Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Allergy Clin Immunol Pract ; 11(1): 149-157.e1, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36154896

RESUMO

Flexible rhinolaryngoscopy is an underused procedure that can provide allergists-immunologists and other physicians with several benefits over existing imaging techniques. In this article, we highlight the many benefits of flexible rhinolaryngoscopy and expand on its safety, cost-effectiveness, and convenience. This article also covers current procedure techniques and assesses the most common indications and relevant clinical findings for which flexible rhinolaryngoscopy can be used to evaluate the nasopharyngeal tract. Videos for the clinician showing some of the most common findings are included.


Assuntos
Alergia e Imunologia , Hipersensibilidade , Médicos , Humanos , Nasofaringe , Alergistas , Tecnologia de Fibra Óptica , Hipersensibilidade/diagnóstico
2.
Biomolecules ; 12(2)2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-35204783

RESUMO

Abnormalities in airway epithelia and lung parenchyma are found in Atp8b1 mutant mice, which develop pulmonary fibrosis after hyperoxic insult. Microarray and ingenuity pathway analysis (IPA) show numerous transcripts involved in ciliogenesis are downregulated in 14-month (14 M) -old Atp8b1 mouse lung compared with wild-type C57BL/6. Lung epithelium of Atp8b1 mice demonstrate apical abnormalities of ciliated and club cells in the bronchial epithelium on transmission electron microscopy (TEM). Matrix metalloproteinase 7 (MMP7) regulates of ciliogenesis and is a biomarker for idiopathic pulmonary fibrosis (IPF) in humans. Mmp7 transcript and protein expression are significantly upregulated in 14 M Atp8b1 mutant mouse lung. MMP7 expression is also increased in bronchoalveolar lavage fluid (BAL). Immunohistochemistry is localized MMP7 to bronchial epithelial cells in the Atp8b1 mutant. In conclusion, MMP7 is upregulated in the aged Atp8b1 mouse model, which displays abnormal ciliated cell and club cell morphology. This mouse model can facilitate the exploration of the role of MMP7 in epithelial integrity and ciliogenesis in IPF. The Atp8b1 mutant mouse is proposed as a model for IPF.


Assuntos
Adenosina Trifosfatases , Fibrose Pulmonar Idiopática , Metaloproteinase 7 da Matriz , Proteínas de Transferência de Fosfolipídeos , Adenosina Trifosfatases/metabolismo , Animais , Líquido da Lavagem Broncoalveolar , Fibrose Pulmonar Idiopática/enzimologia , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/metabolismo , Metaloproteinase 7 da Matriz/genética , Metaloproteinase 7 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Transferência de Fosfolipídeos/metabolismo
3.
FASEB J ; 36(2): e22143, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34985777

RESUMO

Adenosine deaminase acting on RNA 2 (ADAR2), an RNA editing enzyme is involved in a site-selective modification of adenosine (A) to inosine (I) in double-stranded RNA (dsRNA). Its role in the lungs is unknown. The phenotypic characterization of Adarb1 mice that lacked ADAR2 auto-regulation due to the deletion of editing complementary sequence (ΔECS mice) determined the functional role of ADAR2 in the lungs. ADAR2 protein expression increased in the ΔECS mice. These mice display immune cell infiltration and alveolar disorganization. The lung wet by dry ratio indicates there is no lung edema in ΔECS mice. Bronchoalveolar lavage (BAL) analysis of ΔECS mice reveals a significant increase in neutrophils. Interestingly, ΔECS mice spontaneously develop lung fibrosis as indicated by Sirius red staining of collagen fibers in the lung sections and a significant increase in hydroxyproline level in their lungs. ADAR2 expression increased significantly in a bleomycin mouse model, implicating a role of ADAR2 in lung fibrosis. Furthermore, there is a likely possibility that the genetically modified ΔECS mice does not model the physiological or pathophysiological process of lung fibrosis. Nevertheless, this model is useful in interrogating the role of ADAR2 in the lungs. The Ctgf mRNA and connective tissue growth factor (CTGF) protein significantly increased in ΔECS lungs and occurs in bronchial epithelial cells. There is a significant increase in Human antigen R (ELAVL1; HuR) protein levels in ΔECS lungs and suggests a role in stabilizing Ctgf mRNA. Lung mechanics such as total respiratory resistance, Newtonian resistance and tissue damping were increased, whereas inspiratory capacity was decreased in the ΔECS mice. Taken together, these data indicate that overexpression of ADAR2 causes spontaneous lung fibrosis via HuR-mediated CTGF signaling and implicate a role for ADAR2 auto-regulation in lung homeostasis. The identification of ADAR2 target genes in ΔECS mice would facilitate a mechanistic understanding of the role of ADAR2 in the lungs and provide a therapeutic strategy for lung fibrosis.


Assuntos
Adenosina Desaminase/metabolismo , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Pulmão/metabolismo , Fibrose Pulmonar/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais/fisiologia , Animais , Bleomicina/farmacologia , Modelos Animais de Doenças , Feminino , Humanos , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/tratamento farmacológico , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos
4.
Cell Death Discov ; 7(1): 117, 2021 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-34011971

RESUMO

Steroid receptor RNA activator 1 (SRA1) has been described as a novel transcriptional co-activator that affects the migration of cancer cells. Through RT-PCR, we identified that skipping exon 3 of SRA1 produces two isoforms, including the truncated short isoform, SRA1-S, and the long isoform, SRA1-L. However, the effect of these two isomers on the migration of HCC cells, as well as the specific mechanism of exon 3 skipping remain unclear. In this study, we found up regulated expression of SRSF1 and SRA1-L in highly metastatic HCCLM3, as well as in HCCs with SRSF1 demonstrating the strongest correlation with SRA1-L. In contrast, we observed a constitutively low expression of SRA1-S and SRSF1 in lowly metastatic HepG2 cells. Overexpression of SRSF1 or SRA1-L promoted migration and invasion by increasing the expression of CD44, while SRA1-S reversed the effect of SRSF1 and SRA1-L in vitro. In addition, lung metastasis in mice revealed that, knockdown of SRSF1 or SRA1-L inhibited the migration of HCC cells, while SRA1-L overexpression abolished the effect of SRSF1 knockout and instead promoted HCC cells migration in vivo. More importantly, RNA immunoprecipitation and Cross-link immunoprecipitation analyses showed that SRSF1 interacts with exon 3 of SRA1 to up regulate the expression of SRA1-L in HCC cells. RNA pull-down results indicated that SRSF1 could also bind to exon 3 of SRA1 in vitro. Finally, minigene -MS2 mutation experiments showed that mutation of the SRA1 exon 3 binding site for SRSF1 prevented the binding of SRA1 pre-mRNA. In summary, our results provide experimental evidence that SRA1 exon 3 inclusion is up regulated by SRSF1 to promote tumor invasion and metastasis in hepatocellular carcinoma.

5.
Theranostics ; 10(13): 5719-5735, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32483414

RESUMO

The Axl gene is known to encode for a receptor tyrosine kinase involved in the metastasis process of cancer. In this study, we investigated the underlying molecular mechanism of Axl alternative splicing. Methods: The expression levels of PTBP1 in hepatocellular carcinoma (HCC) tissues were obtained from TCGA samples and cell lines. The effect of Axl-L, Axl-S, and PTBP1 on cell growth, migration, invasion tumor formation, and metastasis of liver cancer cells were measured by cell proliferation, wound-healing, invasion, xenograft tumor formation, and metastasis. Interaction between PTBP1 and Axl was explored using cross-link immunoprecipitation, RNA pull-down assays and RNA immunoprecipitation assays. Results: Knockdown of the PTBP1 and exon 10 skipping isoform of Axl (Axl-S), led to impaired invasion and metastasis in hepatoma cells. Immunoprecipitation results indicated that Axl-S protein binds more robustly with Gas6 ligand than Axl-L (exon 10 including) and is more capable of promoting phosphorylation of ERK and AKT proteins. Furthermore, cross-link immunoprecipitation and RNA-pulldown assays revealed that PTBP1 binds to the polypyrimidine sequence(TCCTCTCTGTCCTTTCTTC) on Axl-Intron 9. MS2-GFP-IP experiments demonstrated that PTBP1 competes with U2AF2 for binding to the aforementioned polypyrimidine sequence, thereby inhibiting alternative splicing and ultimately promoting Axl-S production. Conclusion: Our results highlight the biological significance of Axl-S and PTBP1 in tumor metastasis, and show that PTBP1 affects the invasion and metastasis of hepatoma cells by modulating the alternative splicing of Axl exon 10.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Neoplasias Hepáticas/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Processamento Alternativo/genética , Animais , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Éxons/genética , Regulação Neoplásica da Expressão Gênica/genética , Ribonucleoproteínas Nucleares Heterogêneas/genética , Humanos , Fígado/patologia , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Proteínas Proto-Oncogênicas/metabolismo , Precursores de RNA/genética , Splicing de RNA/genética , RNA Mensageiro/genética , Receptores Proteína Tirosina Quinases/metabolismo , Fator de Processamento U2AF/genética , Fator de Processamento U2AF/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Receptor Tirosina Quinase Axl
6.
Eur J Pharmacol ; 852: 77-89, 2019 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-30831079

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a disease with a poor prognosis and high mortality, posing a major threat to human health. Increased levels of inflammatory cytokines, reactive oxygen species and coagulation cascade have been extensively reported in IPF. We previously fused Hirudin and human manganese superoxide dismutase (hSOD2) to generate a dual-feature fusion protein, denoted as rhSOD2-Hirudin fusion protein. In this study, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) and Hydroxyproline (HYP) assays were used to investigate the effects of rhSOD2-Hirudin protein on thrombin-induced fibroblast proliferation and collagen accumulation in vitro. Subsequently, the mice model of pulmonary fibrosis induced by bleomycin was used for evaluating the anti-inflammatory and anti-fibrotic effects of rhSOD2-Hirudin protein in vivo. Results showed that rhSOD2-Hirudin protein could inhibit the proliferation of fibroblasts and reduce the HYP production in vitro by inhibiting the activity of thrombin. In vivo experiments showed that lung inflammation and fibrosis were significantly decreased in rhSOD2-Hirudin protein-treated mice. Furthermore, rhSOD2-Hirudin protein treatment reduced profibrotic protein and gene expression while reducing the number of inflammatory cells in the lung. In conclusion, rhSOD2-Hirudin protein can effectively attenuate pulmonary fibrosis in vitro and in vivo, mainly by inhibiting the activity of thrombin meanwhile increasing SOD2 levels prevent cells from being damaged by reactive oxygen species, thereby mitigating IPF progression. This study provided important information on the feasibility and efficacy of rhSOD2-Hirudin protein as a novel therapeutic agent for IPF.


Assuntos
Bleomicina/farmacologia , Hirudinas/genética , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacologia , Superóxido Dismutase/farmacologia , Actinas/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hidroxiprolina/biossíntese , Camundongos , Células NIH 3T3 , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Proteínas Recombinantes de Fusão/uso terapêutico , Fator de Crescimento Transformador beta1/metabolismo
7.
Aging (Albany NY) ; 11(1): 209-229, 2019 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-30636723

RESUMO

Atp8b1 (ATPase, aminophospholipid transporter, class I, type 8B, member 1) is a cardiolipin transporter in the apical membrane of lung epithelial cells. While the role of Atp8b1 in pneumonia-induced acute lung injury (ALI) has been well studied, its potential role in oxidative stress-induced ALI is poorly understood. We herein show that Atp8b1G308V/G308V mice under hyperoxic conditions display exacerbated cell apoptosis at alveolar epithelium and aberrant proliferation of club cells at bronchiolar epithelium. This hyperoxia-induced ambivalent response in Atp8b1G308V/G308V lungs was followed by patchy distribution of non-uniform interstitial fibrosis at late recovery phase under normoxia. Since this club cell abnormality is commonly observed between Atp8b1G308V/G308V lungs under hyperoxic conditions and IPF lungs, we characterized this mouse fibrosis model focusing on club cells. Intriguingly, subcellular morphological analysis of IPF lungs, using transmission electron microscopy (TEM), revealed that metaplastic bronchiolar epithelial cells in fibrotic lesions and deformed type II alveolar epithelial cells (AECs) in alveoli with mild fibrosis, have common morphological features including cytoplasmic vacuolation and dysmorphic lamellar bodies. In conclusion, the combination of Atp8b1 mutation and hyperoxic insult serves as a novel platform to study unfocused role of club cells in IPF.


Assuntos
Adenosina Trifosfatases/metabolismo , Oxigênio/toxicidade , Proteínas de Transferência de Fosfolipídeos/metabolismo , Fibrose Pulmonar/etiologia , Adenosina Trifosfatases/genética , Animais , Morte Celular , Proliferação de Células , Células Epiteliais/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Mutação , Estresse Oxidativo , Proteínas de Transferência de Fosfolipídeos/genética , Alvéolos Pulmonares/citologia , Uteroglobina/genética , Uteroglobina/metabolismo
8.
Biotechnol Appl Biochem ; 66(1): 21-32, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30192404

RESUMO

Lung cancer is a serious threat to human health. Studies have revealed that human manganese superoxide dismutase (hSOD2) and miRNAs play an essential role in the metastasis process of lung cancer. However, the miRNAs that associated with hSOD2 and involved in metastasis, remain elusive. After databases analysis and dual luciferase reporter validation, we demonstrated that miR-330-3p expression inversely correlated with hSOD2b expression level, and that miR-330-3p directly targeted the 3'untranslated region (3'UTR) of hSOD2b. Furthermore, overexpression of miR-330-3p promoted whereas knockdown of miR-330-3p inhibited invasion/migration and the epithelial-mesenchymal transition (EMT) process of lung cancer cells in vitro. Knockdown of miR-330-3p inhibited metastasis of lung cancer cells in vivo. Moreover, miR-330-3p-mediated enhancement of invasion/migration in 95-D cells could be rescued by over-expression of hSOD2. In conclusion, we demonstrated that miR-330-3p promoted metastasis of lung cancer cells by suppressing hSOD2b expression and unveiled a new clinical application of miR-330-3p in the therapy of lung cancer.


Assuntos
Movimento Celular , Neoplasias Pulmonares/metabolismo , MicroRNAs/metabolismo , Proteínas de Neoplasias/metabolismo , RNA Neoplásico/metabolismo , Superóxido Dismutase/metabolismo , Células A549 , Transição Epitelial-Mesenquimal , Células HeLa , Células Hep G2 , Humanos , Células K562 , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Células MCF-7 , MicroRNAs/genética , Invasividade Neoplásica , Metástase Neoplásica , Proteínas de Neoplasias/genética , RNA Neoplásico/genética , Superóxido Dismutase/genética
9.
Aging (Albany NY) ; 8(11): 3091-3109, 2016 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-27899769

RESUMO

Idiopathic pulmonary fibrosis (IPF) is an age-related multifactorial disease featuring non-uniform lung fibrosis. The decisive cellular events at early stages of IPF are poorly understood. While the involvement of club cells in IPF pathogenesis is unclear, their migration has been associated with lung fibrosis. In this study, we labeled club cells immunohistochemically in IPF lungs using a club cell marker Claudin-10 (Cldn10), a unique protein based on the recent report which demonstrated that the appearance of Cldn10 in developing and repairing lungs precedes other club cell markers including club cell secretory protein (CCSP). Cldn10-positive cells in IPF lungs displayed marked pleomorphism and were found in varied arrangements, suggesting their phenoconversion. These results were corroborated by immunogold labeling for Cldn10. Further, immunohistochemical double-labeling for Cldn10 and α-smooth muscle actin (α-SMA) demonstrated that aberrant α-SMA signals are frequently encountered near disorganized Cldn10-positive cells in hyperplastic bronchiolar epithelium and thickened interstitium of IPF lungs. Collectively, these data indicate that club cells actively participate in the initiation and progression of IPF through phenoconversion involving the acquisition of proliferative and migratory abilities. Thus, our new findings open the possibility for club cell-targeted therapy to become a strategic option for the treatment of IPF.


Assuntos
Actinas/metabolismo , Células Epiteliais Alveolares/metabolismo , Movimento Celular , Claudinas/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , Células Epiteliais Alveolares/citologia , Humanos , Fibrose Pulmonar Idiopática/patologia , Pulmão/metabolismo , Pulmão/patologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/patologia
10.
Aging (Albany NY) ; 8(9): 2232-2252, 2016 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-27689529

RESUMO

OBJECTIVE: Recent studies implicate cardiolipin oxidation in several age-related diseases. Atp8b1 encoding Type 4 P-type ATPases is a cardiolipin transporter. Mutation in Atp8b1 gene or inflammation of the lungs impairs the capacity of Atp8b1 to clear cardiolipin from lung fluid. However, the link between Atp8b1 mutation and age-related gene alteration is unknown. Therefore, we investigated how Atp8b1 mutation alters age-related genes. METHODS: We performed Affymetrix gene profiling of lungs isolated from young (7-9 wks, n=6) and aged (14 months, 14 M, n=6) C57BL/6 and Atp8b1 mutant mice. In addition, Ingenuity Pathway Analysis (IPA) was performed. Differentially expressed genes were validated by quantitative real-time PCR (qRT-PCR). RESULTS: Global transcriptome analysis revealed 532 differentially expressed genes in Atp8b1 lungs, 157 differentially expressed genes in C57BL/6 lungs, and 37 overlapping genes. IPA of age-related genes in Atp8b1 lungs showed enrichment of Xenobiotic metabolism and Nrf2-mediated signaling pathways. The increase in Adamts2 and Mmp13 transcripts in aged Atp8b1 lungs was validated by qRT-PCR. Similarly, the decrease in Col1a1 and increase in Cxcr6 transcripts was confirmed in both Atp8b1 mutant and C57BL/6 lungs. CONCLUSION: Based on transcriptome profiling, our study indicates that Atp8b1 mutant mice may be susceptible to age-related lung diseases.


Assuntos
Adenosina Trifosfatases/genética , Envelhecimento/genética , Pulmão/metabolismo , Proteínas de Transferência de Fosfolipídeos/genética , Proteínas ADAMTS/genética , Proteínas ADAMTS/metabolismo , Adenosina Trifosfatases/metabolismo , Envelhecimento/metabolismo , Animais , Perfilação da Expressão Gênica , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 13 da Matriz/metabolismo , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas de Transferência de Fosfolipídeos/metabolismo , Transcriptoma
11.
J Microbiol Biotechnol ; 26(11): 1881-1890, 2016 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-27435539

RESUMO

Manganese superoxide dismutase (MnSOD) is a vital enzyme that protects cells from free radicals through eliminating superoxide radicals (O²â»). Hirudin, a kind of small active peptide molecule, is one of the strongest anticoagulants that can effectively cure thrombus diseases. In this study, we fused Hirudin to the C terminus of human MnSOD with the GGGGS linker to generate a novel dual-feature fusion protein, denoted as hMnSOD-Hirudin. The hMnSOD-Hirudin gene fragment was cloned into the pET15b (SmaI, CIAP) vector, forming a recombinant pET15b-hMnSOD-Hirudin plasmid, and then was transferred into Escherichia coli strain Rosetta-gami for expression. SDS-PAGE was used to detect the fusion protein, which was expected to be about 30 kDa upon IPTG induction. Furthermore, the hMnSOD-Hirudin protein was heavily detected as a soluble form in the supernatant. The purification rate observed after Ni NTA affinity chromatography was above 95%. The hMnSOD-Hirudin protein yield reached 67.25 mg per liter of bacterial culture. The identity of the purified protein was confirmed by western blotting. The hMnSOD-Hirudin protein activity assay evinced that the antioxidation activity of the hMnSOD-Hirudin protein obtained was 2,444.0 ± 96.0 U/mg, and the anticoagulant activity of the hMnSOD-Hirudin protein was 599.0 ± 35.0 ATU/mg. In addition, in vitro bioactivity assay showed that the hMnSODHirudin protein had no or little cytotoxicity in H9c2, HK-2, and H9 (human CD4⁺, T cell) cell lines. Transwell migration assay and invasion assay showed that the hMnSOD-Hirudin protein could suppress human lung cancer 95-D cell metastasis and invasion in vitro.


Assuntos
Hirudinas/genética , Neoplasias Pulmonares/tratamento farmacológico , Superóxido Dismutase/genética , Superóxido Dismutase/farmacologia , Linhagem Celular Tumoral , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Hirudinas/metabolismo , Hirudinas/farmacologia , Humanos , Neoplasias Pulmonares/patologia , Invasividade Neoplásica , Metástase Neoplásica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Superóxido Dismutase/metabolismo
12.
Oncotarget ; 7(20): 29081-91, 2016 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-27058411

RESUMO

BACKGROUND: Acute lung injury (ALI) is characterized by alveolar damage, increased levels of pro-inflammatory cytokines and impaired alveolar fluid clearance. Recently, we showed that the deletion of Apoptosis signal-regulating kinase 1 (ASK1) protects against hyperoxia-induced acute lung injury (HALI) by suppressing IL-1ß and TNF-α. Previously, our data revealed that the suppressor of cytokine signaling-1 (SOCS-1) overexpression restores alveolar fluid clearance in HALI by inhibiting ASK-1 and suppressing IL-1ß levels. Furthermore, IL-1ß is known to inhibit the expression of epithelial sodium channel α-subunit (ENaC) via a p38 MAPK signaling pathway. OBJECTIVE: To determine whether SOCS-1 overexpression in MLE-12 cells would protect against IL-1ß-mediated depletion of αENaC by suppressing ASK-1 expression. METHODS: We co-transfected MLE-12 cells with SOCS-1 overexpressing plasmid with or without IL-1ß in the presence or absence of sodium channel inhibitor, amiloride. We measured potential difference, transepithelial current, resistance, and sodium uptake levels across MLE-12 cells. We studied the effect of ASK-1 depletion, as well as ASK-1 and SOCS-1 overexpression on αENaC expression. RESULTS: SOCS-1 overexpression sufficiently restored transepithelial current and resistance in MLE-12 cells treated with either IL-1ß or amiloride. The αENaC mRNA levels and sodium transport were increased in SOCS-1 overexpressing MLE-12 cells exposed to IL-1ß. Depletion of ASK-1 in MLE-12 cells increased αENaC mRNA levels. Interestingly, SOCS-1 overexpression restored αENaC expression in MLE-12 cells in the presence of ASK-1 overexpression. CONCLUSION: Collectively, these findings suggest that SOCS-1 may exert its protective effect by rescuing αENaC expression via suppression of ASK-1.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Células Epiteliais/metabolismo , Canais Epiteliais de Sódio/metabolismo , Interleucina-1beta/metabolismo , Proteína 1 Supressora da Sinalização de Citocina/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Linhagem Celular , Células Epiteliais/patologia , Humanos , Camundongos
13.
Am J Physiol Cell Physiol ; 310(8): C625-8, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26825124

RESUMO

In this Perspective, we discuss some recent developments in the study of the mitochondrial scaffolding protein AKAP121 (also known as AKAP1, or AKAP149 as the human homolog), with an emphasis on its role in mitochondrial physiology. AKAP121 has been identified to function as a key regulatory molecule in several mitochondrial events including oxidative phosphorylation, the control of membrane potential, fission-induced apoptosis, maintenance of mitochondrial Ca(2+)homeostasis, and the phosphorylation of various mitochondrial respiratory chain substrate molecules. Furthermore, we discuss the role of hypoxia in prompting cellular stress and damage, which has been demonstrated to mediate the proteosomal degradation of AKAP121, leading to an increase in reactive oxgyen species production, mitochondrial dysfunction, and ultimately cell death.


Assuntos
Proteínas de Ancoragem à Quinase A/fisiologia , Apoptose/fisiologia , Potencial da Membrana Mitocondrial/fisiologia , Mitocôndrias/fisiologia , Modelos Biológicos , Estresse Oxidativo/fisiologia , Animais , Humanos , Oxirredução , Complexo de Endopeptidases do Proteassoma/metabolismo
14.
Oncotarget ; 6(34): 35726-36, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26486088

RESUMO

RNA editing is a post-transcriptional modification of RNA. The majority of these changes result from adenosine deaminase acting on RNA (ADARs) catalyzing the conversion of adenosine residues to inosine in double-stranded RNAs (dsRNAs). Massively parallel sequencing has enabled the identification of RNA editing sites in human transcriptomes. In this study, we sequenced DNA and RNA from human lungs and identified RNA editing sites with high confidence via a computational pipeline utilizing stringent analysis thresholds. We identified a total of 3,447 editing sites that overlapped in three human lung samples, and with 50% of these sites having canonical A-to-G base changes. Approximately 27% of the edited sites overlapped with Alu repeats, and showed A-to-G clustering (>3 clusters in 100 bp). The majority of edited sites mapped to either 3' untranslated regions (UTRs) or introns close to splice sites; whereas, only few sites were in exons resulting in non-synonymous amino acid changes. Interestingly, we identified 652 A-to-G editing events in the 3' UTR of 205 target genes that mapped to 932 potential miRNA target binding sites. Several of these miRNA edited sites were validated in silico. Additionally, we validated several A-to-G edited sites by Sanger sequencing. Altogether, our study suggests a role for RNA editing in miRNA-mediated gene regulation and splicing in human lungs. In this study, we have generated a RNA editome of human lung tissue that can be compared with other RNA editomes across different lung tissues to delineate a role for RNA editing in normal and diseased states.


Assuntos
DNA/análise , Pneumopatias/diagnóstico , Pulmão/fisiologia , MicroRNAs/genética , RNA/análise , Regiões 3' não Traduzidas/genética , Adenina , Adenosina Desaminase/metabolismo , Elementos Alu/genética , Biologia Computacional , Guanosina , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Pneumopatias/genética , Edição de RNA , Transcriptoma
15.
Curr Pharm Des ; 21(24): 3489-95, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26144942

RESUMO

With the current high rates of associated morbidity and mortality, proper management of critical illness is vital in the treatment of severely ill patients. Disruptions of key body systems stemming from these illnesses may be attributed to their severity and limited treatment options. Disruption in circadian rhythms caused by critical illnesses may lead to a decreased patient prognosis, as the biological clocks stemming from circadian rhythms play several key roles in regulating our internal pathways. This review will highlight how the occurrence of hyperoxic injury during the progression of critical illness may severely damage circadian rhymicity, and lead to an increased risk of infection, development of disease, severity of symptoms, morbidity and mortality, and dysregulation of multiple body systems. Overall, there is strong evidence suggesting that the disruption of circadian rhythms may be caused by critical illness. Studies performed on several animal models have shown alterations of key genes associated with circadian clock function may heavily contribute to the increased severity of critical illness linked to circadian disruption. This review will aid in further understanding the link between circadian disruption and critical illness.


Assuntos
Transtornos Cronobiológicos/etiologia , Hiperóxia/complicações , Lesão Pulmonar/etiologia , Animais , Relógios Biológicos/genética , Relógios Biológicos/fisiologia , Transtornos Cronobiológicos/genética , Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Estado Terminal , Modelos Animais de Doenças , Humanos , Lesão Pulmonar/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...