Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Neurochem ; 164(5): 643-657, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36527420

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that selectively attacks motor neurons, and leads to progressive muscle weakness and death. A common pathological feature is the misfolding, aggregation, and cytoplasmic mislocalization of TAR DNA-binding protein 43 (TDP-43) proteins in more than 95% of ALS patients, suggesting a universal role TDP-43 proteinopathy in ALS. Mutations in SQSTM1/p62 have been identified in familial and sporadic cases of ALS. MicroRNAs (miRNAs) are small non-coding RNAs that post-transcriptionally regulate their target genes. Emerging evidence indicates that miRNA dysregulation is associated with neuronal toxicity and mitochondrial dysfunction, and also plays a pivotal role in ALS pathogenesis. Here, we report the first evidence that miR-183-5p is aberrantly upregulated in spinal cords of patients with ALS. Using luciferase reporter assays and miR-183-5p agomirs, we demonstrate that miR-183-5p regulates the SQSTM1/p62 3'-untranslated region to suppress expression. A miR-183-5p agomir attenuated SOSTM1/p62 expression and led to an increase in TDP-43 protein levels in neuronal and non-neuronal cells. In contrast, a miR-183-5p antagomir decreased TDP-43 but increased SQSTM1/p62 protein levels. The antagomir repressed formation of stress granules and aggregated TDP43 protein in neuronal cells under stress-induced conditions and protected against cytotoxicity. Knockdown of SQSTM1/p62 decreased total ubiquitination and increased TDP-43 protein aggregation, indicating that SQSTM1/p62 may play a protective role in cells. In summary, our study reveals a novel mechanism of TDP-43 proteinopathy mediated by the miR-183-5p and provides a molecular link between aberrant RNA processing and protein degradation, two major pillars in ALS pathogenesis.


Assuntos
Esclerose Lateral Amiotrófica , MicroRNAs , Doenças Neurodegenerativas , Humanos , Esclerose Lateral Amiotrófica/metabolismo , Proteína Sequestossoma-1/metabolismo , Doenças Neurodegenerativas/metabolismo , Antagomirs/metabolismo , Neurônios Motores/metabolismo , MicroRNAs/metabolismo , Proteínas de Ligação a DNA/metabolismo
2.
Mol Cell ; 52(2): 193-205, 2013 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-24055345

RESUMO

Fine control of Wnt signaling is essential for various cellular and developmental decision-making processes. However, deregulation of Wnt signaling leads to pathological consequences, one of which is cancer. Here, we identify a function of PAF, a component of translesion DNA synthesis, in modulating Wnt signaling. PAF is specifically overexpressed in colon cancer cells and intestinal stem cells and is required for colon cancer cell proliferation. In Xenopus laevis, ventrovegetal expression of PAF hyperactivates Wnt signaling, developing a secondary axis with ß-catenin target gene upregulation. Upon Wnt signaling activation, PAF dissociates from PCNA and binds directly to ß-catenin. Then, PAF recruits EZH2 to the ß-catenin transcriptional complex and specifically enhances Wnt target gene transactivation, independently of EZH2's methyltransferase activity. In mice, conditional expression of PAF induces intestinal neoplasia via Wnt signaling hyperactivation. Our studies reveal an unexpected role of PAF in regulating Wnt signaling and propose a regulatory mechanism of Wnt signaling during tumorigenesis.


Assuntos
Proteínas de Transporte/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Proliferação de Células , Células Cultivadas , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Proteínas de Ligação a DNA , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste , Regulação da Expressão Gênica no Desenvolvimento , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Immunoblotting , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Complexo Repressor Polycomb 2/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Xenopus laevis , beta Catenina/genética
3.
Nano Lett ; 17(5): 2913-2920, 2017 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-28418672

RESUMO

Ruthenium coordination complexes have the potential to serve as novel theranostic agents for cancer. However, a major limitation in their clinical implementation is effective tumor accumulation. In this study, we have developed a liposome-based theranostic nanodelivery system for [Ru(phen)2dppz](ClO4)2 (Lipo-Ru). This ruthenium polypyridine complex emits a strong fluorescent signal when incorporated in the hydrophobic lipid bilayer of the delivery vehicle or in the DNA helix, enabling visualization of the therapeutic agent in tumor tissues. Incubation of MDA-MB-231 breast cancer cells with Lipo-Ru induced double-strand DNA breaks and triggers apoptosis. In a mouse model of triple-negative breast cancer, treatment with Lipo-Ru dramatically reduced tumor growth. Biodistribution studies of Lipo-Ru revealed that more than 20% of the injected dose accumulated in the tumor. These results suggest that Lipo-Ru could serve as a promising theranostic platform for cancer.


Assuntos
Antineoplásicos/química , Complexos de Coordenação/química , Portadores de Fármacos/química , Compostos Heterocíclicos com 3 Anéis/química , Compostos Heterocíclicos de 4 ou mais Anéis/química , Piridinas/química , Rutênio/química , Neoplasias de Mama Triplo Negativas/diagnóstico por imagem , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Complexos de Coordenação/administração & dosagem , Feminino , Humanos , Lipossomos , Camundongos , Camundongos Nus , Nanomedicina Teranóstica , Distribuição Tecidual
4.
J Cell Biochem ; 113(5): 1547-58, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22174016

RESUMO

Colorectal cancer is the third leading cause of cancer-related mortality in the world; death usually results from uncontrolled metastatic disease. Previously, we developed a novel strategy of TNF-related apoptosis-inducing ligand (Apo2L/TRAIL) in combination with hyperthermia to treat hepatic colorectal metastases. However, previous studies suggest a potential hepatocyte cytotoxicity with TRAIL. Unlike TRAIL, anti-human TRAIL receptor antibody induces apoptosis without hepatocyte toxicity. In this study, we evaluated the anti-tumor efficacy of humanized anti-death receptor 4 (DR4) antibody mapatumumab (Mapa) by comparing it with TRAIL in combination with hyperthermia. TRAIL, which binds to both DR4 and death receptor 5 (DR5), was approximately tenfold more effective than Mapa in inducing apoptosis. However, hyperthermia enhances apoptosis induced by either agent. We observed that the synergistic effect was mediated through elevation of reactive oxygen species, c-Jun N-terminal kinase activation, Bax oligomerization, and translocalization to the mitochondria, loss of mitochondrial membrane potential, release of cytochrome c to cytosol, activation of caspases, and increase in poly(ADP-ribose) polymerase cleavage. We believe that the successful outcome of this study will support the application of Mapa in combination with hyperthermia to colorectal hepatic metastases.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , Ligante Indutor de Apoptose Relacionado a TNF/administração & dosagem , Anticorpos Monoclonais Humanizados , Apoptose , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Terapia Combinada , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Hipertermia Induzida , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/terapia , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Proteína X Associada a bcl-2/deficiência , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
5.
J Cell Biochem ; 112(2): 548-58, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21268076

RESUMO

We identified a chalcone, 2',4'-dihydroxy-6'-methoxy-3'-methylchalcone (stercurensin), as an active compound isolated from the leaves of Syzygium samarangense. In the present study, the anti-inflammatory effects and underlying mechanisms of stercurensin were examined using lipopolysaccharide (LPS)-stimulated RAW264.7 cells and mice. To determine the effects of stercurensin in vitro, inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) expression were analyzed by RT-PCR and immunoblotting. Nuclear factor-κB (NF-κB) activation and its upstream signaling cascades were also investigated using a dual-luciferase reporter assay, electrophoretic mobility shift assay, immunoblotting, immunofluorescence, and immunoprecipitation. To verify the effects of stercurensin in vivo, the mRNA expression levels of iNOS and COX-2 were evaluated in isolated mouse peritoneal macrophages by quantitative real-time PCR, and the production of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and IL-1ß were assessed in serum samples from mice using a Luminex system. Pretreatment with stercurensin reduced LPS-induced iNOS and COX-2 expression, thereby inhibiting nitric oxide (NO) and prostaglandin E(2) production, respectively. In addition, an inhibitory effect of stercurensin on NF-κB activation was shown by the recovery of LPS-induced inhibitor of κB (I-κB) degradation after blocking the transforming growth factor-ß-activated kinase 1 (TAK1)/I-κB kinase signaling pathway. In mouse models, stercurensin negatively regulated NF-κB-dependent pro-inflammatory mediators and cytokines. These results demonstrate that stercurensin modulates NF-κB-dependent inflammatory pathways through the attenuation of TAK1-TAB1 complex formation. Our findings demonstrating the anti-inflammatory effects of stercurensin in vitro and in vivo will aid in understanding the pharmacology and mode of action of stercurensin.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Chalcona/farmacologia , MAP Quinase Quinase Quinases/metabolismo , NF-kappa B/metabolismo , Animais , Western Blotting , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Chalcona/química , Ensaio de Desvio de Mobilidade Eletroforética , Imunoprecipitação , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Microscopia Confocal , Óxido Nítrico/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
6.
J Cell Biochem ; 112(9): 2471-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21538483

RESUMO

Dimethyl cardamonin (2',4'-dihydroxy-6'-methoxy-3',5'-dimethylchalcone; DMC) is a naturally occurring chalcone, and it is the major compound isolated from the leaves of Syzygium samarangense (Blume) Merr. & L.M. Perry (Myrtaceae). Experiments were conducted to determine the effects of DMC on cell proliferation, cell-cycle distribution, and programmed cell death in cultures of human colorectal carcinoma HCT116 and LOVO cells. Results showed that DMC inhibited HCT116 and LOVO cell proliferation and induced G(2) /M cell cycle arrest, which was associated with the conversion of microtubule associated protein light chain 3 (LC3)-I-LC3-II, an autophagosome marker, and the incorporation of monodansylcadaverine (MDC), a marker for the acidic compartment of autolysosomes or acidic vesicular organelles. The treatment of HCT116 and LOVO cells using a combination of DMC with an autophagy inhibitor, such as 3-methyladenine (3-MA), beclin 1 siRNA, or atg5 siRNA, suppressed the effect of DMC-mediated anti-proliferation. These results imply that DMC can suppress colorectal carcinoma HCT116 and LOVO cell proliferation through a G(2) /M phase cell-cycle delay, and can induce autophagy, the hallmark of Type II programmed cell death (PCD). Taken together, our results suggest that DMC may be an effective chemotherapeutic agent for HCT116 and LOVO colorectal carcinoma cells.


Assuntos
Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Chalconas/farmacologia , Adenina/análogos & derivados , Adenina/farmacologia , Apoptose , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 5 Relacionada à Autofagia , Proteína Beclina-1 , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Neoplasias Colorretais , Expressão Gênica , Células HCT116 , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Interferência de RNA
7.
Cell Mol Life Sci ; 67(20): 3499-510, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20473547

RESUMO

We identified CREB3 as a novel HDAC3-interacting protein in a yeast two-hybrid screen for HDAC3-interacting proteins. Among all class I HDACs, CREB3 specifically interacts with HDAC3, in vitro and in vivo. HDAC3 efficiently inhibited CREB3-enhanced NF-κB activation, whereas the other class I HDACs did not alter NF-κB-dependent promoter activities or the expression of NF-κB target genes. Importantly, both knock-down of CREB3 and overexpression of HDAC3 suppressed the transcriptional activation of the novel CREB3-regulated gene, CXCR4. Furthermore, CREB3 was shown to bind to the CRE element in the CXCR4 promoter and to activate the transcription of the CXCR4 gene by causing dissociation of HDAC3 and subsequently increasing histone acetylation. Importantly, both the depletion of HDAC3 and the overexpression of CREB3 substantially increased the migration of MDA-MB-231 metastatic breast cancer cells. Taken together, these findings suggest that HDAC3 selectively represses CREB3-mediated transcriptional activation and chemotactic signalling in human metastatic breast cancer cells.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Movimento Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Histona Desacetilases/metabolismo , Proteínas Repressoras/metabolismo , Transcrição Gênica , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/química , Regulação para Baixo/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Histona Desacetilases/química , Humanos , NF-kappa B/metabolismo , Metástase Neoplásica/genética , Metástase Neoplásica/patologia , Ligação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Receptores CXCR4/genética , Receptores CXCR4/metabolismo
8.
Mol Endocrinol ; 22(5): 1093-104, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18202150

RESUMO

Lis-homology (LisH) motifs are involved in protein dimerization, and the discovery of the conserved N-terminal LisH domain in transducin beta-like protein 1 and its receptor (TBL1 and TBLR1) led us to examine the role of this domain in transcriptional repression. Here we show that multiple beta-transducin (WD-40) repeat-containing proteins interact to form oligomers in solution and that oligomerization depends on the presence of the LisH domain in each protein. Repression of transcription, as assayed using Gal4 fusion proteins, also depended on the presence of the LisH domain, suggesting that oligomerization is a prerequisite for efficient transcriptional repression. Furthermore, we show that the LisH domain is responsible for the binding to the hypoacetylated histone H4 tail and for stable chromatin targeting by the nuclear receptor corepressor complex. Mutations in conserved residues in the LisH motif of TBL1 and TBLR1 block histone binding, oligomerization, and transcriptional repression, supporting the functional importance of the LisH motif in transcriptional repression. Our results indicate that another WD-40 protein, TBL3, also preferentially binds to the N-terminal domain of TBL1 and TBLR1, and forms oligomers with other WD-40 proteins. Finally, we observed that the WD-40 proteins RbAp46 and RbAp48 of the sin3A corepressor complex failed to dimerize. We also found the specific interaction UbcH/E2 with TBL1, but not RbAp46/48. Altogether, our results thus indicate that the presence of multiple LisH/WD-40 repeat containing proteins is exclusive to nuclear receptor corepressor/ silencing mediator for retinoic and thyroid receptor complexes compared with other class 1 histone deacetylase-containing corepessor complexes.


Assuntos
Proteínas Nucleares/metabolismo , Receptores do Ácido Retinoico/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo , Proteínas Repressoras/metabolismo , Transcrição Gênica , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Sítios de Ligação/genética , Western Blotting , Linhagem Celular , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Análise Mutacional de DNA , Células HeLa , Histonas/metabolismo , Humanos , Imunoprecipitação , Proteínas Nucleares/genética , Correpressor 1 de Receptor Nuclear , Ligação Proteica , Receptores do Ácido Retinoico/genética , Receptores dos Hormônios Tireóideos/genética , Sequências Repetitivas de Aminoácidos/genética , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos
9.
Clin Cancer Res ; 24(13): 3176-3185, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29602802

RESUMO

Purpose: Understanding the mechanism of radioresistance could help develop strategies to improve therapeutic response of patients with PDAC. The SMAD4 gene is frequently mutated in pancreatic cancer. In this study, we investigated the role of SMAD4 deficiency in pancreatic cancer cells' response to radiotherapy.Experimental Design: We downregulated SMAD4 expression with SMAD4 siRNA or SMAD4 shRNA and overexpressed SMAD4 in SMAD4 mutant pancreatic cancer cells followed by clonogenic survival assay to evaluate their effects on cell radioresistance. To study the mechanism of radioresistance, the effects of SMAD4 loss on reactive oxygen species (ROS) and autophagy were determined by flow cytometry and immunoblot analysis, respectively. Furthermore, we measured radioresistance by clonogenic survival assay after treatment with autophagy inhibitor (Chloroquine) and ROS inhibitor (N-acetyl-l-cysteine) in SMAD4-depleted pancreatic cancer cells. Finally, the effects of SMAD4 on radioresistance were also confirmed in an orthotopic tumor model derived from SMAD4-depleted Panc-1 cells.Results:SMAD4-depleted pancreatic cancer cells were more resistant to radiotherapy based on clonogenic survival assay. Overexpression of wild-type SMAD4 in SMAD4-mutant cells rescued their radiosensitivity. Radioresistance mediated by SMAD4 depletion was associated with persistently higher levels of ROS and radiation-induced autophagy. Finally, SMAD4 depletion induced in vivo radioresistance in Panc-1-derived orthotopic tumor model (P = 0.038). More interestingly, we observed that the protein level of SMAD4 is inversely correlated with autophagy in orthotopic tumor tissue samples.Conclusions: Our results demonstrate that defective SMAD4 is responsible for radioresistance in pancreatic cancer through induction of ROS and increased level of radiation-induced autophagy. Clin Cancer Res; 24(13); 3176-85. ©2018 AACR.


Assuntos
Autofagia/genética , Autofagia/efeitos da radiação , Mutação , Neoplasias Pancreáticas/genética , Tolerância a Radiação/genética , Proteína Smad4/genética , Animais , Antineoplásicos/farmacologia , Apoptose/genética , Apoptose/efeitos da radiação , Biomarcadores Tumorais , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Dano ao DNA , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Instabilidade Genômica , Humanos , Imuno-Histoquímica , Camundongos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/radioterapia , Espécies Reativas de Oxigênio/metabolismo , Proteína Smad4/metabolismo
10.
Biosci Biotechnol Biochem ; 71(11): 2712-9, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17986787

RESUMO

Histone acetylation depends on the activity of two enzyme families, histone acetyltransferase (HAT) and deacetylase (HDAC). In this study, we screened various plant extracts to find potent HAT inhibitors. Hot water extracts of allspice inhibited HAT activity, especially p300 and CBP (40% at 100 microg/ml). The mRNA levels of two androgen receptor (AR) regulated genes, PSA and TSC22, decreased with allspice treatment (100 microg/ml). Importantly, in IP western analysis, AR acetylation was dramatically decreased by allspice treatment.Furthermore, chromatin immunoprecipitation indicated that the acetylation of histone H3 in the PSA and B2M promoter regions was also repressed. Finally, allspice treatment reduced the growth of human prostate cancer cells, LNCaP (50% growth inhibition at 200 microg/ml). Taken together, our data indicate that the potent HAT inhibitory activity of allspice reduced AR and histone acetylation and led to decreased transcription of AR target genes, resulting in inhibition of prostate cancer cell growth.


Assuntos
Antagonistas de Receptores de Andrógenos , Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Histona Acetiltransferases/antagonistas & inibidores , Pimenta , Neoplasias da Próstata/enzimologia , Acetilação , Carboxipeptidase B/antagonistas & inibidores , Carboxipeptidase B/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Histonas/metabolismo , Humanos , Masculino , Extratos Vegetais/farmacologia , Receptores Androgênicos/metabolismo , Ativação Transcricional/efeitos dos fármacos , Fatores de Transcrição de p300-CBP/antagonistas & inibidores , Fatores de Transcrição de p300-CBP/genética
11.
Nanoscale ; 9(16): 5329-5341, 2017 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-28398453

RESUMO

Drug to carrier ratio is an important consideration in designing drug platforms, since a low loading capacity necessitates the use of high doses of carriers, which can result in side effects. Here, we have engineered a platform to co-deliver small molecule drugs and small interfering RNA (siRNA). This platform consists of cyclodextrin-grafted polyethylenimine (CP) functionalized mesoporous silica nanoparticles (MSNP). A unique multi-step encapsulation procedure was used to obtain a high loading capacity for doxorubicin (DOX) and siRNA oligos specific for the PKM2 gene that encodes pyruvate kinase M2, an enzyme catalyzing the final rate-limiting step in glycolysis. We systematically characterized this platform (CP-MSNP@DOX/PKM2) in vitro and evaluated its therapeutic efficacy in vivo with a mouse model of triple negative breast cancer (TNBC). Exposure of TNBC cells to CP-MSNP@DOX/PKM2 resulted in suppressed target gene expression, reduced cell proliferation, and enhanced apoptosis. Intravenous administration of the drug substantially decreased the tumor burden in comparison to DOX or siRNA monotherapy. In conclusion, we have developed a platform for efficient co-delivery of small molecule drugs and therapeutic siRNA.


Assuntos
Doxorrubicina/administração & dosagem , Portadores de Fármacos , Inativação Gênica , Nanopartículas , Dióxido de Silício , Animais , Antineoplásicos , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Nus , RNA Interferente Pequeno
12.
Adv Healthc Mater ; 3(10): 1629-37, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24692076

RESUMO

Cancer is a complex disease that usually requires several treatment modalities. A multifunctional nanotherapeutic system is designed, incorporating small interfering RNA (siRNA) and gold nanorods (Au NRs) for photothermal therapy. Surface-engineered Au NRs with polyethylenimine are synthesized using a layer-by-layer assembly and siRNA is absorbed on the surface. The siRNA is efficiently delivered into breast cancer cells, resulting in subsequent gene silencing. Cells are then irradiated with near-infrared (NIR) light, causing heat-induced anticancer activity. The combination of gene silencing and photothermal therapy results in effective inhibition of cell proliferation.


Assuntos
Neoplasias da Mama/terapia , Terapia Genética/métodos , Ouro/química , Hipertermia Induzida/métodos , Nanotubos/química , Fototerapia/métodos , RNA Interferente Pequeno/administração & dosagem , Animais , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Feminino , Inativação Gênica , Ouro/administração & dosagem , Humanos , Macrófagos/metabolismo , Camundongos , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética
13.
Theranostics ; 4(5): 487-97, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24672582

RESUMO

Effective delivery holds the key to successful in vivo application of therapeutic small interfering RNA (siRNA). In this work, we have developed a universal siRNA carrier consisting of a mesoporous silica nanoparticle (MSNP) functionalized with cyclodextrin-grafted polyethylenimine (CP). CP provides positive charge for loading of siRNA through electrostatic interaction and enables effective endosomal escape of siRNA. Using intravital microscopy we were able to monitor tumor enrichment of CP-MSNP/siRNA particles in live mice bearing orthotopic MDA-MB-231 xenograft tumors. CP-MSNP delivery of siRNA targeting the M2 isoform of the glycolytic enzyme pyruvate kinase (PKM2) resulted in effective knockdown of gene expression in vitro and in vivo. Suppression of PKM2 led to inhibition of tumor cell growth, invasion, and migration.


Assuntos
Antineoplásicos/uso terapêutico , Produtos Biológicos/uso terapêutico , Portadores de Fármacos/administração & dosagem , Nanopartículas/administração & dosagem , Neoplasias/terapia , RNA Interferente Pequeno/uso terapêutico , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Produtos Biológicos/farmacocinética , Produtos Biológicos/farmacologia , Ciclodextrinas/administração & dosagem , Modelos Animais de Doenças , Feminino , Técnicas de Silenciamento de Genes , Camundongos , Polietilenoimina/administração & dosagem , Piruvato Quinase/antagonistas & inibidores , RNA Interferente Pequeno/farmacocinética , RNA Interferente Pequeno/farmacologia , Dióxido de Silício/administração & dosagem
14.
Cell Rep ; 5(2): 314-22, 2013 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-24209743

RESUMO

Deregulation of mitogen-activated protein kinase (MAPK) signaling leads to development of pancreatic cancer. Although Ras-mutation-driven pancreatic tumorigenesis is well understood, the underlying mechanism of Ras-independent MAPK hyperactivation remains elusive. Here, we have identified a distinct function of PCNA-associated factor (PAF) in modulating MAPK signaling. PAF is overexpressed in pancreatic cancer and required for pancreatic cancer cell proliferation. In mouse models, PAF expression induced pancreatic intraepithelial neoplasia with expression of pancreatic cancer stem cell markers. PAF-induced ductal epithelial cell hyperproliferation was accompanied by extracellular signal-regulated kinase (ERK) phosphorylation independently of Ras or Raf mutations. Intriguingly, PAF transcriptionally activated the expression of late endosomal/lysosomal adaptor, MAPK and mTOR activator 3 (LAMTOR3), which hyperphosphorylates MEK and ERK and is necessary for pancreatic cancer cell proliferation. Our results reveal an unsuspected mechanism of mitogenic signaling activation via LAMTOR3 and suggest that PAF-induced MAPK hyperactivation contributes to pancreatic tumorigenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Transporte/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Carcinogênese , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Camundongos , Células-Tronco Neoplásicas/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Fosforilação , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Ativação Transcricional , Quinases raf/genética , Quinases raf/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
15.
ACS Nano ; 7(11): 9867-80, 2013 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-24131405

RESUMO

Gene silencing agents such as small interfering RNA (siRNA) and microRNA offer the promise to modulate expression of almost every gene for the treatment of human diseases including cancer. However, lack of vehicles for effective systemic delivery to the disease organs has greatly limited their in vivo applications. In this study, we developed a high capacity polycation-functionalized nanoporous silicon (PCPS) platform comprised of nanoporous silicon microparticles functionalized with arginine-polyethyleneimine inside the nanopores for effective delivery of gene silencing agents. Incubation of MDA-MB-231 human breast cancer cells with PCPS loaded with STAT3 siRNA (PCPS/STAT3) or GRP78 siRNA (PCPS/GRP78) resulted in 91 and 83% reduction of STAT3 and GRP78 gene expression in vitro. Treatment of cells with a microRNA-18a mimic in PCPS (PCPS/miR-18) knocked down 90% expression of the microRNA-18a target gene ATM. Systemic delivery of PCPS/STAT3 siRNA in murine model of MDA-MB-231 breast cancer enriched particles in tumor tissues and reduced STAT3 expression in cancer cells, causing significant reduction of cancer stem cells in the residual tumor tissue. At the therapeutic dosage, PCPS/STAT3 siRNA did not trigger acute immune response in FVB mice, including changes in serum cytokines, chemokines, and colony-stimulating factors. In addition, weekly dosing of PCPS/STAT3 siRNA for four weeks did not cause signs of subacute toxicity based on changes in body weight, hematology, blood chemistry, and major organ histology. Collectively, the results suggest that we have developed a safe vehicle for effective delivery of gene silencing agents.


Assuntos
Inativação Gênica , MicroRNAs/genética , Nanomedicina/métodos , RNA Interferente Pequeno/genética , Animais , Arginina/química , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Chaperona BiP do Retículo Endoplasmático , Feminino , Perfilação da Expressão Gênica , Proteínas de Choque Térmico/genética , Humanos , Cinética , Neoplasias Mamárias Experimentais/terapia , Camundongos , Microscopia Eletrônica de Varredura , Nanopartículas/química , Polietilenoimina/química , RNA Interferente Pequeno/metabolismo , Fator de Transcrição STAT3/genética , Silício/química
16.
Food Chem Toxicol ; 50(3-4): 1027-35, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22155221

RESUMO

Aurentiacin is a chalcone isolated from Syzygium samarangense. In the present study, we examined the anti-inflammatory effects of aurentiacin in lipopolysaccharide (LPS)-stimulated mouse macrophages. Aurentiacin significantly inhibited LPS-induced nitric oxide (NO) production in RAW264.7 cells concomitantly with the suppression of inducible nitric oxide synthase (iNOS) expression. Aurentiacin also reduced the mRNA levels of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Electrophoretic mobility shift assays (EMSAs) and reporter gene assays indicated that DNA binding and transcriptional activities of nuclear factor-κB (NF-κB)/p65 were decreased by aurentiacin in LPS-stimulated RAW264.7 cells. Moreover, results from chromatin immunoprecipitation (ChIP) assays over the promoter region of the iNOS gene were in agreement with the EMSA results. Pretreatment with aurentiacin prevented the nuclear translocation of p65 by blocking the phosphorylation of I-κB kinase (IKK). Aurentiacin also attenuated the phosphorylation (Ser536) and acetylation (Lys310) of p65 and phosphorylation of MAPKs. In an inflammatory animal model, the intraperitoneal (i.p.) injection of aurentiacin suppressed the release of pro-inflammatory cytokines. Moreover, the level of iNOS protein ex vivo was decreased by aurentiacin similar to the result in vitro. Taken together, these results suggest that aurentiacin shows anti-inflammatory activity related to the inhibition of NF-κB activation.


Assuntos
Chalconas/uso terapêutico , Inflamação/tratamento farmacológico , Lipopolissacarídeos/antagonistas & inibidores , Macrófagos Peritoneais/efeitos dos fármacos , Syzygium/química , Animais , Western Blotting , Linhagem Celular , Chalconas/farmacologia , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Quinase I-kappa B/metabolismo , Inflamação/induzido quimicamente , Inflamação/imunologia , Interleucina-5/genética , Lipopolissacarídeos/toxicidade , Macrófagos Peritoneais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/genética , Fosforilação , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Fator de Necrose Tumoral alfa/genética
17.
FEBS Lett ; 583(12): 1880-6, 2009 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-19481544

RESUMO

In this study, we discovered that NSD2 specifically interacts with the DNA-binding domain of androgen receptor (AR) via its HMG domain, and the nuclear translocation of both NSD2 and AR is enhanced in the presence of ligand. Furthermore, we also demonstrated that the over expression of NSD2, but not of NSD2 (DeltaSET) HMT-activity defective mutant, enhanced the mRNA level of PSA in a dose-dependent manner. A chromatin immunoprecipitation assay showed that NSD2 protein is recruited to the enhancer region of the PSA gene by AR in an agonist-enhanced manner. Taken together, these results uncover a potential role for NSD2 in AR-mediated transcription, implicating NSD2 in prostate carcinogenesis.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , Receptores Androgênicos/metabolismo , Proteínas Repressoras/metabolismo , Transporte Ativo do Núcleo Celular , Sequência de Bases , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Primers do DNA/genética , Elementos Facilitadores Genéticos , Histona-Lisina N-Metiltransferase/química , Histona-Lisina N-Metiltransferase/genética , Histonas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Ligantes , Masculino , Antígeno Prostático Específico/genética , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/etiologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Domínios e Motivos de Interação entre Proteínas , Receptores Androgênicos/química , Receptores Androgênicos/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA