Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.559
Filtrar
2.
J Transl Med ; 22(1): 479, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773615

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fibrotic interstitial lung diseases, which mainly existed in middle-aged and elderly people. The accumulation of reactive oxygen species (ROS) is a common characteristic of IPF. Previous research also shown that lactate levels can be abnormally elevated in IPF patients. Emerging evidence suggested a relationship between lactate and ROS in IPF which needs further elucidation. In this article, we utilized a mouse model of BLM-induced pulmonary fibrosis to detect alterations in ROS levels and other indicators associated with fibrosis. Lactate could induce mitochondrial fragmentation by modulating expression and activity of DRP1 and ERK. Moreover, Increased ROS promoted P65 translocation into nucleus, leading to expression of lung fibrotic markers. Finally, Ulixertinib, Mdivi-1 and Mito-TEMPO, which were inhibitor activity of ERK, DRP1 and mtROS, respectively, could effectively prevented mitochondrial damage and production of ROS and eventually alleviate pulmonary fibrosis. Taken together, these findings suggested that lactate could promote lung fibrosis by increasing mitochondrial fission-derived ROS via ERK/DRP1 signaling, which may provide novel therapeutic solutions for IPF.


Asunto(s)
Dinaminas , Ratones Endogámicos C57BL , Dinámicas Mitocondriales , Especies Reactivas de Oxígeno , Animales , Especies Reactivas de Oxígeno/metabolismo , Dinámicas Mitocondriales/efectos de los fármacos , Dinaminas/metabolismo , Bleomicina , Transducción de Señal , Ácido Láctico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Mitocondrias/metabolismo , Masculino , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Ratones , Humanos
3.
J Cell Mol Med ; 28(10): e18448, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38774993

RESUMEN

Pulmonary fibrosis represents the final alteration seen in a wide variety of lung disorders characterized by increased fibroblast activity and the accumulation of substantial amounts of extracellular matrix, along with inflammatory damage and the breakdown of tissue architecture. This condition is marked by a significant mortality rate and a lack of effective treatments. The depositing of an excessive quantity of extracellular matrix protein follows the damage to lung capillaries and alveolar epithelial cells, leading to pulmonary fibrosis and irreversible damage to lung function. It has been proposed that the connective tissue growth factor (CTGF) plays a critical role in the advancement of pulmonary fibrosis by enhancing the accumulation of the extracellular matrix and exacerbating fibrosis. In this context, the significance of CTGF in pulmonary fibrosis is examined, and a summary of the development of drugs targeting CTGF for the treatment of pulmonary fibrosis is provided.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo , Fibrosis Pulmonar , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Humanos , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/metabolismo , Animales , Terapia Molecular Dirigida , Matriz Extracelular/metabolismo
4.
Sci Rep ; 14(1): 11131, 2024 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-38750140

RESUMEN

This study aimed to investigate the potential anti-fibrotic activity of vinpocetine in an experimental model of pulmonary fibrosis by bleomycin and in the MRC-5 cell line. Pulmonary fibrosis was induced in BALB/c mice by oropharyngeal aspiration of a single dose of bleomycin (5 mg/kg). The remaining induced animals received a daily dose of pirfenidone (as a standard anti-fibrotic drug) (300 mg/kg/PO) and vinpocetine (20 mg/kg/PO) on day 7 of the induction till the end of the experiment (day 21). The results of the experiment revealed that vinpocetine managed to alleviate the fibrotic endpoints by statistically improving (P ≤ 0.05) the weight index, histopathological score, reduced expression of fibrotic-related proteins in immune-stained lung sections, as well as fibrotic markers measured in serum samples. It also alleviated tissue levels of oxidative stress and inflammatory and pro-fibrotic mediators significantly elevated in bleomycin-only induced animals (P ≤ 0.05). Vinpocetine managed to express a remarkable attenuating effect in pulmonary fibrosis both in vivo and in vitro either directly by interfering with the classical TGF-ß1/Smad2/3 signaling pathway or indirectly by upregulating the expression of Nrf2 enhancing the antioxidant system, activating PPAR-γ and downregulating the NLRP3/NF-κB pathway making it a candidate for further clinical investigation in cases of pulmonary fibrosis.


Asunto(s)
Ratones Endogámicos BALB C , FN-kappa B , Proteína con Dominio Pirina 3 de la Familia NLR , PPAR gamma , Fibrosis Pulmonar , Transducción de Señal , Proteína Smad2 , Proteína smad3 , Factor de Crecimiento Transformador beta1 , Alcaloides de la Vinca , Animales , Alcaloides de la Vinca/farmacología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/inducido químicamente , Factor de Crecimiento Transformador beta1/metabolismo , PPAR gamma/metabolismo , Ratones , FN-kappa B/metabolismo , Proteína smad3/metabolismo , Proteína Smad2/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Transducción de Señal/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos , Humanos , Bleomicina/efectos adversos , Modelos Animales de Enfermedad , Masculino , Línea Celular , Estrés Oxidativo/efectos de los fármacos
5.
Arthritis Res Ther ; 26(1): 94, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702742

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales , Quimiocina CX3CL1 , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped , Fibrosis Pulmonar , Esclerodermia Sistémica , Piel , Animales , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Esclerodermia Sistémica/tratamiento farmacológico , Esclerodermia Sistémica/patología , Esclerodermia Sistémica/inmunología , Ratones , Quimiocina CX3CL1/metabolismo , Quimiocina CX3CL1/antagonistas & inhibidores , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/prevención & control , Piel/patología , Piel/efectos de los fármacos , Piel/metabolismo , Piel/inmunología , Fibrosis , Femenino , Ratones Endogámicos C57BL , Humanos , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/inmunología
6.
Cell Mol Life Sci ; 81(1): 206, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38709307

RESUMEN

The epithelial-mesenchymal transformation (EMT) process of alveolar epithelial cells is recognized as involved in the development of pulmonary fibrosis. Recent evidence has shown that lipopolysaccharide (LPS)-induced aerobic glycolysis of lung tissue and elevated lactate concentration are associated with the pathogenesis of sepsis-associated pulmonary fibrosis. However, it is uncertain whether LPS promotes the development of sepsis-associated pulmonary fibrosis by promoting lactate accumulation in lung tissue, thereby initiating EMT process. We hypothesized that monocarboxylate transporter-1 (MCT1), as the main protein for lactate transport, may be crucial in the pathogenic process of sepsis-associated pulmonary fibrosis. We found that high concentrations of lactate induced EMT while moderate concentrations did not. Besides, we demonstrated that MCT1 inhibition enhanced EMT process in MLE-12 cells, while MCT1 upregulation could reverse lactate-induced EMT. LPS could promote EMT in MLE-12 cells through MCT1 inhibition and lactate accumulation, while this could be alleviated by upregulating the expression of MCT1. In addition, the overexpression of MCT1 prevented LPS-induced EMT and pulmonary fibrosis in vivo. Altogether, this study revealed that LPS could inhibit the expression of MCT1 in mouse alveolar epithelial cells and cause lactate transport disorder, which leads to lactate accumulation, and ultimately promotes the process of EMT and lung fibrosis.


Asunto(s)
Transición Epitelial-Mesenquimal , Ácido Láctico , Lipopolisacáridos , Transportadores de Ácidos Monocarboxílicos , Fibrosis Pulmonar , Simportadores , Transportadores de Ácidos Monocarboxílicos/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/antagonistas & inhibidores , Animales , Transición Epitelial-Mesenquimal/efectos de los fármacos , Lipopolisacáridos/farmacología , Simportadores/metabolismo , Simportadores/genética , Simportadores/antagonistas & inhibidores , Ratones , Ácido Láctico/metabolismo , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/inducido químicamente , Ratones Endogámicos C57BL , Línea Celular , Masculino , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Células Epiteliales Alveolares/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
7.
Respir Res ; 25(1): 212, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38762455

RESUMEN

Paraquat (PQ) is a widely used herbicide and a common cause of poisoning that leads to pulmonary fibrosis with a high mortality rate. However, the underlying mechanisms of PQ-induced pulmonary fibrosis and whether pulmonary epithelial cell senescence is involved in the process remain elusive. In this study, PQ-induced pulmonary epithelial cell senescence and Hippo-YAP/TAZ activation were observed in both C57BL/6 mice and human epithelial cells. PQ-induced senescent pulmonary epithelial cells promoted lung fibroblast transformation through secreting senescence-associated secretory phenotype (SASP) factors. Yap/Taz knockdown in mice lungs significantly decreased the expression of downstream profibrotic protein Ctgf and senescent markers p16 and p21, and alleviated PQ-induced pulmonary fibrosis. Interfering YAP/TAZ in senescent human pulmonary epithelial cells resulted in decreased expression of the anti-apoptosis protein survivin and elevated level of apoptosis. In conclusion, our findings reveal a novel mechanism by which the involvement of Hippo-YAP/TAZ activation in pulmonary epithelial cell senescence mediates the pathogenesis of PQ-induced pulmonary fibrosis, thereby offering novel insights and potential targets for the clinical management of PQ poisoning as well as providing the mechanistic insight of the involvement of Yap/Taz activation in cell senescence in pulmonary fibrosis and its related pulmonary disorders. The YIN YANG balance between cell senescence and apoptosis is important to maintain the homeostasis of the lung, the disruption of which will lead to disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Senescencia Celular , Ratones Endogámicos C57BL , Paraquat , Fibrosis Pulmonar , Factores de Transcripción , Proteínas Señalizadoras YAP , Animales , Senescencia Celular/efectos de los fármacos , Senescencia Celular/fisiología , Proteínas Señalizadoras YAP/metabolismo , Humanos , Ratones , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Paraquat/toxicidad , Masculino , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Transactivadores/metabolismo , Transactivadores/genética
8.
Respir Res ; 25(1): 213, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38762465

RESUMEN

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.


Asunto(s)
Dieta Alta en Grasa , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad , Fibrosis Pulmonar , Animales , Masculino , Dieta Alta en Grasa/efectos adversos , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Ratones , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Fibrosis Pulmonar/prevención & control , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/tratamiento farmacológico , Inhibidores de PCSK9 , Atorvastatina/farmacología , Atorvastatina/uso terapéutico , Ratones Obesos , Proproteína Convertasa 9/metabolismo , Proproteína Convertasa 9/genética , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Hiperreactividad Bronquial/prevención & control , Hiperreactividad Bronquial/tratamiento farmacológico , Hiperreactividad Bronquial/metabolismo , Hiperreactividad Bronquial/fisiopatología , Anticuerpos Monoclonales Humanizados
9.
Lipids Health Dis ; 23(1): 98, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38570797

RESUMEN

Pulmonary fibrosis (PF) is a severe pulmonary disease with limited available therapeutic choices. Recent evidence increasingly points to abnormal lipid metabolism as a critical factor in PF pathogenesis. Our latest research identifies the dysregulation of low-density lipoprotein (LDL) is a new risk factor for PF, contributing to alveolar epithelial and endothelial cell damage, and fibroblast activation. In this study, we first integrative summarize the published literature about lipid metabolite changes found in PF, including phospholipids, glycolipids, steroids, fatty acids, triglycerides, and lipoproteins. We then reanalyze two single-cell RNA-sequencing (scRNA-seq) datasets of PF, and the corresponding lipid metabolomic genes responsible for these lipids' biosynthesis, catabolism, transport, and modification processes are uncovered. Intriguingly, we found that macrophage is the most active cell type in lipid metabolism, with almost all lipid metabolic genes being altered in macrophages of PF. In type 2 alveolar epithelial cells, lipid metabolic differentially expressed genes (DEGs) are primarily associated with the cytidine diphosphate diacylglycerol pathway, cholesterol metabolism, and triglyceride synthesis. Endothelial cells are partly responsible for sphingomyelin, phosphatidylcholine, and phosphatidylethanolamines reprogramming as their metabolic genes are dysregulated in PF. Fibroblasts may contribute to abnormal cholesterol, phosphatidylcholine, and phosphatidylethanolamine metabolism in PF. Therefore, the reprogrammed lipid profiles in PF may be attributed to the aberrant expression of lipid metabolic genes in different cell types. Taken together, these insights underscore the potential of targeting lipid metabolism in developing innovative therapeutic strategies, potentially leading to extended overall survival in individuals affected by PF.


Asunto(s)
Fibrosis Pulmonar , Humanos , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Análisis de Expresión Génica de una Sola Célula , Metabolismo de los Lípidos/genética , Células Endoteliales/metabolismo , Fosfolípidos/metabolismo , Colesterol/metabolismo , Fosfatidilcolinas
10.
J Med Chem ; 67(8): 6624-6637, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38588467

RESUMEN

The increased remodeling of the extracellular matrix (ECM) in pulmonary fibrosis (PF) generates bioactive ECM fragments called matricryptins, which include elastin-derived peptides (EDPs). The interaction between EDPs and their receptors, including elastin-binding protein (EBP), plays a crucial role in exacerbating fibrosis. Here, we present LXJ-02 for the first time, a novel ultralong-acting inhibitor that disrupts the EDPs/EBP peptide-protein interaction, promoting macrophages to secrete matrix metalloproteinase-12 (MMP-12), and showing great promise as a stable peptide. MMP-12 has traditionally been implicated in promoting inflammation and fibrosis in various acute and chronic diseases. However, we reveal a novel role of LXJ-02 that activates the macrophage-MMP-12 axis to increase MMP-12 expression and degrade ECM components like elastin. This leads to the preventing of PF while also improving EDP-EBP interaction. LXJ-02 effectively reverses PF in mouse models with minimal side effects, holding great promise as an excellent therapeutic agent for lung fibrosis.


Asunto(s)
Diseño de Fármacos , Elastina , Fibrosis Pulmonar , Receptores de Superficie Celular , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/metabolismo , Animales , Ratones , Elastina/química , Elastina/metabolismo , Humanos , Metaloproteinasa 12 de la Matriz/metabolismo , Péptidos/farmacología , Péptidos/química , Péptidos/síntesis química , Ratones Endogámicos C57BL , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino
11.
Clin Sci (Lond) ; 138(8): 537-554, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38577922

RESUMEN

Patients with pulmonary fibrosis (PF) often experience exacerbations of their disease, characterised by a rapid, severe deterioration in lung function that is associated with high mortality. Whilst the pathobiology of such exacerbations is poorly understood, virus infection is a trigger. The present study investigated virus-induced injury responses of alveolar and bronchial epithelial cells (AECs and BECs, respectively) from patients with PF and age-matched controls (Ctrls). Air-liquid interface (ALI) cultures of AECs, comprising type I and II pneumocytes or BECs were inoculated with influenza A virus (H1N1) at 0.1 multiplicity of infection (MOI). Levels of interleukin-6 (IL-6), IL-36γ and IL-1ß were elevated in cultures of AECs from PF patients (PF-AECs, n = 8-11), being markedly higher than Ctrl-AECs (n = 5-6), 48 h post inoculation (pi) (P<0.05); despite no difference in H1N1 RNA copy numbers 24 h pi. Furthermore, the virus-induced inflammatory responses of PF-AECs were greater than BECs (from either PF patients or controls), even though viral loads in the BECs were overall 2- to 3-fold higher than AECs. Baseline levels of the senescence and DNA damage markers, nuclear p21, p16 and H2AXγ were also significantly higher in PF-AECs than Ctrl-AECs and further elevated post-infection. Senescence induction using etoposide augmented virus-induced injuries in AECs (but not viral load), whereas selected senotherapeutics (rapamycin and mitoTEMPO) were protective. The present study provides evidence that senescence increases the susceptibility of AECs from PF patients to severe virus-induced injury and suggests targeting senescence may provide an alternative option to prevent or treat the exacerbations that worsen the underlying disease.


Asunto(s)
Células Epiteliales Alveolares , Subtipo H1N1 del Virus de la Influenza A , Fibrosis Pulmonar , Humanos , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Células Epiteliales Alveolares/virología , Células Epiteliales Alveolares/patología , Células Epiteliales Alveolares/metabolismo , Fibrosis Pulmonar/virología , Fibrosis Pulmonar/patología , Masculino , Gripe Humana/virología , Gripe Humana/complicaciones , Gripe Humana/patología , Persona de Mediana Edad , Femenino , Células Cultivadas , Anciano , Senescencia Celular , Estudios de Casos y Controles , Citocinas/metabolismo
12.
Artículo en Chino | MEDLINE | ID: mdl-38678001

RESUMEN

Silicosis is a common occupational disease, and its main characteristic pathological features are the formation of silicon nodules and diffuse pulmonary fibrosis. In the process of silicosis fibrosis, macrophages can be polarized into M1 macrophages and M2 macrophages. M1 macrophages play a pro-inflammatory role in the early stage of silicosis and release a variety of inflammatory factors, which is the core of inflammatory response. M2 macrophages promote inflammation resolution and tissue repair in silicosis fibrosis stage by secreting anti-inflammatory cytokines and pro-fibrotic mediators. M1/M2 polarization balance plays an important role in the occurrence and development of silicosis, and the regulation of macrophage polarization direction may play a positive role in the prevention and treatment of silicosis fibrosis. In this review, the role of macrophage polarization in silicosis fibrosis, the related signaling pathways regulating macrophage polarization in silicosis fibrosis, and the potential therapeutic targets based on macrophage polarization in silicosis fibrosis are reviewed, with a view to further strengthening the understanding of the mechanism of macrophage polarization in the pathogenesis and treatment of silicosis fibrosis.


Asunto(s)
Macrófagos , Fibrosis Pulmonar , Silicosis , Silicosis/patología , Humanos , Fibrosis Pulmonar/patología , Transducción de Señal , Citocinas/metabolismo
13.
Int J Mol Sci ; 25(8)2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38673961

RESUMEN

Mesenchymal stem cell-derived exosomes (MSC-Exos) are nano-sized extracellular vesicles which contain various MSC-sourced anti-fibrotic, immunoregulatory and angio-modulatory proteins (growth factors, immunoregulatory cytokines, chemokines), lipids, and nucleic acids (messenger RNA and microRNAs). Due to their lipid envelope, MSC-Exos easily by-pass all barriers in the body and deliver their cargo directly in target cells, modulating their viability, proliferation, phenotype and function. The results obtained in recently published experimental studies demonstrated beneficial effects of MSC-Exos in the treatment of lung fibrosis. MSC-Exos reduced activation of fibroblasts and prevented their differentiation in myofibroblasts. By delivering MSC-sourced immunoregulatory factors in lung-infiltrated monocytes and T cells, MSC-Exos modulate their function, alleviating on-going inflammation and fibrosis. MSC-Exos may also serve as vehicles for the target delivery of anti-fibrotic and immunomodulatory agents, enabling enhanced attenuation of lung fibrosis. Although numerous pre-clinical studies have demonstrated the therapeutic potential of MSC-Exos in the treatment of pulmonary fibrosis, there are several challenges that currently hinder their clinical implementation. Therefore, in this review article, we summarized current knowledge and we discussed future perspectives regarding molecular and cellular mechanisms which were responsible for the anti-fibrotic, anti-inflammatory and immunoregulatory properties of MSC-Exos, paving the way for their clinical use in the treatment of lung fibrosis.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , Fibrosis Pulmonar , Exosomas/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Fibrosis Pulmonar/terapia , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Animales
14.
Analyst ; 149(10): 2864-2876, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38619825

RESUMEN

Radiation-induced lung injury (RILI) is a dose-limiting toxicity for cancer patients receiving thoracic radiotherapy. As such, it is important to characterize metabolic associations with the early and late stages of RILI, namely pneumonitis and pulmonary fibrosis. Recently, Raman spectroscopy has shown utility for the differentiation of pneumonitic and fibrotic tissue states in a mouse model; however, the specific metabolite-disease associations remain relatively unexplored from a Raman perspective. This work harnesses Raman spectroscopy and supervised machine learning to investigate metabolic associations with radiation pneumonitis and pulmonary fibrosis in a mouse model. To this end, Raman spectra were collected from lung tissues of irradiated/non-irradiated C3H/HeJ and C57BL/6J mice and labelled as normal, pneumonitis, or fibrosis, based on histological assessment. Spectra were decomposed into metabolic scores via group and basis restricted non-negative matrix factorization, classified with random forest (GBR-NMF-RF), and metabolites predictive of RILI were identified. To provide comparative context, spectra were decomposed and classified via principal component analysis with random forest (PCA-RF), and full spectra were classified with a convolutional neural network (CNN), as well as logistic regression (LR). Through leave-one-mouse-out cross-validation, we observed that GBR-NMF-RF was comparable to other methods by measure of accuracy and log-loss (p > 0.10 by Mann-Whitney U test), and no methodology was dominant across all classification tasks by measure of area under the receiver operating characteristic curve. Moreover, GBR-NMF-RF results were directly interpretable and identified collagen and specific collagen precursors as top fibrosis predictors, while metabolites with immune and inflammatory functions, such as serine and histidine, were top pneumonitis predictors. Further support for GBR-NMF-RF and the identified metabolite associations with RILI was found as CNN interpretation heatmaps revealed spectral regions consistent with these metabolites.


Asunto(s)
Aprendizaje Automático , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Espectrometría Raman , Animales , Espectrometría Raman/métodos , Ratones , Metabolómica/métodos , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Neumonitis por Radiación/metabolismo , Neumonitis por Radiación/patología , Pulmón/efectos de la radiación , Pulmón/patología , Pulmón/metabolismo , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Análisis de Componente Principal , Redes Neurales de la Computación
15.
Life Sci ; 346: 122626, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38614295

RESUMEN

AIM: Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive condition with unknown aetiology that causes the lung parenchyma to scar incessantly, lowering the quality of life and hastening death. In this investigation, we studied the anti-fibrotic activity of Geneticin (a derivative of gentamycin) using in vitro and in vivo models. MAIN METHODS: The TGF-ß-mediated differentiation model was adopted to investigate (fibrotic marker's levels/expression) the anti-fibrotic activity of geneticin (GNC) in in-vitro scenarios (LL29 and DHLF cells). In vivo, the bleomycin (BLM)-induced pulmonary fibrosis model was employed by administering BLM intratracheally. Post 14 days of BLM administration, animals were treated with geneticin (6.25, 12.5, and 25 mg·kg-1) for another 14 days, and their therapeutic effect was investigated using a spectrum of techniques. KEY FINDINGS: RTqPCR and western-blot results revealed that geneticin treatment significantly attenuated the TGF-ß/BLM mediated fibrotic cascade of markers in both in-vitro and in-vivo models respectively. Further, the BLM-induced pulmonary fibrosis model revealed, that geneticin dose-dependently reduced the BLM-induced inflammatory cell infiltrations, and thickness of the alveoli walls, improved the structural distortion of the lung, and aided in improving the survival rate of the rats. Picrosirus and Masson's trichrome staining indicated that geneticin therapy reduced collagen deposition and, as a result, lung functional characteristics were improved as assessed by flexivent. Mechanistic studies have shown that geneticin reduced fibrosis by attenuating the TGF-ß/Smad through modulating the AMPK/SIRT1 signaling. SIGNIFICANCE: These findings suggest that geneticin may be a promising therapeutic agent for the treatment of pulmonary fibrosis in clinical settings.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Bleomicina , Fibrosis Pulmonar , Transducción de Señal , Sirtuina 1 , Factor de Crecimiento Transformador beta , Animales , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/inducido químicamente , Ratas , Sirtuina 1/metabolismo , Sirtuina 1/genética , Masculino , Bleomicina/toxicidad , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Smad/metabolismo , Ratas Sprague-Dawley , Modelos Animales de Enfermedad
16.
Int Immunopharmacol ; 133: 112067, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38608444

RESUMEN

Silicosis is one of the most common and severe types of pneumoconiosis and is characterized by lung dysfunction, persistent lung inflammation, pulmonary nodule formation, and irreversible pulmonary fibrosis. The transdifferentiation of fibroblasts into myofibroblasts is one of the main reasons for the exacerbation of silicosis. However, the underlying mechanism of transcription factors regulating silicosis fibrosis has not been clarified. The aim of this study was to investigate the potential mechanism of transcription factor FOXF1 in fibroblast transdifferentiation in silica-induced pulmonary fibrosis. Therefore, a silicosis mouse model was established, and we found that FOXF1 expression level was significantly down-regulated in the silicosis group, and after overexpression of FOXF1 by adeno-associated virus (AAV), FOXF1 expression level was up-regulated, and silicosis fibrosis was alleviated. In order to further explore the specific regulatory mechanism of FOXF1 in silicosis, we established a fibroblasts transdifferentiation model induced by TGF-ß in vitro. In the model, the expression levels of SMAD2/3 and P-SMAD2/3 were up-regulated, but the expression levels of SMAD2/3 and P-SMAD2/3 were down-regulated, inhibiting transdifferentiation and accumulation of extracellular matrix after the overexpressed FOXF1 plasmid was constructed. However, after silencing FOXF1, the expression levels of SMAD2/3 and P-SMAD2/3 were further up-regulated, aggravating transdifferentiation and accumulation of extracellular matrix. These results indicate that the activation of FOXF1 in fibroblasts can slow down the progression of silicosis fibrosis by inhibiting TGF-ß/SMAD2/3 classical pathway, which provides a new idea for further exploration of silicosis treatment.


Asunto(s)
Transdiferenciación Celular , Fibroblastos , Pulmón , Fibrosis Pulmonar , Transducción de Señal , Dióxido de Silicio , Proteína Smad2 , Proteína smad3 , Factor de Crecimiento Transformador beta , Animales , Fibroblastos/metabolismo , Proteína smad3/metabolismo , Proteína smad3/genética , Proteína Smad2/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Factor de Crecimiento Transformador beta/metabolismo , Ratones , Pulmón/patología , Dióxido de Silicio/toxicidad , Ratones Endogámicos C57BL , Silicosis/metabolismo , Silicosis/patología , Masculino , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Modelos Animales de Enfermedad , Humanos , Células Cultivadas
17.
Toxicol Appl Pharmacol ; 486: 116939, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38643951

RESUMEN

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.


Asunto(s)
Antiinflamatorios , Antioxidantes , Bleomicina , Candida parapsilosis , Ratones Endogámicos C57BL , MicroARNs , Fibrosis Pulmonar , Proteína smad3 , Tensoactivos , Factor de Crecimiento Transformador beta1 , Animales , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/prevención & control , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/metabolismo , Bleomicina/toxicidad , Antioxidantes/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Antiinflamatorios/farmacología , Proteína smad3/metabolismo , Ratones , Candida parapsilosis/efectos de los fármacos , Tensoactivos/farmacología , MicroARNs/metabolismo , Masculino , Transducción de Señal/efectos de los fármacos , Bacillus , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ácidos Oléicos
18.
Int J Mol Sci ; 25(8)2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38673771

RESUMEN

Using a lipopolysaccharide model of acute lung injury, we previously showed that endothelin-1 (ET-1), a potent mediator of vasoconstriction, may act as a "gatekeeper" for the influx of inflammatory cells into the lung. These studies provided a rationale for testing the effect of HJP272, an endothelin receptor antagonist (ERA), in hamster models of pulmonary fibrosis induced by intratracheal instillation of either bleomycin (BLM) or amiodarone (AM). To determine the temporal effects of blocking ET-1 activity, animals were given HJP272 either 1 h before initiation of lung injury or 24 h afterward. The results indicated that pretreatment with this agent caused significant reductions in various inflammatory parameters, whereas post-treatment was ineffective. This finding suggests that ERAs are only effective at a very early stage of pulmonary fibrosis and explains their lack of success in clinical trials involving patients with this disease. Nevertheless, ERAs could serve as prophylactic agents when combined with drugs that may induce pulmonary fibrosis. Furthermore, developing a biomarker for the initial changes in the lung extracellular matrix could increase the efficacy of ERAs and other therapeutic agents in preventing the progression of the disease. While no such biomarker currently exists, we propose the ratio of free to peptide-bound desmosine, a unique crosslink of elastin, as a potential candidate for detecting the earliest modifications in lung microarchitecture associated with pulmonary fibrosis.


Asunto(s)
Antagonistas de los Receptores de Endotelina , Fibrosis Pulmonar , Animales , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Antagonistas de los Receptores de Endotelina/farmacología , Antagonistas de los Receptores de Endotelina/uso terapéutico , Bleomicina/efectos adversos , Cricetinae , Modelos Animales de Enfermedad , Masculino , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Humanos , Endotelina-1/metabolismo
19.
Respir Res ; 25(1): 176, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658970

RESUMEN

BACKGROUND: Abnormal lipid metabolism has recently been reported as a crucial signature of idiopathic pulmonary fibrosis (IPF). However, the origin and biological function of the lipid and possible mechanisms of increased lipid content in the pathogenesis of IPF remains undetermined. METHODS: Oil-red staining and immunofluorescence analysis were used to detect lipid accumulation in mouse lung fibrosis frozen sections, Bleomycin-treated human type II alveolar epithelial cells (AECIIs) and lung fibroblast. Untargeted Lipid omics analysis was applied to investigate differential lipid species and identified LysoPC was utilized to treat human lung fibroblasts and mice. Microarray and single-cell RNA expression data sets identified lipid metabolism-related differentially expressed genes. Gain of function experiment was used to study the function of 3-hydroxy-3-methylglutaryl-Coa Synthase 2 (HMGCS2) in regulating AECIIs lipid metabolism. Mice with AECII-HMGCS2 high were established by intratracheally delivering HBAAV2/6-SFTPC- HMGCS2 adeno-associated virus. Western blot, Co-immunoprecipitation, immunofluorescence, site-directed mutation and flow cytometry were utilized to investigate the mechanisms of HMGCS2-mediated lipid metabolism in AECIIs. RESULTS: Injured AECIIs were the primary source of accumulated lipids in response to Bleomycin stimulation. LysoPCs released by injured AECIIs could activate lung fibroblasts, thus promoting the progression of pulmonary fibrosis. Mechanistically, HMGCS2 was decreased explicitly in AECIIs and ectopic expression of HMGCS2 in AECIIs using the AAV system significantly alleviated experimental mouse lung fibrosis progression via modulating lipid degradation in AECIIs through promoting CPT1A and CPT2 expression by interacting with PPARα. CONCLUSIONS: These data unveiled a novel etiological mechanism of HMGCS2-mediated AECII lipid metabolism in the genesis and development of pulmonary fibrosis and provided a novel target for clinical intervention.


Asunto(s)
Regulación hacia Abajo , Fibroblastos , Hidroximetilglutaril-CoA Sintasa , Metabolismo de los Lípidos , Ratones Endogámicos C57BL , Animales , Humanos , Masculino , Ratones , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Bleomicina/toxicidad , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/patología , Hidroximetilglutaril-CoA Sintasa/metabolismo , Hidroximetilglutaril-CoA Sintasa/genética , Hidroximetilglutaril-CoA Sintasa/biosíntesis , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/genética , Metabolismo de los Lípidos/fisiología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/genética
20.
Mol Med ; 30(1): 54, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38649802

RESUMEN

BACKGROUND: Bleomycin, a potent antitumor agent, is limited in clinical use due to the potential for fatal pulmonary toxicity. The accelerated DNA damage and senescence in alveolar epithelial cells (AECs) is considered a key factor in the development of lung pathology. Understanding the mechanisms for bleomycin-induced lung injury is crucial for mitigating its adverse effects. METHODS: Human lung epithelial (A549) cells were exposed to bleomycin and subsequently assessed for cellular senescence, DNA damage, and double-strand break (DSB) repair. The impact of Rad51 overexpression on DSB repair and senescence in AECs was evaluated in vitro. Additionally, bleomycin was intratracheally administered in C57BL/6 mice to establish a pulmonary fibrosis model. RESULTS: Bleomycin exposure induced dose- and time-dependent accumulation of senescence hallmarks and DNA lesions in AECs. These effects are probably due to the inhibition of Rad51 expression, consequently suppressing homologous recombination (HR) repair. Mechanistic studies revealed that bleomycin-mediated transcriptional inhibition of Rad51 might primarily result from E2F1 depletion. Furthermore, the genetic supplement of Rad51 substantially mitigated bleomycin-mediated effects on DSB repair and senescence in AECs. Notably, decreased Rad51 expression was also observed in the bleomycin-induced mouse pulmonary fibrosis model. CONCLUSIONS: Our works suggest that the inhibition of Rad51 plays a pivotal role in bleomycin-induced AECs senescence and lung injury, offering potential strategies to alleviate the pulmonary toxicity of bleomycin.


Asunto(s)
Bleomicina , Senescencia Celular , Reparación del ADN , Recombinasa Rad51 , Bleomicina/efectos adversos , Recombinasa Rad51/metabolismo , Recombinasa Rad51/genética , Animales , Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , Humanos , Ratones , Reparación del ADN/efectos de los fármacos , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Células A549 , Daño del ADN/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de los fármacos , Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F1/genética , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA