Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.005
Filtrar
1.
PLoS One ; 19(8): e0309166, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39172905

RESUMO

Chronic obstructive pulmonary disease is a common chronic lung disease with an ever-increasing incidence. Despite years of drug research and approvals, we are still not able to halt progress or restore normal lung function. Our previous studies have demonstrated that liver growth factor-LGF has an effect on the repair of the affected tissue in a mouse model of cigarette smoke exposure, but by what pathways it achieves this is unknown. The present study aimed to identify differentially expressed genes between emphysematous mice treated with LGF to identify potential therapeutic targets for the treatment of pulmonary emphysema. The emphysema mouse model was induced by prolonged exposure to cigarette smoke. To determine the gene expression profile of the lung in smokers treated or not with LGF, lung messenger RNA gene expression was assessed with the Agilent Array platform. We carried out differentially expressed gene analysis, functional enrichment and validated in treated mouse lung samples. The treated group significantly improved lung function (~35%) and emphysema level (~20%), consistent with our previous published studies. Microarray analysis demonstrated 290 differentially expressed genes in total (2.0-fold over or lower expressed). Injury repair-associated genes and pathways were further enhanced in the lung of LGF treated mice. The expression trends of two genes (Zscan2 and Bag6) were different in emphysematous lungs treated with LGF compared to untreated lungs. Therefore, Zscan2 and Bag6 genes could play a role in regulating inflammation and the immune response in the lung that undergoes partial lung regeneration. However, further studies are necessary to demonstrate this causal relationship.


Assuntos
Modelos Animais de Doenças , Pulmão , Doença Pulmonar Obstrutiva Crônica , Fatores de Transcrição , Animais , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/metabolismo , Camundongos , Pulmão/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Enfisema Pulmonar/genética , Enfisema Pulmonar/metabolismo , Perfilação da Expressão Gênica , Camundongos Endogâmicos C57BL , Masculino , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo
2.
Trans Am Clin Climatol Assoc ; 134: 29-36, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39135587

RESUMO

In this paper, I will discuss recent studies using a cystic fibrosis pig model to better understand the origins of cystic fibrosis lung disease. Specifically, I will review our work investigating how loss of the cystic fibrosis transmembrane conductance regulator function (CFTR) impairs mucociliary transport in the cystic fibrosis airway. These studies reveal new insights into the early, underlying mechanisms of cystic fibrosis lung disease and could lead to novel therapeutic interventions.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística , Fibrose Cística , Modelos Animais de Doenças , Depuração Mucociliar , Fibrose Cística/metabolismo , Fibrose Cística/complicações , Fibrose Cística/fisiopatologia , Fibrose Cística/genética , Animais , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Suínos , Humanos , Pulmão/metabolismo , Pulmão/fisiopatologia , Mutação , Predisposição Genética para Doença , Fenótipo
3.
Trans Am Clin Climatol Assoc ; 134: 94-112, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39135597

RESUMO

Surfactant Protein A (SP-A) is an innate immune modulator produced by the lung with known protective effects against bacteria and viruses. Its role in asthma, an inflammatory lung disease that affects 10% of the world's population, is not entirely known. In this review, we demonstrate that SP-A confers protection against exposure to interleukin-13, a type 2 cytokine integral to eosinophilic asthma, in a mouse model of SP-A deficiency, a house dust mite model of asthma, and in human bronchial epithelial cells from participants with asthma. We also show that small peptides derived from SP-A, such as the major allele of single nucleotide polymorphism (SNP) rs1965708, which includes the carbohydrate recognition domain of SP-A2 at position 223, reduce airway hyperresponsiveness, airway eosinophils, and mucus in a mouse model of asthma. These data suggest that SP-A has beneficial effects relevant to asthma and that an SP-A peptide may have a new therapeutic use in asthma.


Assuntos
Asma , Modelos Animais de Doenças , Imunidade Inata , Proteína A Associada a Surfactante Pulmonar , Asma/imunologia , Asma/tratamento farmacológico , Animais , Proteína A Associada a Surfactante Pulmonar/genética , Proteína A Associada a Surfactante Pulmonar/metabolismo , Proteína A Associada a Surfactante Pulmonar/imunologia , Humanos , Camundongos , Polimorfismo de Nucleotídeo Único , Interleucina-13/metabolismo , Interleucina-13/imunologia , Interleucina-13/genética , Pulmão/imunologia , Pulmão/metabolismo , Antiasmáticos/farmacologia , Antiasmáticos/uso terapêutico , Pyroglyphidae/imunologia
4.
Nat Commun ; 15(1): 6964, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39138195

RESUMO

Though RNAi and RNA-splicing machineries are involved in regulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication, their precise roles in coronavirus disease 2019 (COVID-19) pathogenesis remain unclear. Herein, we show that decreased RNAi component (Dicer and XPO5) and splicing factor (SRSF3 and hnRNPA3) expression correlate with increased COVID-19 severity. SARS-CoV-2 N protein induces the autophagic degradation of Dicer, XPO5, SRSF3, and hnRNPA3, inhibiting miRNA biogenesis and RNA splicing and triggering DNA damage, proteotoxic stress, and pneumonia. Dicer, XPO5, SRSF3, and hnRNPA3 knockdown increases, while their overexpression decreases, N protein-induced pneumonia's severity. Older mice show lower expression of Dicer, XPO5, SRSF3, and hnRNPA3 in their lung tissues and exhibit more severe N protein-induced pneumonia than younger mice. PJ34, a poly(ADP-ribose) polymerase inhibitor, or anastrozole, an aromatase inhibitor, ameliorates N protein- or SARS-CoV-2-induced pneumonia by restoring Dicer, XPO5, SRSF3, and hnRNPA3 expression. These findings will aid in developing improved treatments for SARS-CoV-2-associated pneumonia.


Assuntos
COVID-19 , Carioferinas , Ribonuclease III , SARS-CoV-2 , Fatores de Processamento de Serina-Arginina , Animais , Fatores de Processamento de Serina-Arginina/metabolismo , Fatores de Processamento de Serina-Arginina/genética , Humanos , Ribonuclease III/metabolismo , Ribonuclease III/genética , SARS-CoV-2/genética , COVID-19/metabolismo , COVID-19/virologia , COVID-19/genética , Camundongos , Carioferinas/metabolismo , Carioferinas/genética , RNA Helicases DEAD-box/metabolismo , RNA Helicases DEAD-box/genética , Regulação para Baixo , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Masculino , Feminino , MicroRNAs/genética , MicroRNAs/metabolismo , Splicing de RNA , Autofagia/genética , Dano ao DNA , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B
5.
Matrix Biol ; 132: 47-58, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39147560

RESUMO

BACKGROUND: Lung fibroblasts play a central role in maintaining lung homeostasis and facilitating repair through the synthesis and organization of the extracellular matrix (ECM). This study investigated the cross-talk between interleukin-1 alpha (IL-1α) and transforming growth factor-ß (TGF-ß) signaling, two key regulators in tissue repair and fibrosis, in the context of lung fibroblast repair in the healthy lung. RESULTS: Stimulation of lung fibroblasts with TGF-ß1 and TGF-ß2 induced collagen-I and fibronectin protein expression (p < 0.05), a response inhibited with co-treatment with IL-1α (p < 0.05). Additionally, TGF-ß1 and TGF-ß2 induced myofibroblast differentiation, and collagen-I gel contraction, which were both suppressed by IL-1α (p < 0.05). In contrast, interleukin (IL)-6, IL-8 and thymic stromal lymphopoietin induced by IL-1α, were unaffected by TGF-ß1 or TGF-ß2. Mechanistically, IL-1α administration led to the suppression of TGF-ß1 and TGF-ß2 signaling, through downregulation of mRNA and protein for TGF-ß receptor II and the downstream adaptor protein TRAF6, but not through miR-146a that is known to be induced by IL-1α. DISCUSSION: IL-1α acts as a master regulator, modulating TGF-ß1 and TGF-ß2-induced ECM production, remodeling, and myofibroblast differentiation in human lung fibroblasts, playing a vital role in balancing tissue repair versus fibrosis. Further research is required to understand the dysregulated cross-talk between IL-1α and TGF-ß signaling in chronic lung diseases and the exploration of therapeutic opportunities. METHODS: Primary human lung fibroblasts (PHLF) were treated with media control, or 1 ng/ml IL-1α with or without 50 ng/ml TGF-ß1 or TGF-ß2 for 1, 6 and 72 h. Cell lysates were assessed for the expression of ECM proteins and signaling molecules by western blot, miRNA by qPCR, mRNA by RNA sequencing and cell supernatants for cytokine production by ELISA. PHLFs were also seeded in non-tethered collagen-I gels to measure contraction, and myofibroblast differentiation using confocal microscopy.


Assuntos
Matriz Extracelular , Fibroblastos , Interleucina-1alfa , Pulmão , Transdução de Sinais , Fator de Crescimento Transformador beta1 , Humanos , Interleucina-1alfa/metabolismo , Interleucina-1alfa/genética , Matriz Extracelular/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Pulmão/metabolismo , Pulmão/citologia , Fibroblastos/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/citologia , Diferenciação Celular , Miofibroblastos/metabolismo , Miofibroblastos/efeitos dos fármacos , Células Cultivadas , Colágeno Tipo I/metabolismo , Colágeno Tipo I/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Fibronectinas/metabolismo , Fibronectinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Fator de Crescimento Transformador beta2
6.
BMC Pharmacol Toxicol ; 25(1): 55, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39175070

RESUMO

BACKGROUND: Metamizole is banned in some countries because of its toxicity, although it is widely used in some European countries. In addition, there is limited information on its safety profile, and it is still debated whether it is toxic to the heart, lungs, liver, kidneys, and stomach. AIMS: Our study investigated the effects of metamizole on the heart, lung, liver, kidney, and stomach tissues of rats. METHODS: Eighteen rats were divided into three groups, wassix healthy (HG), 500 mg/kg metamizole (MT-500), and 1000 mg/kg metamizole (MT-1000). Metamizole was administered orally twice daily for 14 days. Meanwhile, the HG group received pure water orally. Biochemical, histopathologic, and macroscopic examinations were performed on blood samples and tissues. RESULTS: Malondialdehyde (MDA), total glutathione (tGSH), superoxide dismutase (SOD), and catalase (CAT) in the lung and gastric tissues of MT-500 and MT-1000 groups were almost the same as those of the HG (p > 0.05). However, MDA levels in the heart and liver tissues of MT-500 and MT-1000 groups were higher (p < 0.05) compared to the HG, while tGSH levels and SOD, and CAT activities were lower (p < 0.05). MDA levels of MT-500 and MT-1000 groups in the kidney tissue increased the most (p < 0.001), and tGSH levels and SOD and CAT activities decreased the most (p < 0.001) compared to HG. Metamizole did not cause oxidative damage in the lung and gastric tissue. While metamizole did not change troponin levels, it significantly increased alanine aminotransferase (ALT), aspartate aminotransferase (AST), blood urea nitrogen (BUN), and creatinine levels compared to HG. Histopathologically, mild damage was detected in heart tissue, moderate damage in liver tissue, and severe damage in renal tissue. However, no histopathologic damage was found in any groups' lung and gastric tissues. CONCLUSION: Metamizole should be used under strict control in patients with cardiac and liver diseases and it would be more appropriate not to use it in patients with renal disease.


Assuntos
Anti-Inflamatórios não Esteroides , Dipirona , Coração , Rim , Fígado , Pulmão , Estômago , Animais , Dipirona/toxicidade , Rim/efeitos dos fármacos , Rim/patologia , Rim/metabolismo , Fígado/efeitos dos fármacos , Fígado/patologia , Fígado/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/metabolismo , Anti-Inflamatórios não Esteroides/toxicidade , Masculino , Ratos , Coração/efeitos dos fármacos , Estômago/efeitos dos fármacos , Estômago/patologia , Malondialdeído/metabolismo , Superóxido Dismutase/metabolismo , Glutationa/metabolismo , Catalase/metabolismo , Miocárdio/patologia , Miocárdio/metabolismo
7.
Biosystems ; 243: 105284, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39103139

RESUMO

In biological systems, solitary organisms or eusocial groups, the metabolic rate often scales allometrically with systems' size, when they are inactive, and the scaling becomes nearly isometric when the systems are active. Here I propose a hypothesis attempting to offer a departing point for a general joint understanding of the difference in the scaling powers between inactive and active states. When the system is inactive, there exist inactive components, which consume less energy than the active ones, and the larger the system is, the larger the fraction of the inactive components, which leads to sublinear scaling. When the system is active, most inactive components are activated, which leads to nearly isometric scaling. I hypothesize that the disproportional fraction of the inactive components is caused by the diffusants screening in the complex transportation network. I.e., when metabolites or information diffuses in the system, due to the physical limitation of the network structure and the diffusant's physical feature, not all the components can equally receive the diffusants so that these components are inactive. Using the mammalian pulmonary system, ant colonies, and other few systems as examples, I discuss how the screening leads to the allometric and isometric metabolic scaling powers in inactive and active states respectively. It is noteworthy that there are a few exceptions, in which the metabolic rate of the system has an isometric scaling relationship with size at rest. I show that these exceptions not only do not disapprove the hypothesis, but actually support it.


Assuntos
Modelos Biológicos , Animais , Metabolismo Energético/fisiologia , Humanos , Formigas/fisiologia , Formigas/metabolismo , Pulmão/metabolismo
8.
Int J Nanomedicine ; 19: 8175-8188, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39157733

RESUMO

Exosomes belong to a subgroup of extracellular vesicles secreted by various cells and are involved in intercellular communication and material transfer. In recent years, exosomes have been used as drug delivery carriers because of their natural origin, high stability, low immunogenicity and high engineering ability. However, achieving targeted drug delivery with exosomes remains challenging. In this paper, a phage display technology was used to screen targeted peptides, and different surface modification strategies of targeted peptide exosomes were reviewed. In addition, the application of peptide-targeted exosomes in pulmonary diseases was also summarised.


Assuntos
Sistemas de Liberação de Medicamentos , Exossomos , Pulmão , Peptídeos , Exossomos/química , Exossomos/metabolismo , Humanos , Peptídeos/química , Peptídeos/farmacologia , Pulmão/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Pneumopatias/tratamento farmacológico , Animais , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Técnicas de Visualização da Superfície Celular/métodos
9.
Nat Commun ; 15(1): 7138, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39164231

RESUMO

Telomere shortening is a prominent hallmark of aging and is emerging as a characteristic feature of Myelodysplastic Syndromes (MDS) and Idiopathic Pulmonary Fibrosis (IPF). Optimal telomerase activity prevents progressive shortening of telomeres that triggers DNA damage responses. However, the upstream regulation of telomerase holoenzyme components remains poorly defined. Here, we identify RIOK2, a master regulator of human blood cell development, as a critical transcription factor for telomere maintenance. Mechanistically, loss of RIOK2 or its DNA-binding/transactivation properties downregulates mRNA expression of both TRiC and dyskerin complex subunits that impairs telomerase activity, thereby causing telomere shortening. We further show that RIOK2 expression is diminished in aged individuals and IPF patients, and it strongly correlates with shortened telomeres in MDS patient-derived bone marrow cells. Importantly, ectopic expression of RIOK2 alleviates telomere shortening in IPF patient-derived primary lung fibroblasts. Hence, increasing RIOK2 levels prevents telomere shortening, thus offering therapeutic strategies for telomere biology disorders.


Assuntos
Proteínas de Ciclo Celular , Fibrose Pulmonar Idiopática , Proteínas Nucleares , Telomerase , Encurtamento do Telômero , Humanos , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Telomerase/metabolismo , Telomerase/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Fibroblastos/metabolismo , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Telômero/metabolismo , Telômero/genética , Regulação da Expressão Gênica , Pulmão/metabolismo , Pulmão/patologia
10.
Sci Signal ; 17(850): eadn3785, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39163389

RESUMO

Coronaviruses rely on host proteases to activate the viral spike protein, which facilitates fusion with the host cell membrane and the release of viral genomic RNAs into the host cell cytoplasm. The distribution of specific host proteases in the host determines the host, tissue, and cellular tropism of these viruses. Here, we identified the kallikrein (KLK) family member KLK5 as a major host protease secreted by human airway cells and exploited by multiple human betacoronaviruses. KLK5 cleaved both the priming (S1/S2) and activation (S2') sites of spike proteins from various human betacoronaviruses in vitro. In contrast, KLK12 and KLK13 displayed preferences for either the S2' or S1/S2 site, respectively. Whereas KLK12 and KLK13 worked in concert to activate SARS-CoV-2 and MERS-CoV spike proteins, KLK5 by itself efficiently activated spike proteins from several human betacoronaviruses, including SARS-CoV-2. Infection of differentiated human bronchial epithelial cells (HBECs) with human betacoronaviruses induced an increase in KLK5 that promoted virus replication. Furthermore, ursolic acid and other related plant-derived triterpenoids that inhibit KLK5 effectively suppressed the replication of SARS-CoV, MERS-CoV, and SARS-CoV-2 in HBECs and mitigated lung inflammation in mice infected with MERS-CoV or SARS-CoV-2. We propose that KLK5 is a pancoronavirus host factor and a promising therapeutic target for current and future coronavirus-induced diseases.


Assuntos
Calicreínas , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Replicação Viral , Humanos , Calicreínas/metabolismo , Calicreínas/genética , Animais , Glicoproteína da Espícula de Coronavírus/metabolismo , Glicoproteína da Espícula de Coronavírus/genética , Camundongos , SARS-CoV-2/metabolismo , Betacoronavirus/metabolismo , Betacoronavirus/fisiologia , COVID-19/metabolismo , COVID-19/virologia , Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/virologia , Coronavírus da Síndrome Respiratória do Oriente Médio/metabolismo , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Células HEK293 , Pulmão/virologia , Pulmão/metabolismo
11.
Biol Sex Differ ; 15(1): 62, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-39107837

RESUMO

BACKGROUND: Lung adenocarcinoma (LUAD) has been observed to have significant sex differences in incidence, prognosis, and response to therapy. However, the molecular mechanisms responsible for these disparities have not been investigated extensively. METHODS: Sample-specific gene regulatory network methods were used to analyze RNA sequencing data from non-cancerous human lung samples from The Genotype Tissue Expression Project (GTEx) and lung adenocarcinoma primary tumor samples from The Cancer Genome Atlas (TCGA); results were validated on independent data. RESULTS: We found that genes associated with key biological pathways including cell proliferation, immune response and drug metabolism are differentially regulated between males and females in both healthy lung tissue and tumor, and that these regulatory differences are further perturbed by tobacco smoking. We also discovered significant sex bias in transcription factor targeting patterns of clinically actionable oncogenes and tumor suppressor genes, including AKT2 and KRAS. Using differentially regulated genes between healthy and tumor samples in conjunction with a drug repurposing tool, we identified several small-molecule drugs that might have sex-biased efficacy as cancer therapeutics and further validated this observation using an independent cell line database. CONCLUSIONS: These findings underscore the importance of including sex as a biological variable and considering gene regulatory processes in developing strategies for disease prevention and management.


Lung adenocarcinoma (LUAD) is a disease that affects males and females differently. Biological sex not only influences chances of developing the disease, but also how the disease progresses and how effective various therapies may be. We analyzed sex-specific gene regulatory networks consisting of transcription factors and the genes they regulate in both healthy lung tissue and in LUAD and identified sex-biased differences. We found that genes associated with cell proliferation, immune response, and drug metabolism are differentially targeted by transcription factors between males and females. We also found that several genes that are drug targets in LUAD, are also regulated differently between males and females. Importantly, these differences are also influenced by an individual's smoking history. Extending our analysis using a drug repurposing tool, we found candidate drugs with evidence that they might work better for one sex or the other. These results demonstrate that considering the differences in gene regulation between males and females will be essential if we are to develop precision medicine strategies for preventing and treating LUAD.


Assuntos
Adenocarcinoma de Pulmão , Redes Reguladoras de Genes , Adenocarcinoma de Pulmão/diagnóstico , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/terapia , Fatores Sexuais , Regulação Neoplásica da Expressão Gênica/genética , Pulmão/metabolismo , Fumar Tabaco/efeitos adversos , Prognóstico , Imunoterapia , Terapia de Alvo Molecular , Linhagem Celular Tumoral , Humanos , Masculino , Feminino , Descoberta de Drogas
12.
Int J Immunopathol Pharmacol ; 38: 3946320241274225, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39140804

RESUMO

OBJECTIVES: Tuberostemonine has several biological activity, the aim of study examined the impact of tuberostemonine on the proliferation of TGF-ß1 induced cell model, and its ability to alleviate pulmonary fibrosis stimulated by bleomycin in mice. METHODS: In vitro, we assessed the effect of tuberostemonine (350, 550 and 750 µM) on the proliferation of cells stimulated by TGF-ß1 (10 µg/L), as well as on parameters such as α-SMA vitality, human fibronectin, collagen, and hydroxyproline levels in cells. In vivo, we analyzed inflammation, hydroxyproline, collagen activity and metabolomics in the lungs of mice. Additionally, a comprehensive investigation into the TGF-ß/smad signaling pathway was undertaken, targeting lung tissue as well as HFL cells. RESULTS: Within the confines of an in vitro setup, the tuberostemonine manifested a discerned IC50 of 1.9 mM. Furthermore, a significant reduction of over fifty percent was ascertained in the secretion levels of hydroxyproline, fibronectin, collagen type I, collagen type III and α-SMA. In vivo, tuberostemonine obviously improved the respiratory function percentage over 50% of animal model and decreased the hydroxyproline, lung inflammation and collagen deposition. A prominent decline in TGF-ß/smad pathway functioning was identified within both the internal and external cellular contexts. CONCLUSIONS: Tuberostemonine is considered as a modulator to alleviate fibrosis and may become a new renovation for pulmonary fibrosis.


Assuntos
Bleomicina , Proliferação de Células , Fibroblastos , Pulmão , Fibrose Pulmonar , Transdução de Sinais , Fator de Crescimento Transformador beta1 , Animais , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Proliferação de Células/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/metabolismo , Humanos , Camundongos , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo , Hidroxiprolina/metabolismo , Proteínas Smad/metabolismo , Camundongos Endogâmicos C57BL , Masculino , Linhagem Celular , Colágeno/metabolismo , Modelos Animais de Doenças , Fibronectinas/metabolismo , Actinas/metabolismo
13.
Part Fibre Toxicol ; 21(1): 33, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-39143599

RESUMO

BACKGROUND: Physiologically based kinetic models facilitate the safety assessment of inhaled engineered nanomaterials (ENMs). To develop these models, high quality datasets on well-characterized ENMs are needed. However, there are at present, several data gaps in the systemic availability of poorly soluble particles after inhalation. The aim of the present study was therefore to acquire two comparable datasets to parametrize a physiologically-based kinetic model. METHOD: Rats were exposed to cerium dioxide (CeO2, 28.4 ± 10.4 nm) and titanium dioxide (TiO2, 21.6 ± 1.5 nm) ENMs in a single nose-only exposure to 20 mg/m3 or a repeated exposure of 2 × 5 days to 5 mg/m3. Different dose levels were obtained by varying the exposure time for 30 min, 2 or 6 h per day. The content of cerium or titanium in three compartments of the lung (tissue, epithelial lining fluid and freely moving cells), mediastinal lymph nodes, liver, spleen, kidney, blood and excreta was measured by Inductively Coupled Plasma-Mass Spectrometry (ICP-MS) at various time points post-exposure. As biodistribution is best studied at sub-toxic dose levels, lactate dehydrogenase (LDH), total protein, total cell numbers and differential cell counts were determined in bronchoalveolar lavage fluid (BALF). RESULTS: Although similar lung deposited doses were obtained for both materials, exposure to CeO2 induced persistent inflammation indicated by neutrophil granulocytes influx and exhibited an increased lung elimination half-time, while exposure to TiO2 did not. The lavaged lung tissue contained the highest metal concentration compared to the lavage fluid and cells in the lavage fluid for both materials. Increased cerium concentrations above control levels in secondary organs such as lymph nodes, liver, spleen, kidney, urine and faeces were detected, while for titanium this was found in lymph nodes and liver after repeated exposure and in blood and faeces after a single exposure. CONCLUSION: We have provided insight in the distribution kinetics of these two ENMs based on experimental data and modelling. The study design allows extrapolation at different dose-levels and study durations. Despite equal dose levels of both ENMs, we observed different distribution patterns, that, in part may be explained by subtle differences in biological responses in the lung.


Assuntos
Líquido da Lavagem Broncoalveolar , Cério , Exposição por Inalação , Pulmão , Titânio , Animais , Titânio/toxicidade , Titânio/farmacocinética , Cério/toxicidade , Cério/farmacocinética , Distribuição Tecidual , Masculino , Pulmão/metabolismo , Pulmão/efeitos dos fármacos , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Ratos , Nanoestruturas/toxicidade , Administração por Inalação , Ratos Wistar , Modelos Biológicos , Tamanho da Partícula , Nanopartículas Metálicas/toxicidade
14.
EBioMedicine ; 106: 105267, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39098109

RESUMO

BACKGROUND: Checkpoint inhibitor pneumonitis (CIP) is a potentially fatal adverse event characterized by new pulmonary infiltrates in cancer patients receiving immune checkpoint inhibitor therapy. This study aims to explore the interplay between lung microbiota, dysregulated metabolites, and host immunity in CIP. METHODS: We recruited thirteen hospitalized CIP patients, eleven idiopathic pulmonary fibrosis (IPF) patients, and ten new-onset non-small cell lung cancer patients. Bronchoalveolar lavage fluid samples were collected for 16S rRNA gene sequencing. The percentages of immune cells were determined using manual counting and flow cytometry. Interactions among microbiota, metabolites, and lymphocytes were analyzed using cultured mouse splenocytes and human T cells. FINDINGS: Proteobacteria emerged as the dominant phylum, notably abundant in both the CIP and IPF groups. Vibrio, Halomonas, Mangrovibacter, and Salinivibrio were the predominant microbiota because of their discriminative abundance patterns. Vibrio (r = 0.72, P-adj = 0.007) and Halomonas (r = 0.65, P-adj = 0.023) demonstrated strong correlations with lymphocytes. Vibrio metschnikovii and Mangrovibacter plantisponsors were more abundant in the CIP group than in the IPF group. Lauroylcarnitine, a key intermediary metabolite co-occurring with the predominant microbiota, exhibited a potent effect on cytokine secretion by mouse and human T cells, notably enhancing IFN-γ and TNF-α production from CD4 and CD8 cells in vitro. INTERPRETATION: Lauroylcarnitine, co-occurring with the predominant lung microbiota in CIP, could activate T cells in vitro. These findings suggest potential involvement of lung microbiota and acylcarnitine metabolism dysregulation in the pathogenesis of CIP. FUNDING: This work was supported by Peking University People's Hospital Scientific Research Development Funds (RDJ2022-15) and Provincial Key Clinical Specialty Capacity Building Project 2020 (Department of the Respiratory Medicine).


Assuntos
Inibidores de Checkpoint Imunológico , Pulmão , Ativação Linfocitária , Microbiota , Pneumonia , Linfócitos T , Humanos , Inibidores de Checkpoint Imunológico/efeitos adversos , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/farmacologia , Animais , Camundongos , Microbiota/efeitos dos fármacos , Masculino , Feminino , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Idoso , Linfócitos T/metabolismo , Linfócitos T/imunologia , Linfócitos T/efeitos dos fármacos , Pulmão/microbiologia , Pulmão/patologia , Pulmão/imunologia , Pulmão/metabolismo , Pneumonia/microbiologia , Pneumonia/etiologia , Pneumonia/metabolismo , Pneumonia/induzido quimicamente , Pneumonia/imunologia , Pessoa de Meia-Idade , Carnitina/análogos & derivados , Carnitina/metabolismo , RNA Ribossômico 16S/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/microbiologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Citocinas/metabolismo
15.
Int J Immunopathol Pharmacol ; 38: 3946320241272642, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39096175

RESUMO

Prolonged exposure to different occupational or environmental toxicants triggered oxidative stress and inflammatory reactions mediated lung damage. This study was designed to explore the influence and protective impact of flavone on lung injury in rats intoxicated with nicotine (NIC) and exposed to radiation (IR). Forty rats were divided into four groups; group I control, group II flavone; rats were administered with flavone (25 mg/kg/day), group III NIC + IR; rats were injected intraperitoneally with NIC (1 mg/kg/day) and exposed to γ-IR (3.5 Gy once/week for 2 weeks) while group IV NIC + IR + flavone; rats were injected with NIC, exposed to IR and administered with flavone. Redox status parameters and histopathological changes in lung tissue were evaluated. Nuclear factor-kappa B (NF-κB), forkhead box O-class1 (FoxO1) and nucleotide-binding domain- (NOD-) like receptor pyrin domain-containing-3 (NLRP3) gene expression were measured in lung tissues. Moreover, nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and phosphatidylinositol three kinase (PI3K) were measured using ELISA kits. Our data demonstrates, for the first time, that flavone protects the lung from NIC/IR-associated cytotoxicity, by attenuating the disrupted redox status and aggravating the antioxidant defence mechanism via activation of the PI3K/Nrf2. Moreover, flavone alleviates pulmonary inflammation by inhibiting the inflammatory signaling pathway FOXO1/NF-κB/NLRP3- Inflammasome. Collectively, the obtained results exhibited a notable efficiency of flavone in alleviating lung injury induced by NIC and IR via modulating PI3K/Nrf2 and FoxO1/NLRP3 Inflammasome.


Assuntos
Flavonas , Inflamassomos , Lesão Pulmonar , Nicotina , Animais , Masculino , Ratos , Flavonas/farmacologia , Proteína Forkhead Box O1 , Raios gama , Inflamassomos/metabolismo , Inflamassomos/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Pulmão/efeitos da radiação , Lesão Pulmonar/metabolismo , Lesão Pulmonar/prevenção & controle , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/metabolismo , Nicotina/farmacologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
16.
Cell Mol Life Sci ; 81(1): 330, 2024 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-39097839

RESUMO

Chronic obstructive pulmonary disease (COPD) is a complex syndrome with poorly understood mechanisms driving its early progression (GOLD stages 1-2). Elucidating the genetic factors that influence early-stage COPD, particularly those related to airway inflammation and remodeling, is crucial. This study analyzed lung tissue sequencing data from patients with early-stage COPD (GSE47460) and smoke-exposed mice. We employed Weighted Gene Co-Expression Network Analysis (WGCNA) and machine learning to identify potentially pathogenic genes. Further analyses included single-cell sequencing from both mice and COPD patients to pinpoint gene expression in specific cell types. Cell-cell communication and pseudotemporal analyses were conducted, with findings validated in smoke-exposed mice. Additionally, Mendelian randomization (MR) was used to confirm the association between candidate genes and lung function/COPD. Finally, functional validation was performed in vitro using cell cultures. Machine learning analysis of 30 differentially expressed genes identified 8 key genes, with CLEC5A emerging as a potential pathogenic factor in early-stage COPD. Bioinformatics analyses suggested a role for CLEC5A in macrophage-mediated inflammation during COPD. Two-sample Mendelian randomization linked CLEC5A single nucleotide polymorphisms (SNPs) with Forced Expiratory Volume in One Second (FEV1), FEV1/Forced Vital Capacity (FVC) and early/later on COPD. In vitro, the knockdown of CLEC5A led to a reduction in inflammatory markers within macrophages. Our study identifies CLEC5A as a critical gene in early-stage COPD, contributing to its pathogenesis through pro-inflammatory mechanisms. This discovery offers valuable insights for developing early diagnosis and treatment strategies for COPD and highlights CLEC5A as a promising target for further investigation.


Assuntos
Progressão da Doença , Inflamação , Lectinas Tipo C , Macrófagos , Polimorfismo de Nucleotídeo Único , Doença Pulmonar Obstrutiva Crônica , Receptores de Superfície Celular , Animais , Humanos , Masculino , Camundongos , Inflamação/genética , Inflamação/patologia , Inflamação/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Pulmão/patologia , Pulmão/metabolismo , Aprendizado de Máquina , Macrófagos/metabolismo , Macrófagos/patologia , Análise da Randomização Mendeliana , Camundongos Endogâmicos C57BL , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/patologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo
17.
Proc Natl Acad Sci U S A ; 121(33): e2406654121, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39116129

RESUMO

Protein therapeutics play a critical role in treating a large variety of diseases, ranging from infections to genetic disorders. However, their delivery to target tissues beyond the liver, such as the lungs, remains a great challenge. Here, we report a universally applicable strategy for lung-targeted protein delivery by engineering Lung-Specific Supramolecular Nanoparticles (LSNPs). These nanoparticles are designed through the hierarchical self-assembly of metal-organic polyhedra (MOP), featuring a customized surface chemistry that enables protein encapsulation and specific lung affinity after intravenous administration. Our design of LSNPs not only addresses the hurdles of cell membrane impermeability of protein and nonspecific tissue distribution of protein delivery, but also shows exceptional versatility in delivering various proteins, including those vital for anti-inflammatory and CRISPR-based genome editing to the lung, and across multiple animal species, including mice, rabbits, and dogs. Notably, the delivery of antimicrobial proteins using LSNPs effectively alleviates acute bacterial pneumonia, demonstrating a significant therapeutic potential. Our strategy not only surmounts the obstacles of tissue-specific protein delivery but also paves the way for targeted treatments in genetic disorders and combating antibiotic resistance, offering a versatile solution for precision protein therapy.


Assuntos
Edição de Genes , Pulmão , Nanopartículas , Animais , Edição de Genes/métodos , Pulmão/metabolismo , Camundongos , Nanopartículas/química , Cães , Coelhos , Humanos , Sistemas CRISPR-Cas , Sistemas de Liberação de Medicamentos/métodos
18.
ACS Nano ; 18(33): 22275-22297, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39105696

RESUMO

Nanomedicine has long pursued the goal of targeted delivery to specific organs and cell types but has yet to achieve this goal with the vast majority of targets. One rare example of success in this pursuit has been the 25+ years of studies targeting the lung endothelium using nanoparticles conjugated to antibodies against endothelial surface molecules. However, here we show that such "endothelial-targeted" nanocarriers also effectively target the lungs' numerous marginated neutrophils, which reside in the pulmonary capillaries and patrol for pathogens. We show that marginated neutrophils' uptake of many of these "endothelial-targeted" nanocarriers is on par with endothelial uptake. This generalizes across diverse nanomaterials and targeting moieties and was even found with physicochemical lung tropism (i.e., without targeting moieties). Further, we observed this in ex vivo human lungs and in vivo healthy mice, with an increase in marginated neutrophil uptake of nanoparticles caused by local or distant inflammation. These findings have implications for nanomedicine development for lung diseases. These data also suggest that marginated neutrophils, especially in the lungs, should be considered a major part of the reticuloendothelial system (RES), with a special role in clearing nanoparticles that adhere to the lumenal surfaces of blood vessels.


Assuntos
Pulmão , Nanopartículas , Neutrófilos , Animais , Neutrófilos/metabolismo , Neutrófilos/imunologia , Humanos , Pulmão/imunologia , Pulmão/metabolismo , Camundongos , Nanopartículas/química , Sistema Fagocitário Mononuclear/metabolismo , Endotélio/metabolismo , Camundongos Endogâmicos C57BL , Nanomedicina
19.
Nat Commun ; 15(1): 6657, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-39143055

RESUMO

Tuberculosis (TB) remains a leading cause of death, but antibiotic treatments for tuberculous meningitis, the deadliest form of TB, are based on those developed for pulmonary TB and not optimized for brain penetration. Here, we perform first-in-human dynamic 18F-pretomanid positron emission tomography (PET) in eight human subjects to visualize 18F-pretomanid biodistribution as concentration-time exposures in multiple compartments (NCT05609552), demonstrating preferential brain versus lung tissue partitioning. Preferential, antibiotic-specific partitioning into brain or lung tissues of several antibiotics, active against multidrug resistant (MDR) Mycobacterium tuberculosis strains, are confirmed in experimentally-infected mice and rabbits, using dynamic PET with chemically identical antibiotic radioanalogs, and postmortem mass spectrometry measurements. PET-facilitated pharmacokinetic modeling predicts human dosing necessary to attain therapeutic brain exposures. These data are used to design optimized, pretomanid-based regimens which are evaluated at human equipotent dosing in a mouse model of TB meningitis, demonstrating excellent bactericidal activity without an increase in intracerebral inflammation or brain injury. Importantly, several antibiotic regimens demonstrate discordant activities in brain and lung tissues in the same animal, correlating with tissue antibiotic exposures. These data provide a mechanistic basis for the compartmentalized activities of antibiotic regimens, with important implications for developing treatments for meningitis and other infections in compartments with unique antibiotic penetration.


Assuntos
Antituberculosos , Encéfalo , Pulmão , Mycobacterium tuberculosis , Adulto , Animais , Feminino , Humanos , Masculino , Camundongos , Coelhos , Antituberculosos/farmacocinética , Antituberculosos/uso terapêutico , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Modelos Animais de Doenças , Pulmão/diagnóstico por imagem , Pulmão/metabolismo , Mycobacterium tuberculosis/efeitos dos fármacos , Tomografia por Emissão de Pósitrons/métodos , Distribuição Tecidual , Tuberculose Meníngea/diagnóstico por imagem , Tuberculose Meníngea/tratamento farmacológico , Tuberculose Resistente a Múltiplos Medicamentos/diagnóstico por imagem , Tuberculose Resistente a Múltiplos Medicamentos/tratamento farmacológico
20.
Am J Respir Cell Mol Biol ; 71(2): 146-153, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39087829

RESUMO

Oxygen as a key element has a high impact on cellular processes. Infection with a pathogen such as SARS-CoV-2 and after inflammation may lead to hypoxic conditions in tissue that impact cellular responses. To develop optimized translational in vitro models for a better understanding of physiologic and pathophysiologic oxygen conditions, it is a prerequisite to determine oxygen concentrations generated in vivo. Our study objective was the establishment of an invasive method for oxygen measurements using a luminescence-based microsensor to determine the dissolved oxygen in the lung tissue of ferrets as animal models for SARS-CoV-2 research. By way of analogy to humans, aged ferrets are more likely to show clinical signs after SARS-CoV-2 infection than are young animals. To investigate oxygen concentrations during a respiratory viral infection, we intratracheally infected nine aged (3-yr-old) ferrets with SARS-CoV-2. The aged SARS-CoV-2-infected ferrets showed mild to moderate clinical signs associated with prolonged viral RNA shedding until 14 days postinfection. SARS-CoV-2-infected ferrets showed histopathologic lung lesion scores that significantly negatively correlated with oxygen concentrations in lung tissue. At 4 days postinfection, oxygen concentrations in lung tissue were significantly lower (mean percentage O2, 3.89 ≙ ≈ 27.78 mm Hg) than in the negative control group (mean percentage O2, 8.65 ≙ ≈ 61.4 mm Hg). In summary, we succeeded in determining the pathophysiologic oxygen conditions in the lung tissue of aged SARS-CoV-2-infected ferrets.


Assuntos
COVID-19 , Modelos Animais de Doenças , Furões , Hipóxia , Pulmão , Oxigênio , SARS-CoV-2 , Animais , COVID-19/metabolismo , COVID-19/virologia , Oxigênio/metabolismo , Pulmão/metabolismo , Pulmão/virologia , Pulmão/patologia , Hipóxia/metabolismo , Hipóxia/virologia , Masculino , Feminino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA