Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cancer Res ; 81(1): 50-63, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33115805

RESUMEN

Metabolic dysregulation is a known hallmark of cancer progression, yet the oncogenic signals that promote metabolic adaptations to drive metastatic cancer remain unclear. Here, we show that transcriptional repression of mitochondrial deacetylase sirtuin 3 (SIRT3) by androgen receptor (AR) and its coregulator steroid receptor coactivator-2 (SRC-2) enhances mitochondrial aconitase (ACO2) activity to favor aggressive prostate cancer. ACO2 promoted mitochondrial citrate synthesis to facilitate de novo lipogenesis, and genetic ablation of ACO2 reduced total lipid content and severely repressed in vivo prostate cancer progression. A single acetylation mark lysine258 on ACO2 functioned as a regulatory motif, and the acetylation-deficient Lys258Arg mutant was enzymatically inactive and failed to rescue growth of ACO2-deficient cells. Acetylation of ACO2 was reversibly regulated by SIRT3, which was predominantly repressed in many tumors including prostate cancer. Mechanistically, SRC-2-bound AR formed a repressive complex by recruiting histone deacetylase 2 to the SIRT3 promoter, and depletion of SRC-2 enhanced SIRT3 expression and simultaneously reduced acetylated ACO2. In human prostate tumors, ACO2 activity was significantly elevated, and increased expression of SRC-2 with concomitant reduction of SIRT3 was found to be a genetic hallmark enriched in prostate cancer metastatic lesions. In a mouse model of spontaneous bone metastasis, suppression of SRC-2 reactivated SIRT3 expression and was sufficient to abolish prostate cancer colonization in the bone microenvironment, implying this nuclear-mitochondrial regulatory axis is a determining factor for metastatic competence. SIGNIFICANCE: This study highlights the importance of mitochondrial aconitase activity in the development of advanced metastatic prostate cancer and suggests that blocking SRC-2 to enhance SIRT3 expression may be therapeutically valuable. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/1/50/F1.large.jpg.


Asunto(s)
Aconitato Hidratasa/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/secundario , Regulación Neoplásica de la Expresión Génica , Mitocondrias/enzimología , Neoplasias de la Próstata/patología , Sirtuina 3/metabolismo , Aconitato Hidratasa/genética , Animales , Apoptosis , Biomarcadores de Tumor/genética , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Proliferación Celular , Humanos , Masculino , Ratones , Ratones Desnudos , Coactivador 3 de Receptor Nuclear/genética , Coactivador 3 de Receptor Nuclear/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Sirtuina 3/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Eur J Med Chem ; 144: 493-503, 2018 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-29288946

RESUMEN

Molecular hybridization is considered as an effective tactic to develop drugs for the treatment of cancer. A series of novel hybrid compounds of isatin and Michael acceptor were designed and synthesized on the basis of association principle. These hybrid compounds were tested for cytotoxic potential against human cancer cell lines namely, BGC-823, SGC-7901 and NCI-H460 by MTT assay. Most compounds showed good anti-growth activities in all tested human cancer cells. SAR and QSAR analysis may provide vital information for the future development of novel anti-cancer inhibitors. Notably, compound 6a showed potent growth inhibition on BGC-823, SGC-7901 and NCI-H460 with the IC50 values of 3.6 ±â€¯0.6, 5.7 ±â€¯1.2, 3.2 ±â€¯0.7 µM, respectively. Besides, colony formation assays, wound healing assays and flow cytometry analysis indicated 6a exhibited a potent anti-growth and anti-migration ability in a concentration-dependence manner through arrested cells in the G2/M phase of cell cycle. Moreover, 6a significantly repressed tumor growth in a NCI-H460 xenograft mouse model. Overall, our findings suggested isatin analogues inspired Michael acceptor may provide promising lead compounds for the development of cancer chemotherapeutics.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Isatina/farmacología , Relación Estructura-Actividad Cuantitativa , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Isatina/síntesis química , Isatina/química , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Células Tumorales Cultivadas
3.
J Vis Exp ; (115)2016 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-27768029

RESUMEN

Intra-iliac artery (IIA) injection is an efficient approach to introduce metastatic lesions of various cancer cells in animals. Compared to the widely used intra-cardiac and intra-tibial injections, IIA injection brings several advantages. First, it can deliver a large quantity of cancer cells specifically to hind limb bones, thereby providing spatiotemporally synchronized early-stage colonization events and allowing robust quantification and swift detection of disseminated tumor cells. Second, it injects cancer cells into the circulation without damaging the local tissues, thereby avoiding inflammatory and wound-healing processes that confound the bone colonization process. Third, IIA injection causes very little metastatic growth in non-bone organs, thereby preventing animals from succumbing to other vital metastases, and allowing continuous monitoring of indolent bone lesions. These advantages are especially useful for the inspection of progression from single cancer cells to multi-cell micrometastases, which has largely been elusive in the past. When combined with cutting-edge approaches of biological imaging and bone histology, IIA injection can be applied to various research purposes related to bone metastases.


Asunto(s)
Neoplasias Óseas , Arteria Ilíaca , Metástasis de la Neoplasia , Animales , Huesos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Miembro Posterior , Humanos
4.
Mol Cell Oncol ; 3(3): e1026526, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27314064

RESUMEN

We recently discovered that bone micrometastases of breast cancer predominantly reside in the microenvironment termed the "osteogenic niche". The heterotypic adherens junctions between cancer cells and osteogenic cells promote early-stage bone colonization by activating the mTOR pathway in cancer cells. Here, we discuss a few questions raised by these findings.

5.
J Mammary Gland Biol Neoplasia ; 20(3-4): 103-8, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26490041

RESUMEN

In breast cancer, the most frequent site of metastasis is bone. Disseminated tumor cells (DTCs) can be detected in the bone marrow of patients by their expression of epithelial oroncogenic markers [1], and the presence and frequency of these DTCs are associated with poor prognosis. However, many of the details behind this process remain elusive, including the biological properties and fates of these apparently indolent cancer cells. To provide pre-clinical models of DTCs, we have developed a procedure that allows for controlled and enhanced delivery of tumor cells to the bone in animal experiments via injection into the iliac artery of the hind limb [2]. To our surprise, we found that most cancer cells became integrated into the solid bone matrix shortly after arriving in the bone, and only a minority can be flushed out with the bone marrow.Here we describe a method that helps to retrieve DTCs homing to the bone in which we achieve an improved recovery of those tumor cells closely associated with the bone microenvironment. In our view it is especially important to analyze these tumor cell subpopulations, as they may take full advantage of growth-, survival- and immune-protective signals provided by neighbor cells. We also show a pilot study on how this approach may be applied to the analysis of cancer dormancy. Our study suggests that the detection and retrieval of DTCs in clinical studies are incomplete because they are conducted exclusively with bone marrow aspirates.


Asunto(s)
Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Separación Celular/métodos , Manejo de Especímenes/métodos , Animales , Médula Ósea/patología , Supervivencia Celular , Modelos Animales de Enfermedad , Femenino , Fémur/patología , Humanos , Arteria Ilíaca , Inyecciones Intraarteriales , Células MCF-7 , Metaloproteinasa 8 de la Matriz , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Tibia/patología
6.
Cancer Cell ; 27(2): 193-210, 2015 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-25600338

RESUMEN

Breast cancer bone micrometastases can remain asymptomatic for years before progressing into overt lesions. The biology of this process, including the microenvironment niche and supporting pathways, is unclear. We find that bone micrometastases predominantly reside in a niche that exhibits features of osteogenesis. Niche interactions are mediated by heterotypic adherens junctions (hAJs) involving cancer-derived E-cadherin and osteogenic N-cadherin, the disruption of which abolishes niche-conferred advantages. We elucidate that hAJ activates the mTOR pathway in cancer cells, which drives the progression from single cells to micrometastases. Human data set analyses support the roles of AJ and the mTOR pathway in bone colonization. Our study illuminates the initiation of bone colonization, and provides potential therapeutic targets to block progression toward osteolytic metastases.


Asunto(s)
Neoplasias Óseas/genética , Neoplasias de la Mama/genética , Osteogénesis/genética , Microambiente Tumoral/genética , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Cadherinas/genética , Línea Celular Tumoral , Femenino , Humanos , Estadificación de Neoplasias , Serina-Treonina Quinasas TOR/genética
7.
Cancer Biol Ther ; 7(8): 1226-31, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18487949

RESUMEN

The second domain of Pseudomonas exotoxin A (PEAII, residues 253-364) has been shown to facilitate translocation of extracellular and vesicular contents into the cytoplasm, and can transport heterologous molecules into target cells. Because full length PEAII may elicit a host immune response, we tried to identify the minimal PEAII translocation motif and use this fragment in combination with an antibody and constitutively active granzyme B (ImmunoGrB) to kill HER2-positive tumor cells. We constructed four ImmunoGrB fusion proteins containing different PEAII deletions and tested their abilities to kill HER2-positive cells. Our data showed that while a complete deletion of PEAII in ImmunoGrB resulted in an inability to kill cancer cells, ImmunoGrBs containing either PEAII (253-358aa) or PEAII (275-358aa) could efficiently kill HER2-positive SK-BR-3 cells. Most interestingly, the construct which contains only a furin cleavage site, named PEAII (275-280aa), could still induce SK-BR-3 apoptosis, although less efficiently. Moreover, delivery of the recombinant proteins by intramuscular plasmid injection led to an apparent tumor regression and prolonged animal survival in a nude mouse xenograft SK-BR-3 tumor model, indicating in vivo antitumor activity of the different PEAII containing ImmunoGrBs. Our results may help in understanding PEAII translocation and may lead to the development of useful tools or alternative therapy.


Asunto(s)
Neoplasias de la Mama/patología , Genes erbB-2 , Receptor ErbB-2/genética , ADP Ribosa Transferasas , Toxinas Bacterianas , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/ultraestructura , Línea Celular Tumoral , Supervivencia Celular , Exotoxinas , Células HeLa , Humanos , Plásmidos , Estructura Terciaria de Proteína/genética , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Proteínas Recombinantes/metabolismo , Transfección , Factores de Virulencia , Exotoxina A de Pseudomonas aeruginosa
8.
Glia ; 56(8): 877-87, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18338795

RESUMEN

Neuregulins (NRGs) are growth factors which bind to Erb receptor tyrosine kinases that localize to Schwann cells (SCs). Although NRGs can promote cell survival, mitogenesis, and myelination in undifferentiated SCs, they also induce demyelination of myelinated co-cultures of SCs and dorsal root ganglion (DRG) neurons. We have shown previously that Erb B2 activity increased in premyelinating SCs in response to hyperglycemia, and that this correlated with the downregulation of the protein caveolin-1 (Cav-1). As myelinated SCs undergo substantial degeneration in diabetic neuropathy, we used myelinated SC/DRG neuron co-cultures to determine if hyperglycemia and changes in Cav-1 expression could enhance NRG-induced demyelination. In basal glucose, NRG1 caused a 2.4-fold increase in the number of damaged myelin segments. This damage reached 3.8-fold under hyperglycemic conditions, and was also associated with a robust decrease in the expression of Cav-1 and compact myelin proteins. The loss of Cav-1 and compact myelin proteins following hyperglycemia and NRG treatment was not due to neuronal loss, since the axons remained intact and there was no loss of PGP 9.5, an axonal marker protein. To examine if changes in Cav-1 were sufficient to alter the extent of NRG-induced demyelination, SC/DRG neurons co-cultures were infected with antisense or dominant-negative Cav-1(P132L) adenoviruses. Either antisense-mediated downregulation or mis-localization of endogenous Cav-1 by Cav-1(P132L) resulted in a 1.5- to 2.4-fold increase in NRG-induced degeneration compared to that present in control cultures. These data support that hyperglycemia and changes in Cav-1 are sufficient to sensitize myelinated SC/DRG co-cultures to NRG-induced demyelination.


Asunto(s)
Caveolina 1/metabolismo , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/fisiopatología , Regulación hacia Abajo/fisiología , Hiperglucemia/fisiopatología , Neurregulina-1 , Animales , Animales Recién Nacidos , Células Cultivadas , Técnicas de Cocultivo , Enfermedades Desmielinizantes/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Ganglios Espinales/citología , Glucosa/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Neurregulina-1/farmacología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Ratas , Células de Schwann/efectos de los fármacos , Células de Schwann/fisiología , Factores de Tiempo , Transfección
9.
Ai Zheng ; 24(2): 160-5, 2005 Feb.
Artículo en Chino | MEDLINE | ID: mdl-15694025

RESUMEN

BACKGROUND & OBJECTIVE: Apoptosis is a kind of evolutional high conservative cell death. Transferring high active pro-apoptotic molecules into cancer cells to induce apoptosis is a potential strategy for cancer gene therapy. Based on our previous generation of reconstructed human caspase-8, which can continuously induce apoptosis of cervical cancer cell line HeLa, by reversing its large and small subunits, this study was designed to investigate the pro-apoptotic efficiencies of 3 reconstructed human caspase-8 (Casp8CD, Rev8, and Rev8L) on HeLa cells, and to explore the feasibility of reconstructed human caspase-8 as potential apoptosis-inducing candidates. METHODS: The eukaryotic expression vectors pIRES2-EGFP carrying Casp8CD, Rev8, and Rev8L genes were transfected into HeLa cells, and breast cancer MCF-7 cells. Expressions and pro-apoptotic effects of Casp8CD, Rev8, and Rev8L genes were observed under fluorescent microscope, and their pro-apoptotic efficiencies were assessed by MTT assay and cells counting. The flexibilities of linking-peptides between subunits of Rev8 and Rev8L were analyzed by bioinformatics. RESULTS: Expressions of the 3 reconstructed caspase-8 genes were observed under fluorescent microscope, and the HeLa and MCF-7 cells expressing Rev8 or Rev8L genes displayed typical apoptotic volume decrease (AVD). MTT assay showed that compared with control cells, A(570) values of Rev8- and Rev8L-transfected cells began to decrease 20 h after transfection. Cell counting results indicated that cell death ratio of Casp8CD-, Rev8-, and Rev8L-transfected cells were 16.9%, 52.3%, and 47.7%, respectively, 24 h after transfection; and 12.9%, 51.6%,and 61.2%,respectively,48 h after transfection. Bioinformatics analysis showed that the linking-peptides between subunits of Rev8 and Rev8L were flexible. CONCLUSION: Rev8 and Rev8L molecules have similar pro-apoptotic effects and efficiencies, but over-expressed Casp8CD had no significant pro-apoptotic effects.


Asunto(s)
Apoptosis , Neoplasias de la Mama/patología , Caspasas/fisiología , Neoplasias de la Mama/metabolismo , Caspasa 8 , Caspasas/biosíntesis , Caspasas/genética , Línea Celular Tumoral , Femenino , Vectores Genéticos , Células HeLa , Humanos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Transfección
10.
J Immunol ; 173(1): 61-7, 2004 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15210759

RESUMEN

Clinical studies have suggested that human epidermal growth factor receptor-2 (HER2) provide a useful target for antitumor therapy. We previously described the generation of a chimeric HER2-targeted immunocasp-3 protein. In this study, we extend the repertoire of chimeric proapoptotic proteins with immunocasp-6, a construct that comprises a HER2-specific single-chain Ab, a single-chain Pseudomonas exotoxin A, and an active caspase-6, which can directly cleave lamin A leading to nucleus damage and inducing programmed cell death. We demonstrate that the secreted immunocasp-6 molecule selectively recognizes and induces apoptosis in HER2-overexpressing tumor cells in vitro, but not in cells with undetectable HER2. The immunocasp-6 gene was next transferred into BALB/c athymic mice bearing human breast SK-BR-3 tumors by i.m. injection of liposome-encapsulated vectors, by intratumor injection of adenoviral vectors, or by i.v. injection of PBMC modified by retroviral infection. Regardless of the method used, expression of immunocasp-6 suppressed tumor growth and prolonged animal survival significantly. Our data show that the chimeric immunocasp-6 molecule can recognize HER2-positive tumor cells, promptly attack their nucleus, and induce their apoptotic death, suggesting the potential of this strategy for the treatment of human cancers that overexpress HER2.


Asunto(s)
Anticuerpos/uso terapéutico , Caspasas/genética , Terapia Genética , Neoplasias Experimentales/terapia , Receptor ErbB-2/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/uso terapéutico , ADP Ribosa Transferasas/uso terapéutico , Adenoviridae/genética , Animales , Apoptosis , Toxinas Bacterianas/uso terapéutico , Caspasa 6 , Línea Celular Tumoral , Exotoxinas/uso terapéutico , Humanos , Células Jurkat , Liposomas , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/química , Receptor ErbB-2/análisis , Receptor ErbB-2/inmunología , Retroviridae/genética , Factores de Virulencia/uso terapéutico , Exotoxina A de Pseudomonas aeruginosa
11.
J Biol Chem ; 279(20): 21343-8, 2004 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-15004021

RESUMEN

Targeted cell killing is required for effective treatment of cancers. We previously described the generation of a chimeric immunocasp-3 protein and its potent selective antitumor activity (Jia, L. T., Zhang, L. H., Yu, C. J., Zhao, J., Xu, Y. M., Gui, J. H., Jin, M., Ji, Z. L., Wen, W. H., Wang, C. J., Chen, S. Y., and Yang, A. G. (2003) Cancer Res. 63, 3257-3262). Here we extend the repertoire of another chimeric pro-apoptotic protein immunoGrB, which comprises an anti-HER2 single-chain antibody, a Pseudomonas exotoxin A translocation domain and active granzyme B. Human lymphoma Jurkat cells transfected with the immunoGrB gene expression vector were able to produce and secrete the chimeric protein. The immunoGrB molecule selectively recognized and destroyed HER2-overexpressing tumor cells both in vitro and in nude mouse after intramuscular injection of the immunoGrB expression plasmid. Further in vivo study showed that intravenous administration of immunoGrB gene-modified lymphocytes led to suppression of HER2-overexpressing tumor growth and prolonged animal survival because of continuous secretion of immunoGrB molecules into blood and lymph fluid. These results demonstrate that the chimeric immunoGrB molecule, which is capable of antibody-directed targeting and granzyme B-mediated killing, has therapeutic potential against HER2 tumors, especially in cases in which caspase-dependent apoptosis is inhibited.


Asunto(s)
Anticuerpos/toxicidad , Neoplasias de la Mama/genética , Receptor ErbB-2/genética , Proteínas Recombinantes de Fusión/toxicidad , Serina Endopeptidasas/toxicidad , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Granzimas , Humanos , Ratones , Ratones Desnudos , Transfección , Trasplante Heterólogo
12.
Sheng Wu Gong Cheng Xue Bao ; 20(4): 501-6, 2004 Jul.
Artículo en Chino | MEDLINE | ID: mdl-15968978

RESUMEN

Translocating protein and translocating peptides have therapeutic potential against tumors by exposing the cytotoxic domains of toxic proteins to the cell cytosol. The aim of this study is to investigate the effect of N-terminally fused PE translocating peptides on granzyme B (GrBa) activity. PE II-GrBa fusion protein genes were constructed by replacing N-terminal signal and acidic dipeptide sequence of human granzyme B gene with two truncated translocating sequences of Pseudomonas exotoxin A (PE II aa 280-364/358) by recombinant PCR, and then cloned into pIND inducible expression vector. The resulting pIND-PE II-GrBa expression vectors were co-transfected with assistant plasmid pVgRXR into HeLa cells through lipofectamine, followed by selection on G418 and zeocin. The resistant cells were collected and induced with ponasterone A. Western blot analysis demonstrated that ponasterone A induction caused the expression of PE II-GrBa fusion proteins, and indirect immunofluorescence detected giant sized multinucleated cells, suggesting cytoskeletal and mitotic abnormalities as reported in our previous studies. Western blot, enzymatic activity assay and cell counting analysis indicated that two types of PE II-GrBa fusion proteins were capable of cleaving both endogenous and exogenous substrates of granzyme B, and inhibiting the growth of cells. The PE II (aa 280-358)-GrBa was shown to have higher serine protease activity and stronger growth inhibitory effect. Such inhibition was presumably associated with G2 arrest as determined by cell cycle analysis. These data prove that PE II-GrBa fusion proteins have cell inhibitory effect similar to GrBa, and that the shorter PE-derived peptide exerts less influence on GrBa activity. This study helps to optimize the construction of recombinant protein comprising translocating peptides and cytotoxic molecules for tumor cell killing.


Asunto(s)
ADP Ribosa Transferasas/farmacología , Toxinas Bacterianas/farmacología , Exotoxinas/farmacología , Granzimas/farmacología , Proteínas Recombinantes de Fusión/farmacología , Factores de Virulencia/farmacología , ADP Ribosa Transferasas/genética , Toxinas Bacterianas/genética , Proliferación Celular/efectos de los fármacos , Exotoxinas/genética , Granzimas/genética , Células HeLa , Humanos , Factores de Virulencia/genética , Exotoxina A de Pseudomonas aeruginosa
13.
Zhonghua Yi Xue Za Zhi ; 83(7): 564-8, 2003 Apr 10.
Artículo en Chino | MEDLINE | ID: mdl-12887745

RESUMEN

OBJECTIVE: To investigate the targeted killing effect to SKBr3 cells due to the expression of a secreted fusion protein consisting of anti-erbB2 antibody and reversed caspase-3. METHODS: A recombinant plasmid pCMV-e23scFv-PEII-revcasp 3 was constructed by subcloning reversed caspase-3 gene to the downstream of anti-erbB2 antibody and transfected into Jurkat cells. The cell lines which secreted expressing fusion protein stably were selected. The fusion protein in media was detected by ELISA and the media was used to culture human breast cancer SKBr3 cells. The recombinant plasmids with liposomes was administrated to BALB/C nude mouses bearing SKBr3 tumor by intramuscular injection. The targetting effect of the recombinant fusion protein caspase-3 was detected by indirect immunofluorescence staining. RESULTS: Fusion protein can be expressed and secreted by Jurkat cells stably and kill SKBr3 cells. Significant prolonged survival time (prolonged by 72%) and inhibition of tumor growth in vivo (within inhibition ratio of 77%) were seen in the group administered with recombinant plasmids. Indirect immunofluorescence staining showed that the recombinant fusion protein caspase-3 has targetting effect. CONCLUSION: Secreted expression of the fusion protein consisting of anti-erbB2 antibody and reversed caspase-3 can targetedly induce SKBr3 cells to death.


Asunto(s)
Anticuerpos/genética , Caspasas/genética , Terapia Genética , Neoplasias Experimentales/terapia , Receptor ErbB-2/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/genética , Animales , Caspasa 3 , Humanos , Células Jurkat , Ratones , Ratones Endogámicos BALB C
14.
Cancer Res ; 63(12): 3257-62, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12810656

RESUMEN

In this study, a novel approach to antitumor therapy was devised by generating a chimeric tumor-targeted killer protein, referred to as immunocasp-3, that comprises a single-chain anti-erbB2/HER2 antibody with a NH(2)-terminal signal sequence, a Pseudomonas exotoxin A translocation domain, and a constitutively active caspase-3 molecule. In principle, cells transfected with the immunocasp-3 gene would express and secrete the chimeric protein, which then binds to HER2-overexpressing tumor cells. Subsequent cleavage of the constitutively active capase-3 domain from the immunocasp-3 molecule and its release from internalized vesicles would lead to apoptotic tumor cell death. To test this strategy, we transduced human lymphoma Jurkat cells with a chimeric immunocasp-3 gene expression vector and showed that they not only expressed and secreted the fusion protein but also selectively killed tumor cells overexpressing HER2 in vitro. i.v. injection of the transduced Jurkat cells led to tumor regression in a mouse xenograft model because of continuous secretion of immunocasp-3 by the transduced cells. The growth of HER2-positive tumor cells in this model was inhibited by i.m. as well as intratumor injection of immunocasp-3 expression plasmid DNA, indicating that the immunocasp-3 molecules secreted by transfected cells have systematic antitumor activity. We conclude that the immunocasp-3 molecule, combining the properties of a tumor-specific antibody with the proapoptotic activity of a caspase, has potent and selective antitumor activity, either as cell-based therapy or as a DNA vaccine. These findings provide a compelling rationale for therapeutic protocols designed for erbB2/HER2-positive tumors.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antineoplásicos/uso terapéutico , Antígenos de Neoplasias/inmunología , Apoptosis/efectos de los fármacos , Caspasas/uso terapéutico , Inmunoconjugados/uso terapéutico , Región Variable de Inmunoglobulina/uso terapéutico , Proteínas de Neoplasias/inmunología , Receptor ErbB-2/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Antineoplásicos/farmacología , Caspasa 3 , Caspasas/farmacología , Activación Enzimática , Inducción Enzimática , Femenino , Células HeLa , Humanos , Inmunoconjugados/farmacología , Región Variable de Inmunoglobulina/farmacología , Células Jurkat/metabolismo , Células Jurkat/trasplante , Ratones , Datos de Secuencia Molecular , Neoplasias Ováricas/patología , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 19(2): 160-2, 2003 Mar.
Artículo en Chino | MEDLINE | ID: mdl-15151756

RESUMEN

AIM: To investigate the killing effect on SKOV3 cells by fusion protein HER2-specific antibody-reversed caspase-3 in the secreted form. METHODS: The reversed human caspase-3 gene was subcloned into pCMV-e23scFv-PEII-PEIII to construct recombinant eukaryotic expression vector pCMV-e23scFv-PEII-revcasp-3 and transfect Jurkat cells. The secreted expression of the in culture supernatant of transfected Jurkat cells was detected by ELISA. The suppression of the fusion protein on growth of SKOV3 cells was examined by co-culture with supernatant of transfected Jurkat cells. RESULTS: There was the secreted expression of the fusion protein in Jurkat cells. Expressed product had the activity of inducing SKOV3 cells to death. CONCLUSION: The fusion protein expressed in the secreted form can targeted towards SKOV3 cells and kill them.


Asunto(s)
Caspasa 3 , Línea Celular Tumoral , Anticuerpos/genética , Apoptosis/genética , Caspasa 3/metabolismo , Humanos , Proteínas Recombinantes de Fusión/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA