKT signaling-related proteins were examined by western blot. In BLM-induced pulmonary fibrosis rats, quercetin exerted its protective effects by reducing histological injury and collagen deposition, lessening cellular senescence, and regulating gut microbiota. In BLM-induced alveolar epithelial cell senescence, quercetin inhibited senescence, lessened SASP cytokine secretion of alveolar epithelial cells, and further ameliorated collagen deposition in fibroblasts. In addition, quercetin might exert its functional effects by regulating the PTEN/PI3K/AKT signaling pathway. Moreover, quercetin regulated intestinal dysbacteriosis in BLM-induced pulmonary fibrosis rats, especially boosting the abundance of Akkermansia. To conclude, our findings provide an in-depth understanding of the potential mechanism behind the protective role of quercetin against pulmonary fibrosis.


Sujet(s)
Pneumocytes , Bléomycine , Vieillissement de la cellule , Dysbiose , Microbiome gastro-intestinal , Phosphohydrolase PTEN , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Fibrose pulmonaire , Quercétine , Transduction du signal , Animaux , Quercétine/pharmacologie , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/prévention et contrôle , Transduction du signal/effets des médicaments et des substances chimiques , Phosphohydrolase PTEN/métabolisme , Mâle , Bléomycine/toxicité , Rats , Phosphatidylinositol 3-kinases/métabolisme , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Pneumocytes/effets des médicaments et des substances chimiques , Pneumocytes/métabolisme , Pneumocytes/anatomopathologie , Rat Sprague-Dawley , Lignée cellulaire , Modèles animaux de maladie humaine
20.
Int J Radiat Oncol Biol Phys ; 118(1): 218-230, 2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-37586613

RÉSUMÉ

PURPOSE: Radiation-induced pulmonary fibrosis (RIPF) is a common side effect of radiation therapy for thoracic tumors without effective prevention and treatment methods at present. The aim of this study was to explore whether glycyrrhetinic acid (GA) has a protective effect on RIPF and the underlying mechanism. METHODS AND MATERIALS: A RIPF mouse model administered GA was used to determine the effect of GA on RIPF. The cocultivation of regulatory T (Treg) cells with mouse lung epithelial-12 cells or mouse embryonic fibroblasts and intervention with GA or transforming growth factor-ß1 (TGF-ß1) inhibitor to block TGF-ß1 was conducted to study the mechanism by which GA alleviates RIPF. Furthermore, injection of Treg cells into GA-treated RIPF mice to upregulate TGF-ß1 levels was performed to verify the roles of TGF-ß1 and Treg cells. RESULTS: GA intervention improved the damage to lung tissue structure and collagen deposition and inhibited Treg cell infiltration, TGF-ß1 levels, epithelial mesenchymal transition (EMT), and myofibroblast (MFB) transformation in mice after irradiation. Treg cell-induced EMT and MFB transformation in vitro were prevented by GA, as well as a TGF-ß1 inhibitor, by decreasing TGF-ß1. Furthermore, reinfusion of Treg cells upregulated TGF-ß1 levels and exacerbated RIPF in GA-treated RIPF mice. CONCLUSIONS: GA can improve RIPF in mice, and the corresponding mechanisms may be related to the inhibition of TGF-ß1 secreted by Treg cells to induce EMT and MFB transformation. Therefore, GA may be a promising therapeutic candidate for the clinical treatment of RIPF.


Sujet(s)
Énoxolone , Lésion pulmonaire , Fibrose pulmonaire , Lésions radiques , Animaux , Souris , Transition épithélio-mésenchymateuse , Fibroblastes/effets des radiations , Énoxolone/pharmacologie , Poumon/effets des radiations , Lésion pulmonaire/anatomopathologie , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/étiologie , Fibrose pulmonaire/prévention et contrôle , Lésions radiques/anatomopathologie , Lymphocytes T régulateurs , Facteur de croissance transformant bêta-1
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE