Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Mol Med ; 27(1): 11, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33541260

RESUMO

BACKGROUND: House dust mite (HDM) inhalation can cause airway epithelial damage which is implicated in the process of airway inflammation in asthma. High mobility group box 1 (HMGB1) is critically required for cellular damage and apoptosis as an important endogenous danger signal. Recently, Clara cell 16KDa protein (CC16) has been identified to exert anti-inflammatory and immunomodulatory influence in various injury-related diseases model. However, little is known about its ability to protect against airway epithelial injury in allergic asthma. This study was aimed to clarify the protective roles of CC16 on airway epithelia in HDM-induced asthma and the regulation of HMGB1 by CC16. METHODS: Mice were sensitized and challenged by HDM extract and administrated intranasally with CC16 (5 µg/g or 10 µg/g) or saline in the challenged period. The BEAS-2B human airway epithelial cell line were cultured with CC16 or the control vehicle and then exposed to HDM. Knockdown or overexpression of HMGB1 was induced by cell transfection or intratracheal injection of recombinant adenovirus. RESULTS: CC16 treatment decreased airway inflammation and histological damage of airway epithelium dose-dependently in HDM-induced asthma model. Airway epithelia apoptosis upon HDM stimulation was noticeably abrogated by CC16 in vivo and in vitro. In addition, upregulation of HMGB1 expression and its related signaling were also detected under HDM conditions, while silencing HMGB1 significantly inhibited the apoptosis of BEAS-2B cells. Furthermore, the activity of HMGB1-mediated signaling was restrained after CC16 treatment whereas HMGB1 overexpression abolished the protective effect of CC16 on HDM-induced airway epithelia apoptosis. CONCLUSIONS: Our data confirm that CC16 attenuates HDM-mediated airway inflammation and damage via suppressing airway epithelial cell apoptosis in a HMGB1-dependent manner, suggesting the role of CC16 as a potential protective option for HDM-induced asthma.


Assuntos
Asma/tratamento farmacológico , Proteína HMGB1/genética , Pyroglyphidae/imunologia , Uteroglobina/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Asma/induzido quimicamente , Asma/genética , Linhagem Celular , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Uteroglobina/farmacologia
2.
J Toxicol Sci ; 45(10): 651-660, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33012733

RESUMO

Inhalation of silica particles leads to pulmonary inflammatory responses. Clara cell protein 16 (CC16) has been reported to played a protective role in inflammatory lung diseases. However, its role on silica particles-induced inflammation has not been fully clarified. In this study, THP-1 macrophages were exposed to 75 µg/cm2 silica particles with or without 2 µg/mL exogenous CC16 (recombinant CC16, rCC16) for 24 hr. The production of inflammatory cytokines, including interleukin (IL)-1ß, tumor necrosis factor (TNF)-α and IL-6, in the cell supernatants of different groups was detected through ELISA kits and real-time RT-PCR, respectively. The nuclear translocation of nuclear factor (NF)-κB, protein levels of pro-IL-1ß, the nucleotide-binding domain-like receptor protein 3 (NLRP3) and caspase-1 were evaluated via immunofluorescence or western blot. Results showed that, at 75 µg/cm2 silica particle concentration, the treatment of rCC16 significantly decreased IL-1ß, TNF-α and IL-6 protein release and mRNA levels in THP-1 macrophages. Compared to those only exposed to silica particles, THP-1 macrophages exposed to both silica particles and rCC16 showed significantly lower nuclear levels and higher cytosol levels of NF-κB p65, as well as lower co-localization coefficients through immunofluorescence. Additionally, the administration of rCC16 significantly attenuated the increase of pro-IL-1ß, NLRP3 and caspase-1 levels induced by silica particle exposure. Our results suggested that exogenous CC16 could inhibit silica particles-induced inflammation in THP-1 macrophages, mainly through suppressing NF-κB pathway and caspase-1 activation.


Assuntos
Caspase 1/metabolismo , Expressão Gênica/efeitos dos fármacos , Inflamação/genética , Macrófagos Alveolares/imunologia , Macrófagos/imunologia , NF-kappa B/metabolismo , Dióxido de Silício/toxicidade , Caspase 1/genética , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Humanos , NF-kappa B/genética , Tamanho da Partícula , Proteínas Recombinantes/farmacologia , Células THP-1 , Uteroglobina/farmacologia
3.
J Neuroinflammation ; 16(1): 239, 2019 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-31775794

RESUMO

BACKGROUND: Sepsis is a critical disease associated with extremely high mortality. Some severe forms of sepsis can induce brain injury, thus causing behavioral and cognitive dysfunction. Pyroptosis is a type of cell death that differs from apoptosis and plays an important role in the occurrence and development of infectious diseases, nervous system-related diseases. A recent study has found that there is pyroptosis in the hippocampus of sepsis-induced brain injury, but its mechanism and treatment scheme have not been evaluated. METHODS: We established immediately a septic rat model by cecal ligation and perforation (CLP) after administration with recombinant club cell protein (rCC16) and/or U46619 in different groups. The clinical performance, survival percentage, vital signs, and neurobehavioral scores were monitored at different time points. Cortical pathological changes were also examined. The expression of cortical nucleotide-binding domain leucine-rich repeat-containing pyrin domain-containing 3 (NLRP3), caspase-1, (p)-p38 mitogen-activated protein kinase (MAPK), and (p)-extracellular signal-related kinase (ERK) was detected by western blotting and immunofluorescence analysis. The levels of interleukin (IL)-1ß, IL-6, and tumor necrosis factor alpha in the cortical supernatant were detected by enzyme-linked immunosorbent assay. RESULTS: Compared with the sham group, the clinical performance, survival percentage, vital signs, and severe cortical pathological changes in the CLP group were worse; NLRP3, caspase-1, and inflammatory factor levels were increased; and phosphorylation of p38 MAPK and ERK was also increased. Meanwhile, multiple indicators were deteriorated further after administration of U46619 in CLP rats. The clinical performance of CLP rats, however, was better after rCC16 administration; cortical pathological changes were attenuated; and NLRP3, caspase-1, and inflammatory factor levels and the phosphorylation of signaling pathway proteins (p38 MAPK and ERK) were reduced. Interestingly, the CLP rats showed the opposite changes in all indicators after administration with both rCC16 and U46619 when compared with those administered rCC16 alone. CONCLUSIONS: In sepsis, rCC16 inhibits cortical pyroptosis through p38 MAPK and ERK signaling pathways. Meanwhile, rCC16 has a protective effect on newborn rats with sepsis, but it is not clear whether its mechanism is directly related to pyroptosis.


Assuntos
Encéfalo/efeitos dos fármacos , Caspase 1/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose/efeitos dos fármacos , Sepse/metabolismo , Transdução de Sinais/efeitos dos fármacos , Uteroglobina/farmacologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Animais , Animais Recém-Nascidos , Encéfalo/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Ratos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
4.
Eur Respir J ; 54(1)2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31023848

RESUMO

Club cell secretory protein (CCSP) knockout mice exhibit increased airway neutrophilia, as found in chronic obstructive pulmonary disease (COPD). We therefore investigated whether treating COPD airway epithelia with recombinant human CCSP (rhCCSP) could dampen exaggerated airway neutrophilia.Control, smoker and COPD air-liquid interface (ALI) cultures exposed to cigarette smoke extract (CSE) were treated with and without rhCCSP. The chemotactic properties of the supernatants were assessed using Dunn chambers. Neutrophil chemotaxis along recombinant human interleukin 8 (rhIL8) gradients (with and without rhCCSP) was also determined. rhCCSP-rhIL8 interactions were tested through co-immunoprecipitation, Biacore surface plasmon resonance (SPR) and in silico modelling. The relationship between CCSP/IL8 concentration ratios in the supernatant of induced sputum from COPD patients versus neutrophilic airway infiltration assessed in lung biopsies was assessed.Increased neutrophilic chemotactic activity of CSE-treated ALI cultures followed IL8 concentrations and returned to normal when supplemented with rhCCSP. rhIL8-induced chemotaxis of neutrophils was reduced by rhCCSP. rhCCSP and rhIL8 co-immunoprecipitated. SPR confirmed this in vitro interaction (equilibrium dissociation constant=8 µM). In silico modelling indicated that this interaction was highly likely. CCSP/IL8 ratios in induced sputum correlated well with the level of small airway neutrophilic infiltration (r2=0.746, p<0.001).CCSP is a biologically relevant counter-balancer of neutrophil chemotactic activity. These different approaches used in this study suggest that, among the possible mechanisms involved, CCSP may directly neutralise IL8.


Assuntos
Bronquíolos/patologia , Quimiotaxia de Leucócito , Neutrófilos/citologia , Doença Pulmonar Obstrutiva Crônica/patologia , Uteroglobina/farmacologia , Humanos , Interleucina-8/metabolismo , Interleucina-8/farmacologia , Neutrófilos/efeitos dos fármacos , Doença Pulmonar Obstrutiva Crônica/metabolismo , Proteínas Recombinantes/farmacologia , Fumar , Escarro/citologia
5.
Mol Med Rep ; 18(2): 2198-2206, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29956762

RESUMO

Club cell protein (CC16) is expressed primarily by club cells possesses anti­inflammatory properties and is located in the bronchiolar epithelium. Previous studies have demonstrated that CC16 deficiency is associated with the progression of chronic obstructive pulmonary disease (COPD). In the present study, the therapeutic effects of recombinant rat CC16 protein in mice with COPD were examined and the underlying mechanisms investigated. A total of 30 adult male C57/BL6 mice were randomly divided into three groups (10 mice/group). A mouse COPD model was generated by exposing 20 mice to cigarette smoke (CS) for 24 weeks. A total of 10 mice were treated intranasally with rCC16 (2.5 µg/g body weight) and control mice were exposed to normal room air. Results indicated that rCC16 treatment ameliorated pathological damage in the lungs and reduced the production of tumor necrosis factor (TNF)­α, interleukin (IL)­6 and IL­8, which were induced by CS exposure. After rCC16 administration, endogenous CC16 was upregulated and the body weight of COPD mice was increased, whereas the opposite was observed in CS­exposed mice. Additionally, rCC16 treatment inhibited the DNA binding of NF­κB/p65 in lung tissues and reduced nuclear translocation of NF­κB/p65 in BALF and epithelial cells. Moreover, rCC16 treatment lead to a decrease in the total number of BALF cells, including macrophages, which was elevated in COPD mice. In conclusion, the present results demonstrate that rCC16 has therapeutic effects on COPD by downregulating pro­inflammatory factors via the NF­κB pathway.


Assuntos
Fumar Cigarros/metabolismo , Pneumonia/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Mucosa Respiratória/metabolismo , Transdução de Sinais/efeitos dos fármacos , Uteroglobina/farmacologia , Animais , Fumar Cigarros/tratamento farmacológico , Fumar Cigarros/patologia , Citocinas/metabolismo , Modelos Animais de Doenças , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Pneumonia/tratamento farmacológico , Pneumonia/patologia , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/patologia , Proteínas Recombinantes/farmacologia , Mucosa Respiratória/patologia , Fator de Transcrição RelA/metabolismo
6.
Exp Biol Med (Maywood) ; 240(10): 1266-78, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25716019

RESUMO

Clara cell protein (CC16) is a well-known anti-inflammatory protein secreted by the epithelial Clara cells of the airways. It is involved in the development of airway inflammatory diseases such as chronic obstructive pulmonary disease and asthma. Previous studies suggest that CC16 gene transfer suppresses expression of interleukin (IL)-8 in bronchial epithelial cells. However, its role in the function of these cells during inflammation is not well understood. In this study, we evaluated the effect of CC16 on the expression of matrix metalloproteinase (MMP)-9 in lipopolysaccharide (LPS)-stimulated rat tracheal epithelial cells and its underlying molecular mechanisms. We generated recombinant rat CC16 protein (rCC16) which was bioactive in inhibiting the activity of phospholipase A2. rCC16 inhibited LPS-induced MMP-9 expression at both mRNA and protein levels in a concentration-dependent (0-2 µg/mL) manner, as demonstrated by real time RT-PCR, ELISA, and zymography assays. Gene transcription and DNA binding studies demonstrated that rCC16 suppressed LPS-induced NF-κB activation and its binding of gene promoters as identified by luciferase reporter and gel mobility shift assays, respectively. Western blotting and immunofluorescence staining analyses further revealed that rCC16 concentration dependently inhibited the effects of LPS on nuclear increase and cytosol reduction of NF-κB, on the phosphorylation and reduction of NF-κB inhibitory IκBα, and on p38 MAPK-dependent NF-κB activation by phosphorylation at Ser276 of its p65 subunit. These data indicate that inhibition of LPS-mediated NF-κB activation by rCC16 involves both translocation- and phosphorylation-dependent signaling pathways. When the tracheal epithelial cells were pretreated with chlorpromazine, an inhibitor of clathrin-mediated endocytosis, cellular uptake of rCC16 and its inhibition of LPS-induced NF-κB nuclear translocation and also MMP-9 production were significantly abolished. Taken together, our data suggest that clathrin-mediated uptake of rCC16 suppresses LPS-mediated inflammatory MMP-9 production through inactivation of NF-κB and p38 MAPK pathways in tracheal epithelial cells.


Assuntos
Lipopolissacarídeos/farmacologia , Metaloproteinase 9 da Matriz/biossíntese , NF-kappa B/metabolismo , Uteroglobina/farmacologia , Animais , Linhagem Celular , Vesículas Revestidas por Clatrina/metabolismo , Endocitose , Células Epiteliais/metabolismo , Proteínas I-kappa B , Inibidor de NF-kappaB alfa , Fosfolipases A2/metabolismo , Fosforilação , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes , Transdução de Sinais , Traqueia/citologia , Traqueia/metabolismo , Uteroglobina/genética , Uteroglobina/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Chest ; 147(6): 1467-1476, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25474370

RESUMO

BACKGROUND: Club cell secretory protein (CCSP) is a protective biomarker associated with annual decline in lung function. COPD progression results from an imbalance between injury and repair initially triggered by cigarette smoking. OBJECTIVE: We investigated the effect of CCSP as a therapeutic strategy to restore the balance between injury and repair in COPD simultaneously, validating an ex vivo air-liquid interface (ALI) culture of human bronchial epithelial cells. METHODS: Endobronchial biopsy specimens (EBBs) were obtained from 13 patients with COPD, eight smokers, and eight control subjects. Morphometric analysis of the initial EBBs was performed. ALI cultures derived from the same EBBs were exposed to cigarette smoke extract (CSE) with or without exogenous recombinant human CCSP (rhCCSP) supplementation. CCSP and IL-8 concentrations were assessed at steady state and after CSE exposure. RESULTS: Morphometric analysis of the initial EBBs showed increased cell density but decreased immunostaining of CCSP+ cells in EBBs of patients with COPD (P = .03 vs control subjects). At steady state, lower CCSP (P = .04) and higher IL-8 levels (P < .0001) were found in COPD ALI epithelium. Exogenous rhCCSP supplementation dampened CSE-induced IL-8-release in patients with COPD and returned to levels similar to those of smokers and control subjects (P = .0001). A negative correlation was found between IL-8-release in ALI and CCSP+ cell density in initial biopsy specimens (P = .0073). CONCLUSIONS: In vitro, rhCCSP exogenous supplementation can reverse CSE-induced IL-8 release in biopsy specimens from patients with COPD, indicating a potential use of this strategy in vivo.


Assuntos
Pneumonia/etiologia , Pneumonia/prevenção & controle , Doença Pulmonar Obstrutiva Crônica/complicações , Uteroglobina/farmacologia , Adulto , Idoso , Estudos de Casos e Controles , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Humanos , Técnicas In Vitro , Interleucina-8/metabolismo , Masculino , Pessoa de Meia-Idade , Nicotina/farmacologia , Pneumonia/patologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/patologia , Proteínas Recombinantes/farmacologia , Fumar/efeitos adversos
8.
Eur Respir J ; 44(4): 1002-10, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24833761

RESUMO

Bacterial lipopolysaccharide (LPS) and interleukin (IL)-13 increase mucus secretion and inflammatory cytokine production in normal human bronchial epithelial (NHBE) cells. We evaluated the effect of club cell 10-kDa protein (CC10), an anti-inflammatory protein produced by epithelial cells, on mucus secretion, cell morphology and inflammatory cytokine production. NHBE cells were cultured at an air-liquid interface with CC10 or vehicle and exposed to LPS on day 14. Mucin MUC5AC, IL-8 and granulocyte-macrophage colony-stimulating factor were measured in cell supernatants. MUC5AC and IL-8 mRNA expression were measured by real-time PCR. Western blotting was used to evaluate nuclear factor (NF)-κB and extracellular signal-regulated kinase (ERK) activation. Cells were evaluated histologically. Additionally, NHBE cells were exposed to IL-13 and CC10 for 14 days, and secretion of the mucins MUC5AC and MUC5B was measured. MUC5AC secretion stimulated either by LPS or by IL-13 was attenuated by CC10 at 20 ng·mL(-1) (p<0.05). CC10 at 20 ng·mL(-1) also attenuated IL-8 secretion (p<0.05). MUC5AC and IL-8 mRNA expression were also decreased by CC10 (p<0.05). CC10 attenuated phosphorylation of NF-κB (p<0.05) and ERK1/2 (p<0.05). CC10 attenuates LPS-induced mucus secretion in airway cells, in part due to inhibition of NF-κB and ERK phosphorylation.


Assuntos
Citocinas/efeitos dos fármacos , Citocinas/metabolismo , Muco/efeitos dos fármacos , Muco/metabolismo , Mucosa Respiratória/citologia , Uteroglobina/farmacologia , Brônquios/citologia , Células Cultivadas , Células Epiteliais/metabolismo , Humanos , Lipopolissacarídeos/farmacologia
9.
Respir Res ; 14: 101, 2013 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-24098933

RESUMO

BACKGROUND: Antiflammin-1 (AF-1), a derivative of uteroglobin (UG), is a synthetic nonapeptide with diverse biological functions. In the present study, we investigated whether AF-1 has a protective effect against bleomycin-induced pulmonary fibrosis. METHODS: C57BL/6 mice were injected with bleomycin intratracheally to create an animal model of bleomycin-induced pulmonary fibrosis. On Day 7 and Day 28, we examined the anti-inflammatory effect and antifibrotic effect, respectively, of AF-1 on the bleomycin-treated mice. The effects of AF-1 on the transforming growth factor-beta 1 (TGF-ß1)-induced proliferation of murine lung fibroblasts (NIH3T3) were examined by a bromodeoxycytidine (BrdU) incorporation assay and cell cycle analysis. RESULTS: Severe lung inflammation and fibrosis were observed in the bleomycin-treated mice on Day 7 and Day 28, respectively. Administration of AF-1 significantly reduced the number of neutrophils in the bronchoalveolar lavage fluid (BALF) and the levels of tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1ß) in the lung homogenates on Day 7. Histological examination revealed that AF-1 markedly reduced the number of infiltrating cells on Day 7 and attenuated the collagen deposition and destruction of lung architecture on Day 28. The hydroxyproline (HYP) content was significantly decreased in the AF-1-treated mice. In vitro, AF-1 inhibited the TGF-ß1-induced proliferation of NIH3T3 cells, which was mediated by the UG receptor. CONCLUSIONS: AF-1 has anti-inflammatory and antifibrotic actions in bleomycin-induced lung injury. We propose that the antifibrotic effect of AF-1 might be related to its suppression of fibroblast growth in bleomycin-treated lungs and that AF-1 has potential as a new therapeutic tool for pulmonary fibrosis.


Assuntos
Anti-Inflamatórios/uso terapêutico , Bleomicina/efeitos adversos , Fragmentos de Peptídeos/uso terapêutico , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/prevenção & controle , Uteroglobina/uso terapêutico , Animais , Anti-Inflamatórios/farmacologia , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Fibroblastos/efeitos dos fármacos , Hidroxiprolina/metabolismo , Técnicas In Vitro , Interleucina-1beta/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Fragmentos de Peptídeos/farmacologia , Fibrose Pulmonar/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Resultado do Tratamento , Fator de Necrose Tumoral alfa/metabolismo , Uteroglobina/farmacologia
10.
Am J Perinatol ; 25(10): 637-45, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18841530

RESUMO

Infant respiratory distress syndrome (IRDS) can lead to impaired alveolarization and dysmorphic vascularization of bronchopulmonary dysplasia. Clara cell secretory protein (CC10) has anti-inflammatory properties but is deficient in the premature infant. Because surfactant and vascular endothelial growth factor (VEGF) profiles are impaired by inflammation and CC10 inhibits lung inflammation, we hypothesized that CC10 may up-regulate surfactant protein (SP) and VEGF expression. Preterm lambs ( N = 24; 126 +/- 3 days [standard error] gestation) with IRDS were randomized to receive 100 mg/kg surfactant, 100 mg/kg surfactant followed by intratracheal 0.5, 1.5, or 5 mg/kg rhCC10 and studied for 4 hours. Gas exchange and lung mechanics were monitored; surfactant protein and VEGF mRNA profiles in lung were assessed. There was a significant rhCC10 dose-dependent increase in respiratory compliance and ventilation efficiency index; both parameters were significantly greater in animals treated with 5 mg/kg rhCC10 than those treated with surfactant alone. Similarly, there was a significant rhCC10 dose and protein-dependent increase in surfactant protein (SP-B > SP-C > SP-A) and dose- and isoform-dependent increase in VEGF (VEGF189 > VEGF165 > VEGF121). These data demonstrate that early intervention with rhCC10 up-regulates surfactant protein and VEGF expression, supporting the role of CC10 to protect against hyperoxia and mechanical ventilation in the immature lung.


Assuntos
Proteínas Associadas a Surfactantes Pulmonares/metabolismo , RNA Mensageiro/metabolismo , Proteínas Recombinantes/farmacologia , Síndrome do Desconforto Respiratório do Recém-Nascido/tratamento farmacológico , Uteroglobina/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Animais Recém-Nascidos , Produtos Biológicos/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Recém-Nascido , Pulmão/metabolismo , Proteínas Associadas a Surfactantes Pulmonares/farmacologia , Surfactantes Pulmonares/farmacologia , Distribuição Aleatória , Respiração Artificial , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Testes de Função Respiratória , Ovinos , Regulação para Cima
11.
Nephrol Dial Transplant ; 23(11): 3437-45, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18558621

RESUMO

BACKGROUND AND METHODS: Tissue transglutaminase (tTG) may induce pro-inflammatory cytokines and produce irreversible end-products, thus promoting renal scarring. It has recently been confirmed that the crescent formation in murine experimental crescentic glomerulonephritis (ecGN) has been inhibited by the administration of recombinant uteroglobin (rUG). However, the ability of UG on tTG modulation has not been thoroughly assessed. In this study, we investigated the feasible protective role of UG in murine ecGN through the modulation of tTG and TGF-beta1 expressions. ecGN was induced by the administration of anti-GBM Ab into C57BL/6 mice. RESULTS: Both proteinuria and BUN levels were distinctively lower in rUG-treated mice compared to those of disease control mice. Glomerular injuries such as mesangial proliferation, matrix production and crescent formation were lessened with the rUG treatment, and these findings were parallel with the attenuated expression of tTG and TGF-beta1. tTG and TGF-beta1 were expressed mainly on mesangial areas by the induction of ecGN and rUG treatment markedly attenuated the expressions of these proteins in glomeruli without spatial changes. With the addition of LPS to mesangial cells, the expressions of tTG and TGF-beta1 were up-regulated, whilst the addition of cysteamine, tTG inhibitor, attenuated the expression of tTG and TGF-beta1 as well as the cellular proliferation which was further induced by LPS. CONCLUSION: We demonstrate for the first time that rUG is able to attenuate the renal injury through the modulation of expressions of tTG and TGF-beta1 in ecGN and further suggest a wide range of feasible molecular targets to reduce the severity of human glomerulonephritis.


Assuntos
Glomerulonefrite/metabolismo , Células Mesangiais/metabolismo , Transglutaminases/metabolismo , Uteroglobina/farmacologia , Animais , Anticorpos Anti-Idiotípicos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Glomerulonefrite/induzido quimicamente , Glomerulonefrite/patologia , Humanos , Células Mesangiais/efeitos dos fármacos , Células Mesangiais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/farmacologia , Índice de Gravidade de Doença , Fator de Crescimento Transformador beta1/metabolismo
12.
Pediatr Res ; 62(6): 684-8, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17957145

RESUMO

Complications from meconium aspiration syndrome (MAS) remain significant despite a variety of therapeutic interventions. Clara cell protein (CC10) is a novel anti-inflammatory agent that can also inhibit phospholipase A2 (PLA2) (an important component of meconium). The present study examined whether administration of recombinant human CC10 (rhCC10) would reduce inflammation and improve lung function in a piglet model of MAS. Following meconium instillation, piglets exhibited significant physiologic dysfunction that improved significantly after surfactant administration. Analysis of tracheal aspirates revealed significant increases in both tumor necrosis factor (TNF) alpha and interleukin (IL)-8 after meconium instillation. rhCC10-treated animals had significantly lower TNF-alpha levels at 24 h (561 +/- 321 versus 1357 +/- 675 pg/mL, p < 0.05) compared with saline controls. There were no differences between rhCC10-treated and untreated groups with respect to other measured physiologic variables or inflammatory markers, including secretory PLA2 activity. Histologic analyses revealed marked inflammatory infiltrates and thickened alveolar walls, but no significant differences among rhCC10 and control animals. Newborn piglets with MAS have significant physiologic dysfunction, marked inflammatory changes and histologic abnormalities, which was partially counteracted by a single dose of exogenous surfactant and rhCC10.


Assuntos
Anti-Inflamatórios/farmacologia , Pulmão/efeitos dos fármacos , Síndrome de Aspiração de Mecônio/tratamento farmacológico , Uteroglobina/farmacologia , Animais , Animais Recém-Nascidos , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/uso terapêutico , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Modelos Animais de Doenças , Quimioterapia Combinada , Inibidores Enzimáticos/farmacologia , Humanos , Recém-Nascido , Interleucina-8/sangue , Interleucina-8/metabolismo , Pulmão/enzimologia , Pulmão/metabolismo , Pulmão/patologia , Pulmão/fisiopatologia , Mecônio/metabolismo , Síndrome de Aspiração de Mecônio/metabolismo , Síndrome de Aspiração de Mecônio/patologia , Síndrome de Aspiração de Mecônio/fisiopatologia , Fosfolipases A2 Secretórias/antagonistas & inibidores , Fosfolipases A2 Secretórias/metabolismo , Surfactantes Pulmonares/farmacologia , Proteínas Recombinantes/farmacologia , Suínos , Fatores de Tempo , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/metabolismo , Uteroglobina/administração & dosagem , Uteroglobina/uso terapêutico
13.
Endocr Rev ; 28(7): 707-25, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17916741

RESUMO

Blastokinin or uteroglobin (UG) is a steroid-inducible, evolutionarily conserved, secreted protein that has been extensively studied from the standpoint of its structure and molecular biology. However, the physiological function(s) of UG still remains elusive. Isolated from the uterus of rabbits during early pregnancy, UG is the founding member of a growing superfamily of proteins called Secretoglobin (Scgb). Numerous studies demonstrated that UG is a multifunctional protein with antiinflammatory/ immunomodulatory properties. It inhibits soluble phospholipase A(2) activity and binds and perhaps sequesters hydrophobic ligands such as progesterone, retinols, polychlorinated biphenyls, phospholipids, and prostaglandins. In addition to its antiinflammatory activities, UG manifests antichemotactic, antiallergic, antitumorigenic, and embryonic growth-stimulatory activities. The tissue-specific expression of the UG gene is regulated by several steroid hormones, although a nonsteroid hormone, prolactin, further augments its expression in the uterus. The mucosal epithelia of virtually all organs that communicate with the external environment express UG, and it is present in the blood, urine, and other body fluids. Although the physiological functions of this protein are still under investigation, a single nucleotide polymorphism in the UG gene appears to be associated with several inflammatory/autoimmune diseases. Investigations with UG-knockout mice revealed that the absence of this protein leads to phenotypes that suggest its critical homeostatic role(s) against oxidative damage, inflammation, autoimmunity, and cancer. Recent studies on UG-binding proteins (receptors) provide further insight into the multifunctional nature of this protein. Based on its antiinflammatory and antiallergic properties, UG is a potential drug target.


Assuntos
Fatores Imunológicos/fisiologia , Uteroglobina/fisiologia , Sequência de Aminoácidos , Animais , Regulação da Expressão Gênica , Hormônios Esteroides Gonadais/metabolismo , Humanos , Fatores Imunológicos/genética , Fatores Imunológicos/farmacologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/farmacologia , Polimorfismo Genético , Conformação Proteica , Coelhos , Proteínas Recombinantes/uso terapêutico , Uteroglobina/genética , Uteroglobina/farmacologia
14.
J Cell Physiol ; 207(2): 553-61, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16453303

RESUMO

Uteroglobin (UG) or Clara Cell 10 kDa protein (CC10) is a small, stable, epithelial secretory anti-inflammatory protein. Uteroglobin has been shown to inhibit neointimal formation in vivo after balloon angioplasty through an unknown mechanism. An interaction between UG and plasma fibronectin (Fn) has been demonstrated in mice. Since Fn plays a key role in endothelial cell (EC) migration and angiogenesis, we investigated whether recombinant human UG (rhUG) affects EC migration via Fn binding. In this report, we show a saturable binding of rhUG to Fn depending on Fn conformation and that rhUG is covalently cross-linked to Fn by transglutaminase (TGase). Additionally, our study highlights that rhUG can also bind to exogenously added or self-secreted Fn on the membrane of human primary microvascular endothelial cells (HMVEC), although these complexes are weakly associated with the plasmalemma. Upon the interaction with Fn in solid phase, rhUG strongly inhibits HMVEC attachment on Fn, but not on other ECM proteins. Consequently, rhUG also inhibits cell migration in a dose dependent fashion (I.C.50 = 65 nM) and hinders the "wound healing" in vitro. The small size, stability and human tolerability of rhUG suggest that rhUG in slow-release form or genetically delivered could be used in humans to modulate cell/Fn interactions in the context of tumor microenvironment or in the context of inflammation and fibrosis.


Assuntos
Movimento Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Fibronectinas/metabolismo , Uteroglobina/farmacologia , Ligação Competitiva , Adesão Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Heparina/química , Heparina/farmacologia , Humanos , Masculino , Ligação Proteica , Conformação Proteica/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Transglutaminases/metabolismo , Uteroglobina/genética , Uteroglobina/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia
15.
J Natl Cancer Inst ; 96(18): 1388-96, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15367572

RESUMO

BACKGROUND: A novel breast cancer-associated antigen, mammaglobin-A, is expressed in 80% of primary breast tumors. The characterization of immune responses against this highly expressed breast cancer-specific antigen would be of value in the development of new therapeutic strategies for breast cancer. METHODS: We developed an in vivo model using human leukocyte antigen-A*0201/human CD8+ (HLA-A2+/hCD8+) double-transgenic mice to define the epitopes and to study the level of protection acquired by mammaglobin-A cDNA vaccination toward mammaglobin-A+/HLA-A2+ breast cancer cell lines. Mammaglobin-A epitopes were identified using an HLA class I peptide binding prediction computer program, and their activity was verified using gamma interferon ELISPOT and cytotoxicity assays. RESULTS: We identified seven mammaglobin-A-derived candidate epitopes that bind the HLA-A*0201 molecule (Mam-A2.1-7). CD8+ cytotoxic T lymphocytes (CTLs) from HLA-A2+/hCD8+ mice reacted to the Mam-A2.1 (amino acids [aa] 83-92, LIYDSSLCDL), Mam-A2.2 (aa 2-10, KLLMVLMLA), Mam-A2.4 (aa 66-74, FLNQTDETL), and Mam-A2.6 (aa 32-40, MQLIYDSSL) epitopes. CD8+ CTLs from breast cancer patients also recognized a similar epitope pattern as did those in the HLA-A2+/hCD8 mice and reacted to the Mam-A2.1, Mam-A2.2, Mam-A2.3, Mam-A2.4, and Mam-A2.7 epitopes. Passive transfer of mammaglobin-A-reactive CTLs into SCID (severe combined immunodeficient) beige mice with actively growing mammaglobin-A+ tumors resulted in statistically significant regression (P<.001) in the growth of the tumors. CONCLUSIONS: The HLA-A2+/hCD8+ mouse represents a valuable animal model to characterize the HLA-A*0201-restricted CD8+ CTL immune response to mammaglobin-A in vivo, and the data reported here demonstrate the immunotherapeutic potential of mammaglobin-A for the treatment and/or prevention of breast cancer.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/imunologia , Vacinas Anticâncer/farmacologia , Imunoterapia/métodos , Proteínas de Neoplasias/farmacologia , Uteroglobina/farmacologia , Animais , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/uso terapêutico , DNA Complementar/uso terapêutico , Modelos Animais de Doenças , Epitopos , Feminino , Humanos , Mamoglobina A , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/uso terapêutico , Linfócitos T Citotóxicos/imunologia , Uteroglobina/genética , Uteroglobina/uso terapêutico
16.
Kidney Int ; 66(3): 1061-7, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15327399

RESUMO

BACKGROUND: Although uteroglobin is known to have an immunomodulatory property and prevents the deposition of immune-complexes on the glomeruli of mice, the therapeutic potential of uteroglobin is uncertain in glomerulonephritis. To test the hypothesis that uteroglobin can prevent glomerulonephritis, we have studied the effects of recombinant uteroglobin on the development of experimental crescentic glomerulonephritis that is induced by anti-glomerular basement membrane (anti-GBM) antibodies. METHODS AND RESULTS: Glomerulonephritis was induced by the intravenous injection of rabbit anti-GBM globulin antibodies into mice (C57BL/6), and renal injury was evaluated 7, 14, and 21 days afterward. Recombinant uteroglobin or phosphate-buffered saline (PBS) were given intravenously to mice for 3 days after anti-GBM antibody injection. Proteinuria was significantly reduced in mice treated with recombinant uteroglobin compared with disease-control mice at 7 and 14 days after an anti-GBM antibody injection, although the serum creatinine concentration was similar in both groups. The amount of proteinuria was similar in recombinant uteroglobin-treated and normal control mice. By histologic analysis, mesangial matrix expansion, mesangial proliferation, and cellular crescents representing crescentic glomerulonephritis were markedly attenuated by injection of recombinant uteroglobin. The in vitro proliferative responses of mesangial cells to lipopolysaccharide (LPS) were blunted by the addition of recombinant uteroglobin in a dose-dependent manner. The preventive effects exerted by recombinant uteroglobin treatment were based on the inhibition of antibodies and complement-3 deposition on the glomeruli. CONCLUSION: This study demonstrates the preventive effects of recombinant uteroglobin in an experimental model of crescentic glomerulonephritis, and suggests the therapeutic implications of uteroglobin for human chronic glomerulonephritis.


Assuntos
Glomerulonefrite/tratamento farmacológico , Glomerulonefrite/prevenção & controle , Uteroglobina/farmacologia , Animais , Anticorpos/farmacologia , Autoanticorpos , Linhagem Celular Tumoral , Feminino , Mesângio Glomerular/efeitos dos fármacos , Mesângio Glomerular/patologia , Glomerulonefrite/patologia , Humanos , Lipopolissacarídeos/farmacologia , Neoplasias Pulmonares , Camundongos , Camundongos Endogâmicos C57BL , Proteinúria/tratamento farmacológico , Proteinúria/patologia , Proteinúria/prevenção & controle , Coelhos , Proteínas Recombinantes/farmacologia
17.
Exp Lung Res ; 29(7): 455-73, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14710438

RESUMO

Clara cell 10-kDa protein (CC10) is a major component of bronchoalveolar lavage fluid and is suggested to be a natural regulator of airway inflammation, possibly through its effects on the proinflammatory enzyme(s), phospholipase A2. We examined the effect of recombinant human (rh) CC10 on endotoxin-induced airway contraction and cytokine release in isolated perfused rat lungs. We found that rhCC10 added to the lung perfusate abolished the endotoxin-induced airway contraction, and that it inhibited both the release of interleukin-1 beta and interleukin-6 into the lung perfusate and the release of tumor necrosis factor-alpha into the pulmonary lavage fluid. By contrast, the levels of interferon-gamma were unaffected by CC10 administration. Rutin, a phospholipase A2 inhibitor, and N omega-nitro-L-arginine methyl ester (L-NAME), a nitric oxide synthase inhibitor, also attenuated the contraction induced by endotoxin. These findings demonstrate that rhCC10 inhibits endotoxin-induced airway contraction and the release of proinflammatory cytokines (interleukin-1 beta, interleukin-6, and tumor necrosis factor-alpha) in isolated perfused rat lungs. The results also indicate that phospholipase A2 and nitric oxide are involved in the airway contraction in this model, possibly through their influence on the production of eicosanoids.


Assuntos
Resistência das Vias Respiratórias/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Pulmão/efeitos dos fármacos , Uteroglobina/farmacologia , Acetilcolina/farmacologia , Animais , Líquido da Lavagem Broncoalveolar/química , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Inibidores Enzimáticos/farmacologia , Pulmão/metabolismo , Pulmão/fisiopatologia , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Perfusão , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes , Rutina/farmacologia
18.
Toxicon ; 40(1): 9-21, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11602274

RESUMO

The mode of action of venom from the ectoparasitic wasp Nasonia vitripennis in eliciting cell death was examined using an in vitro approach with BTI-TN-5B1-4 cells, and the cell responses were compared to those evoked by the extensively studied wasp toxin mastoparan. Wasp venom increased plasma membrane permeability to Na+, resulting in cellular swelling and death due to oncosis. When ouabain was used to disable Na+, K+-ATPases, the effects of venom were enhanced. Measurements of intracellular calcium using fluo-4 AM revealed a rearrangement and an increase in cytosolic [Ca+2]i within 30 min after exposure of BTI-TN-5B1-4 cells to venom. This venom-mediated increase in Ca+2 was apparently due to mobilization of intracellular stores since the changes occurred in the absence of extracellular Ca+2. Phospholipase C (PLC) inhibitors, neomycin and U-73122, blocked the venom-induced death temporarily (<3h), but by 24h, all venom-treated cells swelled and lysed. Pre-treatment of cells with caffeine or theophylline but not ryanodine attenuated the induction of oncosis by wasp venom. Anti-inflammatory peptide 1 (antiflammin 1) but not bromophenacyl bromide, agents that block phospholipase A2 (PLA2) activity, abolished the responsiveness of BTI-TN-5B1-4 cells to venom. These results suggest that venom initiates cell death by inducing Ca+2 release from intracellular stores probably via phospholipase C and IP3. A possible mode of action for venom from N. vitripennis requiring dual activation of PLC and PLA2 is discussed and compared to the pathways known to be activated by mastoparan.


Assuntos
Fosfolipases A/metabolismo , Sódio/metabolismo , Fosfolipases Tipo C/metabolismo , Venenos de Vespas/farmacologia , Vespas , Animais , Cafeína/farmacologia , Cálcio/metabolismo , Morte Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Células Cultivadas , Citosol/efeitos dos fármacos , Citosol/metabolismo , Inibidores Enzimáticos/farmacologia , Estrenos/farmacologia , Feminino , Peptídeos e Proteínas de Sinalização Intercelular , Neomicina/farmacologia , Fragmentos de Peptídeos/farmacologia , Peptídeos , Fosfolipases A/antagonistas & inibidores , Fosfolipases A2 , Pirrolidinonas/farmacologia , Transdução de Sinais , Teofilina/farmacologia , Fosfolipases Tipo C/antagonistas & inibidores , Uteroglobina/farmacologia
19.
Ann N Y Acad Sci ; 923: 128-40, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11193751

RESUMO

Uteroglobin/Clara cell 10-kDa protein (UG/CC10) is a hormonally regulated small secretory protein that has a variety of in vitro and in vivo pharmacological effects. These include a potent anti-inflammatory activity and inhibitory effects on neutrophil migration, thrombin-induced platelet aggregation, in vitro chemoinvasion, as well as "tumor suppressor"-like effects and other properties. Several mechanisms of action have been proposed for these effects. Pharmacological properties suggest that UG itself or substances derived from it may be used as experimental drugs for several indications. The group of oligopeptides collectively known as "antiflammins" (AFs) were originally described in 1988. Their design was derived from the region of highest sequence similarity between UG and another group of proteins with anti-inflammatory properties, the lipocortins or annexins. Nanomolar concentrations of these peptides can reproduce several of the pharmacological activities of UG, including its in vivo anti-inflammatory effects and inhibition of platelet aggregation. The AFs have been safely and effectively used to suppress inflammation and fibrosis in several animal models. Progress in clarifying the mechanism of action of the AFs may facilitate the structure-based design of a novel class of potent anti-inflammatory, antichemotactic drugs.


Assuntos
Inflamação/tratamento farmacológico , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/farmacologia , Uteroglobina/análogos & derivados , Uteroglobina/farmacologia , Animais , Modelos Animais de Doenças , Humanos , Inflamação/metabolismo , Inflamação/fisiopatologia , Oligopeptídeos/metabolismo , Fragmentos de Peptídeos/metabolismo , Estrutura Secundária de Proteína/fisiologia , Uteroglobina/metabolismo
20.
Ann N Y Acad Sci ; 923: 234-48, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11193760

RESUMO

Uteroglobin (UG) is a multifunctional, secreted protein with anti-inflammatory and antichemotactic properties. While its anti-inflammatory effects, in part, stem from the inhibition of soluble phospholipase A2 (sPLA2) activity, the mechanism(s) of its antichemotactic effects is not clearly understood. Although specific binding of UG on microsomal and plasma membranes has been reported recently, how this binding affects cellular function is not clear. Here, we report that recombinant human UG (hUG) binds to both normal and cancer cells with high affinity (20-35 nM, respectively) and specificity. Affinity cross-linking studies revealed that 125I-hUG binds to the NIH 3T3 cell surface with two proteins of apparent molecular masses of 190 and 49 kDa, respectively. UG affinity chromatography yielded similar results. While both the 190- and 49-kDa proteins were expressed in the heart, liver, and spleen, the lung and trachea expressed only the 190-kDa protein. Some cancer cells (e.g., mastocytoma, sarcoma, and lymphoma) expressed both the 190- and 49-kDa proteins. Further, using functional assays, we found that UG dramatically suppressed the motility and extracellular matrix invasion of both NIH 3T3 and some cancer cells. In order to further characterize the anti-ECM-invasive properties of UG, we induced expression of hUG into cancer cell lines derived from organs that, under physiological circumstances, secrete UG at a high level. Interestingly, it has been reported that a high percentage of the adenocarcinomas arising from the same organs fail to express UG. Our results on induced hUG expression in these cells show that inhibition of motility and ECM invasion requires the expression of both UG and its binding proteins. Taken together, our data define receptor-mediated functions of UG in which this protein regulates vital cellular functions by both autocrine and paracrine pathways.


Assuntos
Proteínas de Transporte/metabolismo , Movimento Celular/genética , Invasividade Neoplásica/genética , Ligação Proteica/genética , Receptores de Superfície Celular/metabolismo , Células Tumorais Cultivadas/metabolismo , Uteroglobina/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/farmacologia , Movimento Celular/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Humanos , Camundongos , Invasividade Neoplásica/patologia , Ligação Proteica/efeitos dos fármacos , Ensaio Radioligante/estatística & dados numéricos , Receptores de Superfície Celular/efeitos dos fármacos , Receptores de Superfície Celular/isolamento & purificação , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/patologia , Uteroglobina/genética , Uteroglobina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA