Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.844
Filtrar
1.
Int J Rheum Dis ; 27(5): e15164, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38706209

RESUMO

BACKGROUND: JAK inhibitors are well known for the treatment of rheumatoid arthritis (RA), but whether they can be used to treat pulmonary fibrosis, a common extra-articular disease of RA, remains to be clarified. METHODS: A jak2 inhibitor, CEP33779 (CEP), was administered to a rat model of RA-associated interstitial lung disease to observe the degree of improvement in both joint swelling and pulmonary fibrosis. HFL1 cells were stimulated with TGF-ß1 to observe the expression of p-JAK2. Then, different concentrations of related gene inhibitors (JAK2, TGFß-R1/2, and p-STAT3) or silencers (STAT3, JAK2) were administered to HFL1 cells, and the expression levels of related proteins were detected to explore the underlying mechanisms of action. RESULTS: CEP not only reduced the degree of joint swelling and inflammation in rats but also improved lung function, inhibited the pro-inflammatory factors IL-1ß and IL-6, reduced lung inflammation and collagen deposition, and alleviated lung fibrosis. CEP decreased the expression levels of TGFß-R2, p-SMAD, p-STAT3, and ECM proteins in rat lung tissues. TGF-ß1 induced HFL1 cells to highly express p-JAK2, with the most pronounced expression at 48 h. The levels of p-STAT3, p-SMAD3, and ECM-related proteins were significantly reduced after inhibition of either JAK2 or STAT3. CONCLUSION: JAK2 inhibitors may be an important and novel immunotherapeutic drug that can improve RA symptoms while also delaying or blocking the development of associated pulmonary fibrotic disease. The mechanism may be related to the downregulation of p-STAT3 protein via inhibition of the JAK2/STAT signaling pathway, which affects the phosphorylation of SMAD3.


Assuntos
Modelos Animais de Doenças , Regulação para Baixo , Isoquinolinas , Janus Quinase 2 , Pulmão , Fibrose Pulmonar , Piridinas , Pirróis , Transdução de Sinais , Proteína Smad3 , Animais , Proteína Smad3/metabolismo , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/metabolismo , Janus Quinase 2/metabolismo , Janus Quinase 2/antagonistas & inibidores , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Pulmão/enzimologia , Masculino , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/metabolismo , Humanos , Ratos Sprague-Dawley , Inibidores de Janus Quinases/farmacologia , Inibidores de Janus Quinases/uso terapêutico , Linhagem Celular , Inibidores de Proteínas Quinases/farmacologia , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Artrite Experimental/tratamento farmacológico , Artrite Experimental/metabolismo , Artrite Experimental/enzimologia , Anti-Inflamatórios/farmacologia , Ratos
2.
Respir Res ; 25(1): 175, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38654248

RESUMO

BACKGROUND: Two isoforms of Phosphoinositide 3-kinase (PI3K), p110γ and p110δ, are predominantly expressed in leukocytes and represent attractive therapeutic targets for the treatment of allergic asthma. The study aim was to assess the impact of administration of an inhaled PI3Kγδ inhibitor (AZD8154) in a rat model of asthma. METHODS: Firstly, we checked that the tool compound, AZD8154, inhibited rat PI3K γ & δ kinases using rat cell-based assays. Subsequently, a time-course study was conducted in a rat model of asthma to assess PI3K activity in the lung and how it is temporally associated with other key transcription pathways and asthma like features of the model. Finally, the impact on lung dosed AZD8154 on target engagement, pathway specificity, airway inflammation and lung function changes was assessed. RESULTS: Data showed that AZD8154 could inhibit rat PI3K γ & δ isoforms and, in a rat model of allergic asthma the PI3K pathway was activated in the lung. Intratracheal administration of AZD8154 caused a dose related suppression PI3K pathway activation (reduction in pAkt) and unlike after budesonide treatment, STAT and NF-κB pathways were not affected by AZD8154. The suppression of the PI3K pathway led to a marked inhibition of airway inflammation and reduction in changes in lung function. CONCLUSION: These data show that a dual PI3Kγδ inhibitor suppress key features of disease in a rat model of asthma to a similar degree as budesonide and indicate that dual PI3Kγδ inhibition may be an effective treatment for people suffering from allergic asthma.


Assuntos
Asma , Modelos Animais de Doenças , Animais , Asma/tratamento farmacológico , Asma/metabolismo , Ratos , Masculino , Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , Ratos Sprague-Dawley , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase/uso terapêutico , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Pulmão/enzimologia , Relação Dose-Resposta a Droga , Inibidores de Proteínas Quinases/farmacologia , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Antiasmáticos/farmacologia , Ovalbumina/toxicidade
3.
Biomed Pharmacother ; 174: 116480, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38547765

RESUMO

Sepsis is caused by an inadequate or dysregulated host response to infection. Enzymes causing cellular degradation are matrix metalloproteinases (MMPs). Lipopolysaccharide (LPS) is used in models of sepsis in laboratory settings The aim of the study was to measure MMP 2 and 12 concentrations in spleen and lungs in rats in which septic shock was induced by LPS. The experiment was carried out on 40 male Wistar rats (5 groups of 8): 0. controls 1. administered LPS 2. administered bestatin 3. LPS and bestatin 4.bestatin and after 6 hours LPS Animals were decapitated. Lungs and spleens were collected. Concentrations of MMP-2 and MMP-12 were determined using immunoenzymatic methods. Mean (±SD) MMP-2 in the controls was 43.57 ± 20.53 ng/ml in the lungs and 1.7 ± 0.72 ng/ml in the spleen; Group 1: 31.28 ± 13.13 ng/ml, 0.83 ± 0.8 ng/ml; Group 2: 44.24 ± 22.75 ng /ml, 1.01 ± 0.32 ng/ml; Group 3: 35.94 ± 15.13 ng/ml, 0.41 ± 0.03 ng/ml; Group 4:79.42 ± 44.70 ng/ml, 0.45 ± 0.15, respectively. Mean MMP-12 in controls was 19.79 ± 10.01 ng/ml in lungs and 41.13 ± 15.99 ng/ml in the spleen; Group 1:27.97 ± 15.1 ng/ml; 40.44 ± 11.2 ng/ml; Group 2: 37.93 ± 25.38 ng/ml 41.05 ± 18.08 ng/ml; Group 3: 40.59 ± 11.46 ng/ml, 35.16 ± 12.89 ng/ml; Group 4: 39.4 ± 17.83 ng/ml, 42.04 ± 12.35 ng/ml, respectively. CONCLUSIONS: 1. Bestatin reduces MMP 2 and 12 levels in spleen and lungs. 2. Treatment with bestatin minimizes the effect of LPS.


Assuntos
Modelos Animais de Doenças , Leucina , Leucina/análogos & derivados , Lipopolissacarídeos , Pulmão , Metaloproteinase 12 da Matriz , Metaloproteinase 2 da Matriz , Ratos Wistar , Sepse , Baço , Animais , Baço/efeitos dos fármacos , Baço/metabolismo , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Pulmão/patologia , Pulmão/metabolismo , Sepse/tratamento farmacológico , Sepse/induzido quimicamente , Metaloproteinase 12 da Matriz/metabolismo , Ratos , Leucina/farmacologia , Leucina/uso terapêutico , Inibidores de Metaloproteinases de Matriz/farmacologia
4.
Exp Mol Med ; 56(2): 478-490, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38413821

RESUMO

Idiopathic pulmonary fibrosis (IPF) is characterized by aberrant lung remodeling and the excessive accumulation of extracellular matrix (ECM) proteins. In a previous study, we found that the levels of ornithine aminotransferase (OAT), a principal enzyme in the proline metabolism pathway, were increased in the lungs of patients with IPF. However, the precise role played by OAT in the pathogenesis of IPF is not yet clear. The mechanism by which OAT affects fibrogenesis was assessed in vitro using OAT-overexpressing and OAT-knockdown lung fibroblasts. The therapeutic effects of OAT inhibition were assessed in the lungs of bleomycin-treated mice. OAT expression was increased in fibrotic areas, principally in interstitial fibroblasts, of lungs affected by IPF. OAT levels in the bronchoalveolar lavage fluid of IPF patients were inversely correlated with lung function. The survival rate was significantly lower in the group with an OAT level >75.659 ng/mL than in the group with an OAT level ≤75.659 ng/mL (HR, 29.53; p = 0.0008). OAT overexpression and knockdown increased and decreased ECM component production by lung fibroblasts, respectively. OAT knockdown also inhibited transforming growth factor-ß1 (TGF)-ß1 activity and TGF-ß1 pathway signaling. OAT overexpression increased the generation of mitochondrial reactive oxygen species (ROS) by activating proline dehydrogenase. The OAT inhibitor L-canaline significantly attenuated bleomycin-induced lung injury and fibrosis. In conclusion, increased OAT levels in lungs affected by IPF contribute to the progression of fibrosis by promoting excessive mitochondrial ROS production, which in turn activates TGF-ß1 signaling. OAT may be a useful target for treating patients with fibrotic lung diseases, including IPF.


Assuntos
Fibrose Pulmonar Idiopática , Fator de Crescimento Transformador beta1 , Animais , Humanos , Camundongos , Bleomicina , Proteínas da Matriz Extracelular , Fibrose , Pulmão/enzimologia , Ornitina-Oxo-Ácido Transaminase , Espécies Reativas de Oxigênio
5.
Proc Natl Acad Sci U S A ; 119(24): e2201707119, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35671428

RESUMO

A number of inflammatory lung diseases, including chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, and pneumonia, are modulated by WNT/ß-catenin signaling. However, the underlying molecular mechanisms remain unclear. Here, starting with a forward genetic screen in mouse, we identify the WNT coreceptor Related to receptor tyrosine kinase (RYK) acting in mesenchymal tissues as a cell survival and antiinflammatory modulator. Ryk mutant mice exhibit lung hypoplasia and inflammation as well as alveolar simplification due to defective secondary septation, and deletion of Ryk specifically in mesenchymal cells also leads to these phenotypes. By analyzing the transcriptome of wild-type and mutant lungs, we observed the up-regulation of proapoptotic and inflammatory genes whose expression can be repressed by WNT/RYK signaling in vitro. Moreover, mesenchymal Ryk deletion at postnatal and adult stages can also lead to lung inflammation, thus indicating a continued role for WNT/RYK signaling in homeostasis. Our results indicate that RYK signaling through ß-catenin and Nuclear Factor kappa B (NF-κB) is part of a safeguard mechanism against mesenchymal cell death, excessive inflammatory cytokine production, and inflammatory cell recruitment and accumulation. Notably, RYK expression is down-regulated in the stromal cells of pneumonitis patient lungs. Altogether, our data reveal that RYK signaling plays critical roles as an antiinflammatory modulator during lung development and homeostasis and provide an animal model to further investigate the etiology of, and therapeutic approaches to, inflammatory lung diseases.


Assuntos
Pneumonia , Receptores Proteína Tirosina Quinases , Via de Sinalização Wnt , beta Catenina , Animais , Humanos , Pulmão/enzimologia , Pulmão/crescimento & desenvolvimento , Mesoderma/metabolismo , Camundongos , NF-kappa B/metabolismo , Pneumonia/enzimologia , Pneumonia/genética , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Células Estromais/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
6.
J Pharm Biomed Anal ; 211: 114518, 2022 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-35124452

RESUMO

The inhalation of peptides comes with the advantage of directly targeting the lung as tissue of interest. However, peptides are often rapidly metabolized in lung tissue through proteolytic cleavage. We have developed an assay workflow to obtain half-life and metabolite ID data for peptides incubated with four proteases abundant in lungs of asthma and COPD patients. The assay system has been validated using 28 structurally diverse linear and cyclic peptides with a molecular weight between 708 and 5808 Da. Experimental conditions for incubation, sample preparation, chromatography, data acquisition and analysis are compatible with the required throughput in early stage peptide projects. Together with co-crystal structures and Ala scans, we are using the described assay workflow to guide the first chemical modifications of peptide hits in early respiratory drug discovery projects.


Assuntos
Peptídeo Hidrolases , Peptídeos , Administração por Inalação , Asma/tratamento farmacológico , Asma/enzimologia , Ensaios de Triagem em Larga Escala , Humanos , Pulmão/enzimologia , Peptídeo Hidrolases/metabolismo , Peptídeos/administração & dosagem , Peptídeos/química , Peptídeos/farmacocinética , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/enzimologia
7.
Respir Res ; 23(1): 20, 2022 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-35130915

RESUMO

BACKGROUND: Tyrosine kinase activation plays an important role in the progression of pulmonary fibrosis. In this study, we analyzed the expression of 612 kinase-coding and cancer-related genes using next-generation sequencing to identify potential therapeutic targets for idiopathic pulmonary fibrosis (IPF). METHODS: Thirteen samples from five patients with IPF (Cases 1-5) and eight samples from four patients without IPF (control) were included in this study. Six of the thirteen samples were obtained from different lung segments of a single patient who underwent bilateral pneumonectomy. Gene expression analysis of IPF lung tissue samples (n = 13) and control samples (n = 8) was performed using SureSelect RNA Human Kinome Kit. The expression of the selected genes was further confirmed at the protein level by immunohistochemistry (IHC). RESULTS: Gene expression analysis revealed a correlation between the gene expression signatures and the degree of fibrosis, as assessed by Ashcroft score. In addition, the expression analysis indicated a stronger heterogeneity among the IPF lung samples than among the control lung samples. In the integrated analysis of the 21 samples, DCLK1 and STK33 were found to be upregulated in IPF lung samples compared to control lung samples. However, the top most upregulated genes were distinct in individual cases. DCLK1, PDK4, and ERBB4 were upregulated in IPF case 1, whereas STK33, PIM2, and SYK were upregulated in IPF case 2. IHC revealed that these proteins were expressed in the epithelial layer of the fibrotic lesions. CONCLUSIONS: We performed a comprehensive kinase expression analysis to explore the potential therapeutic targets for IPF. We found that DCLK1 and STK33 may serve as potential candidate targets for molecular targeted therapy of IPF. In addition, PDK4, ERBB4, PIM2, and SYK might also serve as personalized therapeutic targets of IPF. Additional large-scale studies are warranted to develop personalized therapies for patients with IPF.


Assuntos
Regulação da Expressão Gênica , Fibrose Pulmonar Idiopática/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Pulmão/enzimologia , Fosfotransferases/genética , RNA/genética , Idoso , Feminino , Seguimentos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Fibrose Pulmonar Idiopática/metabolismo , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Fosfotransferases/biossíntese , Estudos Retrospectivos
8.
Am J Physiol Regul Integr Comp Physiol ; 322(2): R126-R135, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34984926

RESUMO

Many lung diseases are caused by an excessive inflammatory response, and inflammatory lung diseases are often modeled using lipopolysaccharide (LPS) in mice. Cyclooxygenase-2 (COX-2) encoded by the Ptgs2 gene is induced in response to inflammatory stimuli including LPS. The objective of this study was to test the hypothesis that mice deficient in COX-2 (Ptgs2-/-) will be protected from LPS-induced lung injury. Wild-type (WT; CD1 mice) and Ptgs2-/- mice (on a CD1 background) were treated with LPS or vehicle for 24 h. LPS treatment resulted in histological evidence of lung injury, which was attenuated in the Ptgs2-/- mice. LPS treatment increased the mRNA levels for tumor necrosis factor-α, interleukin-10, and monocyte chemoattractant protein-1 in the lungs of WT mice, and the LPS-induced increases in these levels were attenuated in the Ptgs2-/- mice. The protein levels of active caspase-3 and caspase-9 were lower in the LPS-treated lungs of Ptgs2-/- mice than in LPS-treated WT mice, as were the number of terminal deoxynucleotide transferase dUTP nick end labeling-positive cells in lung sections. LPS exposure resulted in a greater lung wet-to-dry weight ratio (W/D) in WT mice, suggestive of pulmonary edema, while in LPS-treated Ptgs2-/- mice, the W/D was not different from controls and less than in LPS-treated WT mice. These results demonstrate that COX-2 is involved in the inflammatory response to LPS and suggest that COX-2 not only acts as a downstream participant in the inflammatory response, but also acts as a regulator of the inflammatory response likely through a feed-forward mechanism following LPS stimulation.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Apoptose , Ciclo-Oxigenase 2/deficiência , Pulmão/enzimologia , Pneumonia/prevenção & controle , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/enzimologia , Lesão Pulmonar Aguda/patologia , Animais , Caspase 3/metabolismo , Caspase 9/metabolismo , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Ciclo-Oxigenase 2/genética , Modelos Animais de Doenças , Feminino , Interleucina-10/genética , Interleucina-10/metabolismo , Lipopolissacarídeos , Pulmão/patologia , Masculino , Camundongos Knockout , Pneumonia/induzido quimicamente , Pneumonia/enzimologia , Pneumonia/patologia , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
9.
Nat Commun ; 13(1): 318, 2022 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-35031603

RESUMO

Lung emphysema and chronic bronchitis are the two most common causes of chronic obstructive pulmonary disease. Excess macrophage elastase MMP-12, which is predominantly secreted from alveolar macrophages, is known to mediate the development of lung injury and emphysema. Here, we discovered the endolysosomal cation channel mucolipin 3 (TRPML3) as a regulator of MMP-12 reuptake from broncho-alveolar fluid, driving in two independently generated Trpml3-/- mouse models enlarged lung injury, which is further exacerbated after elastase or tobacco smoke treatment. Mechanistically, using a Trpml3IRES-Cre/eR26-τGFP reporter mouse model, transcriptomics, and endolysosomal patch-clamp experiments, we show that in the lung TRPML3 is almost exclusively expressed in alveolar macrophages, where its loss leads to defects in early endosomal trafficking and endocytosis of MMP-12. Our findings suggest that TRPML3 represents a key regulator of MMP-12 clearance by alveolar macrophages and may serve as therapeutic target for emphysema and chronic obstructive pulmonary disease.


Assuntos
Macrófagos Alveolares/enzimologia , Metaloproteinase 12 da Matriz/metabolismo , Elastase Pancreática/metabolismo , Enfisema Pulmonar/enzimologia , Canais de Potencial de Receptor Transitório/deficiência , Animais , Modelos Animais de Doenças , Endossomos/metabolismo , Feminino , Humanos , Pulmão/enzimologia , Metaloproteinase 12 da Matriz/genética , Camundongos , Camundongos Knockout , Elastase Pancreática/genética , Enfisema Pulmonar/genética , Enfisema Pulmonar/metabolismo , Canais de Potencial de Receptor Transitório/genética
10.
Biosci Rep ; 42(1)2022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-34750610

RESUMO

BACKGROUND: Acute lung injury (ALI) is a respiratory disease with high morbidity and mortality rates. Currently, there is no effective treatment to complement mechanical ventilation. Exosomes and microRNAs (miRNAs) are promising agents for the management of this disease. METHODS: Exosomes were isolated from mouse bone marrow stromal stem cells (BMSCs). The levels of two miRNAs, miR-542-3P and miR-150, in exosomes were determined using RT-PCR, and miR-150 was selected for further study. ALI model was established in mice using lipopolysaccharides, and then, they were treated with saline, exosomes, miRNA agomirs, or miRNA antagomirs. The concentrations of TNF-α, IL-6, and IL-1ß and the number of neutrophils and macrophages in the bronchoalveolar lavage fluid were measured. The wet/dry weight ratio of the lung tissue was calculated, and tissue pathology and apoptosis were observed using hematoxylin and eosin and terminal deoxynucleotidyl transferase dUTP nick-end labeling staining. CD34 and VE-cadherin expression was detected using immunofluorescence. Proteins associated with apoptosis and MAPK signaling were detected using Western blotting, and miR-150 expression in lung tissue was evaluated using RT-PCR. RESULTS: We successfully isolated BMSCs and exosomes and showed that the level of miR-150 was significantly higher than that of miR-542-3p. Exosomes and miR-150 reduced inflammation and lung edema while maintaining the integrity of the alveolar structure. They also mitigated microvascular endothelial cell injury by regulating the caspase-3, Bax/Bcl-2, and MAPK signaling. CONCLUSIONS: Exosomal miR-150 attenuates lipopolysaccharide-induced ALI through the MAPK pathway.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Exossomos/transplante , Pulmão/irrigação sanguínea , Transplante de Células-Tronco Mesenquimais , MicroRNAs/metabolismo , Microvasos/enzimologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/enzimologia , Lesão Pulmonar Aguda/patologia , Animais , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Exossomos/genética , Exossomos/metabolismo , Lipopolissacarídeos , Pulmão/enzimologia , Pulmão/patologia , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Transdução de Sinais
11.
J Mol Cell Cardiol ; 164: 13-16, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34774871

RESUMO

Aged males disproportionately succumb to increased COVID-19 severity, hospitalization, and mortality compared to females. Angiotensin-converting enzyme 2 (ACE2) and transmembrane protease, serine 2 (TMPRSS2) facilitate SARS-CoV-2 viral entry and may have sexually dimorphic regulation. As viral load dictates disease severity, we investigated the expression, protein levels, and activity of ACE2 and TMPRSS2. Our data reveal that aged males have elevated ACE2 in both mice and humans across organs. We report the first comparative study comprehensively investigating the impact of sex and age in murine and human levels of ACE2 and TMPRSS2, to begin to elucidate the sex bias in COVID-19 severity.


Assuntos
Envelhecimento/metabolismo , Enzima de Conversão de Angiotensina 2/biossíntese , COVID-19/epidemiologia , Regulação Enzimológica da Expressão Gênica , Receptores Virais/biossíntese , SARS-CoV-2/fisiologia , Caracteres Sexuais , Envelhecimento/genética , Enzima de Conversão de Angiotensina 2/genética , Animais , Suscetibilidade a Doenças , Feminino , Coração/virologia , Humanos , Intestino Delgado/enzimologia , Intestino Delgado/virologia , Rim/enzimologia , Rim/virologia , Pulmão/enzimologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Miocárdio/enzimologia , Especificidade de Órgãos , Receptores Virais/genética , Serina Endopeptidases/biossíntese , Serina Endopeptidases/genética , Adulto Jovem
12.
Am J Physiol Lung Cell Mol Physiol ; 322(2): L204-L223, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34878944

RESUMO

During newborn lung injury, excessive activity of lysyl oxidases (LOXs) disrupts extracellular matrix (ECM) formation. Previous studies indicate that TGFß activation in the O2-injured mouse pup lung increases lysyl oxidase (LOX) expression. But how TGFß regulates this, and whether the LOXs generate excess pulmonary aldehydes are unknown. First, we determined that O2-mediated lung injury increases LOX protein expression in TGFß-stimulated pup lung interstitial fibroblasts. This regulation appeared to be direct; this is because TGFß treatment also increased LOX protein expression in isolated pup lung fibroblasts. Then using a fibroblast cell line, we determined that TGFß stimulates LOX expression at a transcriptional level via Smad2/3-dependent signaling. LOX is translated as a pro-protein that requires secretion and extracellular cleavage before assuming amine oxidase activity and, in some cells, reuptake with nuclear localization. We found that pro-LOX is processed in the newborn mouse pup lung. Also, O2-mediated injury was determined to increase pro-LOX secretion and nuclear LOX immunoreactivity particularly in areas populated with interstitial fibroblasts and exhibiting malformed ECM. Then, using molecular probes, we detected increased aldehyde levels in vivo in O2-injured pup lungs, which mapped to areas of increased pro-LOX secretion in lung sections. Increased activity of LOXs plays a critical role in the aldehyde generation; an inhibitor of LOXs prevented the elevation of aldehydes in the O2-injured pup lung. These results reveal new mechanisms of TGFß and LOX in newborn lung disease and suggest that aldehyde-reactive probes might have utility in sensing the activation of LOXs in vivo during lung injury.


Assuntos
Aldeídos/metabolismo , Lesão Pulmonar/metabolismo , Pulmão/enzimologia , Pulmão/patologia , Proteína-Lisina 6-Oxidase/metabolismo , Aldeídos/química , Animais , Animais Recém-Nascidos , Embrião de Mamíferos/patologia , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Regulação Enzimológica da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Sondas Moleculares/metabolismo , Células NIH 3T3 , Proteína-Lisina 6-Oxidase/genética , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Transdução de Sinais , Proteínas Smad/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo
13.
J Pathol ; 256(3): 256-261, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34859884

RESUMO

COVID-19 is a pandemic with high morbidity and mortality. In an autopsy cohort of COVID-19 patients, we found extensive accumulation of the tryptophan degradation products 3-hydroxy-anthranilic acid and quinolinic acid in the lungs, heart, and brain. This was not related to the expression of the tryptophan-catabolizing indoleamine 2,3-dioxygenase (IDO)-1, but rather to that of its isoform IDO-2, which otherwise is expressed rarely. Bioavailability of tryptophan is an absolute requirement for proper cell functioning and synthesis of hormones, whereas its degradation products can cause cell death. Markers of apoptosis and severe cellular stress were associated with IDO-2 expression in large areas of lung and heart tissue, whereas affected areas in brain were more restricted. Analyses of tissue, cerebrospinal fluid, and sequential plasma samples indicate early initiation of the kynurenine/aryl-hydrocarbon receptor/IDO-2 axis as a positive feedback loop, potentially leading to severe COVID-19 pathology. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Assuntos
Encéfalo/enzimologia , COVID-19/enzimologia , Indolamina-Pirrol 2,3,-Dioxigenase/análise , Pulmão/enzimologia , Miocárdio/enzimologia , Ácido 3-Hidroxiantranílico/análise , Adulto , Idoso , Apoptose , Autopsia , Encéfalo/patologia , COVID-19/mortalidade , COVID-19/patologia , COVID-19/virologia , Humanos , Cinurenina/análise , Pulmão/patologia , Pessoa de Meia-Idade , Miocárdio/patologia , Estudos Prospectivos , Ácido Quinolínico/análise , Índice de Gravidade de Doença , Triptofano/análise
14.
Int J Mol Sci ; 22(22)2021 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-34830188

RESUMO

Cytochrome P450 2A13 is an omitted brother of CYP2A6 that has an important role in the drug metabolism of liver. Due to extrahepatic expression, it has gained less attention than CYP2A6, despite the fact that it plays a significant role in toxicant-induced pulmonary lesions and, therefore, lung cancer. The purpose of this mini-review is to summarize the basic knowledge about this enzyme in relation to the substrates, inhibitors, genetic polymorphisms, and transcriptional regulation that are known so far (September 2021).


Assuntos
Hidrocarboneto de Aril Hidroxilases/genética , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Pulmão/metabolismo , Polimorfismo Genético , Animais , Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Hidrocarboneto de Aril Hidroxilases/metabolismo , Humanos , Pulmão/enzimologia , Pulmão/patologia , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/patologia , Metoxaleno/farmacologia , Especificidade por Substrato
15.
Cell Rep ; 37(6): 109921, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34758300

RESUMO

Regulatory T (Treg) cells are critical for immunological tolerance and immune homeostasis. Treg cells strongly rely on mitochondrial metabolism and show a lower level of glycolysis. However, little is known about the role of lipid metabolism in the regulation of Treg cell homeostasis. Some members of the ACSL family of acyl-coenzyme A (CoA) synthases are expressed in T cells, but their function remains unclear. A combination of RNA-sequencing and proteome analyses shows that Acsbg1, a member of ACSL, is selectively expressed in Treg cells. We show that the genetic deletion of Acsbg1 not only causes mitochondrial dysfunction, but it also dampens other metabolic pathways. The extrinsic supplementation of Acsbg1-deficient Treg cells with oleoyl-CoA restores the phenotype of the Treg metabolic signature. Furthermore, this pathway in ST2+ effector Treg cells enhances immunosuppressive capacity in airway inflammation. Thus, Acsbg1 serves as a metabolic checkpoint governing Treg cell homeostasis and the resolution of lung inflammation.


Assuntos
Coenzima A Ligases/metabolismo , Metabolismo Energético , Pulmão/enzimologia , Mitocôndrias/enzimologia , Pneumonia/enzimologia , Linfócitos T Reguladores/enzimologia , Animais , Coenzima A Ligases/genética , Modelos Animais de Doenças , Ácidos Graxos/metabolismo , Regulação Enzimológica da Expressão Gênica , Homeostase , Interleucina-33 , Pulmão/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/imunologia , Biogênese de Organelas , Pneumonia/genética , Pneumonia/imunologia , Transdução de Sinais , Linfócitos T Reguladores/imunologia
16.
Biomed Pharmacother ; 144: 112340, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34678729

RESUMO

Fibrosis is an outcome of tissue repair after different types of injuries. The homeostasis of extracellular matrix is broken, and excessive deposition occurs, affecting the normal function of tissues and organs, which could become prostrated in serious cases.Finding a suitable target to regulate the repair process and reduce the damage caused by fibrosis is a hot research topic at present. The TRIM family is number of one of the E3 ubiquitin ligase subfamilies and participates in various biological processes including intracellular signal transduction, apoptosis, autophagy, and immunity by regulating the ubiquitination of target proteins. For the past few years, the important role of TRIM in the occurrence and development of fibrosis has been gradually revealed. In this review, we focus on the recent emerging topics on TRIM proteins in the regulation of fibrosis, fibrosis-related cytokines and pathways.


Assuntos
Matriz Extracelular/metabolismo , Cirrose Hepática/enzimologia , Fígado/enzimologia , Pulmão/enzimologia , Fibrose Pulmonar/enzimologia , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Antifibróticos/uso terapêutico , Citocinas/metabolismo , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/genética , Matriz Extracelular/patologia , Fibrose , Humanos , Mediadores da Inflamação/metabolismo , Fígado/efeitos dos fármacos , Fígado/patologia , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/genética , Cirrose Hepática/patologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Conformação Proteica , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/genética , Fibrose Pulmonar/patologia , Transdução de Sinais , Relação Estrutura-Atividade , Proteínas com Motivo Tripartido/antagonistas & inibidores , Proteínas com Motivo Tripartido/química , Proteínas com Motivo Tripartido/genética , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/genética
17.
Respir Res ; 22(1): 267, 2021 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-34666758

RESUMO

BACKGROUND: The non-cancerous functions of Akt in the airway are understudied. In some tissues, Akt phosphorylates and activates endothelial nitric oxide synthase (eNOS) to produce nitric oxide (NO) that has anti-inflammatory effects. NO production has antibacterial and antiviral effects in the airway, and increasing NO may be a useful anti-pathogen strategy. Akt also stimulates the nuclear factor erythroid 2-related factor 2 (Nrf-2) transcription factor, which transcribes antioxidant genes. Therefore, we hypothesized that activation of the Akt/eNOS pathway, which also activates Nrf-2, may have protective effects in human airway cells against injury. METHODS: To directly test the effects of Akt signaling in the airway, we treated A549 and 16HBE cells as well as primary bronchial, nasal, and type II alveolar epithelial cells with small molecule Akt activator SC79. We examined the effects of SC79 on eNOS activation, NO production, Nrf-2 target levels, and interleukin-8 (IL-8) transcription during exposure to TNF-α or Pseudomonas flagellin (TLR5 agonist). Additionally, air-liquid interface bronchial cultures were treated with cadmium, an oxidative stressor that causes airway barrier breakdown. RESULTS: SC79 induced a ~ twofold induction of p-eNOS and Nrf-2 protein levels blocked by PI3K inhibitor LY294002. Live cell imaging revealed SC79 increased acute NO production. Quantitative RT-PCR showed a ~ twofold increase in Nrf-2 target gene transcription. TNF-α or flagellin-induced IL-8 levels were also significantly reduced with SC79 treatment. Moreover, the transepithelial electrical resistance decrease observed with cadmium was ameliorated by SC79, likely by an acute increase in tight junction protein ZO-1 levels. CONCLUSIONS: Together, the data presented here demonstrate SC79 activation of Akt induces potentially anti-pathogenic NO production, antioxidant gene transcription, reduces IL-8 transcription, and may protect against oxidative barrier dysfunction in a wide range of airway epithelial cells.


Assuntos
Acetatos/farmacologia , Anti-Inflamatórios/farmacologia , Benzopiranos/farmacologia , Ativadores de Enzimas/farmacologia , Células Epiteliais/efeitos dos fármacos , Interleucina-8/metabolismo , Pulmão/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Óxido Nítrico/metabolismo , Pneumonia/prevenção & controle , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células A549 , Impedância Elétrica , Ativação Enzimática , Células Epiteliais/enzimologia , Células Epiteliais/imunologia , Humanos , Interleucina-8/genética , Pulmão/enzimologia , Pulmão/imunologia , Fator 2 Relacionado a NF-E2/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação , Pneumonia/enzimologia , Pneumonia/genética , Pneumonia/imunologia , Transdução de Sinais , Transcrição Gênica , Proteína da Zônula de Oclusão-1/metabolismo
18.
Toxicol Appl Pharmacol ; 432: 115757, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34673086

RESUMO

An increasing number of studies have shown that arsenic exposure increases the risk of lung cancer as well as a variety of non-malignant respiratory diseases, including bronchitis and tracheobronchitis. HMGB1 is widely expressed in a variety of tissues and cells and is involved in the pathological processes of many lung diseases through binding to the corresponding receptors and activating the downstream signaling pathways. However, the exact role of HMGB1/RAGE in arsenic-induced lung injury remains unknown. The aim of this study was to investigate whether HMGB1/RAGE and its activated downstream pathways are involved in the process of arsenic exposure-induced lung injury in rats. In this study, an animal model of oral exposure to arsenic was induced using 2.5, 5 and 10 mg/kg NaAsO2. The results showed that capillary permeability (LDH, TP, ACP, and AKP) was increased in the arsenic exposure groups, resulting in cell damage; this was accompanied by acute inflammation marked by significant neutrophil infiltration. Meanwhile, obvious histopathological damage, including thickening of the lung epithelium, increased infiltration of inflammatory cells, rupture of the alveolar wall, swelling of the mitochondria, and chromatin agglutination was observed by H&E staining and transmission electron microscopy. Furthermore, the results confirmed that the expressions of HMGB1 and RAGE in lung tissue were enhanced, and protein expression of PI3K, p-AKT, IL-1ß, IL-18, and MMP-9 was increased in lung homogenates from the arsenic-exposed groups compared to the control group. Finally, Masson's staining results revealed arsenic-induced fibrosis and collagen deposition. Moreover, a significant increase in key fibrosis factors, including TGF-ß1, p-SMAD2, p-SMAD3, and SMAD4 was observed in the lung homogenates in arsenic-exposed groups. In conclusion, the current study demonstrates that sub-chronic arsenic exposure triggers the inflammatory response and collagen fiber deposition in rat lung tissue. The potential mechanism may be closely related to activation of the pro-inflammatory-related HMGB1/RAGE pathway and initiation of the PI3K/AKT and TGF-ß1/SMAD pathways.


Assuntos
Proteína HMGB1/metabolismo , Pulmão/enzimologia , Fosfatidilinositol 3-Quinase/metabolismo , Pneumonia/enzimologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fibrose Pulmonar/enzimologia , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Arsenitos , Colágeno/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Pulmão/ultraestrutura , Masculino , Fosforilação , Pneumonia/induzido quimicamente , Pneumonia/patologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Ratos Wistar , Transdução de Sinais , Compostos de Sódio
19.
Front Immunol ; 12: 718136, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34646263

RESUMO

Angiotensin-converting enzyme 2 (ACE2) is a receptor for the spike protein of SARS-COV-2 that allows viral binding and entry and is expressed on the surface of several pulmonary and non-pulmonary cell types, with induction of a "cytokine storm" upon binding. Other cell types present the receptor and can be infected, including cardiac, renal, intestinal, and endothelial cells. High ACE2 levels protect from inflammation. Despite the relevance of ACE2 levels in COVID-19 pathogenesis, experimental studies to comprehensively address the question of ACE2 regulations are still limited. A relevant observation from the clinic is that, besides the pro-inflammatory cytokines, such as IL-6 and IL-1ß, the anti-inflammatory cytokine IL-10 is also elevated in worse prognosis patients. This could represent somehow a "danger signal", an alarmin from the host organism, given the immuno-regulatory properties of the cytokine. Here, we investigated whether IL-10 could increase ACE2 expression in the lung-derived Calu-3 cell line. We provided preliminary evidence of ACE2 mRNA increase in cells of lung origin in vitro, following IL-10 treatment. Endothelial cell infection by SARS-COV-2 is associated with vasculitis, thromboembolism, and disseminated intravascular coagulation. We confirmed ACE2 expression enhancement by IL-10 treatment also on endothelial cells. The sartans (olmesartan and losartan) showed non-statistically significant ACE2 modulation in Calu-3 and endothelial cells, as compared to untreated control cells. We observed that the antidiabetic biguanide metformin, a putative anti-inflammatory agent, also upregulates ACE2 expression in Calu-3 and endothelial cells. We hypothesized that IL-10 could be a danger signal, and its elevation could possibly represent a feedback mechanism fighting inflammation. Although further confirmatory studies are required, inducing IL-10 upregulation could be clinically relevant in COVID-19-associated acute respiratory distress syndrome (ARDS) and vasculitis, by reinforcing ACE2 levels.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , Anti-Inflamatórios/farmacologia , COVID-19/enzimologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Interleucina-10/farmacologia , Pulmão/efeitos dos fármacos , RNA Mensageiro/metabolismo , SARS-CoV-2/patogenicidade , Enzima de Conversão de Angiotensina 2/genética , COVID-19/genética , COVID-19/imunologia , Linhagem Celular , Interações Hospedeiro-Patógeno , Células Endoteliais da Veia Umbilical Humana/enzimologia , Células Endoteliais da Veia Umbilical Humana/imunologia , Humanos , Pulmão/enzimologia , Pulmão/imunologia , Metformina/farmacologia , RNA Mensageiro/genética , SARS-CoV-2/imunologia , Regulação para Cima
20.
Respir Res ; 22(1): 258, 2021 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-34602075

RESUMO

BACKGROUND: Diabetes mellitus (DM) is a key contributing factor to poor survival in lung transplantation recipients. Mitochondrial dysfunction is recognized as a critical mediator in the pathogenesis of diabetic lung ischemia-reperfusion (IR) injury. The protective effects of adiponectin have been demonstrated in our previous study, but the underlying mechanism remains unclear. Here we demonstrated an important role of mitophagy in the protective effect of adiponectin during diabetic lung IR injury. METHODS: High-fat diet-fed streptozotocin-induced type 2 diabetic rats were exposed to adiponectin with or without administration of the SIRT1 inhibitor EX527 following lung transplantation. To determine the mechanisms underlying the action of adiponectin, rat pulmonary microvascular endothelial cells were transfected with SIRT1 small-interfering RNA or PINK1 small-interfering RNA and then subjected to in vitro diabetic lung IR injury. RESULTS: Mitophagy was impaired in diabetic lungs subjected to IR injury, which was accompanied by increased oxidative stress, inflammation, apoptosis, and mitochondrial dysfunction. Adiponectin induced mitophagy and attenuated subsequent diabetic lung IR injury by improving lung functional recovery, suppressing oxidative damage, diminishing inflammation, decreasing cell apoptosis, and preserving mitochondrial function. However, either administration of 3-methyladenine (3-MA), an autophagy antagonist or knockdown of PINK1 reduced the protective action of adiponectin. Furthermore, we demonstrated that APN affected PINK1 stabilization via the SIRT1 signaling pathway, and knockdown of SIRT1 suppressed PINK1 expression and compromised the protective effect of adiponectin. CONCLUSION: These data demonstrated that adiponectin attenuated reperfusion-induced oxidative stress, inflammation, apoptosis and mitochondrial dysfunction via activation of SIRT1- PINK1 signaling-mediated mitophagy in diabetic lung IR injury.


Assuntos
Adiponectina/farmacologia , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Lesão Pulmonar/prevenção & controle , Transplante de Pulmão/efeitos adversos , Pulmão/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitofagia/efeitos dos fármacos , Traumatismo por Reperfusão/prevenção & controle , Animais , Células Cultivadas , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/enzimologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Mediadores da Inflamação/metabolismo , Pulmão/enzimologia , Pulmão/patologia , Lesão Pulmonar/enzimologia , Lesão Pulmonar/etiologia , Lesão Pulmonar/patologia , Masculino , Mitocôndrias/enzimologia , Mitocôndrias/patologia , Estresse Oxidativo/efeitos dos fármacos , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Ratos Sprague-Dawley , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Sirtuína 1/genética , Sirtuína 1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA