Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
Biomed Pharmacother ; 138: 111465, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34311522

RESUMO

Acidic mammalian chitinase (CHIA) belongs to the 18-glycosidase family and is expressed in epithelial cells and certain immune cells (such as neutrophils and macrophages) in various organs. Under physiological conditions, as a hydrolase, CHIA can degrade chitin-containing pathogens, participate in Type 2 helper T (Th2)-mediated inflammation, and enhance innate and adaptive immunity to pathogen invasion. Under pathological conditions, such as rhinitis, ocular conjunctivitis, asthma, chronic atrophic gastritis, type 2 diabetes, and pulmonary interstitial fibrosis, CHIA expression is significantly changed. In addition, studies have shown that CHIA has an anti-apoptotic effect, promotes epithelial cell proliferation and maintains organ integrity, and these effects are not related to chitinase degradation. CHIA can also be used as a biomolecular marker in diseases such as chronic atrophic gastritis, dry eye, and acute kidney damage caused by sepsis. Analysis of the authoritative TCGA database shows that CHIA expression in gastric adenocarcinoma, liver cancer, renal clear cell carcinoma and other tumors is significantly downregulated compared with that in normal tissues, but the specific mechanism is unclear. This review is based on all surveys conducted to date and summarizes the expression patterns and functional diversity of CHIA in various organs. Understanding the physiological and pathophysiological relevance of CHIA in multiple organs opens new possibilities for disease treatment.


Assuntos
Encéfalo/enzimologia , Quitinases/metabolismo , Sistema Digestório/enzimologia , Olho/enzimologia , Rim/enzimologia , Sistema Respiratório/enzimologia , Animais , Encéfalo/fisiopatologia , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/fisiopatologia , Sistema Digestório/fisiopatologia , Olho/fisiopatologia , Humanos , Rim/fisiopatologia , Sistema Respiratório/fisiopatologia , Transdução de Sinais
2.
Elife ; 92020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-33164751

RESUMO

Pandemic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus 19 disease (COVID-19) which presents a large spectrum of manifestations with fatal outcomes in vulnerable people over 70-years-old and with hypertension, diabetes, obesity, cardiovascular disease, COPD, and smoking status. Knowledge of the entry receptor is key to understand SARS-CoV-2 tropism, transmission and pathogenesis. Early evidence pointed to angiotensin-converting enzyme 2 (ACE2) as SARS-CoV-2 entry receptor. Here, we provide a critical summary of the current knowledge highlighting the limitations and remaining gaps that need to be addressed to fully characterize ACE2 function in SARS-CoV-2 infection and associated pathogenesis. We also discuss ACE2 expression and potential role in the context of comorbidities associated with poor COVID-19 outcomes. Finally, we discuss the potential co-receptors/attachment factors such as neuropilins, heparan sulfate and sialic acids and the putative alternative receptors, such as CD147 and GRP78.


Assuntos
Betacoronavirus/fisiologia , Infecções por Coronavirus/virologia , Peptidil Dipeptidase A/fisiologia , Pneumonia Viral/virologia , Ligação Viral , Enzima de Conversão de Angiotensina 2 , Basigina/fisiologia , COVID-19 , Comorbidade , Infecções por Coronavirus/epidemiologia , Chaperona BiP do Retículo Endoplasmático , Regulação Enzimológica da Expressão Gênica , Heparitina Sulfato/fisiologia , Humanos , Hipertensão/epidemiologia , Hipertensão/fisiopatologia , Neuropilina-1/fisiologia , Oligopeptídeos/fisiologia , Especificidade de Órgãos , Pandemias , Pneumonia Viral/epidemiologia , Ligação Proteica , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Virais , Sistema Renina-Angiotensina/fisiologia , Sistema Respiratório/enzimologia , SARS-CoV-2 , Ácidos Siálicos/fisiologia , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/fisiologia , Internalização do Vírus
3.
Am J Physiol Lung Cell Mol Physiol ; 319(5): L843-L847, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32996784

RESUMO

The incidence, severity, and mortality of ongoing coronavirus infectious disease 19 (COVID-19) is greater in men compared with women, but the underlying factors contributing to this sex difference are still being explored. In the current study, using primary isolated human airway smooth muscle (ASM) cells from normal males versus females as a model, we explored the effect of estrogen versus testosterone in modulating the expression of angiotensin converting enzyme 2 (ACE2), a cell entry point for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Using confocal imaging, we found that ACE2 is expressed in human ASM. Furthermore, Western analysis of ASM cell lysates showed significantly lower ACE2 expression in females compared with males at baseline. In addition, ASM cells exposed to estrogen and testosterone for 24 h showed that testosterone significantly upregulates ACE2 expression in both males and females, whereas estrogen downregulates ACE2, albeit not significant compared with vehicle. These intrinsic and sex steroids induced differences may help explain sex differences in COVID-19.


Assuntos
Infecções por Coronavirus/metabolismo , Peptidil Dipeptidase A/biossíntese , Pneumonia Viral/metabolismo , Sistema Respiratório/metabolismo , Adulto , Idoso , Enzima de Conversão de Angiotensina 2 , COVID-19 , Células Cultivadas , Infecções por Coronavirus/enzimologia , Estrogênios/metabolismo , Estrogênios/farmacologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/metabolismo , Pandemias , Peptidil Dipeptidase A/metabolismo , Pneumonia Viral/enzimologia , Sistema Respiratório/citologia , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/enzimologia , Fatores Sexuais , Testosterona/metabolismo , Testosterona/farmacologia
4.
PLoS Pathog ; 15(8): e1007963, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31381610

RESUMO

Human respiratory syncytial virus (RSV) is the leading viral cause of acute pediatric lower respiratory tract infections worldwide, with no available vaccine or effective antiviral drug. To gain insight into virus-host interactions, we performed a genome-wide siRNA screen. The expression of over 20,000 cellular genes was individually knocked down in human airway epithelial A549 cells, followed by infection with RSV expressing green fluorescent protein (GFP). Knockdown of expression of the cellular ATP1A1 protein, which is the major subunit of the Na+,K+-ATPase of the plasma membrane, had one of the strongest inhibitory effects on GFP expression and viral titer. Inhibition was not observed for vesicular stomatitis virus, indicating that it was RSV-specific rather than a general effect. ATP1A1 formed clusters in the plasma membrane very early following RSV infection, which was independent of replication but dependent on the attachment glycoprotein G. RSV also triggered activation of ATP1A1, resulting in signaling by c-Src-kinase activity that transactivated epidermal growth factor receptor (EGFR) by Tyr845 phosphorylation. ATP1A1 signaling and activation of both c-Src and EGFR were found to be required for efficient RSV uptake. Signaling events downstream of EGFR culminated in the formation of macropinosomes. There was extensive uptake of RSV virions into macropinosomes at the beginning of infection, suggesting that this is a major route of RSV uptake, with fusion presumably occurring in the macropinosomes rather than at the plasma membrane. Important findings were validated in primary human small airway epithelial cells (HSAEC). In A549 cells and HSAEC, RSV uptake could be inhibited by the cardiotonic steroid ouabain and the digitoxigenin derivative PST2238 (rostafuroxin) that bind specifically to the ATP1A1 extracellular domain and block RSV-triggered EGFR Tyr845 phosphorylation. In conclusion, we identified ATP1A1 as a host protein essential for macropinocytic entry of RSV into respiratory epithelial cells, and identified PST2238 as a potential anti-RSV drug.


Assuntos
Pinocitose , Infecções por Vírus Respiratório Sincicial/complicações , Vírus Sincicial Respiratório Humano/patogenicidade , Infecções Respiratórias/prevenção & controle , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Proteínas Virais/metabolismo , Internalização do Vírus , Células A549 , Cardiotônicos/farmacologia , Digitoxigenina/química , Digitoxigenina/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Células Epiteliais/virologia , Receptores ErbB/metabolismo , Ensaios de Triagem em Larga Escala , Humanos , Ouabaína/farmacologia , RNA Interferente Pequeno/genética , Infecções por Vírus Respiratório Sincicial/virologia , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/enzimologia , Sistema Respiratório/virologia , Infecções Respiratórias/epidemiologia , Infecções Respiratórias/virologia , Transdução de Sinais , ATPase Trocadora de Sódio-Potássio/genética , Proteínas Virais/genética
5.
Transbound Emerg Dis ; 66(2): 831-841, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30520548

RESUMO

This study investigated the co-localization of the Middle East respiratory syndrome coronavirus (MERS-CoV) and its receptor dipeptidyl peptidase-4 (DPP4) by immunohistochemistry (IHC) across respiratory and lymphoid organs of experimentally MERS-CoV infected pigs and llamas. Also, scanning electron microscopy was performed to assess the ciliary integrity of respiratory epithelial cells in both species. In pigs, on day 2 post-inoculation (p.i.), DPP4-MERS-CoV co-localization was detected in medial turbinate epithelium. On day 4 p.i., the virus/receptor co-localized in frontal and medial turbinate epithelial cells in pigs, and epithelial cells distributed unevenly through the whole nasal cavity and in the cervical lymph node in llamas. MERS-CoV viral nucleocapsid was mainly detected in upper respiratory tract sites on days 2 and 4 p.i. in pigs and day 4 p.i. in llamas. No MERS-CoV was detected on day 24 p.i. in any tissue by IHC. While pigs showed severe ciliary loss in the nasal mucosa both on days 2 and 4 p.i. and moderate loss in the trachea on days 4 and 24 p.i., ciliation of respiratory organs in llamas was not significantly affected. Obtained data confirm the role of DPP4 for MERS-CoV entry in respiratory epithelial cells of llamas. Notably, several nasal epithelial cells in pigs were found to express viral antigen but not DPP4, suggesting the possible existence of other molecule/s facilitating virus entry or down regulation of DPP4 upon infection.


Assuntos
Camelídeos Americanos/virologia , Infecções por Coronavirus/veterinária , Dipeptidil Peptidase 4/metabolismo , Tecido Linfoide/enzimologia , Coronavírus da Síndrome Respiratória do Oriente Médio/patogenicidade , Sistema Respiratório/enzimologia , Doenças dos Suínos/virologia , Animais , Infecções por Coronavirus/virologia , Imuno-Histoquímica/veterinária , Microscopia Eletrônica de Varredura/veterinária , RNA Viral/genética , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Receptores Virais/metabolismo , Suínos
6.
Clin Sci (Lond) ; 132(15): 1711-1723, 2018 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-29980604

RESUMO

Rhinovirus (RV) infection is involved in acute exacerbations of asthma and chronic obstructive pulmonary disease (COPD). RV primarily infects upper and lower airway epithelium. Immunoproteasomes (IP) are proteolytic machineries with multiple functions including the regulation of MHC class I antigen processing during viral infection. However, the role of IP in RV infection has not been explored. We sought to investigate the expression and function of IP during airway RV infection. Primary human tracheobronchial epithelial (HTBE) cells were cultured at air-liquid interface (ALI) and treated with RV16, RV1B, or interferon (IFN)-λ in the absence or presence of an IP inhibitor (ONX-0914). IP gene (i.e. LMP2) deficient mouse tracheal epithelial cells (mTECs) were cultured for the mechanistic studies. LMP2-deficient mouse model was used to define the in vivo role of IP in RV infection. IP subunits LMP2 and LMP7, antiviral genes MX1 and OAS1 and viral load were measured. Both RV16 and RV1B significantly increased the expression of LMP2 and LMP7 mRNA and proteins, and IFN-λ mRNA in HTBE cells. ONX-0914 down-regulated MX1 and OAS1, and increased RV16 load in HTBE cells. LMP2-deficient mTECs showed a significant increase in RV1B load compared with the wild-type (WT) cells. LMP2-deficient (compared with WT) mice increased viral load and neutrophils in bronchoalveolar lavage (BAL) fluid after 24 h of RV1B infection. Mechanistically, IFN-λ induction by RV infection contributed to LMP2 and LMP7 up-regulation in HTBE cells. Our data suggest that IP are induced during airway RV infection, which in turn may serve as an antiviral and anti-inflammatory mechanism.


Assuntos
Células Epiteliais/imunologia , Infecções por Picornaviridae/imunologia , Complexo de Endopeptidases do Proteassoma/imunologia , Rhinovirus/imunologia , Animais , Asma/enzimologia , Asma/imunologia , Asma/virologia , Linhagem Celular , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/imunologia , Cisteína Endopeptidases/metabolismo , Células Epiteliais/enzimologia , Células Epiteliais/virologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/imunologia , Humanos , Camundongos Knockout , Oligopeptídeos/farmacologia , Infecções por Picornaviridae/enzimologia , Infecções por Picornaviridae/virologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Sistema Respiratório/enzimologia , Sistema Respiratório/imunologia , Sistema Respiratório/virologia , Rhinovirus/fisiologia
7.
Am J Respir Crit Care Med ; 198(9): 1188-1198, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29864375

RESUMO

RATIONALE: In lung cancer, upregulation of the PI3K (phosphoinositide 3-kinase) pathway is an early event that contributes to cell proliferation, survival, and tissue invasion. Upregulation of this pathway was recently described as associated with enrichment of the lower airways with bacteria identified as oral commensals. OBJECTIVES: We hypothesize that host-microbe interactions in the lower airways of subjects with lung cancer affect known cancer pathways. METHODS: Airway brushings were collected prospectively from subjects with lung nodules at time of diagnostic bronchoscopy, including 39 subjects with final lung cancer diagnoses and 36 subjects with noncancer diagnoses. In addition, samples from 10 healthy control subjects were included. 16S ribosomal RNA gene amplicon sequencing and paired transcriptome sequencing were performed on all airway samples. In addition, an in vitro model with airway epithelial cells exposed to bacteria/bacterial products was performed. MEASUREMENTS AND MAIN RESULTS: The composition of the lower airway transcriptome in the patients with cancer was significantly different from the control subjects, which included up-regulation of ERK (extracellular signal-regulated kinase) and PI3K signaling pathways. The lower airways of patients with lung cancer were enriched for oral taxa (Streptococcus and Veillonella), which was associated with up-regulation of the ERK and PI3K signaling pathways. In vitro exposure of airway epithelial cells to Veillonella, Prevotella, and Streptococcus led to upregulation of these same signaling pathways. CONCLUSIONS: The data presented here show that several transcriptomic signatures previously identified as relevant to lung cancer pathogenesis are associated with enrichment of the lower airway microbiota with oral commensals.


Assuntos
Neoplasias Pulmonares/enzimologia , Microbiota/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Sistema Respiratório/enzimologia , Regulação para Cima/fisiologia , Adulto , Idoso , Broncoscopia , Estudos Transversais , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/microbiologia , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Sistema Respiratório/metabolismo , Sistema Respiratório/microbiologia
8.
Am J Respir Cell Mol Biol ; 58(4): 530-541, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29262264

RESUMO

Two cAMP signaling compartments centered on adenylyl cyclase (AC) exist in human airway smooth muscle (HASM) cells, one containing ß2-adrenergic receptor AC6 and another containing E prostanoid receptor AC2. We hypothesized that different PDE isozymes selectively regulate cAMP signaling in each compartment. According to RNA-sequencing data, 18 of 24 PDE genes were expressed in primary HASM cells derived from age- and sex-matched donors with and without asthma. PDE8A was the third most abundant of the cAMP-degrading PDE genes, after PDE4A and PDE1A. Knockdown of PDE8A using shRNA evoked twofold greater cAMP responses to 1 µM forskolin in the presence of 3-isobutyl-1-methylxanthine. Overexpression of AC2 did not alter this response, but overexpression of AC6 increased cAMP responses an additional 80%. We examined cAMP dynamics in live HASM cells using a fluorescence sensor. PF-04957325, a PDE8-selective inhibitor, increased basal cAMP concentrations by itself, indicating a significant basal level of cAMP synthesis. In the presence of an AC inhibitor to reduce basal signaling, PF-04957325 accelerated cAMP production and increased the inhibition of cell proliferation induced by isoproterenol, but it had no effect on cAMP concentrations or cell proliferation regulated by prostaglandin E2. Lipid raft fractionation of HASM cells revealed PDE8A immunoreactivity in buoyant fractions containing caveolin-1 and AC5/6 immunoreactivity. Thus, PDE8 is expressed in lipid rafts of HASM cells, where it specifically regulates ß2-adrenergic receptor AC6 signaling without effects on signaling by the E prostanoid receptors 2/4-AC2 complex. In airway diseases such as asthma and chronic obstructive pulmonary disease, PDE8 may represent a novel therapeutic target to modulate HASM responsiveness and airway remodeling.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Adenilil Ciclases/metabolismo , Asma/enzimologia , AMP Cíclico/metabolismo , Músculo Liso/enzimologia , Miócitos de Músculo Liso/enzimologia , Receptores Adrenérgicos beta 2/metabolismo , Sistema Respiratório/enzimologia , 3',5'-AMP Cíclico Fosfodiesterases/genética , Adenilil Ciclases/genética , Remodelação das Vias Aéreas , Asma/genética , Asma/patologia , Asma/fisiopatologia , Estudos de Casos e Controles , Proliferação de Células , Células Cultivadas , Humanos , Microdomínios da Membrana/enzimologia , Microdomínios da Membrana/patologia , Músculo Liso/patologia , Músculo Liso/fisiopatologia , Miócitos de Músculo Liso/patologia , Receptores Adrenérgicos beta 2/genética , Sistema Respiratório/patologia , Sistema Respiratório/fisiopatologia , Sistemas do Segundo Mensageiro , Fatores de Tempo
9.
PLoS One ; 11(12): e0167169, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27935962

RESUMO

Chronic obstructive pulmonary disease (COPD) is characterized by persistent airflow limitation and lung inflammation resulting in a progressive decline in lung function whose principle cause is cigarette smoke. MAP3K19 is a novel kinase expressed predominantly by alveolar and interstitial macrophages and bronchial epithelial cells in the lung. We found that MAP3K19 mRNA was overexpressed in a limited sampling of lung tissue from COPD patients, and a closer examination found it to be overexpressed in bronchoalveolar macrophages from COPD patients, as well as the bronchial epithelium and inflammatory cells in the lamina propria. We further found MAP3K19 to be induced in various cell lines upon environmental stress, such as cigarette smoke, oxidative and osmotic stress. Exogenous expression of MAP3K19 in cells caused an upregulation of transcriptionally active NF-κB, and secretion of the chemokines CXCL-8, CCL-20 and CCL-7. Inhibition of MAP3K19 activity by siRNA or small molecular weight inhibitors caused a decrease in cigarette smoke-induced inflammation in various murine models, which included a decrease in pulmonary neutrophilia and KC levels. In a chronic cigarette smoke model, inhibition of MAP3K19 significantly attenuated emphysematous changes in airway parenchyma. Finally, in a viral exacerbation model, mice exposed to cigarette smoke and influenza A virus showed a decrease in pulmonary neutrophilia, pro-inflammatory cytokines and viral load upon inhibition of MAP3K19. Collectively, these results suggest that inhibition of MAP3K19 may represent a novel strategy to target COPD that promises to have a potential therapeutic benefit for patients.


Assuntos
Mediadores da Inflamação/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Pneumonia/enzimologia , Doença Pulmonar Obstrutiva Crônica/enzimologia , Sistema Respiratório/enzimologia , Fumar/efeitos adversos , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Western Blotting , Linhagem Celular Tumoral , Citocinas/metabolismo , Células Epiteliais/enzimologia , Células Epiteliais/metabolismo , Feminino , Expressão Gênica , Células HEK293 , Humanos , MAP Quinase Quinase Quinases/genética , Macrófagos/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Pneumonia/etiologia , Pneumonia/genética , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/metabolismo , Interferência de RNA , Sistema Respiratório/metabolismo , Sistema Respiratório/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Am J Pathol ; 186(1): 78-86, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26597880

RESUMO

Dipeptidyl peptidase 4 (DPP4, CD26), a type II transmembrane ectopeptidase, is the receptor for the Middle Eastern respiratory syndrome coronavirus (MERS-CoV). MERS emerged in 2012 and has a high mortality associated with severe lung disease. A lack of autopsy studies from MERS fatalities has hindered understanding of MERS-CoV pathogenesis. We investigated the spatial and cellular localization of DPP4 to evaluate an association MERS clinical disease. DPP4 was rarely detected in the surface epithelium from nasal cavity to conducting airways with a slightly increased incidence in distal airways. DPP4 was also found in a subset of mononuclear leukocytes and in serous cells of submucosal glands. In the parenchyma, DPP4 was found principally in type I and II cells and alveolar macrophages and was also detected in vascular endothelium (eg, lymphatics) and pleural mesothelia. Patients with chronic lung disease, such as chronic obstructive pulmonary disease and cystic fibrosis, exhibited increased DPP4 immunostaining in alveolar epithelia (type I and II cells) and alveolar macrophages with similar trends in reactive mesothelia. This finding suggests that preexisting pulmonary disease could increase MERS-CoV receptor abundance and predispose individuals to MERS morbidity and mortality, which is consistent with current clinical observations. We speculate that the preferential spatial localization of DPP4 in alveolar regions may explain why MERS is characterized by lower respiratory tract disease.


Assuntos
Infecções por Coronavirus , Dipeptidil Peptidase 4/biossíntese , Sistema Respiratório/enzimologia , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Feminino , Humanos , Imuno-Histoquímica , Lactente , Masculino , Pessoa de Meia-Idade , Adulto Jovem
11.
Am J Respir Cell Mol Biol ; 54(2): 241-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26181301

RESUMO

Airway hyperresponsiveness (AHR) is a hallmark feature in asthma characterized by exaggerated airway contractile response to stimuli due to increased airway sensitivity and chronic airway remodeling. We have previously shown that allergen-induced AHR in mice is associated with aberrant DNA methylation in the lung genome, suggesting that AHR could be epigenetically regulated, and these changes might predispose the animals to asthma. Previous studies demonstrated that overexpression of phosphodiesterase 4D (PDE4D) is associated with increased AHR. However, epigenetic regulation of this gene in asthmatic airway smooth muscle cells (ASMCs) has not been examined. In this study, we aimed to examine the relationship between epigenetic regulation of PDE4D and ASMC phenotypes. We identified CpG site-specific hypomethylation at PDE4D promoter in human asthmatic ASMCs. We next used methylated oligonucleotides to introduce CpG site-specific methylation at PDE4D promoter and examined its effect on ASMCs. We showed that PDE4D methylation decreased cell proliferation and migration of asthmatic ASMCs. We further elucidated that methylated PDE4D decreased PDE4D expression in asthmatic ASMCs, increased cAMP level, and inhibited the aberrant increase in Ca(2+) level. Moreover, PDE4D methylation reduced the phosphorylation level of downstream effectors of Ca(2+) signaling, including myosin light chain kinase and p38. Taken together, our findings demonstrate that gene-specific epigenetic changes may predispose ASMCs to asthma through alterations in cell phenotypes. Modulation of ASMC phenotypes by methylated PDE4D oligonucleotides can reverse the aberrant ASMC functions to normal phenotypes. This has provided new insight to the development of novel therapeutic options for this debilitative disease.


Assuntos
Asma/enzimologia , Asma/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Metilação de DNA , Epigênese Genética , Miócitos de Músculo Liso/enzimologia , Sistema Respiratório/enzimologia , Remodelação das Vias Aéreas , Asma/patologia , Cálcio/metabolismo , Estudos de Casos e Controles , Movimento Celular , Proliferação de Células , Células Cultivadas , Ilhas de CpG , AMP Cíclico/metabolismo , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Miócitos de Músculo Liso/patologia , Quinase de Cadeia Leve de Miosina/metabolismo , Fenótipo , Fosforilação , Regiões Promotoras Genéticas , Sistema Respiratório/patologia , Transdução de Sinais , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Mucosal Immunol ; 9(1): 112-23, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25993443

RESUMO

Infections in cystic fibrosis (CF), often involving Pseudomonas aeruginosa, result from a dysregulated airway immunity where one hallmark is the accumulation of necrotic and apoptotic immune cells, in particular neutrophils. In addition, neutrophils actively release DNA, forming neutrophil extracellular traps (NETs) that contain antimicrobial proteins. Altogether, free DNA in complex with actin accumulates in the airway lumen, resulting in highly viscous sputum that provides an anionic matrix, binding cationic antimicrobial proteins. In this study, granulocyte chemotactic protein 2 (GCP-2)/CXCL6, a neutrophil-activating chemokine with bactericidal properties, was detected in the airway epithelium of CF patients and was also present in azurophilic and specific granules of neutrophils. Elastase of neutrophils, but not of P. aeruginosa, completely degraded CXCL6 (chemokine (C-X-C motif) ligand 6). In addition, CXCL6 colocalized with extracellular DNA in both CF sputa and in in vitro-formed NETs. In vitro, CXCL6 bound DNA with a KD of 2,500 nM. Interestingly, both the bactericidal and the receptor-activating properties of CXCL6 (against neutrophils) remained largely unaffected in the presence of DNA. However, the chemotactic properties of CXCL6 were reduced by the presence of DNA. Taken together, CXCL6 is expressed in CF, retaining its functional properties even after binding to the anionic scaffold that extracellular DNA provides in CF.


Assuntos
Fibrose Cística/imunologia , DNA/imunologia , Armadilhas Extracelulares/imunologia , Elastase de Leucócito/imunologia , Neutrófilos/imunologia , Sistema Respiratório/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Estudos de Casos e Controles , Fibrose Cística/enzimologia , Fibrose Cística/genética , Fibrose Cística/patologia , DNA/metabolismo , Armadilhas Extracelulares/química , Expressão Gênica , Humanos , Elastase de Leucócito/genética , Elastase de Leucócito/metabolismo , Ativação de Neutrófilo , Infiltração de Neutrófilos , Neutrófilos/enzimologia , Neutrófilos/patologia , Ligação Proteica , Proteólise , Pseudomonas aeruginosa/enzimologia , Sistema Respiratório/enzimologia , Sistema Respiratório/patologia , Especificidade da Espécie , Escarro/química , Escarro/imunologia
13.
J Cyst Fibros ; 14(3): 324-33, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25286826

RESUMO

BACKGROUND: Balanced levels of proteases and anti-proteases are essential in host defense systems. In CF patients' lungs, elevated protease/anti-protease-ratios contribute to damage of airway tissue and premature death with the inherited disease. Little is known about upper airway protease equilibrium in CF. METHODS: Neutrophil elastase (NE), Secretory leukocyte protease inhibitor (SLPI), matrix metalloproteinase (MMP)9, tissue inhibitors of metalloproteinase (TIMP)1, cathepsin S (CTSS) and the corresponding cellular distribution were assessed in the nasal lavage (NL) and sputum of 40 CF patients. RESULTS: Concentrations of all proteases and anti-proteases were markedly higher in sputum than in NL (NE: 10-fold, SLPI: 5000-fold). Interestingly, the NE/SLPI ratio was 726-fold higher in NL compared to sputum, while the MMP9/TIMP1 ratio was 4.5-fold higher in sputum compared to NL. DISCUSSION: This first study to compare protease/anti-protease networks of CF upper and lower airways by NL and sputum reveals substantial differences between both compartments' immunological responses. This finding may have implications for sinonasal and pulmonary treatment, possibly leading to new therapeutic approaches.


Assuntos
Fibrose Cística/metabolismo , Elastase de Leucócito/metabolismo , Líquido da Lavagem Nasal/química , Sistema Respiratório/enzimologia , Inibidor Secretado de Peptidases Leucocitárias/metabolismo , Escarro/enzimologia , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Fibrose Cística/patologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Líquido da Lavagem Nasal/citologia , Sistema Respiratório/patologia , Escarro/citologia , Adulto Jovem
14.
Int J Mol Sci ; 15(9): 16975-97, 2014 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-25250913

RESUMO

Many, if not all, environmental pollutants/chemicals and infectious agents increase intracellular levels of reactive oxygen species (ROS) at the site of exposure. ROS not only function as intracellular signaling entities, but also induce damage to cellular molecules including DNA. Among the several dozen ROS-induced DNA base lesions generated in the genome, 8-oxo-7,8-dihydroguanine (8-oxoG) is one of the most abundant because of guanine's lowest redox potential among DNA bases. In mammalian cells, 8-oxoG is repaired by the 8-oxoguanine DNA glycosylase-1 (OGG1)-initiated DNA base excision repair pathway (OGG1-BER). Accumulation of 8-oxoG in DNA has traditionally been associated with mutagenesis, as well as various human diseases and aging processes, while the free 8-oxoG base in body fluids is one of the best biomarkers of ongoing pathophysiological processes. In this review, we discuss the biological significance of the 8-oxoG base and particularly the role of OGG1-BER in the activation of small GTPases and changes in gene expression, including those that regulate pro-inflammatory chemokines/cytokines and cause inflammation.


Assuntos
DNA Glicosilases/fisiologia , Reparo do DNA/fisiologia , Guanina/análogos & derivados , Inflamação/enzimologia , Animais , Líquidos Corporais/química , Doença Crônica , Citocinas/biossíntese , Citocinas/genética , Dano ao DNA , DNA Glicosilases/deficiência , DNA Glicosilases/genética , Poluentes Ambientais/toxicidade , Ativação Enzimática , Células Epiteliais/enzimologia , Células Epiteliais/patologia , GTP Fosfo-Hidrolases/metabolismo , Regulação da Expressão Gênica , Guanina/metabolismo , Humanos , Inflamação/genética , Inflamação/patologia , Pneumopatias/enzimologia , Pneumopatias/etiologia , Pneumopatias/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Mutagênese , Estresse Oxidativo , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Sistema Respiratório/enzimologia , Sistema Respiratório/patologia
15.
Oxid Med Cell Longev ; 2014: 389629, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24803981

RESUMO

Pseudomonas aeruginosa and Staphylococcus aureus cause chronic lung infection in cystic fibrosis (CF) patients, inducing chronic oxidative stress. Several markers of plasma protein oxidative damage and glycoxidation and activities of erythrocyte antioxidant enzymes have been compared in stable CF patients chronically infected with Pseudomonas aeruginosa (n = 12) and Staphylococcus aureus (n = 10) in relation to healthy subjects (n = 11). Concentration of nitric oxide was also measured in the exhaled air from the lower respiratory tract of patients with CF. Elevated glycophore (4.22 ± 0.91 and 4.19 ± 1.04 versus control 3.18 ± 0.53 fluorescence units (FU)/mg protein; P < 0.05) and carbonyl group levels (1.9 ± 0.64, 1.87 ± 0.45 versus control 0.94 ± 0.19 nmol/mg protein; P < 0.05) as well as increased glutathione S-transferase activity (2.51 ± 0.88 and 2.57 ± 0.79 U/g Hb versus 0.77 ± 0.16 U/g Hb; P < 0.05) were noted in Pseudomonas aeruginosa and Staphylococcus aureus infected CF. Kynurenine level (4.91 ± 1.22 versus 3.89 ± 0.54 FU/mg protein; P < 0.05) was elevated only in Staphylococcus aureus infected CF. These results confirm oxidative stress in CF and demonstrate the usefulness of the glycophore level and protein carbonyl groups as markers of oxidative modifications of plasma proteins in this disease.


Assuntos
Infecções Bacterianas/diagnóstico , Proteínas Sanguíneas/metabolismo , Fibrose Cística/diagnóstico , Pseudomonas aeruginosa/patogenicidade , Staphylococcus aureus/patogenicidade , Adolescente , Infecções Bacterianas/complicações , Infecções Bacterianas/microbiologia , Proteínas Sanguíneas/química , Criança , Fibrose Cística/complicações , Fibrose Cística/metabolismo , Eritrócitos/citologia , Eritrócitos/enzimologia , Feminino , Glutationa Transferase/metabolismo , Humanos , Cinurenina/metabolismo , Masculino , Óxido Nítrico/metabolismo , Estresse Oxidativo , Carbonilação Proteica , Sistema Respiratório/enzimologia , Sistema Respiratório/metabolismo , Sistema Respiratório/microbiologia
16.
J Pharmacol Exp Ther ; 349(1): 85-93, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24513870

RESUMO

Inhaled long-acting ß(2)-adrenoceptor agonists (LABA) that act as bronchodilators and the oral anti-inflammatory phosphodiesterase 4 (PDE4) inhibitor roflumilast are both approved therapies for chronic obstructive pulmonary disease (COPD). Here we describe the activity of a novel, inhaled, bifunctional, small molecule (R)-6-[(3-{[4-(5-{[2-hydroxy-2-(8-hydroxy-2-oxo-1,2-dihydroquinolin-5-yl)ethyl]amino}pent-1-yn-1-yl)phenyl]carbamoyl}phenyl)sulfonyl]-4-[(3-methoxyphenyl)amino]-8-methylquinoline-3-carboxamide (GS-5759), which has specific ß(2) agonist and PDE4 inhibitory activity. GS-5759 demonstrated potent and full agonist activity at ß(2) adrenoceptors (EC(50) = 8 ± 4 nM) and is a potent inhibitor of the PDE4 enzyme (IC(50) = 5 ± 3 nM). In cell assays, GS-5759 inhibited lipopolysaccharide (LPS)-induced tumor necrosis factor α (TNFα) production in human peripheral mononuclear cells (PBMC) with an IC(50) = 0.3 nM [confidence interval (CI) 0.1-0.6] and in human neutrophils formyl-methionyl-leucyl-phenylalanine (fMLP)-induced super oxide anion production with an IC(50) = 3 nM (CI 0.8-8). The addition of the ß(2) antagonist ICI 118551 shifted the IC(50) in these cell assays to 4 and 38 nM, respectively, demonstrating the contribution of both ß(2) agonist and PDE4 inhibitory activity to GS-5759. GS-5759 was also a potent inhibitor of profibrotic and proinflammatory mediator release from human lung fibroblasts. GS-5759 relaxed guinea pig airway smooth muscle strips precontracted with carbachol in a concentration-dependent manner with an EC(50) = 0.5 µM (CI 0.2-2) and had slow dissociation kinetics with an Off T(1/2) > 720 minutes at an EC(80) concentration of 3 µM. GS-5759 is a novel bifunctional molecule with both potent ß(2) agonist and PDE4 inhibitor activity that could provide inhaled bronchodilator and anti-inflammatory therapy for COPD.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Fibroblastos/efeitos dos fármacos , Leucócitos Mononucleares/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Inibidores da Fosfodiesterase 4/farmacologia , Quinolonas/farmacologia , Sistema Respiratório/efeitos dos fármacos , Sulfonas/farmacologia , Agonistas de Receptores Adrenérgicos beta 2/síntese química , Agonistas de Receptores Adrenérgicos beta 2/química , Animais , Técnicas de Cultura de Células , Citocinas/antagonistas & inibidores , Citocinas/imunologia , Fibroblastos/enzimologia , Fibroblastos/imunologia , Fibroblastos/metabolismo , Cobaias , Humanos , Leucócitos Mononucleares/enzimologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Estrutura Molecular , Músculo Liso/enzimologia , Músculo Liso/imunologia , Músculo Liso/metabolismo , Inibidores da Fosfodiesterase 4/síntese química , Inibidores da Fosfodiesterase 4/química , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Quinolonas/síntese química , Quinolonas/química , Sistema Respiratório/enzimologia , Sistema Respiratório/imunologia , Sistema Respiratório/metabolismo , Sulfonas/síntese química , Sulfonas/química , Fatores de Tempo
17.
Mol Genet Metab ; 111(2): 63-72, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24388732

RESUMO

The mucopolysaccharidoses (MPS), a group of rare genetic disorders caused by defects in glycosaminoglycan (GAG) catabolism, are progressive, multi-systemic diseases with a high burden of morbidity. Enzyme replacement therapy (ERT) is available for MPS I, II, and VI, and may improve walking ability, endurance, and pulmonary function as evidenced by data from pivotal trials and extension studies. Despite these demonstrable benefits, cardiac valve disease, joint disease, and skeletal disease, all of which cause significant morbidity, do not generally improve with ERT if pathological changes are already established. Airway disease improves, but usually does not normalize. These limitations can be well understood by considering the varied functions of GAG in the body. Disruption of GAG catabolism has far-reaching effects due to the triggering of secondary pathogenic cascades. It appears that many of the consequences of these secondary pathogenic events, while they may improve on treatment, cannot be fully corrected even with long-term exposure to enzyme, thereby supporting the treatment of patients with MPS before the onset of clinical disease. This review examines the data from clinical trials and other studies in human patients to explore the limits of ERT as currently used, then discusses the pathophysiology, fetal tissue studies, animal studies, and sibling reports to explore the question of how early to treat an MPS patient with a firm diagnosis. The review is followed by an expert opinion on the rationale for and the benefits of early treatment.


Assuntos
Disostoses/tratamento farmacológico , Terapia de Reposição de Enzimas , Iduronato Sulfatase/uso terapêutico , Mucopolissacaridoses/tratamento farmacológico , N-Acetilgalactosamina-4-Sulfatase/uso terapêutico , Prevenção Secundária , Pré-Escolar , Ensaios Clínicos como Assunto , Disostoses/complicações , Disostoses/enzimologia , Disostoses/fisiopatologia , Glicosaminoglicanos/metabolismo , Valvas Cardíacas/efeitos dos fármacos , Valvas Cardíacas/enzimologia , Valvas Cardíacas/fisiopatologia , Humanos , Articulações/efeitos dos fármacos , Articulações/enzimologia , Articulações/fisiopatologia , Mucopolissacaridoses/complicações , Mucopolissacaridoses/enzimologia , Mucopolissacaridoses/fisiopatologia , Proteínas Recombinantes/uso terapêutico , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/enzimologia , Sistema Respiratório/fisiopatologia
18.
J Infect Dis ; 207(10): 1544-55, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23402824

RESUMO

Biofilm microcolonies of Pseudomonas aeruginosa chronically infect the airways of patients with cystic fibrosis and fuel ongoing destructive inflammation, yet the impact of the switch from planktonic to biofilm growth on host responses is poorly understood. We report that in airway epithelial cells a threshold of p38α mitogen-activated protein kinase (MAPK) activation was required to trigger neutrophil recruitment, which is influenced by extrinsic and intrinsic factors. Planktonic P. aeruginosa diffusible material (PsaDM) induced stronger p38α MAPK activation as compared to biofilm PsaDM. Biofilm PsaDM activated p38α MAPK in a Toll-like receptor-independent fashion via the lasI/lasR quorum-sensing system, but this activation was insufficient to recruit neutrophils. However, in airway epithelial cells from patients with cystic fibrosis with hypersensitivity to injurious stimuli, biofilm PsaDM activated p38α MAPK strongly enough to recruit neutrophils, which can contribute to lung injury.


Assuntos
Biofilmes/crescimento & desenvolvimento , Células Epiteliais/imunologia , Imunidade Inata , Proteína Quinase 14 Ativada por Mitógeno/imunologia , Pseudomonas aeruginosa/imunologia , Pseudomonas aeruginosa/patogenicidade , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Biópsia , Células Cultivadas , Fibrose Cística/imunologia , Fibrose Cística/microbiologia , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/microbiologia , Humanos , Immunoblotting , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Mucosa Nasal/citologia , Infiltração de Neutrófilos , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Plâncton/metabolismo , Percepção de Quorum , Sistema Respiratório/citologia , Sistema Respiratório/enzimologia , Sistema Respiratório/imunologia , Sistema Respiratório/microbiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores/genética , Transativadores/metabolismo
19.
Pharmacol Ther ; 137(2): 248-65, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23089371

RESUMO

Asthma and chronic obstructive pulmonary disease (COPD) are obstructive lung diseases characterized by airway obstruction, airway inflammation and airway remodelling. Next to inflammatory cells and airway epithelial cells, airway mesenchymal cells, including airway smooth muscle cells and (myo)fibroblasts, substantially contribute to disease features by the release of inflammatory mediators, smooth muscle contraction, extracellular matrix deposition and structural changes in the airways. Current pharmacological treatment of both diseases intends to target the dynamic features of the endogenous intracellular suppressor cyclic AMP (cAMP). This review will summarize our current knowledge on cAMP and will emphasize on key discoveries and paradigm shifts reflecting the complex spatio-temporal nature of compartmentalized cAMP signalling networks in health and disease. As airway fibroblasts and airway smooth muscle cells are recognized as central players in the development and progression of asthma and COPD, we will focus on the role of cAMP signalling in their function in relation to airway function and plasticity. We will recapture on the recent identification of cAMP-sensing multi-protein complexes maintained by cAMP effectors, including A-kinase anchoring proteins (AKAPs), proteins kinase A (PKA), exchange protein directly activated by cAMP (Epac), cAMP-elevating seven-transmembrane (7TM) receptors and phosphodiesterases (PDEs) and we will report on findings indicating that the pertubation of compartmentalized cAMP signalling correlates with the pathopysiology of obstructive lung diseases. Future challenges include studies on cAMP dynamics and compartmentalization in the lung and the development of novel drugs targeting these systems for therapeutic interventions in chronic obstructive inflammatory diseases.


Assuntos
Asma/tratamento farmacológico , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Sistema Respiratório/efeitos dos fármacos , Asma/enzimologia , Asma/metabolismo , Asma/fisiopatologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Humanos , Relaxamento Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Músculo Liso/enzimologia , Músculo Liso/metabolismo , Doença Pulmonar Obstrutiva Crônica/enzimologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Sistema Respiratório/enzimologia , Sistema Respiratório/metabolismo , Transdução de Sinais/efeitos dos fármacos
20.
Am J Physiol Lung Cell Mol Physiol ; 303(4): L327-33, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22683572

RESUMO

The airway is kept sterile by an efficient innate defense mechanism. The cornerstone of airway defense is mucus containing diverse antimicrobial factors that kill or inactivate pathogens. Most of the mucus in the upper airways is secreted by airway submucosal glands. In patients with cystic fibrosis (CF), airway defense fails and the lungs are colonized by bacteria, usually Pseudomonas aeruginosa. Accumulating evidence suggests that airway submucosal glands contribute to CF pathogenesis by failing to respond appropriately to inhalation of bacteria. However, the regulation of submucosal glands by the innate immune system remains poorly understood. We studied the response of submucosal glands to the proinflammatory cytokines interleukin-1ß and tumor necrosis factor-α. These are released into the airway submucosa in response to infection with the bacterium P. aeruginosa and are elevated in CF airways. Stimulation with IL-1ß and TNF-α increased submucosal gland secretion in a concentration-dependent manner with a maximal secretion rate of 240 ± 20 and 190 ± 40 pl/min, respectively. The half maximal effective concentrations were 11 and 20 ng/ml, respectively. The cytokine effect was dependent on cAMP but was independent of cGMP, nitric oxide, Ca(2+), or p38 MAP kinase. Most importantly, IL-1ß- and TNF-α-stimulated secretion was blocked by the CF transmembrane conductance regulator (CFTR) blocker, CFTRinh172 (100 µmol/l) but was not affected by the Ca(2+)-activated Cl(-) channel blocker, niflumic acid (1 µmol/l). The data suggest, that during bacterial infections and resulting release of proinflammatory cytokines, the glands are stimulated to secrete fluid, and this response is mediated by cAMP-activated CFTR, a process that would fail in patients with CF.


Assuntos
Líquidos Corporais/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Interleucina-1beta/farmacologia , Muco/metabolismo , Sistema Respiratório/anatomia & histologia , Sistema Respiratório/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Líquidos Corporais/efeitos dos fármacos , Cálcio/metabolismo , AMP Cíclico/farmacologia , GMP Cíclico/farmacologia , Feminino , Técnicas In Vitro , Masculino , Muco/efeitos dos fármacos , Ácido Niflúmico/farmacologia , Óxido Nítrico/metabolismo , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/enzimologia , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Suínos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA