Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Cancer ; 120(4): 480-91, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24496866

RESUMO

BACKGROUND: Patients with pancreatic ductal adenocarcinoma (PDAC) have limited therapeutic options and poor response to the standard gemcitabine (GCB)-based chemotherapy. In the current study, the authors investigated the feasibility of noninvasive short-wave radiofrequency (RF) electric fields to improve the cytotoxic effect of GCB on PDAC cells and determined its mechanism of action. METHODS: The cytotoxicity of RF alone and in combination with GCB was studied in vitro on normal pancreatic human pancreatic ductal epithelial cells and different PDAC cell lines by flow cytometry, and in vivo on ectopic and orthotopic human PDAC xenograft models in mice. The mechanism of RF activity was studied by Western blot analysis and immunohistochemistry. Toxicity was determined by histopathology. RESULTS: Exposure of different PDAC cells to 13.56-megahertz radio waves resulted in a substantial cytotoxic effect, which was accompanied by the induction of autophagy but not apoptosis. These effects of RF were found to be absent in normal cells. Excessive numbers of autophagosomes in cancer cells persisted 24 to 48 hours after RF exposure and then declined. The addition of a subtoxic dose of GCB to RF treatment inhibited the recovery of cancer cells from the RF-induced autophagy and enhanced the cytotoxic effect of the latter on cancer cells. The treatment of PDAC in situ in mice with the combination of noninvasive RF and GCB was found to have a superior antitumor effect compared with the use of RF or GCB alone, yet there was no evidence of systemic toxicity. CONCLUSIONS: Noninvasive RF treatment induced autophagy but not apoptosis in cancer cells and demonstrated potential as an enhancer of chemotherapy for treating patients with pancreatic cancer without toxicity to normal cells.


Assuntos
Adenocarcinoma/radioterapia , Autofagia/efeitos da radiação , Neoplasias Pancreáticas/radioterapia , Ondas de Rádio , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Animais , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Humanos , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
2.
Nanomedicine ; 8(7): 1096-105, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22349096

RESUMO

The use of noninvasive radiofrequency (RF) electric fields as an energy source for thermal activation of nanoparticles within cancer cells could be a valuable addition to the emerging field of nano-mediated cancer therapies. Based on investigations of cell death through hyperthermia, and offering the ability for total-body penetration by RF fields, this technique is thought to complement and possibly outperform existing nano-heat treatments that utilize alternative heat production via optical or magnetic stimuli. However, it remains a challenge to understand fully the complex RF-nanoparticle-intracellular interactions before full system optimization can be engineered. Herein we have shown that liver cancer cells can selectively internalize antibody-conjugated gold nanoparticles (AuNPs) through receptor-mediated endocytosis, with the nanoparticles predominantly accumulating and aggregating within cytoplasmic endolysosomes. After exposure to an external RF field, nonaggregated AuNPs absorbed and dissipated energy as heat, causing thermal damage to the targeted cancer cells. We also observed that RF absorption and heat dissipation is dependent on solubility of AuNPs in the colloid, which is pH dependent. Furthermore, by modulating endolysosomal pH it is possible to prevent intracellular AuNP aggregation and enhance thermal cytotoxicity in hepatocellular cancer cells. FROM THE CLINICAL EDITOR: Gold nanoparticles absorb energy from RF fields and can exert hyperthermic effects leading to cell death. Combining this known effect with antibody-based targeting of the nanoparticles, selective cancer specific hyperthermia induced cell death therapies can be designed, as demonstrated in this article.


Assuntos
Ouro/uso terapêutico , Hipertermia Induzida/métodos , Imunoconjugados/uso terapêutico , Neoplasias Hepáticas/terapia , Nanopartículas/uso terapêutico , Terapia por Radiofrequência , Anticorpos/química , Anticorpos/uso terapêutico , Linhagem Celular Tumoral , Ouro/química , Humanos , Concentração de Íons de Hidrogênio , Imunoconjugados/química , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Lisossomos/metabolismo , Lisossomos/patologia , Nanopartículas/química , Solubilidade
3.
J Exp Ther Oncol ; 7(4): 313-26, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19227011

RESUMO

Shortwave (MHz range) radiofrequency (RF) energy is nonionizing, penetrates deeply into biologic tissues with no adverse side effects, and heats gold nanoparticles efficiently. Targeted delivery of gold nanoparticles to cancer cells should result in hyperthermic cytotoxicity upon exposure to a focused, noninvasive RF field. In this report we demonstrate that gold nanoparticles conjugated with cetuximab (C225) are quickly internalized by Panc-1 (pancreatic adenocarcinoma) and Difi (colorectal adenocarcinoma) cancer cells overexpressing epidermal growth factor receptor (EGFR). Panc-1 or Difi cells treated with naked gold nanoparticles or nonspecific IgG-conjugated gold nanoparticles demonstrated minimal intracellular uptake of gold nanoparticles by transmission electron microscopy (TEM). In contrast, there were dense concentrations of cytoplasmic vesicles containing gold nanoparticles following treatment with cetuximab-conjugated gold nanoparticles. Exposure of cells to a noninvasive RF field produced nearly 100% cytotoxicity in cells treated with the cetuximab-conjugated gold nanoparticles, but significantly lower levels of cytotoxicity in the two control groups (P < 0.00012). Treatment of a breast cancer cell line (CAMA-1) that does not express EGFR with cetuximab-conjugated gold nanoparticles produced no enhanced cytotoxicity following treatment in the RF field. Conjugation of cancer cell-directed targeting agents to gold nanoparticles may represent an effective and cancer-specific therapy to treat numerous types of human malignant disease using noninvasive RF hyperthermia.


Assuntos
Adenocarcinoma/tratamento farmacológico , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Ouro/química , Nanopartículas Metálicas/química , Nanotecnologia/métodos , Neoplasias Pancreáticas/tratamento farmacológico , Adenocarcinoma/radioterapia , Anticorpos Monoclonais Humanizados , Apoptose , Linhagem Celular Tumoral , Cetuximab , Receptores ErbB/metabolismo , Humanos , Hipertermia Induzida , Microscopia Eletrônica de Transmissão , Necrose , Neoplasias Pancreáticas/radioterapia , Ondas de Rádio
4.
Arch Otolaryngol Head Neck Surg ; 134(2): 190-7, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18283163

RESUMO

OBJECTIVE: To develop a reproducible orthotopic model of papillary thyroid carcinoma for the BRAF(V600E) mutation (GenBank NM004333) and an RET/PTC rearrangement (GenBank M31213) that recapitulates the clinical picture in humans. DESIGN: In vitro and in vivo study. SETTING: Department of Head and Neck Surgery, M. D. Anderson Cancer Center. SUBJECTS: Eight- to 12-week-old athymic female nude mice. INTERVENTIONS: Either BRAF-mutated or RET/PTC1-rearranged papillary thyroid carcinoma cells were injected into the thyroid glands of athymic female nude mice. The mice were euthanized when the tumor burden exceeded 1.0 cm or when they exhibited significant morbidity. MAIN OUTCOME MEASURES: Tumorigenicity, extent of tumor invasion and metastasis, cell invasion and migration, and median survival. RESULTS: All the BRAF-mutated cell lines and 1 selected RET/PTC1-rearranged cell line were 100% tumorigenic in mice. These mouse tumor models exhibited a wide range of biological potential, including laryngeal invasion, lymph node metastasis, and pulmonary metastasis, thus reflecting the clinical spectrum of papillary carcinoma. CONCLUSIONS: An orthotopic model of papillary thyroid carcinoma was successfully established in nude mice using BRAF-mutated and RET/PTC1-rearranged cell lines. These models mimic the human disease and will thus be useful for evaluating the clinical potential of novel targeted therapies.


Assuntos
Carcinoma Papilar , Modelos Animais de Doenças , Neoplasias da Glândula Tireoide , Animais , Carcinoma Papilar/genética , Carcinoma Papilar/metabolismo , Carcinoma Papilar/patologia , Ensaios de Migração Celular , Feminino , Imuno-Histoquímica , Técnicas In Vitro , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Invasividade Neoplásica , Proteínas de Fusão Oncogênica/genética , Mutação Puntual , Proteínas Proto-Oncogênicas B-raf/genética , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia
5.
Cancer Res ; 65(24): 11501-9, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16357159

RESUMO

Headpin is a novel serine proteinase inhibitor (serpin) with constitutive mRNA expression in histologically normal oral mucosa but with lost or down-regulated expression in head and neck squamous cell carcinoma. Several serpin family members are similarly lost in multiple cancer types and hold tumor suppressor functions including the inhibition of angiogenesis. However, the functional significance for the loss of headpin expression in cancer is not known. Using immunohistochemical analysis of invasive squamous cell carcinoma and matched normal squamous mucosa of patient specimens, headpin expression was lost or down-regulated in the vast majority of tumor specimens. We investigated the functions of exogenous recombinant headpin and endogenously expressed headpin related to angiogenesis. In a rat corneal assay of neovascularization, recombinant headpin protein blocked in vivo angiogenesis mediated by interleukin 8 (IL-8) and vascular endothelial growth factor (VEGF). In assays of cellular events in angiogenesis, headpin blocked the invasion, migration, and tube formation of endothelial cells. In light of our findings of nuclear subcellular localization of headpin, we investigated the expression and secretion of angiogenic factors and found reduced mRNA, protein, and promoter activities of IL-8 and VEGF. Finally, using a murine flank tumor model, headpin expression reduced growth and microvessel density in tumors derived from headpin-expressing UMSCC1 cells relative to those from vector control cells. These findings of nuclear regulatory functions of a serpin in the inhibition of angiogenesis bring new understanding to the cellular and molecular mechanisms of serpins. Therefore, this novel serpin targets diverse mechanisms against tumor angiogenesis on which to base therapeutic strategies.


Assuntos
Carcinoma de Células Escamosas/irrigação sanguínea , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/irrigação sanguínea , Interleucina-8 , Neovascularização Patológica/prevenção & controle , Serpinas/metabolismo , Fator A de Crescimento do Endotélio Vascular , Animais , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Movimento Celular , Córnea/irrigação sanguínea , Córnea/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Camundongos , Camundongos Nus , Microcirculação/patologia , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serpinas/genética , Frações Subcelulares , Veias Umbilicais/citologia , Veias Umbilicais/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
6.
Oncogene ; 22(55): 8902-11, 2003 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-14654786

RESUMO

Head and neck squamous cell carcinoma (HNSCC) is the fifth most frequent cancer in the US. Several genetic and epigenetic alterations are associated with HNSCC tumorigenesis, including inactivation of CDKN2A, which encodes the p16 tumor suppressor, in cell lines and primary tumors by DNA methylation. Reactivation of tumor suppressor genes by DNA-demethylating agents and histone deacetylase (HDAC) inhibitors shows therapeutic promise for other cancers. Therefore, we investigated the ability of these agents to reactivate p16 in Tu159 HNSCC cells. Treatment of cells with 5-aza-2'deoxycytidine (5-aza-dC) increases CDKN2A expression and slightly increases histone H3 acetylation at this gene. No reactivation of CDKN2A is observed upon treatment with the HDAC inhibitor trichostatin A (TSA), but synergistic reactivation of CDKN2A is observed upon sequential treatment of Tu159 cells with both 5-aza-dC and TSA. Silencing of CDKN2A in Tu159 cells is correlated with increased methylation of histone H3 at lysine 9 and decreased methylation at lysine 4 relative to the upstream p15 gene promoter. Interestingly, global levels of H3-K9 methylation are decreased upon treatment with 5-aza-dC. Together these data indicate that DNA methylation is a dominant epigenetic mark for silencing of CDKN2A in Tu159 tumor cells. Moreover, changes in DNA methylation can reset the histone code by impacting multiple H3 modifications.


Assuntos
Azacitidina/análogos & derivados , Carcinoma de Células Escamosas/genética , Metilação de DNA , Genes p16 , Neoplasias de Cabeça e Pescoço/genética , Histonas/metabolismo , Animais , Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/farmacologia , Carcinoma de Células Escamosas/metabolismo , Decitabina , Inativação Gênica , Genes p16/efeitos dos fármacos , Genes p16/fisiologia , Neoplasias de Cabeça e Pescoço/metabolismo , Inibidores de Histona Desacetilases , Humanos , Ácidos Hidroxâmicos/farmacologia
7.
Hepatol Res ; 42(7): 706-13, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22414001

RESUMO

AIM: Although thioredoxin 1 (TXN) has pleiotropic cellular functions as a redox-sensitive protein, very little is known about its role in tumor survival and growth under hypoxia. MHCC97H hepatocellular carcinoma cells have a high metastatic potential and high thioredoxin expression levels compared with their parent cell line, MHCC97. Thus, we used this cell line to explore the functional connections between TXN and hypoxia. METHODS: MHCC97H cells were cultured under normoxia and hypoxia for specific periods after nucleofection with TXN siRNA or control siRNA. We assessed the ß-phenylethyl isothiocyanate (PEITC) sensitivity of the cells, cell proliferation, cell cycle and senescence, and DNA damage response by using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays, colony formation assays, flow cytometry, ß-galactosidase staining, western blotting, and immunohistochemistry. RESULTS: ß-phenylethyl isothiocyanate treatment shifted reduced TXN to oxidized TXN in MHCC97H cells. Although silencing of TXN via siRNA had no effect on the PEITC sensitivity of the cells, it suppressed cell proliferation and colony formation under both normoxia and hypoxia. Under hypoxia, silencing TXN did not induce apoptosis but induced DNA damage response and cellular senescence. CONCLUSIONS: High TXN levels in MHCC97H cells protect them from DNA damage and cellular senescence under hypoxia. Targeting TXN might enhance the chemotherapeutic effects of some DNA-damaging agents against hepatocellular carcinoma.

8.
Arch Biochem Biophys ; 435(1): 89-102, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15680911

RESUMO

The secretory lympho-epithelial Kazal-type-inhibitor (LEKTI) is synthesized as a pro-LEKTI protein containing an N-terminal signal peptide and 15 potentially inhibitory domains. This inhibitor is of special interest because of its pathophysiological importance for the severe congenital disease Netherton syndrome. We showed that LEKTI is a potent inhibitor of a family of serine proteinases involved in extracellular matrix remodeling and its expression is downregulated in head and neck squamous cell carcinomas. To assess the role of C-terminal domains and N-terminal signal peptide in LEKTI secretion, we constructed deletion mutants of LEKTI, expressed them in HEK 293T cells, and analyzed their secretion behavior, stability, subcellular distribution, and proteinase inhibitory function. Pro-LEKTI is processed and secreted into the medium. On the basis of partial N-terminal sequencing and immunoblotting, the cleavage products are ordered from amino- to carboxy-terminal as follows: 37, 40, and 60kDa. Inhibitors of furin lead to enhanced secretion of unprocessed LEKTI, suggesting that processing was not required for secretion. Deletion of the N-terminal signal peptide of pro-LEKTI caused altered distribution of LEKTI from endoplasmic reticulum (ER) to cytoplasm and markedly reduced its stability, consistent with its failure to become secreted into the medium. Interestingly, when we deleted the C-terminal domains, stable partial LEKTI (LD-1-6) accumulated and still retained its association with ER but was not secreted. Recombinant LD-1-6 specifically inhibited the trypsin activity. We conclude that N-terminal signal peptide is required for LEKTI import into ER and elements present in C-terminal domains may have a role in regulating LEKTI secretion.


Assuntos
Proteínas de Transporte/metabolismo , Rim/metabolismo , Sinais Direcionadores de Proteínas/fisiologia , Frações Subcelulares/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Proteínas de Transporte/genética , Células Cultivadas , Deleção de Genes , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Secretadas Inibidoras de Proteinases , Proteínas Recombinantes/metabolismo , Inibidor de Serinopeptidase do Tipo Kazal 5 , Relação Estrutura-Atividade
9.
Protein Expr Purif ; 35(1): 93-101, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15039071

RESUMO

The precursor lympho-epithelial Kazal-type-related inhibitor (LEKTI), containing two Kazal-type and 13 nonKazal-type domains, is an efficient inhibitor of multiple serine proteinases, among them plasmin, subtilisin A, cathepsin G, elastase, and trypsin. To gain insight into the structure and function of some of these domains, a portion of the cDNA coding for LEKTI domains 6-9' was cloned and expressed in Sf9 cells using the baculovirus expression vector system (BEVS). Through a single purification step using a Co2+ column, 3-4 mg of purified recombinant LEKTI-domains 6-9' (rLEKTI6-9') with the predicted molecular mass of 34.6 kDa was obtained from the cell pellet of a 1-L culture. Unlike full-length LEKTI, rLEKTI6-9' inhibited trypsin and subtilisin A but not plasmin, cathepsin G, or elastase. The inhibition of trypsin and subtilisin A by rLEKTI6-9' occurred through a noncompetitive mechanism, with inhibitory constants (Ki) of 356 +/- 12 and 193 +/- 10 nM, respectively. On the basis of the Ki values, rLEKTI6-9' was determined to be a more potent trypsin inhibitor and a less potent subtilisin A inhibitor than the full-length LEKTI. In contrast to LEKTI domains 6-9', recombinant LEKTI domain 6 does not inhibit subtilisin A but competitively inhibited trypsin with a Ki of 200 +/- 10 nM. Taking LEKTI6-9' as an example, the BEVS should facilitate the structure-function analysis of naturally occurring processed LEKTI forms that have physiological relevance.


Assuntos
Baculoviridae/metabolismo , Proteínas de Transporte/metabolismo , Inibidores de Serina Proteinase/metabolismo , Subtilisinas/metabolismo , Tripsina/metabolismo , Animais , Baculoviridae/genética , Proteínas de Transporte/genética , Proteínas de Transporte/isolamento & purificação , Linhagem Celular , Dissulfetos/química , Insetos , Estrutura Terciária de Proteína , Proteínas Secretadas Inibidoras de Proteinases , Inibidor de Serinopeptidase do Tipo Kazal 5 , Inibidores de Serina Proteinase/genética , Inibidores de Serina Proteinase/isolamento & purificação , Subtilisinas/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA