Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cell Signal ; 115: 111029, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38163576

RESUMO

Sirtuin 3 (SIRT3) regulates mitochondrial function as a mitochondrial deacetylase during oxidative stress. However, the specific regulatory mechanism and function of SIRT3 in radioresistant cancer cells are unclear. In this study, we aim to investigate how SIRT3 determines the susceptibility to glucose deprivation and its regulation in p53-based radioresistant head and neck cancer cells. We observed mitochondrial function using two established isogenic radioresistant subclones (HN3R-A [p53 null] and HN3R-B [p53 R282W]) with intratumoral p53 heterogeneity. Cell counting analysis was performed to evaluate cell proliferation and cell death. The correlation between the regulation of SIRT3 and enhancer of zeste homolog 2 (EZH2) was confirmed by immunoblotting and chromatin immunoprecipitation assay. p53-deficient radioresistant cells (HN3R-A) expression reduced SIRT3 levels and increased sensitivity to glucose deprivation due to mitochondrial dysfunction compared to other cells. In these cells, activation of SIRT3 significantly prevented glucose deprivation-induced cell death, whereas the loss of SIRT3 increased the susceptibility to glucose deficiency. We discovered that radiation-induced EZH2 directly binds to the SIRT3 promoter and represses the expression. Conversely, inhibiting EZH2 increased the expression of SIRT3 through epigenetic changes. Our findings indicate that p53-deficient radioresistant cells with enhanced EZH2 exhibit increased sensitivity to glucose deprivation due to SIRT3 suppression. The regulation of SIRT3 by EZH2 plays a critical role in determining the cell response to glucose deficiency in radioresistant cancer cells. Therefore, EZH2-dependent SIRT3 could be used as a predictive biomarker to select treatment options for patients with radiation-resistance.


Assuntos
Neoplasias de Cabeça e Pescoço , Sirtuína 3 , Humanos , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Sirtuína 3/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Neoplasias de Cabeça e Pescoço/radioterapia , Estresse Oxidativo
2.
Mod Pathol ; 26(5): 626-39, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23328978

RESUMO

WNT inhibitory factor-1 (WIF1) is an antagonist of the WNT signaling pathway. We investigated the relationship between WIF1 promoter methylation and regulation of the WNT/ß-catenin signaling pathway, tumor grade, and survival in patients with astrocytoma. This study included 86 cases of astrocytoma, comprising 20 diffuse astrocytomas and 66 glioblastomas. In addition, 17 temporal lobectomy specimens from patients with epilepsy were included as controls. The ratio of methylated DNA to total methylated and unmethylated DNA (% methylation) was measured by methylation- and unmethylation-specific PCR. Representative tumor tissue was immunostained for WIF1, ß-catenin, cyclin D1, c-myc, and isocitrate dehydrogenase 1. Levels of WIF1 promoter methylation, mRNA expression, and protein expression in a glioblastoma cell line were compared before and after demethylation treatment. The mean percent methylation of the WIF1 promoter in astrocytomas was higher than that in control brain tissue. WIF1 protein expression was lower in the tumor group with >5% methylation than in the group with <5% methylation. Cytoplasmic ß-catenin staining was more frequently observed in tumors with a low WIF1 protein expression level. Demethylation treatment of a glioblastoma cell line increased WIF1 mRNA and protein expression. Increased WIF1 promoter methylation and decreased WIF1 protein expression were not related to patient survival. In conclusion, WIF1 expression is downregulated by promoter methylation and is an important mechanism of aberrant WNT/ß-catenin pathway activation in astrocytoma pathogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Astrocitoma/genética , Astrocitoma/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Via de Sinalização Wnt/fisiologia , Adolescente , Adulto , Idoso , Astrocitoma/mortalidade , Neoplasias Encefálicas/mortalidade , Criança , Metilação de DNA/genética , Feminino , Imunofluorescência , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos , Adulto Jovem
3.
Exp Mol Med ; 55(1): 43-54, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36596853

RESUMO

Glioblastoma multiforme (GBM), the most aggressive and malignant glioma, has a poor prognosis. Although patients with GBM are treated with surgery, chemotherapy, and radiation therapy, GBM is highly resistant to treatment, making it difficult and expensive to treat. In this study, we analyzed the Gene Expression Profiling Interactive Analysis dataset, the Cancer Genome Atlas dataset, and Gene Expression Omnibus array data. ZBTB7A (also called FBI1/POKEMON/LRF) was found to be highly expressed in low-grade glioma but significantly downregulated in patients with GBM. ZBTB7A is a transcription factor that plays an important role in many developmental stages, including cell proliferation. The activation of epithelial-mesenchymal transition (EMT) is a key process in cancer progression and metastasis. Erythrocyte membrane protein band 4.1 like 5 (EPB41L5) is an essential protein for EMT progression and metastasis in various types of cancer. We found that ZBTB7A depletion in U87 cells induced GBM progression and metastasis. Based on RNA sequencing data, ZBTB7A directly binds to the promoter of the EPB41L5 gene, reducing its expression and inhibiting GBM progression. We demonstrated that ZBTB7A dramatically inhibits GBM tumor growth through transcriptional repression of EPB41L5. Thus, both ZBTB7A and EPB41L5 may be potential biomarkers and novel therapeutic targets for GBM treatment. Overall, we discovered the role of a novel tumor suppressor that directly inhibits GBM progression (ZBTB7A) and identified EPB41L5 as a therapeutic target protein for patients with GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Glioblastoma/metabolismo , Linhagem Celular Tumoral , Glioma/genética , Transformação Celular Neoplásica/genética , Carcinogênese/genética , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Proliferação de Células/genética , Proteínas de Membrana/metabolismo
4.
Biochem Biophys Res Commun ; 418(2): 260-6, 2012 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-22266309

RESUMO

The issue of whether aberrant expression of ß1-integrin is associated with cancer progression and development of resistance to cytotoxic therapy is of considerable interest. Studies to date have shown that the anchorage-independent survival of cancer is attributed, in part, to epithelial-to-mesenchymal transition (EMT). Here, we have reported a novel alternative mechanism of anchorage-independent survival of cancer cells. Cell lines derived from head and neck cancer patients (AMC-HN-3 and AMC-HN-9) and the well-known EMT cancer cell line, MDA-MB231, were examined. The EMT features of AMC-HN-9 cells were comparable to those of MDA-MB231, whereas AMC-HN-3 cells showed no EMT characteristics. Although the pattern and degree of ß1-integrin expression were similar in all three cell lines, sensitivities of the cells to ß1-integrin knockdown with small interfering RNA (siRNA) were different. Cancer cells with no EMT features underwent cell death to a more significant extent following ß1-integrin silencing than those with EMT. Intriguingly, we observed reactive activation of the p53-p21 pathway after ß1-integrin silencing in AMC-HN-9 cells lacking an apparent cell death response. Simultaneous knockdown of wild-type p53 and ß1-integrin in this cell line promoted cell death. Our data collectively indicate that ß1-integrin-related cell death is closely associated with EMT phenotypes and activation of the p53-p21 pathway is partly involved in the acquisition of resistance to apoptosis induced by ß1-integrin silencing. Further clarification of the mechanisms underlying p53 integration with ß1-integrin signaling may facilitate the development of novel anti-cancer strategies.


Assuntos
Apoptose/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Transição Epitelial-Mesenquimal/genética , Neoplasias de Cabeça e Pescoço/patologia , Integrina beta1/genética , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Interferência de RNA , RNA Interferente Pequeno/genética
5.
Cell Mol Life Sci ; 67(11): 1845-58, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20165901

RESUMO

GD2 ganglioside has been identified as a key determinant of bone marrow-derived mesenchymal stem cells (BM-MSCs). Here, we characterized GD2 ganglioside expression and its implications in umbilical cord blood-derived MSCs (UCB-MSCs). Using immune-selection analysis, we showed that both GD2-positive and GD2-negative UCB-MSCs expressed general stem cell markers and possessed mesodermal lineage differentiation potential. Although the fraction of GD2-expressing cells was lower in UCB-MSC than in BM-MSC populations, inhibition of GD2 synthesis in UCB-MSCs suppressed neuronal differentiation and down-regulated basic helix-loop-helix (bHLH) transcription factors, which are involved in early stage neuronal differentiation. In addition, the levels of bHLH factors in neuronally induced UCB-MSCs were significantly higher in GD2-positive than GD2-negative cells. Our data demonstrate that GD2 ganglioside expression is associated with regulation of bHLH factors and identify neurogenic-capable UCB-MSCs, providing new insights into the potential clinical applications of MSC-based therapy.


Assuntos
Sangue Fetal/citologia , Sangue Fetal/metabolismo , Gangliosídeos/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Proliferação de Células , Separação Celular , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Primers do DNA/genética , Humanos , Recém-Nascido , Transplante de Células-Tronco Mesenquimais , N-Acetilgalactosaminiltransferases/antagonistas & inibidores , N-Acetilgalactosaminiltransferases/genética , N-Acetilgalactosaminiltransferases/metabolismo , Neurogênese , RNA Interferente Pequeno/genética
6.
Cell Signal ; 77: 109820, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33137455

RESUMO

The manner in which p53 maintains redox homeostasis and the means by which two key metabolic elements, glucose and glutamine, contribute to p53-dependent redox stability remain unclear. To elucidate the manner in which p53 deals with glucose-deprived, reactive oxygen species (ROS)-prone conditions in this regard, two isogenic cancer subclones (HN3R-A and HN3R-B) bearing distinct p53 mutations as an in vitro model of intratumoral p53 heterogeneity were identified. Following cumulative irradiation, the subclones showed a similar metabolic shift to aerobic glycolysis and increasing NADPH biogenesis for cellular defense against oxidative damage irrespective of p53 status. The radioresistant cancer cells became more sensitive to glycolysis-targeting drugs. However, in glucose-deprived and ROS-prone conditions, HN3R-B, the subclone with the original p53 increased the utilization of glutamine by GLS2, thereby maintaining redox homeostasis and ATP. Conversely, HN3R-A, the p53-deficient radioresistant subclone displayed an impairment in glutamine usage and high susceptibility to metabolic stresses as well as ROS-inducing agents despite the increased ROS scavenging system. Collectively, our findings suggest that p53 governs the alternative utilization of metabolic ingredients, such as glucose and glutamine, in ROS-prone conditions. Thus, p53 status may be an important biomarker for selecting cancer treatment strategies, including metabolic drugs and ROS-inducing agents, for recurrent cancers after radiotherapy.


Assuntos
Glutamina/metabolismo , Estresse Oxidativo/genética , Proteína Supressora de Tumor p53/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Glucose/farmacologia , Glutaminase/metabolismo , Glutationa/metabolismo , Glicólise , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , NADP/metabolismo , Oxirredução , Tolerância a Radiação , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/genética , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
7.
Cell Death Dis ; 12(3): 250, 2021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33674559

RESUMO

Tumors are composed of subpopulations of cancer cells with functionally distinct features. Intratumoral heterogeneity limits the therapeutic effectiveness of cancer drugs. To address this issue, it is important to understand the regulatory mechanisms driving a subclonal variety within a therapy-resistant tumor. We identified tumor subclones of HN9 head and neck cancer cells showing distinct responses to radiation with different levels of p62 expression. Genetically identical grounds but epigenetic heterogeneity of the p62 promoter regions revealed that radioresistant HN9-R clones displayed low p62 expression via the creation of repressive chromatin architecture, in which cooperation between DNMT1 (DNA methyltransferases 1) and HDAC1 (histone deacetylases 1) resulted in DNA methylation and repressive H3K9me3 and H3K27me3 marks in the p62 promoter. Combined inhibition of DNMT1 and HDAC1 by genetic depletion or inhibitors enhanced the suppressive effects on proliferative capacity and in vivo tumorigenesis following irradiation. Importantly, ectopically p62-overexpressed HN9-R clones increased the induction of senescence along with p62-dependent autophagy activation. These results demonstrate the heterogeneous expression of p62 as the key component of clonal variation within a tumor against irradiation. Understanding the epigenetic diversity of p62 heterogeneity among subclones allows for improved identification of the functional state of subclones and provides a novel treatment option to resolve resistance to current therapies.


Assuntos
Autofagia/efeitos dos fármacos , Senescência Celular/efeitos da radiação , Epigênese Genética , Neoplasias de Cabeça e Pescoço/radioterapia , Tolerância a Radiação , Proteína Sequestossoma-1/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/radioterapia , Acetilação , Animais , Linhagem Celular Tumoral , Montagem e Desmontagem da Cromatina , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Heterogeneidade Genética , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Histona Desacetilase 1/metabolismo , Humanos , Masculino , Camundongos Nus , Regiões Promotoras Genéticas , Tolerância a Radiação/genética , Proteína Sequestossoma-1/genética , Transdução de Sinais , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Arch Pharm Res ; 32(5): 639-46, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19471876

RESUMO

The genetic materials for systemic administration meet a number of hurdles before they reach the nucleus of the target cells, such as enzymatic degradation in the bloodstream, extravascularization around the target tissue, endocytosis by the target cells, and endosomal escape of the genes. Therefore, there have been tremendous needs of effective gene carriers that can deliver the genetic materials to the target site. Of numerous approaches, recent studies have demonstrated that the lipid-based emulsion systems have the high potential as non-viral gene carriers: 1 lipid emulsions are biocompatible because their major constituents are composed of the non-toxic oils and amphiphilic lipids; 2 the cationic lipid emulsions can form nano-sized complexes with negatively charged DNAs, through which the genetic materials can be protected from the enzymatic degradation in the body fluids; 3 The emulsion/DNA complexes are shown to be stable in the bloodstream since their surfaces are rarely recognized by the immune-related cells and serum proteins; and 4 the surfaces of the emulsion complexes are readily modified by varying the lipid composition. In this review, highlighted are the recent advances in the emulsion-based gene carriers.


Assuntos
Núcleo Celular/metabolismo , DNA/metabolismo , Técnicas de Transferência de Genes , Terapia Genética/métodos , Óleos/metabolismo , Transporte Ativo do Núcleo Celular , DNA/química , Desoxirribonucleases/metabolismo , Emulsificantes/química , Emulsões , Nanopartículas , Óleos/química , Óleos/toxicidade , Distribuição Tecidual
9.
Exp Mol Med ; 51(11): 1-11, 2019 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-31784505

RESUMO

Branched-chain amino acid (BCAA) catabolism and high levels of enzymes in the BCAA metabolic pathway have recently been shown to be associated with cancer growth and survival. However, the precise roles of BCAA metabolism in cancer growth and survival remain largely unclear. Here, we found that BCAA metabolism has an important role in human pancreatic ductal adenocarcinoma (PDAC) growth by regulating lipogenesis. Compared with nontransformed human pancreatic ductal (HPDE) cells, PDAC cells exhibited significantly elevated BCAA uptake through solute carrier transporters, which were highly upregulated in pancreatic tumor tissues compared with normal tissues. Branched-chain amino-acid transaminase 2 (BCAT2) knockdown markedly impaired PDAC cell proliferation, but not HPDE cell proliferation, without significant alterations in glutamate or reactive oxygen species levels. Furthermore, PDAC cell proliferation, but not HPDE cell proliferation, was substantially inhibited upon knockdown of branched-chain α-keto acid dehydrogenase a (BCKDHA). Interestingly, BCKDHA knockdown had no significant effect on mitochondrial metabolism; that is, neither the level of tricarboxylic acid cycle intermediates nor the oxygen consumption rate was affected. However, BCKDHA knockdown significantly inhibited fatty-acid synthesis, indicating that PDAC cells may utilize BCAAs as a carbon source for fatty-acid biosynthesis. Overall, our findings show that the BCAA metabolic pathway may provide a novel therapeutic target for pancreatic cancer.


Assuntos
Aminoácidos de Cadeia Ramificada/metabolismo , Metabolismo dos Lipídeos/fisiologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Trifosfato de Adenosina/metabolismo , Animais , Carcinoma Ductal Pancreático/metabolismo , Proliferação de Células/fisiologia , Feminino , Ácido Glutâmico/metabolismo , Humanos , Lentivirus/genética , Metabolômica/métodos , Camundongos SCID , Antígenos de Histocompatibilidade Menor/metabolismo , Consumo de Oxigênio/fisiologia , Proteínas da Gravidez/metabolismo , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transaminases/metabolismo
10.
Head Neck ; 41(9): 3362-3371, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31268585

RESUMO

BACKGROUND: CIP2A may activate multiple oncogenic proteins and promote the proliferation of various cancer cells. METHODS: We investigated that the role of CIP2A in radioresistant head and neck cancer (HNC) cell line with TP53 mutation and the effect of the rapamycin on the response of HN31 with TP53 mutation cells to irradiation related to CIP2A expression. RESULTS: CIP2A expression was stimulated by p53 mutation and critical for the inhibition of senescence induction in response to radiation. The treatment with radiation alone neither induced cytotoxicity in HN31 cells nor completely suppressed the activation of CIP2A. However, the combination of radiation and rapamycin increase the radiosensitivity through the induction of senescence with downregulation of CIP2A expression both in vivo and in vitro. CONCLUSION: Our results suggest that CIP2A may serve as a therapeutic target of rapamycin through induction of senescence in radioresistant HNC with TP53 mutation.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Autoantígenos/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/terapia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Sirolimo/farmacologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Animais , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Neoplasias de Cabeça e Pescoço/genética , Humanos , Masculino , Camundongos , Mutação/genética , Transplante de Neoplasias , Tolerância a Radiação , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/terapia , Proteína Supressora de Tumor p53/genética
11.
Oncogene ; 38(19): 3729-3742, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30664690

RESUMO

The role of p53 in genotoxic therapy-induced metabolic shift in cancers is not yet known. In this study, we investigated the role of p53 in the glycolytic shift in head and neck squamous cell carcinoma cell lines following irradiation. Isogenic p53-null radioresistant cancer cells established through cumulative irradiation showed decreased oxygen consumption and increased glycolysis with compromised mitochondria, corresponding with their enhanced sensitivity to drugs that target glycolysis. In contrast, radioresistant cancer cells with wild-type p53 preserved their primary metabolic profile with intact mitophagic processes and maintained their mitochondrial integrity. Moreover, we identified a previously unappreciated link between p53 and mitophagy, which limited the glycolytic shift through the BNIP3-dependent clearance of abnormal mitochondria. Thus, drugs targeting glycolysis could be used as an alternative strategy for overcoming radioresistant cancers, and the p53 status could be used as a biomarker for selecting participants for clinical trials.


Assuntos
Neoplasias de Cabeça e Pescoço/metabolismo , Proteínas de Membrana/metabolismo , Mitofagia/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Linhagem Celular Tumoral , Glicólise/fisiologia , Neoplasias de Cabeça e Pescoço/radioterapia , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos Endogâmicos NOD , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Proto-Oncogênicas/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/radioterapia , Proteína Supressora de Tumor p53/genética , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Biomaterials ; 29(12): 1920-30, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18289669

RESUMO

Antiangiogenic peptide drugs have received much attention in the fields of tumor therapy and tumor imaging because they show promise in the targeting of integrins such as alpha(v)beta(3) on angiogenic endothelial cells. However, systemic antiangiogenic peptide drugs have short half-lives in vivo, resulting in fast serum clearance via the kidney, and thus the therapeutic effects of such drugs remain modest. In this study, we prepared self-assembled glycol chitosan nanoparticles and explored whether this construct might function as a prolonged and sustained drug delivery system for RGD peptide, used as an antiangiogenic model drug in cancer therapy. Glycol chitosan hydrophobically modified with 5beta-cholanic acid (HGC) formed nanoparticles with a diameter of 230 nm, and RGD peptide was easily encapsulated into HGC nanoparticles (yielding RGD-HGC nanoparticles) with a high loading efficiency (>85%). In vitro work demonstrated that RGD-HGC showed prolonged and sustained release of RGD, lasting for 1 week. RGD-HGC also inhibited HUVEC adhesion to a beta ig-h3 protein-coated surface, indicating an antiangiogenic effect of the RGD peptide in the HGC nanoparticles. In an in vivo study, the antiangiogenic peptide drug formulation of RGD-HGC markedly inhibited bFGF-induced angiogenesis and decreased hemoglobin content in Matrigel plugs. Intratumoral administration of RGD-HGC significantly decreased tumor growth and microvessel density compared to native RGD peptide injected either intravenously or intratumorally, because the RGD-HGC formulation strongly enhanced the antiangiogenic and antitumoral efficacy of RGD peptide by affording prolonged and sustained RGD peptide delivery locally and regionally in solid tumors.


Assuntos
Quitosana/química , Preparações de Ação Retardada/química , Portadores de Fármacos/química , Melanoma/tratamento farmacológico , Nanopartículas/uso terapêutico , Neovascularização Patológica/tratamento farmacológico , Oligopeptídeos/administração & dosagem , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/química , Animais , Preparações de Ação Retardada/administração & dosagem , Portadores de Fármacos/administração & dosagem , Glicóis/administração & dosagem , Glicóis/química , Masculino , Teste de Materiais , Melanoma/irrigação sanguínea , Melanoma/patologia , Camundongos , Camundongos Nus , Nanopartículas/química , Nanopartículas/ultraestrutura , Neovascularização Patológica/patologia , Oligopeptídeos/química , Resultado do Tratamento
13.
Stem Cells Transl Med ; 6(6): 1491-1503, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28244243

RESUMO

Data are limited on the mechanisms underlying memory impairment in heart failure (HF). We hypothesized that angiotensin II (Ang II) may determine the fate of adult hippocampal neural stem cells (HCNs), a cause of memory impairment in HF. HCNs with neurogenesis potential were isolated and cultured from adult rat hippocampi. Ang II decreased HCN proliferation in dose- and time-dependent manners. Moreover, Ang II treatment (1 µM) for 48 hours induced apoptotic death, which was attenuated by pretreatment with Ang II receptor blockers (ARBs). Ang II increased mitochondrial reactive oxygen species (ROS) levels, which was related to mitochondrial morphological changes and functional impairment. Moreover, ROS activated the AMP-activated protein kinase (AMPK) and consequent peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) expression, causing cell apoptosis. In the HF rat model induced by left anterior descending artery ligation, ARB ameliorated the spatial memory ability which decreased 10 weeks after ischemia. In addition, neuronal cell death, especially of newly born mature neurons, was observed in HF rat hippocampi. ARB decreased cell death and promoted the survival of newly born neural precursor cells and mature neurons. In conclusion, Ang II caused HCN apoptosis through mitochondrial ROS formation and subsequent AMPK-PGC1α signaling. ARB improved learning and memory behaviors impaired by neuronal cell death in the HF animal model. These findings suggest that HCN is one treatment target for memory impairment in HF and that ARBs have additional benefits in HF combined with memory impairment. Stem Cells Translational Medicine 2017;6:1491-1503.


Assuntos
Angiotensina II/metabolismo , Apoptose , Insuficiência Cardíaca/complicações , Hipocampo/metabolismo , Transtornos da Memória/metabolismo , Células-Tronco Neurais/metabolismo , Transdução de Sinais , Quinases Proteína-Quinases Ativadas por AMP , Animais , Células Cultivadas , Feminino , Insuficiência Cardíaca/metabolismo , Hipocampo/patologia , Masculino , Transtornos da Memória/etiologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Proteínas Quinases/metabolismo , Ratos , Ratos Sprague-Dawley
14.
Part Fibre Toxicol ; 3: 9, 2006 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-16723032

RESUMO

BACKGROUND: Human beta-defensin (hBD)-2, antimicrobial peptide primarily induced in epithelial cells, is a key factor in the innate immune response of the respiratory tract. Several studies showed increased defensin levels in both inflammatory lung diseases, such as cystic fibrosis, diffuse panbronchiolitis, idiopathic pulmonary fibrosis and acute respiratory distress syndrome, and infectious diseases. Recently, epidemiologic studies have demonstrated acute and serious adverse effects of particulate air pollution on respiratory health, especially in people with pre-existing inflammatory lung disease. To elucidate the effect of diesel exhaust particles (DEP) on pulmonary innate immune response, we investigated the hBD-2 and interleukin-8 (IL-8) expression to DEP exposure in interleukin-1 beta (IL-1beta)-stimulated A549 cells. RESULTS: IL-1beta markedly up-regulated the hBD-2 promoter activity, and the subsequent DEP exposure increased dose-dependently the expression of hBD-2 and inflammatory cytokine IL-8 at the transcriptional level. In addition, DEP further induced the NF-kappaB activation in IL-1beta-stimulated A549 cells more rapidly than in unstimulated control cells, which was showed by nuclear translocation of p65 NF-kappaB and degradation of IkappaB-alpha. The experiment using two NF-kappaB inhibitors, PDTC and MG132, confirmed that this increase of hBD-2 expression following DEP exposure was regulated through NF-kappaB-mediated pathway. CONCLUSION: These results demonstrated that DEP exposure increases the expression of antimicrobial peptide and inflammatory cytokine at the transcriptional level in IL-1beta-primed A549 epithelial cells and suggested that the increase is mediated at least partially through NF-kappaB activation. Therefore, DEP exposure may contribute to enhance the airway-responsiveness especially on the patients suffering from chronic respiratory disease.

15.
Head Neck ; 38 Suppl 1: E1909-17, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26713771

RESUMO

BACKGROUND: We attempted to elucidate the mechanism of cell death after radiation by studying how ß-catenin silencing controls the radiation sensitivity of radioresistant head and neck cancer cells. METHODS: The most radioresistant cancer cell line (AMC-HN-9) was selected for study. Targeted silencing of ß-catenin was used on siRNAs. Sensitivity to radiation was examined using clonogenic and methylthiazol tetrazolium (MTT) assays. RESULTS: A combination of irradiation plus ß-catenin silencing led to a significant reduction in the inherent radioresistance of AMC-HN-9 cells. Although expression of Ku70/80 was upregulated in AMC-HN-9 cells after irradiation, Ku70/80 was dramatically decreased in a combination of irradiation and ß-catenin silencing. Interestingly, irradiation-induced Ku70/80 was completely prevented by ß-catenin silencing-induced LKB1/AMP-activated protein kinase (LKB1/AMPK) signal. CONCLUSION: The LKB1/AMPK pathway might relay the signal between the Wnt/ß-catenin pathway and the Ku70/Ku80 DNA repair machinery, and play a decisive role in fine-tuning the responses of cancer cells to irradiation. © 2015 Wiley Periodicals, Inc. Head Neck 38: E1909-E1917, 2016.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Inativação Gênica , Neoplasias de Cabeça e Pescoço/radioterapia , Autoantígeno Ku/metabolismo , Tolerância a Radiação , beta Catenina/genética , Quinases Proteína-Quinases Ativadas por AMP , Linhagem Celular Tumoral , Neoplasias de Cabeça e Pescoço/genética , Humanos , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno , Via de Sinalização Wnt
16.
Antioxid Redox Signal ; 24(9): 471-85, 2016 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-26573462

RESUMO

AIMS: Cellular senescence and its secretory phenotype (senescence-associated secretory phenotype [SASP]) develop after long-term expansion of mesenchymal stromal cells (MSCs). Further investigation of this phenotype is required to improve the therapeutic efficacy of MSC-based cell therapies. In this study, we show that positive feedback between SASP and inherent senescence processes plays a crucial role in the senescence of umbilical cord blood-derived MSCs (UCB-MSCs). RESULTS: We found that monocyte chemoattractant protein-1 (MCP-1) was secreted as a dominant component of the SASP during expansion of UCB-MSCs and reinforced senescence via its cognate receptor chemokine (c-c motif) receptor 2 (CCR2) by activating the ROS-p38-MAPK-p53/p21 signaling cascade in both an autocrine and paracrine manner. The activated p53 in turn increased MCP-1 secretion, completing a feed-forward loop that triggered the senescence program in UCB-MSCs. Accordingly, knockdown of CCR2 in UCB-MSCs significantly improved their therapeutic ability to alleviate airway inflammation in an experimental allergic asthma model. Moreover, BMI1, a polycomb protein, repressed the expression of MCP-1 by binding to its regulatory elements. The reduction in BMI1 levels during UCB-MSC senescence altered the epigenetic status of MCP-1, including the loss of H2AK119Ub, and resulted in derepression of MCP-1. INNOVATION: Our results provide the first evidence supporting the existence of the SASP as a causative contributor to UCB-MSC senescence and reveal a so far unappreciated link between epigenetic regulation and SASP for maintaining a stable senescent phenotype. CONCLUSION: Senescence of UCB-MSCs is orchestrated by MCP-1, which is secreted as a major component of the SASP and is epigenetically regulated by BMI1.


Assuntos
Quimiocina CCL2/metabolismo , Células-Tronco Mesenquimais/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Animais , Asma/tratamento farmacológico , Asma/genética , Asma/imunologia , Asma/metabolismo , Comunicação Autócrina , Células Cultivadas , Senescência Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Sangue Fetal/citologia , Humanos , Estresse Oxidativo , Comunicação Parácrina , Fenótipo , Análise Serial de Proteínas , Ligação Proteica , Receptores CCR2/genética , Receptores CCR2/metabolismo , Transcrição Gênica , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
Toxicol Lett ; 155(1): 65-71, 2005 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-15585361

RESUMO

Asbestos is a very important material for industrial use. However, the need for a substitute for asbestos fiber is currently on the rise due to its high disease causing potential. This study evaluated the potential bio-hazardous effects of TAFMAG, a natural fibrous silicate produced in China, in comparison with chrysotile, a typical toxic asbestos. The physicochemical properties of TAFMAG were very similar to those of chrysotile when it was examined by a scanning electron microscope (SEM) and X-ray diffraction (XRD) analyses. Both of TAFMAG and chrysotile showed high content of magnetite and Fenton activity when compared with wollastonite, a non-asbestos fiber with a known low toxicity. When their cellular toxicity was assessed, TAFMAG showed no or less comparable to that of chrysotile in the hemolysis and lipid peroxidation of erythrocytes, and also on a MTT assay in RLE-6TN, a rat alveolar epithelial cell line. Pre-treatment of fibers with desferrioxamine, an iron chelator, showed that iron content of TAFMAG and chrysotile might be important in their cellular toxicity. These results suggest that TAFMAG is potentially toxic when inhaled into the lung and appropriate laws and regulations should be established for its use.


Assuntos
Fibras Minerais/toxicidade , Silicatos/toxicidade , Animais , Asbestos Serpentinas/toxicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Fenômenos Químicos , Físico-Química , Células Epiteliais/efeitos dos fármacos , Membrana Eritrocítica/efeitos dos fármacos , Eritrócitos/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Peróxido de Hidrogênio , Técnicas In Vitro , Ferro , Quelantes de Ferro/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Tamanho da Partícula , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/efeitos dos fármacos , Ratos , Sais de Tetrazólio , Tiazóis
18.
Toxicol Lett ; 155(2): 337-42, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15603929

RESUMO

Diesel exhaust particles (DEPs) are known to induce allergic responses in airway epithelial cells, such as the production of various cytokines via nuclear factor-kappa B (NF-kappaB). However, the intracellular signal transduction pathways underlying this phenomenon have not been fully examined. This study showed that DEP induced NF-kappaB activity via transforming growth factor-beta activated kinase 1 (TAK1) and NF-kappaB-inducing kinase (NIK) in L2 rat lung epithelial cells. DEP induced the NF-kB dependent reporter activity approximately two- to three-fold in L2 cells. However, this effect was abolished by the expression of the dominant negative forms of TAK1 or NIK. Furthermore, it was shown that DEP induced TAK1 phosphorylation in the L2 cells. These results suggest that TAK1 and NIK are important mediators of DEP-induced NF-kappaB activation.


Assuntos
Células Epiteliais/efeitos dos fármacos , MAP Quinase Quinase Quinases/metabolismo , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Emissões de Veículos/toxicidade , Animais , Linhagem Celular , Células Epiteliais/enzimologia , Células Epiteliais/metabolismo , Tamanho da Partícula , Fosforilação , Ratos , Emissões de Veículos/análise , Quinase Induzida por NF-kappaB
19.
J Environ Pathol Toxicol Oncol ; 24(3): 163-74, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16050801

RESUMO

Silica is a causative factor of acute cell injury in pulmonary fibrosis. Inducible cyclooxygenase-2 (COX-2) was suggested to play a role in the process of inflammation and fibrosis. We report that silica induces COX-2 expression in WI-38 fibroblasts. Further analysis showed that silica activated the transcription of COX-2 gene primarily via a nuclear factor (NF)-kB binding site in the promoter. NF-kB-inducing kinase (NIK) and TGF-k activated kinase 1 (TAK1), the upstream signaling molecules of NF-kB, are involved in the silica-mediated COX-2 expression. The Electrophoretic Mobility Shift Assay (EMSA) showed that silica induced the direct binding of NF-kB on the putative binding site in COX-2 promoter. These results suggest that silica activates the human COX-2 gene transcription through the induction of NF-kB activity.


Assuntos
Fibroblastos/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , NF-kappa B/metabolismo , Prostaglandina-Endoperóxido Sintases/genética , Dióxido de Silício/toxicidade , Sítios de Ligação/efeitos dos fármacos , Linhagem Celular , Ciclo-Oxigenase 2 , Relação Dose-Resposta a Droga , Indução Enzimática , Fibroblastos/enzimologia , Humanos , MAP Quinase Quinase Quinases/metabolismo , Proteínas de Membrana , Prostaglandina-Endoperóxido Sintases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Quinase Induzida por NF-kappaB
20.
Nat Commun ; 6: 7930, 2015 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-26246279

RESUMO

For therapeutic applications of siRNA, there are technical challenges with respect to targeted and systemic delivery. We here report a new siRNA carrier, RNAtr NPs, in a way that multiple tandem copies of RNA hairpins as a result of rolling circle transcription (RCT) can be readily adapted in tumour-targeted and systemic siRNA delivery. RNAtr NPs provide a means of condensing large amounts of multimeric RNA transcripts into the compact nanoparticles, especially without the aid of polycationic agents, and thus reduce the risk of immunogenicity and cytotoxicity by avoiding the use of synthetic polycationic reagents. This strategy allows the design of a platform technology for systemic delivery of siRNA to tumour sites, because RCT reaction, which enzymatically generates RNA polymers in multiple copy numbers at low cost, can lead to directly accessible routes to targeted and systemic delivery. Therefore, RNAtr NPs suggest great potentials as the siRNA therapeutics for cancer treatment.


Assuntos
DNA/química , Sistemas de Liberação de Medicamentos , Nanopartículas , RNA Interferente Pequeno/administração & dosagem , RNA/química , Animais , Colesterol/química , Feminino , Ácido Fólico/química , Inativação Gênica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Transcrição Gênica , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA