Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Carcinog ; 62(3): 348-359, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36453714

RESUMO

Adenosine-to-inosine (A-to-I) RNA editing is the most common posttranscriptional editing to create somatic mutations and increase proteomic diversity. However, the functions of the edited mutations are largely underexplored. To identify novel targets in lung adenocarcinoma (LUAD), we conducted a genome-wide somatic A-to-I RNA editing analysis of 23 paired adjacent normal and LUAD transcriptomes and identified 26,280 events, including known nonsynonymous AZIN1-S367G and novel RHOAiso2 (RHOA isoform 2)-R176G, tubulin gamma complex associated protein 2 (TUBGCP2)-N211S, and RBMXL1-I40 M mutations. We validated the edited mutations in silico in multiple databases and in newly collected LUAD tissue pairs with the SEQUENOM MassARRAY® and TaqMan PCR Systems. We selected RHOAiso2-R176G due to its significant level, isoform-specificity, and being the most common somatic edited nonsynonymous mutation of RHOAiso2 to investigate its roles in LUAD tumorigenesis. RHOAiso2 is a ubiquitous but low-expression alternative spliced isoform received a unique Alu-rich exon at the 3' RHOA mRNA to become an editing RNA target, leading to somatic hypermutation and protein diversity. Interestingly, LUAD patients harboring the RHOAiso2-R176G mutation were associated with aberrant RHOA functions, cancer cell proliferation and migration, and poor clinical outcomes in transcriptome analysis. Mechanistically, RHOAiso2-R176G mutation-expressing LUAD cells potentiate RHOA-guanosine triphosphate (GTP) activity to phosphorylate ROCK1/2 effectors and enhance cell proliferation and migration in vitro and increase tumor growth in xenograft and systemic metastasis models in vivo. Taken together, the RHOAiso2-R176G mutation is a common somatic A-to-I edited mutation of the hypermutated RHOA isoform 2. It is an oncogenic and isoform-specific theranostic target that activates RHOA-GTP/p-ROCK1/2 signaling to promote tumor progression.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Humanos , RNA , Proteômica , Adenosina , Adenocarcinoma de Pulmão/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Neoplasias Pulmonares/genética , Guanosina Trifosfato , Inosina , Mutação , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Linhagem Celular Tumoral , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
2.
J Hematol Oncol ; 15(1): 85, 2022 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-35794621

RESUMO

BACKGROUND: Metastasis and chemoresistance are major culprits of cancer mortality, but factors contributing to these processes are incompletely understood. METHODS: Bioinformatics methods were used to identify the relations of Smyca expression to clinicopathological features of human cancers. RNA-sequencing analysis was used to reveal Smyca-regulated transcriptome. RNA pull-down and RNA immunoprecipitation were used to examine the binding of Smyca to Smad3/4 and c-Myc/Max. Chromatin immunoprecipitation and chromatin isolation by RNA purification were used to determine the binding of transcription factors and Smyca to various gene loci, respectively. Real-time RT-PCR and luciferase assay were used to examine gene expression levels and promoter activities, respectively. Xenograft mouse models were performed to evaluate the effects of Smyca on metastasis and chemoresistance. Nanoparticle-assisted gapmer antisense oligonucleotides delivery was used to target Smyca in vivo. RESULTS: We identify lncRNA Smyca for its association with poor prognosis of many cancer types. Smyca potentiates metabolic reprogramming, migration, invasion, cancer stemness, metastasis and chemoresistance. Mechanistically, Smyca enhances TGF-ß/Smad signaling by acting as a scaffold for promoting Smad3/Smad4 association and further serves as a Smad target to amplify/prolong TGF-ß signaling. Additionally, Smyca potentiates c-Myc-mediated transcription by enhancing the recruitment of c-Myc/Max complex to a set of target promoters and c-Myc binding to TRRAP. Through potentiating TGF-ß and c-Myc pathways, Smyca synergizes the Warburg effect elicited by both pathways but evades the anti-proliferative effect of TGF-ß. Targeting Smyca prevents metastasis and overcomes chemoresistance. CONCLUSIONS: This study uncovers a lncRNA that coordinates tumor-relevant pathways to orchestra a pro-tumor program and establishes the clinical values of Smyca in cancer prognosis and therapy.


Assuntos
Neoplasias , RNA Longo não Codificante , Animais , Humanos , Camundongos , Regiões Promotoras Genéticas , RNA Longo não Codificante/genética , Fator de Crescimento Transformador beta/metabolismo
3.
J Biomed Sci ; 28(1): 57, 2021 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-34340703

RESUMO

Dysregulation of nucleocytoplasmic shuttling is commonly observed in cancers and emerging as a cancer hallmark for the development of anticancer therapeutic strategies. Despite its severe adverse effects, selinexor, a selective first-in-class inhibitor of the common nuclear export receptor XPO1, was developed to target nucleocytoplasmic protein shuttling and received accelerated FDA approval in 2019 in combination with dexamethasone as a fifth-line therapeutic option for adults with relapsed refractory multiple myeloma (RRMM). To explore innovative targets in nucleocytoplasmic shuttling, we propose that the aberrant contextual determinants of nucleocytoplasmic shuttling, such as PSPC1 (Paraspeckle component 1), TGIF1 (TGF-ß Induced Factor Homeobox 1), NPM1 (Nucleophosmin), Mortalin and EBP50, that modulate shuttling (or cargo) proteins with opposite tumorigenic functions in different subcellular locations could be theranostic targets for developing anticancer strategies. For instance, PSPC1 was recently shown to be the contextual determinant of the TGF-ß prometastatic switch and PTK6/ß-catenin reciprocal oncogenic nucleocytoplasmic shuttling during hepatocellular carcinoma (HCC) progression. The innovative nucleocytoplasmic shuttling inhibitor PSPC1 C-terminal 131 polypeptide (PSPC1-CT131), which was developed to target both the shuttling determinant PSPC1 and the shuttling protein PTK6, maintained their tumor-suppressive characteristics and exhibited synergistic effects on tumor suppression in HCC cells and mouse models. In summary, targeting the contextual determinants of nucleocytoplasmic shuttling with cargo proteins having opposite tumorigenic functions in different subcellular locations could be an innovative strategy for developing new therapeutic biomarkers and agents to improve cancer therapy.


Assuntos
Progressão da Doença , Neoplasias/genética , Oncogenes , Proteínas de Ligação a RNA/genética , Translocação Genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Nucleofosmina , Proteínas de Ligação a RNA/metabolismo
4.
Cells ; 9(6)2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32570949

RESUMO

Paraspeckle protein 1 (PSPC1) overexpression in cancers is known to be the pro-metastatic switch of tumor progression associated with poor prognosis of cancer patients. However, the detail molecular mechanisms to facilitate cancer cell migration remain elusive. Here, we conducted integrated analysis of human phospho-kinase antibody array, transcriptome analysis with RNA-seq, and proteomic analysis of protein pulldown to study the molecular detail of PSPC1-potentiated phenotypical transformation, adhesion, and motility in human hepatocellular carcinoma (HCC) cells. We found that PSPC1 overexpression re-assembles and augments stress fiber formations to promote recruitment of focal adhesion contacts at the protruding edge to facilitate cell migration. PSPC1 activated focal adhesion-associated kinases especially FAK/Src signaling to enhance cell adhesion and motility toward extracellular matrix (ECM). Integrated transcriptome and gene set enrichment analysis indicated that PSPC1 modulated receptor tyrosine kinase IGF1R involved in the focal adhesion pathway and induction of diverse integrins expression. Knockdown IGF1R expression and treatment of IGF1R inhibitor suppressed PSPC1-induced cell motility. Interestingly, knockdown PSPC1-interacted paraspeckle components including NONO, FUS, and the lncRNA Neat1 abolished PSPC1-activated IGF1R expression. Together, PSPC1 overexpression induced focal adhesion formation and facilitated cell motility via activation of IGF1R signaling. PSPC1 overexpression in tumors could be a potential biomarker of target therapy with IGF1R inhibitor for improvement of HCC therapy.


Assuntos
Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/fisiopatologia , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/fisiopatologia , Proteínas de Ligação a RNA/fisiologia , Receptor IGF Tipo 1/fisiologia , Carcinoma Hepatocelular/genética , Adesão Celular/genética , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/fisiologia , Quimiotaxia , Adesões Focais/genética , Adesões Focais/fisiologia , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Hepáticas/genética , Proteômica , Motivo de Reconhecimento de RNA , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/genética , Receptor IGF Tipo 1/antagonistas & inibidores , Receptor IGF Tipo 1/genética , Transdução de Sinais
5.
Mol Cell Oncol ; 7(2): 1721253, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32158931

RESUMO

Protein-tyrosine kinase 6 (PTK6) sequestrated by its substrate paraspeckle component 1 (PSPC1) in nucleus is a tumor suppressor. PSPC1 functions as a contextual determinant of oncogenic subcellular translocations to synergize PSPC1/ß-catenin and PTK6 tumorigenesis. Besides, PSPC1 C-terminal interacting domain (PSPC1-CT131), a dual inhibitor of PSPC1 and PTK6, suppresses cancer progression.

6.
Nucleic Acids Res ; 48(3): 1175-1191, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31853539

RESUMO

Precise noncoding RNA (ncRNA)-based network prediction is necessary to reveal ncRNA functions and pathological mechanisms. Here, we established a systemic pipeline to identify prognostic ncRNAs, predict their functions and explore their pathological mechanisms in lung adenocarcinoma (LUAD). After in silico and experimental validation based on evaluations of prognostic value in multiple LUAD cohorts, we selected the PTTG3P pseudogene from among other prognostic ncRNAs (MIR497HG, HSP078, TBX5-AS1, LOC100506990 and C14orf64) for mechanistic studies. PTTG3P upregulation in LUAD cells shortens the metaphase to anaphase transition in mitosis, increases cell viability after cisplatin or paclitaxel treatment, facilitates tumor growth that leads to poor survival in orthotopic lung models, and is associated with a poor survival rate in LUAD patients in the TCGA cohort who received chemotherapy. Mechanistically, PTTG3P acts as an ncRNA that interacts with the transcription factor FOXM1 to regulate the transcriptional activation of the mitotic checkpoint kinase BUB1B, which augments tumor growth and chemoresistance and leads to poor outcomes for LUAD patients. Overall, we established a systematic strategy to uncover prognostic ncRNAs with functional prediction methods suitable for pan-cancer studies. Moreover, we revealed that PTTG3P, due to its upregulation of the PTTG3P/FOXM1/BUB1B axis, could be a therapeutic target for LUAD patients.


Assuntos
Adenocarcinoma/genética , Neoplasias Pulmonares/genética , RNA não Traduzido/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidade , Adenocarcinoma/patologia , Animais , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Cromatina/genética , Simulação por Computador , Resistencia a Medicamentos Antineoplásicos/genética , Proteína Forkhead Box M1/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Mitose , Prognóstico , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais
7.
Nat Commun ; 10(1): 5716, 2019 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-31844057

RESUMO

Hepatocellular carcinoma (HCC) is one of the most lethal cancers worldwide due to metastasis. Paraspeckle component 1 (PSPC1) upregulation has been identified as an HCC pro-metastatic activator associated with poor patient prognosis, but with a lack of targeting strategy. Here, we report that PSPC1, a nuclear substrate of PTK6, sequesters PTK6 in the nucleus and loses its metastasis driving capability. Conversely, PSPC1 upregulation or PSPC1-Y523F mutation promotes epithelial-mesenchymal transition, stemness, and metastasis via cytoplasmic translocation of active PTK6 and nuclear translocation of ß-catenin, which interacts with PSPC1 to augment Wnt3a autocrine signaling. The aberrant nucleocytoplasmic shuttling of active PTK6/ß-catenin is reversed by expressing the PSPC1 C-terminal interacting domain (PSPC1-CT131), thereby suppressing PSPC1/PTK6/ß-catenin-activated metastasis to prolong the survival of HCC orthotopic mice. Thus, PSPC1 is the contextual determinant of the oncogenic switch of PTK6/ß-catenin subcellular localizations, and PSPC1-CT131 functions as a dual inhibitor of PSPC1 and PTK6 with potential for improving cancer therapy.


Assuntos
Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas de Ligação a RNA/metabolismo , beta Catenina/metabolismo , Adulto , Idoso , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/mortalidade , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Progressão da Doença , Transição Epitelial-Mesenquimal/genética , Feminino , Humanos , Estimativa de Kaplan-Meier , Fígado/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/mortalidade , Masculino , Camundongos , Pessoa de Meia-Idade , Mutação , Proteínas de Ligação a RNA/genética , Transdução de Sinais , Regulação para Cima , Proteína Wnt3A/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem
8.
Sci Rep ; 9(1): 9687, 2019 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-31273221

RESUMO

Cancer stemness drives tumor initiation, progression, metastasis, recurrence, and therapy resistance. However, mechanisms that potentiate the acquisition and maintenance of stemness fate of cancer cells remain incompletely understood. Here, we show that miR-103/107 stimulate multiple stem-like features in colorectal cancer, including expression of stem-like markers, appearance of side-population cells, and capabilities in self-renewal, tumor initiation, recurrence, and chemoresistance. Mechanistically, these stemness-promoting functions are mediated by miR-103/107-dependent repression of Axin2, a negative feedback regulator of Wnt/ß-catenin signaling. Through inhibiting Axin2, miR-103/107 trigger a prolonged duration of Wnt/ß-catenin signaling and a sustained induction of Wnt responsive genes. In colorectal cancer patients, miR-103/107 expression correlates inversely with Axin2 expression and a signature of miR-103/107 high and Axin2 low expression profile correlates with poor prognosis. Together, our study identifies a novel function of miR-103/107 in promoting colorectal cancer stemness by targeting Axin2 and elucidates the clinical relevance and prognostic value of this axis in colorectal cancer.


Assuntos
Proteína Axina/metabolismo , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Células-Tronco Neoplásicas/patologia , Proteína Wnt1/metabolismo , beta Catenina/metabolismo , Animais , Apoptose , Proteína Axina/genética , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Prognóstico , Células Tumorais Cultivadas , Via de Sinalização Wnt , Proteína Wnt1/genética , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/genética
9.
Cancer Res ; 79(15): 3797-3805, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31300476

RESUMO

The TGFß cytokine plays dichotomous roles during tumor progression. In normal and premalignant cancer cells, the TGFß signaling pathway inhibits proliferation and promotes cell-cycle arrest and apoptosis. However, the activation of this pathway in late-stage cancer cells could facilitate the epithelial-to-mesenchymal transition, stemness, and mobile features to enhance tumorigenesis and metastasis. The opposite functions of TGFß signaling during tumor progression make it a challenging target to develop anticancer interventions. Nevertheless, the recent discovery of cellular contextual determinants, especially the binding partners of the transcription modulators Smads, is critical to switch TGFß responses from proapoptosis to prometastasis. In this review, we summarize the recently identified contextual determinants (such as PSPC1, KLF5, 14-3-3ζ, C/EBPß, and others) and the mechanisms of how tumor cells manage the context-dependent autonomous TGFß responses to potentiate tumor progression. With the altered expression of some contextual determinants and their effectors during tumor progression, the aberrant molecular prometastatic switch might serve as a new class of theranostic targets for developing anticancer strategies.


Assuntos
Neoplasias/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Humanos , Neoplasias/genética , Neoplasias/patologia , Fator de Crescimento Transformador beta/genética
10.
Nat Cell Biol ; 20(4): 479-491, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29593326

RESUMO

Activation of metastatic reprogramming is critical for tumour metastasis. However, more detailed knowledge of the underlying mechanism is needed to enable targeted intervention. Here, we show that paraspeckle component 1 (PSPC1), identified in an aberrant 13q12.11 locus, is upregulated and associated with poor survival in patients with cancer. PSPC1 promotes tumorigenesis, epithelial-to-mesenchymal transition (EMT), stemness and metastasis in multiple cell types and in spontaneous mouse cancer models. PSPC1 is the master activator for transcription factors of EMT and stemness and accompanies c-Myc activation to facilitate tumour growth. PSPC1 increases transforming growth factor-ß1 (TGF-ß1) secretion through an interaction with phosphorylated and nuclear Smad2/3 to potentiate TGF-ß1 autocrine signalling. Moreover, PSPC1 acts as a contextual determinant of the TGF-ß1 pro-metastatic switch to alter Smad2/3 binding preference from tumour-suppressor to pro-metastatic genes. Having validated the PSPC1-Smads-TGF-ß1 axis in various cancers, we conclude that PSPC1 is a master activator of pro-metastatic switches and a potential target for anti-metastasis drugs.


Assuntos
Comunicação Autócrina , Movimento Celular , Transição Epitelial-Mesenquimal , Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Células A549 , Animais , Feminino , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Metástase Neoplásica , Neoplasias/genética , Neoplasias/patologia , Células-Tronco Neoplásicas/patologia , Proteínas Nucleares/genética , Células PC-3 , Fenótipo , Fosforilação , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas de Ligação a RNA/genética , Proteína Smad2/genética , Proteína Smad3/genética , Fatores de Tempo , Fator de Crescimento Transformador beta1/genética
11.
Cancer Res ; 72(14): 3631-41, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22593189

RESUMO

Metastasis is the major cause of poor prognosis in colorectal cancer (CRC), and increasing evidence supports the contribution of miRNAs to cancer progression. Here, we found that high expression of miR-103 and miR-107 (miR-103/107) was associated with metastasis potential of CRC cell lines and poor prognosis in patients with CRC. We showed that miR-103/107 targeted the known metastasis suppressors death-associated protein kinase (DAPK) and Krüppel-like factor 4 (KLF4) in CRC cells, resulting in increased cell motility and cell-matrix adhesion and decreased cell-cell adhesion and epithelial marker expression. miR-103/107 expression was increased in the presence of hypoxia, thereby potentiating DAPK and KLF4 downregulation and hypoxia-induced motility and invasiveness. In mouse models of CRC, miR-103/107 overexpression potentiated local invasion and liver metastasis effects, which were suppressed by reexpression of DAPK or KLF4. miR-103/107-mediated downregulation of DAPK and KLF4 also enabled the colonization of CRC cells at a metastatic site. Clinically, the signature of a miR-103/107 high, DAPK low, and KLF4 low expression profile correlated with the extent of lymph node and distant metastasis in patients with CRC and served as a prognostic marker for metastasis recurrence and poor survival. Our findings therefore indicate that miR-103/107-mediated repression of DAPK and KLF4 promotes metastasis in CRC, and this regulatory circuit may contribute in part to hypoxia-stimulated tumor metastasis. Strategies that disrupt this regulation might be developed to block CRC metastasis.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição Kruppel-Like/metabolismo , MicroRNAs/fisiologia , Metástase Neoplásica/genética , Animais , Linhagem Celular Tumoral , Movimento Celular , Junções Célula-Matriz/metabolismo , Neoplasias Colorretais/mortalidade , Proteínas Quinases Associadas com Morte Celular , Regulação para Baixo , Humanos , Fator 4 Semelhante a Kruppel , Masculino , Camundongos , Camundongos Nus , Prognóstico
12.
J Pharmacol Exp Ther ; 340(1): 169-75, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22005041

RESUMO

Bronchopulmonary dysplasia (BPD) remains a major cause of morbidity and mortality during the first year of life, and many infants have significant respiratory problems throughout childhood. Currently no effective therapy is clinically available to prevent the long-term pulmonary sequelae of BPD. Previous research has demonstrated that the renin-angiotensin system is up-regulated in human lung fibroblasts. Angiotensin II type 1 receptor (AT1R) antagonists and AT1R short interfering RNA diminished hyperoxia-increased collagen expression, whereas AT2R antagonists did not have any effects on these hyperoxia-induced changes. The in vivo therapeutic effects of AT1R antagonists on hyperoxia-induced lung fibrosis remain unknown. The present study assessed the effects of an AT1R antagonist (losartan) on preventing hyperoxia-induced lung fibrosis in newborn rats. Rat pups were exposed to 7 days of > 95% O2 and an additional 2 weeks of 60% O2. AT1R antagonist-treated pups were injected intraperitoneally with losartan at a dose of 10 mg/kg/day from postnatal days 1 to 7 and a dose of 5 mg/kg/day from postnatal days 8 to 21. Control group pups were injected with an equal volume of normal saline. AT1R antagonist treatment attenuated the hyperoxia-induced lung fibrosis on postnatal days 7 and 21 and also decreased the hyperoxia-induced expression of extracellular signal-regulated protein kinase and α-smooth muscle actin. AT1R antagonist treatment did not affect body weight or lung weight of the rats. These data suggest that AT1R antagonist may offer a novel therapeutic strategy for preventing hyperoxia-induced lung fibrosis.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Hiperóxia/complicações , Fibrose Pulmonar/tratamento farmacológico , Actinas/biossíntese , Animais , Animais Recém-Nascidos , Western Blotting , Peso Corporal/efeitos dos fármacos , Colágeno/biossíntese , Colágeno/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/biossíntese , Feminino , Hiperóxia/patologia , Losartan/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Tamanho do Órgão/efeitos dos fármacos , Gravidez , Fibrose Pulmonar/etiologia , Fibrose Pulmonar/patologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real
13.
Neonatology ; 101(1): 47-54, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21791939

RESUMO

BACKGROUND: Oxygen toxicity plays an important role in lung injury and may lead to bronchopulmonary dysplasia. We previously demonstrated that hyperoxia activated the renin-angiotensin system (RAS) in cultured human fetal lung fibroblasts. OBJECTIVE: To examine whether the upregulation of RAS components is associated with hyperoxia-induced lung fibrosis in neonatal Sprague-Dawley rats. METHODS: Experimental rat pups were exposed to 1 week of >95% O(2) and a further 2 weeks of 60% O(2). Control pups were exposed to room air over the same periods. Lung tissues were taken for biochemical and histochemical assays on postnatal days 7 and 21. RESULTS: Hyperoxia significantly increased total collagen content and the expression of type I collagen and alpha smooth muscle actin when compared to control rats. RAS components including angiotensinogen, angiotensin-converting enzyme, angiotensin II, and angiotensin II type 1 receptor were significantly upregulated by hyperoxia. The results also demonstrated that only the extracellular signal-regulated kinase (ERK) signaling pathway was activated by hyperoxia exposure. p38 mitogen-activated protein kinase and c-Jun N-terminal kinase were not activated. CONCLUSIONS: Local RAS activation is involved in the pathogenesis of hyperoxia-induced lung fibrosis in neonatal rats. ERK phosphorylation might mediate angiotensin II type 1 receptor activation.


Assuntos
Hiperóxia/induzido quimicamente , Oxigênio/efeitos adversos , Fibrose Pulmonar/induzido quimicamente , Sistema Renina-Angiotensina/fisiologia , Angiotensina II/genética , Angiotensina II/metabolismo , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/genética , Receptor Tipo 2 de Angiotensina/metabolismo
14.
Pediatr Neonatol ; 52(1): 5-10, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21385650

RESUMO

BACKGROUND: Airway remodeling refers to the structural changes in the airways of asthma. Caveolin-1 reduces cell growth and negatively regulates smooth muscle cell proliferation. The aim was to investigate lung caveolin-1 status in a murine model of acute allergic airway disease. METHODS: Six- to eight-week-old female BALB/c mice were sensitized by intraperitoneal injections of phosphate-buffered saline or ovalbumin (OVA) and aluminium hydroxide on Days 0 and 14, challenged with aerosolized saline or OVA (1%) on Days 21-25, 28-32, and 35. The mice were killed 1 day after the last OVA/saline challenge. Serum OVA-specific immunoglobulin E (IgE) was measured by enzyme-linked immunosorbent assay. Peribronchial inflammation was quantified by morphometric analysis. Lung caveolin-1 and Type I collagen mRNA expression was determined by real-time reverse-transcription polymerase chain reaction. Total lung collagen was measured using Sircol Assay Kit. RESULTS: Serum OVA-specific IgE levels were significantly elevated in OVA-challenged mice when compared with saline-challenged mice. Percentage of inflammatory cells in the bronchoalveolar lavage was significantly higher in the OVA-challenged animals. The animals' lungs that were sensitized and challenged with OVA contained large numbers of inflammatory cells concentrated near the airways and in the perivascular areas. The thickness of the bronchial epithelial layer and smooth muscle layer and the numbers of total inflammatory cells and eosinophils significantly increased in OVA-challenged mice. Caveolin-1 mRNA expression significantly decreased and Type I collagen mRNA expression significantly increased in the lung tissue of OVA-challenged mice. CONCLUSION: These results suggest that caveolin-1 seems to be involved in the pathogenesis of airway remodeling of acute allergic airway disease.


Assuntos
Asma/metabolismo , Caveolina 1/metabolismo , Doença Aguda , Remodelação das Vias Aéreas , Animais , Biomarcadores/metabolismo , Lavagem Broncoalveolar , Colágeno/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Free Radic Biol Med ; 49(1): 88-95, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20353822

RESUMO

A high concentration of oxygen can cause lung injury and lead to pulmonary fibrosis. Angiotensin (Ang) II induces human lung fibroblast proliferation and stimulates collagen synthesis. However, the role of the renin-angiotensin system (RAS) in the pathogenesis of hyperoxia-induced collagen production is unclear. The aims of this study were to investigate the effects of hyperoxia on the components of the RAS and collagen expression in human lung fibroblasts (MRC-5). Hyperoxia increased total collagen, collagen type I, and alpha-smooth muscle actin (alpha-SMA) mRNA and protein expression. RAS components and Ang II production were also significantly increased after hyperoxic exposure. Hyperoxia induced Ang II type 1 receptor (AT1R) expression but did not alter AT2R expression, furthermore, silencing of AT1R signaling with small interfering RNA suppressed hyperoxia-induced phosphorylated-ERK (p-ERK) 1/2, alpha-SMA, and collagen type I expression. Ang II increased p-ERK 1/2 and collagen type I expression, and these increases were inhibited by the AT1R inhibitor, losartan, but not by the AT2R inhibitor, PD123319 under both normoxic and hyperoxic conditions. These data suggest Ang II-mediated signaling transduction via AT1R is involved in hyperoxia-induced collagen synthesis in human lung fibroblasts.


Assuntos
Actinas/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Colágeno Tipo I/biossíntese , Fibroblastos/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Actinas/genética , Bloqueadores do Receptor Tipo 2 de Angiotensina II , Células Cultivadas , Colágeno Tipo I/genética , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Humanos , Hiperóxia , Imidazóis/farmacologia , Losartan/farmacologia , Pulmão/patologia , Piridinas/farmacologia , RNA Interferente Pequeno/genética , Receptor Tipo 1 de Angiotensina/genética , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/genética , Transdução de Sinais
16.
Respiration ; 80(3): 246-53, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20110652

RESUMO

BACKGROUND: Mechanical ventilation with a high tidal volume (VT) increases lung and systemic plasminogen activator inhibitor (PAI)-1 levels and alveolar fibrin deposition. Activated protein C (APC) may decrease PAI activity in endothelial cell-conditioned medium and thus enhance fibrinolysis. OBJECTIVES: The aims of this study were to test the hypothesis that APC can neutralize PAI-1 activity and improve lung function in an animal model of ventilator-induced lung injury. METHODS: Rats were ventilated with a high-volume zero positive end-expiratory pressure (PEEP; HVZP) protocol by a volume-cycled ventilator for 2 h at a VT of 30 ml/kg, a respiratory rate of 25 breaths/min, and an FiO(2) of 0.21. Fifteen minutes before ventilation, the rats received intravenous APC (250 microg/kg, HVZP+APC group) or normal saline (vehicle; HVZP group). Another group that received no ventilation served as the control group. RESULTS: Levels of arterial blood gas tension were comparable between the two ventilation groups throughout the study period. Rats treated with the HVZP protocol exhibited significantly higher total protein and macrophage inflammatory protein-2 concentrations in bronchoalveolar lavage fluid (BALF) and higher lung PAI-1 mRNA expression and plasma active PAI-1 levels than did the control group. Administration of APC tended to reduce the BALF protein content and systemic PAI-1 activity but did not improve the lung histology in the HVZP+APC group. Plasma levels of D-dimers were comparable among the three study groups. CONCLUSIONS: These results suggest that APC administered at a higher dosage might improve lung function by reducing alveolar protein leakage and systemic coagulation.


Assuntos
Fibrinólise/efeitos dos fármacos , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Proteína C/uso terapêutico , Lesão Pulmonar Induzida por Ventilação Mecânica/prevenção & controle , Animais , Líquido da Lavagem Broncoalveolar/química , Quimiocina CXCL2/metabolismo , Produtos de Degradação da Fibrina e do Fibrinogênio/metabolismo , Imuno-Histoquímica , Pulmão/patologia , Masculino , Proteína C/farmacologia , Troca Gasosa Pulmonar , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Volume de Ventilação Pulmonar , Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia
17.
Toxicol Lett ; 193(1): 19-25, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19969051

RESUMO

Survivors of paraquat poisoning may be left with pulmonary fibrosis and a restrictive type of pulmonary dysfunction. Chymase converts angiotensin (Ang) I to Ang II, which is closely involved with lung fibrosis. The role played by chymase in paraquat-induced lung fibrosis is unclear. We examined the effects of paraquat on chymase, renin-angiotensin system components, and collagen expression in murine and human lung fibroblasts (MRC-5). Lung chymase and collagen type I mRNA and protein expression were significantly increased and angiotensin-converting enzyme (ACE) mRNA and protein expression were comparable between the control and paraquat-treated mice 1 and 3 weeks after administration. Paraquat significantly upregulated angiotensinogen mRNA expression in a dose-dependent manner while ACE activity and protein expression were similar in MRC-5 cells. Furthermore, paraquat enhanced Ang II and collagen type I mRNA and protein expression, alpha-smooth muscle actin, and chymase protein and chymase small interfering RNA inhibited these effects. The cDNA sequence of chymase in MRC-5 cells is identical to that in human mast cells. This study found increased chymase expression in paraquat-treated human lung fibroblasts and confirmed in vitro and in an in vivo paraquat model of lung fibrosis that chymase generates Ang II and enhances collagen expression. These data suggest a role for chymase in the pathogenesis of paraquat-induced lung fibrosis.


Assuntos
Quimases/metabolismo , Colágeno Tipo I/biossíntese , Fibroblastos/metabolismo , Herbicidas/toxicidade , Paraquat/toxicidade , Actinas/metabolismo , Angiotensina II/metabolismo , Animais , Western Blotting , Linhagem Celular , DNA Complementar/biossíntese , DNA Complementar/genética , Fibroblastos/efeitos dos fármacos , Humanos , Pulmão/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptidil Dipeptidase A/metabolismo , RNA Interferente Pequeno/genética , Sistema Renina-Angiotensina/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta1/metabolismo
18.
Toxicol In Vitro ; 24(3): 803-8, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20035857

RESUMO

Survivors of paraquat poisoning are left with pulmonary fibrosis which results in a restrictive type of long-term pulmonary dysfunction. Connective tissue growth factor (CTGF) is a key growth factor that initiates tissue repair and underlies the development of lung fibrosis. Angiotensin (ANG) II may induce CTGF expression in the heart and kidney and plays an important role in the pathogenesis of lung fibrosis. The biological effects of ANG II are mediated by ANG II type 1 receptor (AT1R) and AT2R. The aims of this study were to investigate the effects of paraquat on ANG II, ANG II receptors, CTGF, and collagen expressions and to assess the role of ANG II receptors in paraquat-induced collagen synthesis in human lung fibroblasts (MRC-5). MRC-5 cells were incubated with various concentrations of paraquat with or without the ANG II receptor antagonist, saralasin. Paraquat increased ANG II production and AT1R mRNA and protein expression and decreased AT2R mRNA expression. Furthermore, paraquat treatment increased CTGF and collagen mRNA and protein expression in a dose-dependent manner and saralasin inhibited these effects. These results indicate that paraquat increases CTGF and collagen expression by activating angiotensin signaling pathway in human lung fibroblasts.


Assuntos
Angiotensina II/fisiologia , Colágeno/biossíntese , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fibroblastos/efeitos dos fármacos , Herbicidas/toxicidade , Paraquat/toxicidade , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Western Blotting , Colágeno Tipo I/biossíntese , Colágeno Tipo II/biossíntese , Relação Dose-Resposta a Droga , Imunofluorescência , Herbicidas/antagonistas & inibidores , Humanos , Paraquat/antagonistas & inibidores , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptor Tipo 2 de Angiotensina/biossíntese , Receptor Tipo 2 de Angiotensina/genética , Receptores de Angiotensina/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Saralasina/farmacologia , Transdução de Sinais/efeitos dos fármacos
19.
Exp Biol Med (Maywood) ; 233(11): 1334-40, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18791129

RESUMO

Neonates with premature rupture of the membrane and oligohydramnios have an increased risk of acute respiratory morbidity. The aims of this study are to investigate the effects of experimental oligohydramnios on transforming growth factor (TGF)-beta1 and connective tissue growth factor (CTGF) expressions and collagen level in fetal rat lungs. On day 16 of gestation, we anesthetized timed pregnant Sprague-Dawley dams, punctured the uterine wall and fetal membranes of each amniotic sac which resulted in oligohydramnios. Fetuses in the opposite uterine horn served as controls. On days 19 and 21 of gestation, fetuses were delivered by cesarean section. Rats exposed to oligohydramnios exhibited significantly lower lung weight/body weight ratios on days 19 and 21 of gestation than did the control rats. Lung type I collagen and TGF-beta1 mRNA expressions and lung collagen levels were significantly decreased in rats exposed to oligohydramnios on days 19 and 21 of gestation. Type I collagen and inhibitors of metalloproteinase-1 (TIMP-1) proteins were decreased and matrix metalloproteinase-1 (MMP-1) was increased in oligohydramnios-exposed rats on days 19 and 21 of gestation. CTGF mRNA expressions were comparable between control and oligohydramnios-exposed rats on days 19 and 21 of gestation. These data suggest that downregulation of collagen might be involved in the pathogenesis of oligohydramnios-induced respiratory morbidity.


Assuntos
Colágeno/metabolismo , Feto/metabolismo , Pulmão/embriologia , Oligo-Hidrâmnio/metabolismo , Animais , Western Blotting , Peso Corporal , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Modelos Animais de Doenças , Feminino , Imuno-Histoquímica , Pulmão/metabolismo , Pulmão/patologia , Metaloproteinase 1 da Matriz/metabolismo , Tamanho do Órgão , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
20.
Crit Care Med ; 36(6): 1880-5, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18520644

RESUMO

OBJECTIVE: To test the hypotheses that high tidal-volume ventilation increases plasminogen activator inhibitor (PAI)-1, and the angiotensin-converting enzyme inhibitor, captopril (CAP), may attenuate these effects. SETTING: University research facility. SUBJECTS: Twenty adult male Sprague-Dawley rats. INTERVENTIONS: All rats were randomized to receive two ventilation strategies for 2 h: 1) a high-volume zero positive end-expiratory pressure (PEEP) (HVZP) group at a tidal volume of 40 mL/kg, a respiratory rate of 25 breaths/min, and an FiO2 of 0.21; and 2) an HVZP + CAP group which received an intraperitoneal injection of CAP (100 mg/kg) 30 min before HVZP ventilation. Another group that was not subjected to ventilation served as the control. MEASUREMENTS AND MAIN RESULTS: Total protein recovered from bronchoalveolar lavage fluid was significantly higher in rats ventilated with the HVZP protocols than in control rats. Rats treated with HVZP ventilation had significantly higher lung angiotensin (ANG) II and PAI-1 messenger RNA expression levels and a higher plasma active PAI-1 level than did the control and HVZP + CAP groups. Lung ANG II levels were positively correlated with plasma PAI-1. Representative lung tissue of the HVZP + CAP group showed mild inflammatory cell infiltration and less hemorrhage and fibrin deposition than did the HVZP group. The HVZP and HVZP + CAP groups had significantly higher lung injury scores than did the control group and rats treated with HVZP + CAP ventilation exhibited significantly lower lung injury scores than did the HVZP group. CONCLUSIONS: Mechanical ventilation with a high tidal volume and no PEEP increases alveolar fibrin deposition and systemic PAI-1 activity, which are attenuated by captopril, an angiotensin-converting enzyme inhibitor. These results imply that local ANG II is involved in the pathogenesis of disordered coagulation in ventilator-induced lung injury (VILI) and suggest that the protective mechanism of captopril's attenuation of VILI is related to a reduction in PAI-1.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Captopril/farmacologia , Inibidor 1 de Ativador de Plasminogênio/sangue , Pneumonia Associada à Ventilação Mecânica/sangue , Respiração com Pressão Positiva/efeitos adversos , Volume de Ventilação Pulmonar/fisiologia , Angiotensina II/sangue , Angiotensina II/genética , Animais , Fibrinólise/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/patologia , Masculino , Inibidor 1 de Ativador de Plasminogênio/genética , Pneumonia Associada à Ventilação Mecânica/patologia , Reação em Cadeia da Polimerase , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...