Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Gynecol Oncol ; 156(1): 211-221, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31776040

RESUMO

OBJECTIVE: Cyclin-dependent kinase 7 (CDK7) engages tumor growth by acting as a direct link between the regulation of transcription and the cell cycle. Here, we investigated the clinical significance of CDK7 expression and its potential as a therapeutic target in epithelial ovarian cancer (EOC). METHODS: CDK7 expression was examined in 436 ovarian tissues including normal to metastatic ovarian tumors using immunohistochemistry, and its clinical implications were analyzed. Furthermore, we performed in vitro and in vivo experiments using CDK7 siRNA or a covalent CDK7 inhibitor (THZ1) to elucidate the effect of CDK7 inhibition on tumorigenesis in EOC cells. RESULTS: The patient incidence of high CDK7 expression (CDK7High) gradually increased from normal ovarian epithelium to EOC (P < 0.001). Moreover, CDK7High was associated with an advanced stage and high-grade histology (P = 0.035 and P = 0.011, respectively) in EOC patients and had an independent prognostic significance in EOC recurrence (P = 0.034). CDK7 inhibition with siRNA or THZ1 decreased cell proliferation and migration, and increased apoptosis in EOC cells, and this anti-cancer mechanism is caused by G0/G1 cell cycle arrest. In in vivo therapeutic experiments using cell-line xenograft and PDX models, CDK7 inhibition significantly decreased the tumor weight, which was mediated by cell proliferation and apoptosis. CONCLUSION: Mechanistic interrogation of CDK7 revealed that it is significantly associated with an aggressive phenotype of EOC, and it has independent prognostic power for EOC recurrence. Furthermore, CDK7 may be a potential therapeutic target for patients with EOC, whether platinum sensitive or resistant.


Assuntos
Carcinoma Epitelial do Ovário/enzimologia , Quinases Ciclina-Dependentes/biossíntese , Neoplasias Ovarianas/enzimologia , Animais , Biomarcadores Tumorais/antagonistas & inibidores , Biomarcadores Tumorais/biossíntese , Carcinoma Epitelial do Ovário/tratamento farmacológico , Carcinoma Epitelial do Ovário/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quinases Ciclina-Dependentes/antagonistas & inibidores , Feminino , Xenoenxertos , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/enzimologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Fenilenodiaminas/farmacologia , Prognóstico , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Quinase Ativadora de Quinase Dependente de Ciclina
2.
Mol Pharm ; 16(12): 4940-4953, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31651174

RESUMO

Tumor-infiltrating T lymphocytes highly express programmed cell death protein-1 (PD-1) that interacts with its ligand, programmed cell death protein ligand-1 (PD-L1) on tumors. PD-1/PD-L1 interactions cause functional exhaustion of effector T cells and impair antitumor immunity, allowing tumors to escape immune surveillance. In addition to such extrinsic interactions, tumors proliferate by transmitting intrinsic PD-L1 signals via the mTOR pathway. Here, we simultaneously silenced PD-1 and PD-L1 expressions on CTLs and colon tumors using PD-1 siRNA/PD-L1 siRNA-loaded PLGA nanoparticles and investigated functional activation of tumor-specific CTLs. When compared to a single PD-1 silencing on CTLs or a single PD-L1 silencing on tumors, cosilencing of PD-1/PD-L1 on CTLs and tumors more efficiently promoted effector functions of tumor-specific CTLs. Moreover, PD-L1-silenced tumors inhibited mTOR signaling and showed an antiproliferative response independent of the adaptive immune response. Ultimately, systemic administration of PD-1 and PD-L1 siRNA via PLGA nanoparticles restored the effector functions of tumor-specific CTLs in MC38 tumor-bearing mice. Compared with antitumor effects of single silencing of PD-1 or PD-L1 alone, cosilencing of PD-1 and PD-L1 showed more significant tumor growth suppression and long-term tumor inhibition in colon cancer. Thus, this study provides an efficient therapeutic strategy for achieving immunotherapy in colon cancer.


Assuntos
Neoplasias do Colo/metabolismo , Neoplasias do Colo/terapia , Nanopartículas/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Receptor de Morte Celular Programada 1/metabolismo , RNA Interferente Pequeno/fisiologia , Animais , Apoptose/genética , Apoptose/fisiologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/antagonistas & inibidores , RNA Interferente Pequeno/química
3.
Dermatol Surg ; 45(2): 203-209, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30286001

RESUMO

BACKGROUND: Minimally invasive aesthetic procedures of the neck are becoming more popular. However, anatomical studies on the venous structures of the neck in relation to these procedures are lacking. OBJECTIVE: The aims of this study were to identify the locations and communication patterns of the anterior jugular vein and external jugular vein (AJV and EJV) and the communicating vein (CV) based on superficial anatomical landmarks and to determine dangerous areas for dermal filler injections into the neck. MATERIALS AND METHODS: Thirty sides of the neck from Korean adult cadavers were dissected for this study. RESULTS: Four anatomical variants were identified. In Type Ia, the CV ran along the anterior border of the sternocleidomastoid muscle (SCM) (33.4%); in Type Ib, a single vein was observed connecting the CV and the EJV at the level of laryngeal prominence (23.3%); in Type Ic, the CV proceeded separately from the medial side of the anterior border of the SCM (13.3%); and in Type II, the CV was absent while the EJV and AJV were observed (30%). CONCLUSION: Given the 4 anatomical variants identified in this study, the authors recommend exerting caution when performing dermal filler injections approximately 10, 30, and 60 mm lateral to the midsagittal line to avoid iatrogenic side effects.


Assuntos
Preenchedores Dérmicos/administração & dosagem , Pescoço/irrigação sanguínea , Veias/anatomia & histologia , Idoso , Pontos de Referência Anatômicos , Variação Anatômica , Cadáver , Feminino , Humanos , Injeções , Veias Jugulares/anatomia & histologia , Masculino , República da Coreia
4.
Biochem Biophys Res Commun ; 503(3): 1716-1722, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30049442

RESUMO

Although siRNA-mediated downregulation technology has been highly successful in suppressing the expression of any disease-related gene, systemic delivery of siRNA for the clinical applications remains challenging, especially in the use of cancer therapy. DC-Chol/DOPE cationic liposomes as one of the most attractive vehicles for gene delivery have been widely exploited for transfection of siRNA into cells, but complexity of systemic delivery has allowed only their direct injection into local targets due to the formation of aggregations with negatively-charged blood components. Herein, we demonstrate the effects of PEGylation on DC-Chol/DOPE cationic liposomes for systemic siRNA delivery in cancer therapy. In contrast to non-PEGylated DC-Chol/DOPE-siRNA lipoplexes, PEGylated DC-Chol/DOPE-siRNA lipoplexes reduce the excretion by kidneys and scavenging in liver, prolonging the circulation time in vivo, and ultimately increase their preferential tumor accumulation. Therefore, systemic injection of PEGylated DC-Chol/DOPE liposomes loaded with siRNA against kinesin spindle protein (KSP) gene exhibited a high level of target gene silencing at tumor sites and substantial suppression of tumor growth. Furthermore, systemically administered PEGylated lipoplexes did not lead to any activation of innate immune responses in the immunocompetent mice. These results suggest the potential of PEGylated DC-Chol/DOPE liposomes as a systemic delivery carrier for siRNA-mediated cancer therapy.


Assuntos
Colesterol/análogos & derivados , Portadores de Fármacos/química , Inativação Gênica , Cinesinas/genética , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , RNA Interferente Pequeno/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Colesterol/química , Feminino , Humanos , Cinesinas/metabolismo , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/patologia , Polietilenoglicóis/química , Ribonucleases/antagonistas & inibidores , Ribonucleases/sangue
5.
J Gen Virol ; 97(1): 134-143, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26490105

RESUMO

Previous reports have demonstrated that hepatitis B virus (HBV) X protein (HBx) represses E-cadherin expression to induce epithelial-mesenchymal transition (EMT), an essential component of cancer progression to more aggressive phenotypes characterized by tumour invasion, migration and metastasis; however, the underlying mechanism for this phenomenon is still unclear. In this study, we found that ectopic expression of HBx in human hepatocytes using overexpression and 1.2-mer WT HBV replicon systems upregulated levels of the transcriptional repressors E12 and E47, resulting in inactivation of the E-cadherin promoter, containing three E-box motifs, and subsequent repression of its expression. E12/E47 knockdown using a specific small interfering RNA almost completely abolished the potential of HBx to repress E-cadherin expression. HBx inhibited the ubiquitin-dependent proteasomal degradation of E12/E47 without affecting their expression at the transcriptional level. Upregulation of E12/E47 by HBx ultimately led to EMT in human hepatocytes, as demonstrated by morphological changes, altered protein levels of EMT markers, including E-cadherin, plakoglobin, fibronectin, vimentin and N-cadherin, and increased capacity for cell detachment and migration.


Assuntos
Caderinas/antagonistas & inibidores , Transição Epitelial-Mesenquimal , Regulação da Expressão Gênica/efeitos dos fármacos , Vírus da Hepatite B/fisiologia , Interações Hospedeiro-Patógeno , Transativadores/metabolismo , Fator 3 de Transcrição/metabolismo , Linhagem Celular , Hepatócitos/metabolismo , Humanos , Proteínas Virais Reguladoras e Acessórias
6.
Mol Pharm ; 10(1): 18-25, 2013 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-22663765

RESUMO

Recently, we reported that a chimeric capsid protein assembled into a macromolecular container-like structure with capsid shell and the resulting siRNA/capsid nanocarrier complexes efficiently suppressed RFP gene expression in the cell culture system. To extend RNAi to the in vivo applications, we here demonstrated that the siRNA/capsid nanocarrier complexes could have tumor-specific targeting ability in vivo as well as the increased stability of siRNA during body circulation. When systemically administered, our siRNA/capsid nanocarrier complexes delivered siRNA to tumor tissues and efficiently suppressed RFP gene expression in tumor-bearing mice. The enhanced longevity of siRNA in vivo could be explained by shielding effect derived from the capsid shell, where the encapsulated siRNAs are protected from nucleases in plasma. The multivalent RGD peptides on shell surface, as a result of self-assembling of capsid protein subunits, showed efficient delivery of siRNA to the tumor tissues in vivo, due to the RGD-mediated binding to integrin receptors overexpressed on tumor cells. Moreover, the prolonged in vivo circulation time of our siRNA/capsid nanocarrier complexes increased the potential to serve as siRNA carriers for optimal in vivo RNAi. These results provide an alternative approach to systemically deliver siRNA to the tumor sites as well as to enhance the stability of siRNA in vivo. Therefore, our results revealed the promising potential of our capsid nanocarrier system as a therapeutic siRNA carrier for cancer treatment.


Assuntos
Proteínas do Capsídeo/administração & dosagem , Proteínas do Capsídeo/genética , Melanoma Experimental/tratamento farmacológico , Interferência de RNA/fisiologia , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Animais , Capsídeo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Portadores de Fármacos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Masculino , Melanoma Experimental/genética , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/administração & dosagem , Proteínas Nucleares/genética , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Ubiquitina-Proteína Ligases
7.
Mol Pharm ; 10(2): 763-73, 2013 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-23276207

RESUMO

For the efficient cytoplasmic delivery of siRNA in a receptor-specific fashion, we designed a p19-YSA fusion protein composed of p19 RNA binding protein and ephrin mimetic peptide (YSA peptide). The resulting recombinant protein had the high affinity for EphA2 receptor overexpressed on cancer cells as well as the complexing ability with siRNA, thus leading to tumor-targeted delivery of siRNA. The buried structure of siRNA within p19-YSA/siRNA complexes allowed the bound siRNAs to be protected from the external RNases, resulting in the enhanced stability of siRNA in serum conditions. The p19-YSA carriers could complex with siRNA in a size-dependent and sequence-independent manner and showed the pH-dependent complexing/dissocation behaviors with siRNA. In contrast to electrostatic interaction-mediated siRNA delivery systems such as cationic polymers/siRNA or cationic polypeptides/siRNA complexes, the bound siRNA within p19-YSA/siRNA complexes showed enhanced stability against large polyanions found outside cells, due to the nanomolar levels of affinity. Here, we demonstrated the superior efficiency of p19-YSA/siRNA complexes in RFP gene silencing, compared to untreated cells. These results provide an alternative approach to enhance the stability of siRNA as well as to achieve the targeted siRNA delivery.


Assuntos
RNA Interferente Pequeno/química , Proteínas Recombinantes de Fusão/química , Linhagem Celular , Humanos , Estrutura Secundária de Proteína , Receptor EphA2
8.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 5): 531-40, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22525751

RESUMO

When the Z-type variant of human α(1)-antitrypsin was overexpressed in Saccharomyces cerevisiae, proteomics analysis identified YLR301w as one of the up-regulated proteins. YLR301w is a 27.5 kDa protein with no sequence homology to any known protein and has been reported to interact with Sec72 and Hrr25. The crystal structure of S. cerevisiae YLR301w has been determined at 2.3 Å resolution, revealing a novel ß-structure. It consists of an N-terminal ten-stranded ß-barrel with two short α-helices connected by a 23-residue linker to a seven-stranded half-barrel with two short helices at the C-terminus. The N-terminal barrel has a highly conserved hydrophobic channel that can bind hydrophobic molecules such as PEG. It forms a homodimer both in the crystal and in solution. YLR301w binds Sec72 with a K(d) of 6.2 µM, but the biological significance of this binding requires further investigation.


Assuntos
Proteínas de Transporte/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/química , Sequência de Aminoácidos , Proteínas de Transporte/metabolismo , Cristalografia por Raios X , Modelos Moleculares , Dados de Sequência Molecular , Polietilenoglicóis/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Alinhamento de Sequência
9.
Amino Acids ; 41(5): 1113-22, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20953646

RESUMO

Among the classical collagenases, matrix metalloproteinase-13 (called MMP-13, collagenase-3) is one of the most important components for cartilage destruction of osteoarthritis (OA) developments. Despite many efforts, the detection methods of MMP-13 activity have been met with limited success in vivo, in part, due to the low sensitivity and low selectivity by homology of MMP family. Previously, we demonstrated the use of strongly dark-quenched fluorogenic probe allowed for the visual detection of MMP-13 in vitro and in OA-induced rat models. In this study, we described the optimization of MMP-13 fluorogenic probe for OA detection in vivo. Three candidate probes demonstrated recovered fluorescent intensity proportional with MMP-13 concentrations, respectively; however, Probe 2 exhibited both high signal amplification and selective recognition for MMP-13, not MMP-2 and MMP-9 in vitro. When Probe 2 was applied to OA-induced rat models, clear visualization of MMP-13 activity in OA-induced cartilage was obtained. Optimized MMP-13 fluorogenic probe can be applied to detect and image OA and have potential for evaluating the in vivo efficacy of MMP-13 inhibitors which are being tested for therapeutic treatment of OA.


Assuntos
Carbocianinas/metabolismo , Corantes Fluorescentes/metabolismo , Metaloproteinase 13 da Matriz/análise , Metaloproteinase 13 da Matriz/metabolismo , Imagem Molecular/métodos , Sondas Moleculares/metabolismo , Osteoartrite/enzimologia , Animais , Carbocianinas/química , Modelos Animais de Doenças , Corantes Fluorescentes/química , Humanos , Cinética , Sondas Moleculares/química , Osteoartrite/diagnóstico , Peptídeos/química , Peptídeos/metabolismo , Ratos , Ratos Sprague-Dawley , Sensibilidade e Especificidade , Espectroscopia de Luz Próxima ao Infravermelho
10.
Biomacromolecules ; 12(8): 3099-106, 2011 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-21728293

RESUMO

Protein cages have the potential to serve as biomaterials for the targeted therapeutic and imaging systems. As an effort to exploit small heat shock protein (Hsp) cages as multifunctional biomaterials, we demonstrate that chemically and genetically modified Hsp cages permeate the cells via cancer cell binding and subsequent endocytic internalization and can image caspase activity in the live cells. Moreover, we report here that these functional Hsp cages can be specifically accumulated to tumor tissues of tumor-bearing mice when administered intravenously through the lateral tail vein. These tumor-targeting properties could be explained by the prolonged in vivo circulation and enhanced permeability and retention (EPR) effect as well as the ligand-mediated binding to cancer cells. Furthermore, when combined with the caspase sensing ability, our Hsp cage allows us to monitor the therapeutic evaluation after anticancer drug treatment by imaging the caspase activity within tumors. Therefore, we demonstrate that the Hsp cages have multifunctional scaffolds amenable to genetic and chemical modifications without loss of the cagelike architecture and can be exploited as biomedical materials including drug or imaging agent carriers.


Assuntos
Caspases/metabolismo , Proteínas de Choque Térmico/metabolismo , Neoplasias/metabolismo , Animais , Endocitose , Escherichia coli/genética , Células HeLa , Proteínas de Choque Térmico/genética , Humanos , Mathanococcus/genética , Camundongos , Microscopia Eletrônica de Transmissão
11.
Biomaterials ; 272: 120793, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33836291

RESUMO

Therapeutic application of CRISPR/Cas9 nucleases remains a challenge due to the lack of efficient in vivo delivery carriers. Here, we examine the ability of lentiviral vectors pseudotyped with hepatitis C virus (HCV)/E1E2 envelope glycoproteins to systemically deliver CRISPR/Cas9 to hepatic tumors in vivo. We demonstrated that systemic administration of E1E2-pseudotyped lentiviral vectors can selectively deliver Cas9 and sgRNA specific for kinesin spindle protein (KSP) to Huh7 tumors in the orthotopic Huh7 mice due to the specific interactions between E1E2 and their cellular receptors. This specific delivery leads to effective KSP gene disruption, potently inhibiting tumor growth. Furthermore, we demonstrated that E1E2-pseudotyping is more suitable for systemic delivery of CRISPR/Cas9 in cancer therapy than vesicular stomatitis virus-pseudotyping, the most widely used pseudotyping, because of stability in human serum, little transduction to DCs, low innate immune response, and cell-specific targeting ability. This study suggests that E1E2-pseudotyped lentivirus carrying CRISPR/Cas9 can substantially benefit the treatment of Huh7 tumors.


Assuntos
Sistemas CRISPR-Cas , Neoplasias Hepáticas , Animais , Sistemas CRISPR-Cas/genética , Vetores Genéticos , Lentivirus/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Camundongos , Transdução Genética , Tropismo , Proteínas do Envelope Viral/genética
12.
Cells ; 10(10)2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34685714

RESUMO

Pancreatic cancer is characterized by late detection, frequent drug resistance, and a highly metastatic nature, leading to poor prognosis. Antibody-based immunotherapy showed limited success for pancreatic cancer, partly owing to the low delivery rate of the drug into the tumor. Herein, we describe a poly(lactic-co-glycolic acid;PLGA)-based siRNA nanoparticle targeting PD-L1 (siPD-L1@PLGA). The siPD-L1@PLGA exhibited efficient knockdown of PD-L1 in cancer cells, without affecting the cell viability up to 6 mg/mL. Further, 99.2% of PDAC cells uptake the nanoparticle and successfully blocked the IFN-gamma-mediated PD-L1 induction. Consistently, the siPD-L1@PLGA sensitized cancer cells to antigen-specific immune cells, as exemplified by Ovalbumin-targeting T cells. To evaluate its efficacy in vivo, we adopted a pancreatic PDX model in humanized mice, generated by grafting CD34+ hematopoeitic stem cells onto NSG mice. The siPD-L1@PLGA significantly suppressed pancreatic tumor growth in this model with upregulated IFN-gamma positive CD8 T cells, leading to more apoptotic tumor cells. Multiplex immunofluorescence analysis exhibited comparable immune cell compositions in control and siPD-L1@PLGA-treated tumors. However, we found higher Granzyme B expression in the siPD-L1@PLGA-treated tumors, suggesting higher activity of NK or cytotoxic T cells. Based on these results, we propose the application of siPD-L1@PLGA as an immunotherapeutic agent for pancreatic cancer.


Assuntos
Antígeno B7-H1/metabolismo , Imunidade , Nanopartículas/química , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , RNA Interferente Pequeno/metabolismo , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Animais , Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Proliferação de Células , Granzimas/metabolismo , Humanos , Evasão da Resposta Imune , Camundongos , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Microambiente Tumoral/imunologia , Regulação para Cima , Neoplasias Pancreáticas
13.
J Control Release ; 321: 184-197, 2020 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-32035195

RESUMO

Despite the promising anticancer effects of kinesin spindle protein (KSP) inhibition, functional plasticity of kinesins induced resistance against KSP inhibitors in a variety of cancers, leading to clinical failure. Additionally, paclitaxel is a widely used anticancer agent, but drug resistance has limited its use in the recurrent cancers. To overcome resistance against KSP inhibitors, we paired KSP inhibition with microtubule stabilization using KSP siRNA and paclitaxel. To enable temporal co-localization of both drugs in tumor cells in vivo, we exploited PEGylated cationic liposomes carrying both simultaneously. Drug synergism study shows that resistance against KSP inhibition can be suppressed by the action of microtubule-stabilizing paclitaxel, because microtubule stabilization prevents a different kinesin Kif15 from replacing all essential functions of KSP when KSP is inhibited. Our combination therapy showed more effective antiproliferative activity in vitro and in vivo than either paclitaxel or KSP siRNA alone. Ultimately, we could observe significantly improved therapeutic effects in the drug-resistant in vivo models, including cell line and patient-derived xenografts. Taken together, our combination therapy provides a potential anticancer strategy to overcome resistance against KSP inhibitors. Particularly, this strategy also provides an efficient approach to improve the therapeutic effects of paclitaxel in the drug-resistant cancers.


Assuntos
Cinesinas , Neoplasias Ovarianas , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Lipossomos , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel , Polietilenoglicóis , RNA Interferente Pequeno
14.
Biochem Pharmacol ; 171: 113735, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31786264

RESUMO

Unusual activation or overexpression of epidermal growth factor receptor (EGFR) has been found in various cancers, and therefore down-regulation of EGFR expression is recognized as a promising strategy for cancer treatment. For decades, RNAi has emerged as an effective solution to regulate gene overexpression, but transient effects of exogenous siRNA have limited the development of EGFR-targeting siRNA therapeutics. Recently, we developed T7 autogene-based hybrid mRNA/DNA system as a non-viral vector for shRNA production and reported the feasibility of long-term silencing for RFP expression as a concept of proof. To investigate its therapeutic availability in cancer therapy, we here modified the hybrid system to stably express EGFR shRNA and confirmed the antitumor effects. When autocatalytic production of T7pol protein in cytoplasm was combined with pT7-driven cytoplasmic transcription for EGFR shRNA, a single transfection lead to stable EGFR silencing in SKOV3 cells for more than 13 days. Also, liposomal systemic administration at two-week intervals resulted in significant inhibition of tumor growth in both SKOV3-bearing mice and PDX models, contrast to the conventional siRNA approach. Our results show an efficient strategy to overcome the temporary effects of synthetic EGFR siRNA in cancer treatment and ultimately provide a potential candidate as an anticancer drug.


Assuntos
DNA/genética , Receptores ErbB/genética , Interferência de RNA , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Terapêutica com RNAi/métodos , Animais , Linhagem Celular Tumoral , Citoplasma/genética , Citoplasma/metabolismo , DNA/metabolismo , Receptores ErbB/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , RNA Mensageiro/metabolismo , Carga Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
15.
Sci Rep ; 10(1): 4904, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32184452

RESUMO

Axitinib, small molecule tyrosine kinase inhibitor, demonstrates anti-cancer activity for various solid tumors. We investigated anti-cancer effect of axitinib in epithelial ovarian cancer (EOC). We treated EOC cells (A2780, HeyA8, RMG1, and HeyA8-MDR) with axitinib to evaluate its effects on cell viabilty, apoptosis and migration. Western blots were performed to assess VEGFR2, ERK, and AKT levels, and ELISA and FACS to evaluate apoptosis according to axitinib treatment. In addition, in vivo experiments in xenografts using A2780, RMG1, and HeyA8-MDR cell lines were performed. We repeated the experiment with patient-derived xenograft models (PDX) of EOC. Axitinib significantly inhibited cell survival and migration, and increased apoptosis in EOC cells. The expression of VEGFR2 and phosphorylation of AKT and ERK in A2780, RMG1, and HeyA8 were decreased with axitinib treatment in dose-dependent manner, but not in HeyA8-MDR. In in vivo experiments, axitinib significantly decreased tumor weight in xenograft models of drug-sensitive (A2780), and clear cell carcinoma (RMG1) and PDX models for platinum sensitive EOC compared to control, but was not effective in drug-resistant cell line (HeyA8-MDR) or heavily pretreated refractory PDX model. Axitinib showed significant anti-cancer effects in drug-sensitive or clear cell EOC cells via inhibition of VEGFR signals associated with cell proliferation, apoptosis and migration, but not in drug-resistant cells.


Assuntos
Axitinibe/uso terapêutico , Carcinoma Epitelial do Ovário/tratamento farmacológico , Carcinoma Epitelial do Ovário/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Inibidores de Proteínas Quinases/uso terapêutico , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo
17.
Acta Biomater ; 91: 258-269, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31026519

RESUMO

Adeno-associated virus (AAV) is a promising vector for systemic delivery of siRNA because of its long-term expression ability without immunogenicity and pathogenicity. However, its broad host tropism and lack of tissue specificity have limited clinical applications such as cancer therapy. Therefore, redirecting the natural tropism of AAV vectors to unique cell surface antigens is an important requirement for in vivo RNAi-based cancer therapy. To use the overexpression property of epithelial cell adhesion molecule (EpCAM) in specific cancer types, we herein created anti-EpCAM antibody-conjugated AAV serotype 2 (AAV2) vectors through a streptavidin-biotin bridge. Upon intravenous injection, anti-EpCAM-conjugated AAV2 vectors showed prominent tumor-specific accumulation in EpCAM-positive tumor-bearing mice without undesirable sequestration in liver. In addition, when loaded with transgenes to express shRNA against epidermal growth factor receptor (EGFR), systemically injected anti-EpCAM-conjugated AAV2/shEGFR vectors induced significant downregulation of EGFR expression in tumors and eventually suppressed tumor growth even at the long dosing interval of two weeks. This in vivo antitumor effect represents the increased infection efficacy of tropism-modified AAV2 vectors and prolonged expression of EGFR shRNA in tumor tissues. Thus, this study suggests the great potential of anti-EpCAM-conjugated AAV2/shEGFR vectors as RNAi-based cancer therapeutics. STATEMENT OF SIGNIFICANCE: Adeno-associated virus (AAV) is a promising vector for systemic delivery of siRNA, but its broad host tropism has limited clinical applications. By using the overexpression property of epithelial cell adhesion molecule (EpCAM) on tumors, we demonstrate that anti-EpCAM-conjugated AAV2 vectors through a streptavidin-biotin bridge are redirected to EpCAM-positive tumors in vivo. In addition, when loaded with transgenes to express shRNA against epidermal growth factor receptor (EGFR), systemically injected anti-EpCAM-conjugated AAV2/shEGFR vectors significantly downregulate EGFR expression in tumors, eventually suppressing tumor growth for long periods. We herein suggest the potential of anti-EpCAM-AAV2/shEGFR vectors as an antitumor agent. Furthermore, redirection of AAV2 infection through EpCAM would provide a powerful means for systemic delivery of short hairpin RNA to tumor sites.


Assuntos
Antineoplásicos Imunológicos , Molécula de Adesão da Célula Epitelial/antagonistas & inibidores , Terapia Genética , Parvovirinae , RNA Interferente Pequeno , Transdução Genética , Linhagem Celular Tumoral , Dependovirus , Molécula de Adesão da Célula Epitelial/metabolismo , Receptores ErbB/biossíntese , Receptores ErbB/genética , Feminino , Humanos , Neoplasias Ovarianas , RNA Interferente Pequeno/biossíntese , RNA Interferente Pequeno/genética
18.
Biochem Pharmacol ; 166: 192-202, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31129050

RESUMO

Duration of gene silencing due to the short-term silencing effects induced by exogenous siRNA have limited the therapeutic applications of RNAi and the development of RNAi-based therapeutics. We here generated Eg5 shRNA-expressing plasmids using the inverted terminal repeats (ITRs) sequences to produce Eg5 hairpin RNA under the control of U6 promoter. Using PEGylated DC-Chol/DOPE cationic liposomes, we demonstrated that a single systemic administration of Eg5 shRNA-expressing plasmid/liposome lipoplexes induced the long-term Eg5 silencing in the tumor sites of tumor-bearing mice, and ultimately lead to more sustained anticancer effects than standard synthetic siEg5/liposome lipoplexes. This non-viral Eg5 shRNA expression system had no risk of immunogenicity anticipated in the use of viral vectors, and could reduce the potential of off-target effects by scaling down the administration dose of RNAi therapeutics in patient. Therefore, the sustainable shRNA expression properties in the tumor sites suggest an efficient strategy to overcome the limitations caused by chemically synthesized siRNA methods such as short-term silencing effects and off-target effects. Herein, this study provides a non-viral silencing strategy for inducing long-term Eg5 silencing in vivo and suggests the great potential of Eg5 shRNA-expressing lipoplexes as a DNA-based RNAi therapeutics for cancer treatment.


Assuntos
Colesterol/análogos & derivados , Inativação Gênica/fisiologia , Cinesinas/biossíntese , Fosfatidiletanolaminas/biossíntese , Plasmídeos/biossíntese , RNA Interferente Pequeno/biossíntese , Animais , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Colesterol/administração & dosagem , Colesterol/biossíntese , Colesterol/genética , Feminino , Expressão Gênica , Inativação Gênica/efeitos dos fármacos , Técnicas de Transferência de Genes , Humanos , Cinesinas/administração & dosagem , Cinesinas/genética , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Fosfatidiletanolaminas/administração & dosagem , Fosfatidiletanolaminas/genética , Plasmídeos/administração & dosagem , Plasmídeos/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Distribuição Aleatória , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
19.
Sci Rep ; 9(1): 2993, 2019 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-30816180

RESUMO

The transient silencing effects currently demonstrated by nonviral siRNA delivery systems limit the therapeutic utility of RNAi, but it remains a technical challenge to prolong duration of gene silencing. We have developed a T7 autogene-based hybrid mRNA/DNA system to enable long-term expression of shRNA in cytoplasm in vitro and in vivo. This hybrid mRNA/DNA system consists of T7 polymerase (T7pol) mRNA, pT7/shRNA-encoding DNA fragment and T7 autogene plasmid, and it can generate higher levels of T7pol proteins, compared to pCMV-triggering T7 autogene system, especially without the need of nuclear entry of any gene. A large amount of T7pol proteins produced are used to induce pT7-driven expression of shRNA in cytoplasm, and through cellular processing of RNA hairpins, mature siRNAs are generated for more than 13 days. We here demonstrate that a single liposomal delivery of this hybrid system leads to the long-term silencing effects in vitro and in vivo, in contrast to the conventional siRNA methods relying on the repeated administrations every 2 or 3 days. These sustainable shRNA expression properties in cytoplasm can provide an efficient strategy to address the limitations caused by shRNA-encoding plasmid DNA systems such as low nuclear entry efficiency and short-term silencing effect. The development of long-term shRNA expression system in vivo could scale down administration frequency of RNAi therapeutics in the treatment of chronic diseases, thereby increasing its clinical utility.


Assuntos
RNA Polimerases Dirigidas por DNA/genética , Técnicas de Transferência de Genes , Neoplasias Experimentais/terapia , RNA Interferente Pequeno/genética , Terapêutica com RNAi/métodos , Proteínas Virais/genética , Animais , Linhagem Celular Tumoral , Citoplasma/metabolismo , DNA/genética , DNA/metabolismo , RNA Polimerases Dirigidas por DNA/metabolismo , Feminino , Lipossomos/química , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas Virais/metabolismo
20.
Materials (Basel) ; 12(18)2019 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-31489908

RESUMO

Clay ball is a pavement surface defect which refers to a clump in which clay or dirt is mixed with hot asphalt mixture. Clay ball is typically caused by a combination of aggregate contamination of clay or soil, high aggregate moisture, and low production temperature at the asphalt plant. It usually appears a few weeks or months after paving under traffic load, after being liquefied and knocked from the pavement surface. Clay balls can be the source of potholing, raveling, and other issues such as moisture infiltration and reduced ride quality. This paper presents an investigation of the clay balls on US-31 one winter after construction in Hamilton County, Indiana. In order to understand the pavement condition, their severity was measured using both visual observation and infrared image collection system. In addition, a clay ball amount, its distribution pattern, and cores condition were evaluated. A precipitation effect on clay ball formation was investigated for finding a cause of the clay balls. The investigation found that infrared image collection system was appropriate in detecting the clay balls. The clay balls were elliptic in shape with 2.5 cm to 10 cm in diameter, and the maximum clay ball depth was almost penetrating the entire surface course. It was also found that the asphalt paving on the raining days or right after raining could increase the potential of clay balls. Monitoring of aggregate moisture during construction on or after raining days should be able to reduce the risk of clay balls.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA