Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.673
Filtrar
1.
J Immunotoxicol ; 21(1): 2332172, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38563602

RESUMO

Efficacious therapeutic options capable of resolving inflammatory lung disease associated with environmental and occupational exposures are lacking. This study sought to determine the preclinical therapeutic potential of lung-delivered recombinant interleukin (IL)-10 therapy following acute organic dust exposure in mice. Here, C57BL/6J mice were intratracheally instilled with swine confinement organic dust extract (ODE) (12.5%, 25%, 50% concentrations) with IL-10 (1 µg) treatment or vehicle control intratracheally-administered three times: 5 hr post-exposure and then daily for 2 days. The results showed that IL-10 treatment reduced ODE (25%)-induced weight loss by 66% and 46% at Day 1 and Day 2 post-exposure, respectively. IL-10 treatment reduced ODE (25%, 50%)-induced lung levels of TNFα (-76%, -83% [reduction], respectively), neutrophil chemoattractant CXCL1 (-51%, -60%), and lavage fluid IL-6 (-84%, -89%). IL-10 treatment reduced ODE (25%, 50%)-induced lung neutrophils (-49%, -70%) and recruited CD11cintCD11b+ monocyte-macrophages (-49%, -70%). IL-10 therapy reduced ODE-associated expression of antigen presentation (MHC Class II, CD80, CD86) and inflammatory (Ly6C) markers and increased anti-inflammatory CD206 expression on CD11cintCD11b+ cells. ODE (12.5%, 25%)-induced lung pathology was also reduced with IL-10 therapy. In conclusion, the studies here showed that short-term, lung-delivered IL-10 treatment induced a beneficial response in reducing inflammatory consequences (that were also associated with striking reduction in recruited monocyte-macrophages) following acute complex organic dust exposure.


Assuntos
Pneumopatias , Pneumonia , Animais , Camundongos , Suínos , Interleucina-10/metabolismo , Camundongos Endogâmicos C57BL , Pneumonia/tratamento farmacológico , Pulmão/patologia , Pneumopatias/induzido quimicamente , Pneumopatias/tratamento farmacológico , Poeira
2.
Ecotoxicol Environ Saf ; 275: 116247, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38520808

RESUMO

The epidemiological evidences for the association between cooking fuel exposure and respiratory health were inconsistent, and repeated-measures prospective evaluation of cooking fuel exposure was still lacking. We assessed the longitudinal association of chronic lung disease (CLD) and lung function with cooking fuel types among Chinese adults aged ≥ 40 years. In this prospective, nationwide representative cohort of the China Health and Retirement Longitudinal Study from 2011 to 2018, 9004 participants from 28 provinces in China were included. CLD was identified based on self-reported physician diagnosis in 2018. Lung function was assessed by peak expiratory flow (PEF) in 2011, 2013 and 2015. Multivariable logistic and linear mixed-effects repeated-measures models were conducted to measure the associations of CLD and PEF with cooking fuel types. Three-level mixed-effects model was performed as sensitivity analysis. Among the participants, 3508 and 3548 participants used persistent solid and clean cooking fuels throughout the survey, and 1948 participants who used solid cooking fuels at baseline switched to clean cooking fuels. Use of persistent clean cooking fuels (adjusted odds ratio [aOR] = 0.73, 95 % confidence interval [CI]: 0.61, 0.88) and switch of solid fuels to clean fuels (aOR = 0.81, 95 % CI: 0.67, 0.98) were associated with lower risk of CLD. The use of clean cooking fuels throughout the survey and switch of solid fuels to clean fuels in 2013 were also significantly associated with higher PEF level. Similar results were observed in stratified analyses and different statistical models. The evidence from CHARLS cohort suggested that reducing solid cooking fuel exposure was associated with lower risk of CLD and better lung function. Given the recent evidence, improving household air quality will reduce the burden of chronic lung diseases.


Assuntos
Poluição do Ar em Ambientes Fechados , Pneumopatias , Adulto , Humanos , Estudos Longitudinais , Aposentadoria , Estudos Prospectivos , Pneumopatias/induzido quimicamente , Pneumopatias/epidemiologia , Culinária/métodos , Poluição do Ar em Ambientes Fechados/efeitos adversos , Poluição do Ar em Ambientes Fechados/análise , China/epidemiologia
3.
Environ Int ; 186: 108580, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38507932

RESUMO

Desert dust exposure is associated with adverse respiratory health effects. Desert dust is a complex pollutant mixtures that includes respirable crystalline and amorphous particles, metals, and microbial constituents. Given the health effects of desert dust and its heterogeneity, as yet unidentified harmful biological pathways may be triggered. Therefore, we exposed human in vitro air-liquid interface co-cultures of alveolar epithelial A549 cells and THP-1 macrophages to Saharan dust (SD). For comparison, we used the known pulmonary toxicant DQ12 quartz dust. Via RNA sequencing, we identified that SD but not DQ12 increased the gene expression of granulocyte-macrophage colony-stimulating factor (GMCSF) and granulocyte colony-stimulating factor (GCSF). These findings were confirmed by quantitative reverse transcriptase PCR. SD dose-dependently upregulated GMCSF and GCSF expression with significant 7 and 9-fold changes, respectively, at the highest tested concentration of 31 µg/cm2. Furthermore, we observed that SD significantly enhanced the secretion of GM-CSF and G-CSF by 2-fold. Both cytokines have previously been associated with lung diseases such as asthma and fibrosis. Hence, we present two molecular messengers that may contribute to the adverse health effects of desert dust and might serve as drug targets for this globally relevant non-anthropogenic air pollutant.


Assuntos
Poeira , Fator Estimulador de Colônias de Granulócitos , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Pneumopatias , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Fator Estimulador de Colônias de Granulócitos/metabolismo , Pneumopatias/induzido quimicamente , Células A549 , Células THP-1 , Citocinas/metabolismo
4.
Inhal Toxicol ; 36(2): 57-74, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38422051

RESUMO

Many inhalation exposures induce pulmonary inflammation contributing to disease progression. Inflammatory processes are actively regulated via mediators including bioactive lipids. Bioactive lipids are potent signaling molecules involved in both pro-inflammatory and resolution processes through receptor interactions. The formation and clearance of lipid signaling mediators are controlled by multiple metabolic enzymes. An imbalance of these lipids can result in exacerbated and sustained inflammatory processes which may result in pulmonary damage and disease. Dysregulation of pulmonary bioactive lipids contribute to inflammation and pulmonary toxicity following exposures. For example, inhalation of cigarette smoke induces activation of pro-inflammatory bioactive lipids such as sphingolipids, and ceramides contributing to chronic obstructive pulmonary disease. Additionally, exposure to silver nanoparticles causes dysregulation of inflammatory resolution lipids. As inflammation is a common consequence resulting from inhaled exposures and a component of numerous diseases it represents a broadly applicable target for therapeutic intervention. With new appreciation for bioactive lipids, technological advances to reliably identify and quantify lipids have occurred. In this review, we will summarize, integrate, and discuss findings from recent studies investigating the impact of inhaled exposures on pro-inflammatory and resolution lipids within the lung and their contribution to disease. Throughout the review current knowledge gaps in our understanding of bioactive lipids and their contribution to pulmonary effects of inhaled exposures will be presented. New methods being employed to detect and quantify disruption of pulmonary lipid levels following inhalation exposures will be highlighted. Lastly, we will describe how lipid dysregulation could potentially be addressed by therapeutic strategies to address inflammation.


Assuntos
Pneumopatias , Nanopartículas Metálicas , Humanos , Exposição por Inalação/efeitos adversos , Prata , Inflamação/induzido quimicamente , Pneumopatias/induzido quimicamente , Ceramidas , Mediadores da Inflamação/metabolismo
5.
Respir Investig ; 62(2): 284-290, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38277865

RESUMO

BACKGROUND: CONVERT, a randomized, active-controlled, global, Phase 3 trial demonstrated that patients with treatment-refractory Mycobacterium avium complex (MAC) pulmonary disease were more likely to achieve culture conversion with amikacin liposome inhalation suspension (ALIS) plus guideline-based therapy (GBT) versus those continuing on GBT alone. This subgroup analysis reports the efficacy and safety of ALIS in Japanese patients enrolled in CONVERT. METHODS: Japanese patients aged ≥20 years with treatment-refractory MAC pulmonary disease from Japanese sites were included. Patients were randomized to receive once-daily 590 mg ALIS + GBT or GBT alone; patients converting by Month 6 remained in the study to complete 12-month treatment followed by a 12-month off-treatment period. Nonconverters exited the study at Month 8. The primary endpoint was the proportion of patients achieving culture conversion by Month 6. RESULTS: Of the 59 Japanese patients screened, 48 were randomized to receive ALIS + GBT (n = 34) or GBT alone (n = 14), and 41/48 (85.4 %) were women. The mean (standard deviation) age of patients was 64.5 (8.6) years, and 83.3 % of patients had bronchiectasis at baseline. By Month 6, sputum culture conversion was cumulatively achieved in 9/34 (26.5 %) patients receiving ALIS + GBT versus none receiving GBT alone. Treatment-emergent adverse events were reported in 94.1 % and 100.0 % of patients receiving ALIS + GBT and GBT alone, respectively. No deaths were reported. CONCLUSIONS: The efficacy observed in the Japanese subpopulation was largely consistent with that in the overall CONVERT study population, with more patients achieving culture conversion with ALIS + GBT versus GBT alone. Safety profiles were similar between the overall population and the Japanese subpopulation. CLINICAL TRIAL REGISTRATION: NCT02344004.


Assuntos
Pneumopatias , Infecção por Mycobacterium avium-intracellulare , Feminino , Humanos , Masculino , Amicacina/efeitos adversos , Antibacterianos/efeitos adversos , Japão , Lipossomos/uso terapêutico , Pneumopatias/induzido quimicamente , Complexo Mycobacterium avium , Infecção por Mycobacterium avium-intracellulare/tratamento farmacológico , Pessoa de Meia-Idade , Idoso
6.
Sci Total Environ ; 912: 169352, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38110102

RESUMO

Silicon dioxide nanoparticles (SiNPs) are one of the major forms of silicon dioxide and are composed of the most-abundant compounds on earth. Based on their excellent properties, SiNPs are widely used in food production, synthetic processes, medical diagnostics, drug delivery, and other fields. The mass production and wide application of SiNPs increases the risk of human exposure to SiNPs. In the workplace and environment, SiNPs mainly enter the human body through the respiratory tract and reach the lungs; therefore, the lungs are the most important and most toxicologically affected target organ of SiNPs. An increasing number of studies have shown that SiNP exposure can cause severe lung toxicity. However, studies on the toxicity of SiNPs in ex vivo and in vivo settings are still in the exploratory phase. The molecular mechanisms underlying the lung toxicity of SiNPs are varied and not yet fully understood. As a result, this review summarizes the possible mechanisms of SiNP-induced lung toxicity, such as oxidative stress, endoplasmic reticulum stress, mitochondrial damage, and cell death. Moreover, this study provides a summary of the progression of diseases caused by SiNPs, thereby establishing a theoretical basis for future studies on the mechanisms of SiNP-induced lung toxicity.


Assuntos
Pneumopatias , Nanopartículas , Humanos , Dióxido de Silício/toxicidade , Nanopartículas/toxicidade , Estresse Oxidativo , Pulmão , Pneumopatias/induzido quimicamente
7.
Ann Clin Microbiol Antimicrob ; 22(1): 106, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-38057841

RESUMO

OBJECTIVE: To evaluate the effectiveness and safety of linezolid-containing regimens for treatment of M. abscessus pulmonary disease. METHODS: The records of 336 patients with M. abscessus pulmonary disease who were admitted to Shanghai Pulmonary Hospital from January 2018 to December 2020 were retrospectively analyzed. A total of 164 patients received a linezolid-containing regimen and 172 controls did not. The effectiveness, safety, antibiotic susceptibility profiles, outcomes, culture conversion, cavity closure, and adverse reactions were compared in these two groups. RESULTS: The two groups had similar treatment success (56.1% vs. 48.8%; P > 0.05), but treatment duration was shorter in the linezolid group (16.0 months [inter-quartile ranges, IQR: 15.0-17.0] vs. 18.0 months [IQR: 16.0-18.0]; P < 0.01). The rates of sputum culture conversion were similar (53.7% vs. 46.5%, P > 0.05), but time to conversion was shorter in the linezolid group (3.5 months [IQR: 2.5-4.4] vs. 5.5 months [IQR: 4.0-6.8]; P < 0.01). The linezolid group had a higher rate of cavity closure (55.2% vs. 28.6%, P < 0.05) and a shorter time to cavity closure (3.5 months [IQR: 2.5-4.4] vs. 5.5 months [IQR: 4.0-6.8]; P < 0.01). Anemia and peripheral neuropathy were more common in the linezolid group (17.7% vs. 1.7%, P < 0.01; 12.8% vs. 0.6%, P < 0.01). CONCLUSIONS: The linezolid and control groups had similar treatment success rates. The linezolid group had a shorter treatment duration, shorter time to sputum culture conversion, and higher rate and shorter time to lung cavity closure. More patients receiving linezolid developed anemia and peripheral neuropathy.


Assuntos
Anemia , Pneumopatias , Infecções por Mycobacterium não Tuberculosas , Mycobacterium abscessus , Doenças do Sistema Nervoso Periférico , Humanos , Linezolida/efeitos adversos , Estudos Retrospectivos , China , Pneumopatias/tratamento farmacológico , Pneumopatias/induzido quimicamente , Pneumopatias/microbiologia , Resultado do Tratamento , Anemia/induzido quimicamente , Anemia/tratamento farmacológico , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Antibacterianos/efeitos adversos
8.
Ren Fail ; 45(2): 2282710, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37975167

RESUMO

A 68-year-old male, who was undergoing XELOX plus trastuzumab therapy for gastric cancer, developed proteinuria, hematuria, and progressive increase in creatinine after 3 months. Subsequently, the patient also experienced hemoptysis, nasal bleeding. Chest CT examination shown pulmonary hemorrhage. The MRI of the nasopharynx ruled out nasopharyngeal cancer recurrence. The MPO and PR3 were elevated, and renal biopsy confirmed ANCA-related vasculitis, which affected the lungs, kidneys, and nasopharynx. Based on the review of the patient''s medical history and medication, it is believed that ANCA-related vasculitis was caused by XELOX plus trastuzumab chemotherapy, but it is difficult to confirm which specific drug caused it. After stopping XELOX plus trastuzumab chemotherapy, glucocorticoids and cyclophosphamide was given, the patient''s pulmonary hemorrhage and nasal bleeding stopped, and the lung lesions were absorbed. The renal function also improved. The patient later experienced pulmonary infection again, and tNGS indicated Legionella pneumophila and pulmonary tuberculosis infection. Despite anti-infection treatment, steroid dose was rapidly reduced. Ultimately, the patient gave up on treatment and eventually died.


Assuntos
Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos , Nefropatias , Pneumopatias , Neoplasias Nasofaríngeas , Masculino , Humanos , Idoso , Oxaliplatina , Anticorpos Anticitoplasma de Neutrófilos , Trastuzumab/efeitos adversos , Capecitabina , Epistaxe/complicações , Neoplasias Nasofaríngeas/complicações , Recidiva Local de Neoplasia/complicações , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/induzido quimicamente , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/tratamento farmacológico , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/complicações , Pneumopatias/induzido quimicamente , Nefropatias/complicações , Peroxidase
9.
Toxicol Sci ; 196(2): 141-151, 2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-37740395

RESUMO

Inhaled toxicants drive the onset of and exacerbate preexisting chronic pulmonary diseases, however, the biological mechanisms by which this occurs are largely unknown. Exposure to inhaled toxicants, both environmental and occupational, drives pulmonary inflammation and injury. Upon activation of the inflammatory response, polyunsaturated fatty acids (PUFAs) are metabolized into predominately proinflammatory lipid mediators termed eicosanoids which recruit immune cells to the site of injury, perpetuating inflammation to clear the exposed toxicants. Following inflammation, lipid mediator class-switching occurs, a process that leads to increased metabolism of hydroxylated derivates of PUFAs. These mediators, which include mono-hydroxylated PUFA derivatives and specialized proresolving lipid mediators, initiate an active process of inflammation resolution by inhibiting the inflammatory response and activating resolution pathways to return the tissue to homeostasis. Exposure to inhaled toxicants leads to alterations in the synthesis of these proinflammatory and proresolving lipid mediator pathways, resulting in greater pulmonary inflammation and injury, and increasing the risk for the onset of chronic lung diseases. Recent studies have begun utilizing supplementation of PUFAs and their metabolites as potential therapeutics for toxicant-induced pulmonary inflammation and injury. Here we will review the current understanding of the lipid mediators in pulmonary inflammation and resolution as well as the impact of dietary fatty acid supplementation on lipid mediator-driven inflammation following air pollution exposure.


Assuntos
Pneumopatias , Pneumonia , Humanos , Metabolismo dos Lipídeos , Pulmão/metabolismo , Inflamação/metabolismo , Ácidos Graxos Insaturados/metabolismo , Pneumonia/metabolismo , Eicosanoides/metabolismo , Pneumopatias/induzido quimicamente , Mediadores da Inflamação/metabolismo
10.
J Investig Med High Impact Case Rep ; 11: 23247096231196698, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37649377

RESUMO

Diffuse alveolar hemorrhage (DAH) syndrome is characterized by bleeding into the alveolar spaces of the lungs and occurs when there is an injury to the alveolar microcirculation that leads to hemorrhage from the alveolar capillaries. We report a case of an 82-year-old woman who presented with acute respiratory distress. The patient had a history of rheumatoid arthritis (RA) and was on amiodarone for atrial fibrillation therapy. Initial diagnostic workup, including bronchoscopy and imaging studies, revealed diffuse opacities and the bronchoalveolar lavage fluid consistent with DAH. The patient required aggressive management with supportive care and corticosteroids. Laboratory work showed a synergistic effect between amiodarone and RA in inducing DAH. This makes the report unique as no reports in the literature described a synergic effect of amiodarone and RA in inducing DAH. The primary objective of this report is to guide physicians and remind them to keep DAH at the top of their differential diagnosis in the setting of an RA patient taking amiodarone.


Assuntos
Amiodarona , Artrite Reumatoide , Pneumopatias , Feminino , Humanos , Idoso de 80 Anos ou mais , Amiodarona/efeitos adversos , Alvéolos Pulmonares/irrigação sanguínea , Hemorragia/induzido quimicamente , Hemorragia/diagnóstico , Pneumopatias/induzido quimicamente , Pneumopatias/diagnóstico por imagem , Artrite Reumatoide/complicações , Artrite Reumatoide/tratamento farmacológico
11.
Respir Res ; 24(1): 187, 2023 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-37443038

RESUMO

As the public health burden of air pollution continues to increase, new strategies to mitigate harmful health effects are needed. Dietary antioxidants have previously been explored to protect against air pollution-induced lung injury producing inconclusive results. Inhaled (pulmonary or nasal) administration of antioxidants presents a more promising approach as it could directly increase antioxidant levels in the airway surface liquid (ASL), providing protection against oxidative damage from air pollution. Several antioxidants have been shown to exhibit antioxidant, anti-inflammatory, and anti-microbial properties in in vitro and in vivo models of air pollution exposure; however, little work has been done to translate these basic research findings into practice. This narrative review summarizes these findings and data from human studies using inhaled antioxidants in response to air pollution, which have produced positive results, indicating further investigation is warranted. In addition to human studies, cell and murine studies should be conducted using more relevant models of exposure such as air-liquid interface (ALI) cultures of primary cells and non-aqueous apical delivery of antioxidants and pollutants. Inhalation of antioxidants shows promise as a protective intervention to prevent air pollution-induced lung injury and exacerbation of existing lung disease.


Assuntos
Poluentes Atmosféricos , Poluição do Ar , Pneumopatias , Lesão Pulmonar , Humanos , Camundongos , Animais , Antioxidantes/farmacologia , Poluição do Ar/efeitos adversos , Poluição do Ar/análise , Pneumopatias/induzido quimicamente , Pneumopatias/prevenção & controle , Pulmão , Poluentes Atmosféricos/efeitos adversos
12.
Adv Pharmacol ; 98: 55-81, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37524492

RESUMO

Phosphodiesterase enzymes (PDE) have long been known as regulators of cAMP and cGMP, second messengers involved in various signaling pathways and expressed in a variety of cell types implicated in respiratory diseases such as airway smooth muscle and inflammatory cells making them a key target for the treatment of lung diseases as chronic obstructive pulmonary disease (COPD), asthma, cystic fibrosis, and pulmonary hypertension (PH). The first reported PDE inhibitor was the xanthine, theophylline, described as a non-specific PDE inhibitor and whilst this drug is effective, it also has a range of unwanted side effects. In an attempt to improve the therapeutic window of xanthines, a number of selective PDE inhibitors have been developed for the treatment of respiratory diseases with only the selective PDE 4 inhibitor, roflumilast, being approved for the treatment of severe COPD. However, roflumilast also has a very narrow therapeutic window due to a number of important doses limiting side effects, particularly in the gastrointestinal tract. However, there continues to be research carried out in this field to identify improved selective PDE inhibitors, both by targeting other PDE subtypes (e.g., PDE 7 found in a number of inflammatory and immune cells) and through development of selective PDE inhibitors for pulmonary administration to reduce systemic exposure and improve the side effect profile. This approach has been exemplified by the development of ensifentrine, a dual PDE 3-PDE 4 inhibitor, an inhaled drug that has recently completed two successful Phase III clinical trials in patients with COPD.


Assuntos
Pneumopatias , Inibidores da Fosfodiesterase 4 , Doença Pulmonar Obstrutiva Crônica , Humanos , Inibidores da Fosfodiesterase 4/efeitos adversos , Pneumopatias/tratamento farmacológico , Pneumopatias/induzido quimicamente , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/induzido quimicamente , Aminopiridinas/efeitos adversos
14.
Sci Rep ; 13(1): 5955, 2023 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-37045933

RESUMO

Particulate matter (PM), an environmental risk factor, is linked with health risks such as respiratory diseases. This study aimed to establish an animal model of PM-induced lung injury with artificial PM (APM) and identify the potential of APM for toxicological research. APM was generated from graphite at 600 °C and combined with ethylene. We analyzed diesel exhaust particulate (DEP) and APM compositions and compared toxicity and transcriptomic profiling in lungs according to the exposure. For the animal study, C57BL/6 male mice were intratracheally administered vehicle, DEP, or APM. DEP or APM increased relative lung weight, inflammatory cell numbers, and inflammatory protein levels compared with the vehicle control. Histological assessments showed an increase in particle-pigment alveolar macrophages and slight inflammation in the lungs of DEP and APM mice. In the only APM group, granulomatous inflammation, pulmonary fibrosis, and mucous hyperplasia were observed in the lungs of some individuals. This is the first study to compare pulmonary toxicity between DEP and APM in an animal model. Our results suggest that the APM-treated animal model may contribute to understanding the harmful effects of PM in toxicological studies showing that APM can induce various lung diseases according to different doses of APM.


Assuntos
Pneumopatias , Material Particulado , Camundongos , Masculino , Animais , Material Particulado/toxicidade , Material Particulado/metabolismo , Transcriptoma , Emissões de Veículos/toxicidade , Camundongos Endogâmicos C57BL , Pulmão/patologia , Pneumopatias/induzido quimicamente , Pneumopatias/genética
16.
Respir Res ; 24(1): 95, 2023 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-36978106

RESUMO

The lungs, in addition to participating in gas exchange, represent the first line of defense against inhaled pathogens and respiratory toxicants. Cells lining the airways and alveoli include epithelial cells and alveolar macrophages, the latter being resident innate immune cells important in surfactant recycling, protection against bacterial invasion and modulation of lung immune homeostasis. Environmental exposure to toxicants found in cigarette smoke, air pollution and cannabis can alter the number and function of immune cells in the lungs. Cannabis (marijuana) is a plant-derived product that is typically inhaled in the form of smoke from a joint. However, alternative delivery methods such as vaping, which heats the plant without combustion, are becoming more common. Cannabis use has increased in recent years, coinciding with more countries legalizing cannabis for both recreational and medicinal purposes. Cannabis may have numerous health benefits owing to the presence of cannabinoids that dampen immune function and therefore tame inflammation that is associated with chronic diseases such as arthritis. The health effects that could come with cannabis use remain poorly understood, particularly inhaled cannabis products that may directly impact the pulmonary immune system. Herein, we first describe the bioactive phytochemicals present in cannabis, with an emphasis on cannabinoids and their ability to interact with the endocannabinoid system. We also review the current state-of-knowledge as to how inhaled cannabis/cannabinoids can shape immune response in the lungs and discuss the potential consequences of altered pulmonary immunity. Overall, more research is needed to understand how cannabis inhalation shapes the pulmonary immune response to balance physiological and beneficial responses with potential deleterious consequences on the lungs.


Assuntos
Canabinoides , Cannabis , Pneumopatias , Humanos , Cannabis/efeitos adversos , Pulmão , Pneumopatias/induzido quimicamente , Pneumopatias/tratamento farmacológico , Canabinoides/farmacologia , Imunidade
17.
Int. j. morphol ; 41(1): 167-174, feb. 2023. ilus, tab, graf
Artigo em Inglês | LILACS | ID: biblio-1430531

RESUMO

SUMMARY: The present study investigated the possible protective effects of melatonin on Bleomycin, Cisplatin and etoposide (BEP) chemotherapy regimens using immunohistochemistry. Forty male Wistar rats were divided into four groups of ten as; group 1 as untreated control; group 2 as BEP group which received the three cycles of 21 days' regimen each of 0.5¥ dose levels ofBEP (bleomycin 0.75 mg/kg, etoposide 7.5 mg/kg and cisplatin 1.5 mg/kg). Rats in the group 3 (MEL group) received 10 mg/kg/day melatonin once daily. Group 4 received the melatonin (30 min before the BEP injections) and BEP as in groups 2. Proliferating cell nuclear antigen (PCNA) staining was used to detect cell proliferation and caspase-3, caspase-9 and Caspase-8 were detected to investigate apoptosis. PCNA immunostaining in alveolar epithelium, alveolar macrophages and bronchus was weak to moderate in BEP group. However, diffuse and strong caspase immunoreactions for caspase-3, caspase 8- and caspase-9 were detected in the bronchioles epithelium, vascular endothelium, alveolar luminal macrophages in the BEP group. PCNA and caspase immunoreactivities in MEL and Mel + BEP groups were close to the control one. The surface are in the BEP group was significantly reduced as compared to the control one ((P0.05). It can be concluded that BEP regimen can affects negatively on lung tissue and melatonin inhibits lung tissue injuries during BEP chemotherapy.


El presente estudio investigó los posibles efectos protectores de la melatonina en los regímenes de quimioterapia con bleomicina, etopósido y cisplatino (BEP) mediante inmunohistoquímica. Cuarenta ratas Wistar macho se dividieron en cuatro grupos de diez: grupo 1, control sin tratar; grupo 2, quimioterapia con una dosis de 0,5x de BEP (0,75 mg/kg de bleomicina, 7,5 mg/ kg de etopósido y 1,5 mg/kg de cisplatino) con tres ciclos de 21 días cada uno. Las ratas del grupo 3 (grupo MEL) recibieron 10 mg/kg/día de melatonina una vez al día. El grupo 4 (Mel + BEP) recibió melatonina (30 minutos antes de las inyecciones de BEP) y BEP, como en los grupos 2. Se usó la tinción del antígeno nuclear de células en proliferación (PCNA) para detectar la proliferación celular y, caspasa- 3, caspasa-9 y caspasa-8 para investigar apoptosis. La inmunotinción de PCNA en el epitelio alveolar, los macrófagos alveolares y los bronquios varió de débil a moderada en el grupo BEP. Sin embargo, se detectaron inmunorreacciones difusas y fuertes para caspasa-3, caspasa 8- y caspasa-9 en el epitelio de los bronquiolos, endotelio vascular y macrófagos luminales alveolares. Las inmunorreactividades de PCNA y caspasa en los grupos MEL y Mel + BEP fueron similares a las del control. El área de superficie en el grupo BEP se redujo significativamente en comparación con el control (P0,05). Se puede concluir que la quimioterapia con BEP puede afectar negativamente al tejido pulmonar y la melatonina inhibe las lesiones durante la quimioterapia.


Assuntos
Animais , Masculino , Ratos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Pneumopatias/prevenção & controle , Melatonina/administração & dosagem , Antioxidantes/administração & dosagem , Bleomicina/efeitos adversos , Imuno-Histoquímica , Cisplatino/efeitos adversos , Ratos Wistar , Apoptose/efeitos dos fármacos , Antígeno Nuclear de Célula em Proliferação , Substâncias Protetoras , Etoposídeo/efeitos adversos , Pneumopatias/induzido quimicamente
18.
Toxicol Sci ; 191(1): 61-78, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36303316

RESUMO

Air pollution accounts for more than 7 million premature deaths worldwide. Using ultrafine carbon black (CB) and ozone (O3) as a model for an environmental co-exposure scenario, the dose response relationships in acute pulmonary injury and inflammation were determined by generating, characterizing, and comparing stable concentrations of CB aerosols (2.5, 5.0, 10.0 mg/m3), O3 (0.5, 1.0, 2.0 ppm) with mixture CB + O3 (2.5 + 0.5, 5.0 + 1.0, 10.0 + 2.0). C57BL6 male mice were exposed for 3 h by whole body inhalation and acute toxicity determined after 24 h. CB itself did not cause any alteration, however, a dose response in pulmonary injury/inflammation was observed with O3 and CB + O3. This increase in response with mixtures was not dependent on the uptake but was due to enhanced reactivity of the particles. Benchmark dose modeling showed several-fold increase in potency with CB + O3 compared with CB or O3 alone. Principal component analysis provided insight into response relationships between various doses and treatments. There was a significant correlation in lung responses with charge-based size distribution, total/alveolar deposition, oxidant generation, and antioxidant depletion potential. Lung tissue gene/protein response demonstrated distinct patterns that are better predicted by either particle dose/aerosol responses (interleukin-1ß, keratinocyte chemoattractant, transforming growth factor beta) or particle reactivity (thymic stromal lymphopoietin, interleukin-13, interleukin-6). Hierarchical clustering showed a distinct signature with high dose and a similarity in mRNA expression pattern of low and medium doses of CB + O3. In conclusion, we demonstrate that the biological outcomes from CB + O3 co-exposure are significantly greater than individual exposures over a range of aerosol concentrations and aerosol characteristics can predict biological outcome.


Assuntos
Poluentes Atmosféricos , Pneumopatias , Lesão Pulmonar , Ozônio , Pneumonia , Camundongos , Animais , Masculino , Ozônio/toxicidade , Fuligem/toxicidade , Lesão Pulmonar/metabolismo , Aerossóis e Gotículas Respiratórios , Pneumopatias/induzido quimicamente , Pulmão , Pneumonia/metabolismo , Inflamação/metabolismo , Poluentes Atmosféricos/toxicidade , Poluentes Atmosféricos/metabolismo
19.
Biometals ; 36(3): 603-615, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-35976499

RESUMO

Lactoferrin (LTF), an iron binding protein, is known to exhibit immune modulatory effects on pulmonary pathology during insult-induced models of primary Mycobacterium tuberculosis (Mtb) infection. The effects of LTF correlate with modulation of the immune related development of the pathology, and altering of the histological nature of the physically compact and dense lung granuloma in mice. Specifically, a recombinant human version of LTF limits immediate progression of granulomatous severity following administration of the Mtb cell wall mycolic acid, trehalose 6,6'-dimycolate (TDM), in part through reduced pro-inflammatory responses known to control these events. This current study investigates a limited course of LTF to modulate not only initiation, but also maintenance and resolution of pathology post development of the granulomatous response in mice. Comparison is made to a fusion of LTF with the Fc domain of IgG2 (FcLTF), which is known to extend LTF half-life in circulation. TDM induced granulomas were examined at extended times post insult (day 7 and 14). Both LTF and the novel FcLTF exerted sustained effects on lung granuloma pathology. Reduction of pulmonary pro-inflammatory cytokines TNF-α and IL-1ß occurred, correlating with reduced pathology. Increase in IL-6, known to regulate granuloma maintenance, was also seen with the LTFs. The FcLTF demonstrated greater impact than the recombinant LTF, and was superior in limiting damage to pulmonary tissues while limiting residual inflammatory cytokine production.


Assuntos
Fatores Corda , Granuloma do Sistema Respiratório , Lactoferrina , Pneumopatias , Animais , Humanos , Camundongos , Fatores Corda/metabolismo , Fatores Corda/toxicidade , Lactoferrina/uso terapêutico , Mycobacterium tuberculosis/metabolismo , Granuloma do Sistema Respiratório/induzido quimicamente , Granuloma do Sistema Respiratório/tratamento farmacológico , Pneumopatias/induzido quimicamente , Pneumopatias/tratamento farmacológico
20.
Toxicol Mech Methods ; 33(3): 239-247, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36482745

RESUMO

Methotrexate (MTX) is an anticancer agent widely used in clinical practice for various oncological, rheumatological, autoimmune, and inflammatory diseases. However, the side effects of MTX limit its usage for treatment. In addition, diffuse alveolar damage, interstitial pneumonia, fibrosis, and pleural reactions may be encountered in MTX-induced pulmonary toxicity. Ramelteon (RML), a melatonin receptor agonist, has antioxidant, anti-inflammatory, and protective effects are shown by several studies. This study aimed to show the antioxidant, anti-inflammatory, and antiapoptotic effects of RML and its effect on the airway surface liquid volume homeostasis via aquaporins (AQP) in MTX-induced lung injury. Thirty-two female Wistar Albino rats were grouped into four groups as control, MTX (20 mg/kg, intraperitoneally, a single dose), MTX + RML, and RML (10 mg/kg, via oral gavage, for seven days) groups. Once the experiment ended, the rats' lung tissues were taken for biochemical, genetic, histopathological, and immunohistochemical examinations. MTX significantly increased oxidative stress index and total oxidative status, and decreased total antioxidant status levels by 202.0%, 141.4%, 20.2%, respectively, relative to the control (p ˂ 0.001 for all). AQP-1/5, which is an indicator of lung damage, was also found to decrease significantly (p ˂ 0.001). In addition, a significant increase was observed in interleukin-1ß, interferon-beta, and caspase-8 expressions and histopathological changes as a result of immunohistochemical and histochemical examinations (p ˂ 0.001). RML treatment ameliorated all these changes and significantly regressed lung damage. Our results suggest that RML might be used as a lung-protective agent in various models of lung and tissue injury.


Assuntos
Antioxidantes , Pneumopatias , Animais , Ratos , Feminino , Antioxidantes/metabolismo , Ratos Wistar , Metotrexato/toxicidade , Estresse Oxidativo , Pneumopatias/induzido quimicamente , Anti-Inflamatórios/uso terapêutico , Anti-Inflamatórios/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...