Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell Death Dis ; 13(8): 687, 2022 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-35933456

RESUMO

A significant proportion of breast cancers are driven by ErbB2/Her2 oncoprotein that they overexpress. These malignancies are typically treated with various ErbB2-targeted drugs, but many such cancers develop resistance to these agents and become incurable. Conceivably, treatment of ErbB2-positive cancers could be facilitated by use of agents blocking oncogenic signaling mechanisms downstream of ErbB2. However, current understanding of these mechanisms is limited. The ability of solid tumor cells to resist anoikis, cell death triggered by cell detachment from the extracellular matrix (ECM), is thought to be critical for 3D tumor growth. In an effort to understand the mechanisms of ErbB2-driven breast cancer cell anoikis resistance we found that detachment of non-malignant breast epithelial cells from the ECM upregulates a cell death-promoting tumor suppressor adapter protein BLNK and that ErbB2 blocks this upregulation by reducing tumor cell levels of transcription factor IRF6. We further observed that trastuzumab, a therapeutic anti-ErbB2 antibody, upregulates BLNK in human trastuzumab-sensitive but not trastuzumab-resistant ErbB2-positive breast cancer cells. Moreover, we established that BLNK promotes anoikis by activating p38 MAP kinase and that ErbB2-dependent BLNK downregulation blocks breast cancer cell anoikis. In search for pharmacological approaches allowing to upregulate BLNK in tumor cells we found that clinically approved proteasome inhibitor bortezomib upregulates IRF6 and BLNK in human breast cancer cells and inhibits their 3D growth in a BLNK-dependent manner. In addition, we found that BLNK upregulation in human ErbB2-positive breast cancer cells blocks their ability to form tumors in mice. Furthermore, we used publicly available data on mRNA levels in multiple breast cancers to demonstrate that increased BLNK mRNA levels correlate with increased relapse-free survival in a cohort of approximately 400 patients with ErbB2-positive breast cancer. In summary, we discovered a novel mechanism of ErbB2-driven 3D breast tumor growth mediated by ErbB2-dependent BLNK downregulation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias da Mama , Animais , Anoikis , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Regulação para Baixo , Feminino , Humanos , Fatores Reguladores de Interferon/metabolismo , Camundongos , RNA Mensageiro , Receptor ErbB-2/metabolismo , Trastuzumab/farmacologia
2.
Breast Cancer Res Treat ; 187(3): 743-758, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33728523

RESUMO

PURPOSE: Patients with ErbB2/Her2 oncoprotein-positive breast cancers often receive neoadjuvant therapies (NATs) containing the anti-ErbB2 antibody trastuzumab. Tumors that are still present after NATs are resected, and patients continue receiving trastuzumab. These cancers are associated with high relapse risk. Whether relapse will occur cannot be presently reliably predicted. The ability to make such predictions could improve disease management. We found previously that ErbB2 blocks breast tumor cell anoikis, apoptosis induced by cell detachment from the extracellular matrix, by downregulating the pro-apoptotic protein Irf6 and upregulating the anti-apoptotic protein Epidermal Growth Factor Receptor (EGFR) in the cells and, thus, promotes their three-dimensional growth. We now tested whether tumor levels of these proteins before and after NATs correlate with patients' relapse-free survival (RFS) and overall survival (OS). METHODS: We selected archival breast tumor samples collected from 37 women with ErbB2-positive stages II and III breast cancer before and after NATs. We used immunohistochemistry to test whether levels of the indicated proteins in respective tumors correlate with RFS and OS. RESULTS: We observed that the presence of high Irf6 levels in the tumors following NATs correlated with reduced RFS and OS. Perhaps not by coincidence, we noticed that trastuzumab-sensitive ErbB2-positive breast cancer cells selected for the ability to overproduce exogenous Irf6 in culture acquired trastuzumab resistance. Finally, EGFR presence in patients' tumors before or after NATs was associated with decreased RFS and OS. CONCLUSIONS: This study could help identify patients with ErbB2-positive tumors that are at increased risk of disease relapse following NATs.


Assuntos
Neoplasias da Mama , Anoikis , Mama/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Feminino , Humanos , Fatores Reguladores de Interferon , Recidiva Local de Neoplasia/tratamento farmacológico , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Trastuzumab
3.
Breast Cancer Res ; 22(1): 105, 2020 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-33023655

RESUMO

BACKGROUND: ErbB2/HER2 oncoprotein often drives breast cancers (BCs) which are treated with the anti-ErbB2 antibody trastuzumab. The efficacy of trastuzumab-based metastatic BC therapies is routinely assessed by imaging studies. Trastuzumab typically becomes ineffective in the case of this disease and is then replaced by other drugs. Biomarkers of BC trastuzumab response could allow imaging studies and the switch to other drugs to occur earlier than is now possible. Moreover, bone-only BC metastases can be hard to measure, and biomarkers of their trastuzumab response could facilitate further treatment decisions. Such biomarkers are presently unavailable. In this study, we searched for proteins whose levels in BC cell-emitted extracellular vesicles (EVs) potentially correlate with BC trastuzumab sensitivity. METHODS: We isolated EVs from cultured trastuzumab-sensitive and trastuzumab-resistant human BC cells before and after trastuzumab treatment and characterized these EVs by nanoparticle tracking analysis and electron microscopy. We found previously that ErbB2 drives BC by downregulating a pro-apoptotic protein PERP. We now tested whether trastuzumab-induced PERP upregulation in EVs emitted by cultured human BC cells correlates with their trastuzumab sensitivity. We also used mass spectrometry to search for additional proteins whose levels in such EVs reflect BC cell trastuzumab sensitivity. Once we identified proteins whose EV levels correlate with this sensitivity in culture, we explored the feasibility of testing whether their levels in the blood EVs of trastuzumab-treated metastatic BC patients correlate with patients' response to trastuzumab-based treatments. RESULTS: We found that neither trastuzumab nor acquisition of trastuzumab resistance by BC cells affects the size or morphology of EVs emitted by cultured BC cells. We established that EV levels of proteins PERP, GNAS2, GNA13, ITB1, and RAB10 correlate with BC cell trastuzumab response. Moreover, these proteins were upregulated during trastuzumab-based therapies in the blood EVs of a pilot cohort of metastatic BC patients that benefited from these therapies but not in those derived from patients that failed such treatments. CONCLUSIONS: Upregulation of a protein set in EVs derived from cultured breast tumor cells correlates with tumor cell trastuzumab sensitivity. It is feasible to further evaluate these proteins as biomarkers of metastatic BC trastuzumab response.


Assuntos
Biomarcadores Farmacológicos/metabolismo , Neoplasias da Mama/tratamento farmacológico , Vesículas Extracelulares/metabolismo , Proteínas de Membrana/metabolismo , Receptor ErbB-2/metabolismo , Trastuzumab/uso terapêutico , Adulto , Antineoplásicos Imunológicos/uso terapêutico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Cromograninas/metabolismo , Estudos de Coortes , Feminino , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Genes Supressores de Tumor , Humanos , Pessoa de Meia-Idade , Metástase Neoplásica , Proteômica/métodos , Receptor ErbB-2/antagonistas & inibidores , Regulação para Cima , Proteínas rab de Ligação ao GTP/metabolismo
5.
Breast Cancer Res ; 20(1): 151, 2018 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-30545388

RESUMO

BACKGROUND: The ability of solid tumor cells to resist anoikis, apoptosis triggered by cell detachment from the extracellular matrix (ECM), is thought to be critical for 3D tumor growth. ErbB2/Her2 oncoprotein is often overproduced by breast tumor cells and blocks their anoikis by partially understood mechanisms. In our effort to understand them better, we observed that detachment of nonmalignant human breast epithelial cells from the ECM upregulates the transcription factor Irf6. Irf6 is thought to play an important role in mammary gland homeostasis and causes apoptosis by unknown mechanisms. We noticed that ErbB2, when overproduced by detached breast epithelial cells, downregulates Irf6. METHODS: To test whether ErbB2 downregulates Irf6 in human ErbB2-positive breast cancer cells, we examined the effect of ErbB2 inhibitors, such as the anti-ErbB2 antibody trastuzumab or the ErbB2/epidermal growth factor receptor small-molecule inhibitor lapatinib, on Irf6 in these cells. Moreover, we performed Irf6 IHC analysis of tumor samples derived from the locally advanced ErbB2-positive breast cancers before and after neoadjuvant trastuzumab-based therapies. To examine the role of Irf6 in anoikis of nonmalignant and ErbB2-overproducing breast epithelial cells, we studied anoikis after knocking down Irf6 in the former cells by RNA interference and after overproducing Irf6 in the latter cells. To examine the mechanisms by which cell detachment and ErbB2 control Irf6 expression in breast epithelial cells, we tested the effects of genetic and pharmacological inhibitors of the known ErbB2-dependent signaling pathways on Irf6 in these cells. RESULTS: We observed that trastuzumab and lapatinib upregulate Irf6 in ErbB2-positive human breast tumor cells and that neoadjuvant trastuzumab-based therapies tend to upregulate Irf6 in human breast tumors. We found that detachment-induced Irf6 upregulation in nonmalignant breast epithelial cells requires the presence of the transcription factor ∆Np63α and that Irf6 mediates their anoikis. We showed that ErbB2 blocks Irf6 upregulation in ErbB2-overproducing cells by activating the mitogen-activated protein kinases that inhibit ∆Np63α-dependent signals required for Irf6 upregulation. Finally, we demonstrated that ErbB2-driven Irf6 downregulation in ErbB2-overproducing breast epithelial cells blocks their anoikis and promotes their anchorage-independent growth. CONCLUSIONS: We have demonstrated that ErbB2 blocks anoikis of breast epithelial cells by downregulating Irf6.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Transformação Celular Neoplásica/patologia , Fatores Reguladores de Interferon/metabolismo , Receptor ErbB-2/metabolismo , Anoikis/efeitos dos fármacos , Antineoplásicos/uso terapêutico , Biópsia , Mama/citologia , Mama/patologia , Neoplasias da Mama/tratamento farmacológico , Técnicas de Cultura de Células , Linhagem Celular , Estudos de Coortes , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Feminino , Humanos , Fatores Reguladores de Interferon/genética , Terapia Neoadjuvante/métodos , Projetos Piloto , RNA Interferente Pequeno/metabolismo , Receptor ErbB-2/antagonistas & inibidores , Trastuzumab/farmacologia , Trastuzumab/uso terapêutico , Regulação para Cima/efeitos dos fármacos
6.
Autophagy ; 14(1): 134-151, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28933585

RESUMO

Activating mutations of RAS GTPase contribute to the progression of many cancers, including colorectal carcinoma. So far, attempts to develop treatments of mutant RAS-carrying cancers have been unsuccessful due to insufficient understanding of the salient mechanisms of RAS signaling. We found that RAS downregulates the protein ATG12 in colon cancer cells. ATG12 is a mediator of autophagy, a process of degradation and reutilization of cellular components. In addition, ATG12 can kill cells via autophagy-independent mechanisms. We established that RAS reduces ATG12 levels in cancer cells by accelerating its proteasomal degradation. We further observed that RAS-dependent ATG12 loss in these cells is mediated by protein kinases MAP2K/MEK and MAPK1/ERK2-MAPK3/ERK1, known effectors of RAS. We also demonstrated that the reversal of the effect of RAS on ATG12 achieved by the expression of exogenous ATG12 in cancer cells triggers both apoptotic and nonapoptotic signals and efficiently kills the cells. ATG12 is known to promote autophagy by forming covalent complexes with other autophagy mediators, such as ATG5. We found that the ability of ATG12 to kill oncogenic RAS-carrying malignant cells does not require covalent binding of ATG12 to other proteins. In summary, we have identified a novel mechanism by which oncogenic RAS promotes survival of malignant intestinal epithelial cells. This mechanism is driven by RAS-dependent loss of ATG12 in these cells.


Assuntos
Proteína 12 Relacionada à Autofagia/metabolismo , Autofagia , Neoplasias Colorretais/metabolismo , Mucosa Intestinal/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Apoptose , Proteína 5 Relacionada à Autofagia/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Neoplasias Colorretais/patologia , Regulação para Baixo , Humanos , Mucosa Intestinal/patologia , MAP Quinase Quinase Quinases/metabolismo , Mutação , Ligação Proteica , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais
7.
Autophagy ; 11(8): 1230-46, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26061804

RESUMO

Detachment of nonmalignant intestinal epithelial cells from the extracellular matrix (ECM) triggers their growth arrest and, ultimately, apoptosis. In contrast, colorectal cancer cells can grow without attachment to the ECM. This ability is critical for their malignant potential. We found previously that detachment-induced growth arrest of nonmalignant intestinal epithelial cells is driven by their detachment-triggered autophagy, and that RAS, a major oncogene, promotes growth of detached cells by blocking such autophagy. In an effort to identify the mechanisms of detachment-induced autophagy and growth arrest of nonmalignant cells we found here that detachment of these cells causes upregulation of ATG3 and that ATG3 upregulation contributes to autophagy and growth arrest of detached cells. We also observed that when ATG3 expression is artificially increased in the attached cells, ATG3 promotes neither autophagy nor growth arrest but triggers their apoptosis. ATG3 upregulation likely promotes autophagy of the detached but not that of the attached cells because detachment-dependent autophagy requires other detachment-induced events, such as the upregulation of ATG7. We further observed that those few adherent cells that do not die by apoptosis induced by ATG3 become resistant to apoptosis caused by cell detachment, a property that is critical for the ability of normal epithelial cells to become malignant. We conclude that cell-ECM adhesion can switch ATG3 functions: when upregulated in detached cells in the context of other autophagy-promoting events, ATG3 contributes to autophagy. However, when overexpressed in the adherent cells, in the circumstances not favoring autophagy, ATG3 triggers apoptosis.


Assuntos
Apoptose , Autofagia , Células Epiteliais/metabolismo , Matriz Extracelular/metabolismo , Intestinos/citologia , Enzimas de Conjugação de Ubiquitina/metabolismo , Animais , Proteína 7 Relacionada à Autofagia , Proteínas Relacionadas à Autofagia , Adesão Celular , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Regulação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Intestino Delgado/patologia , Proteínas Associadas aos Microtúbulos/metabolismo , Peptídeo Sintases/metabolismo , Interferência de RNA , Ratos , Enzimas Ativadoras de Ubiquitina/metabolismo , Regulação para Cima
8.
Curr Chem Genom Transl Med ; 8(Suppl 1): 27-35, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24596682

RESUMO

The multi cellular tumor spheroid (MCTS) model has been used for decades with proven superiority over monolayer cell culture models at recapitulating in vivo tumor growth. Yet its use in high-throughput drug discovery has been limited, particularly with image based screening, due to practical and technical hurdles. Here we report a significant advance in utilizing live MCTS models for high-content image based drug discovery. Using a validated GFP reporter (CK5Pro-GFP) of luminal breast cancer stem cells (CSC), we developed an algorithm to quantify changes in CK5Pro-GFP expression levels for individual Z-stack planes (local) or as maximal projections of the summed Z-stacks (global) of MCTS. From these image sets, we can quantify the cross-sectional area of GFP positive cells, the fluorescence intensity of the GFP positive cells, and the percent of spheroid cross-sectional area that expresses CK5Pro-GFP.We demonstrate that acquiring data in this manner can be done in real time and is statistically robust (Z'=0.85) for use in primary high-content screening cancer drug discovery.

9.
Horm Cancer ; 4(1): 36-49, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23184698

RESUMO

Progestins play a deleterious role in the onset of breast cancer, yet their influence on existing breast cancer and tumor progression is not well understood. In luminal estrogen receptor (ER)- and progesterone receptor (PR)-positive breast cancer, progestins induce a fraction of cells to express cytokeratin 5 (CK5), a marker of basal epithelial and progenitor cells in the normal breast. CK5(+) cells lose expression of ER and PR and are relatively quiescent, increasing their resistance to endocrine and chemotherapy compared to intratumoral CK5(-)ER(+)PR(+) cells. Characterization of live CK5(+) cells has been hampered by a lack of means for their direct isolation. Here, we describe optical (GFP) and bioluminescent (luciferase) reporter models to quantitate and isolate CK5(+) cells in luminal breast cancer cell lines utilizing the human KRT5 gene promoter and a viral vector approach. Using this system, we confirmed that the induction of GFP(+)/CK5(+) cells is specific to progestins, is dependent on PR, can be blocked by antiprogestins, and does not occur with other steroid hormones. Progestin-induced, fluorescence-activated cell sorting-isolated CK5(+) cells had lower ER and PR mRNA, were slower cycling, and were relatively more invasive and sphere forming than their CK5(-) counterparts in vitro. Repeated progestin treatment and selection of GFP(+) cells enriched for a persistent population of CK5(+) cells, suggesting that this transition can be semi-permanent. These data support that in PR(+) breast cancers, progestins induce a subpopulation of CK5(+)ER(-)PR(-) cells with enhanced progenitor properties and have implications for treatment resistance and recurrence in luminal breast cancer.


Assuntos
Neoplasias da Mama/patologia , Queratina-5/biossíntese , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Progesterona/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo/métodos , Células HEK293 , Humanos , Queratina-5/genética , Queratina-5/metabolismo , Células MCF-7 , Células-Tronco Neoplásicas/metabolismo , Progestinas/farmacologia , Regiões Promotoras Genéticas/genética , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
10.
J Biomol Screen ; 17(9): 1211-20, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22751729

RESUMO

Breast cancers expressing hormone receptors for estrogen (ER) and progesterone (PR) represent ~70% of all cases and are treated with both ER-targeted and chemotherapies, with near 40% becoming resistant. We have previously described that in some ER(+) tumors, the resistant cells express cytokeratin 5 (CK5), a putative marker of breast stem and progenitor cells. CK5(+) cells have lost expression of ER and PR, express the tumor-initiating cell surface marker CD44, and are relatively quiescent. In addition, progestins, which increase breast cancer incidence, expand the CK5(+) subpopulation in ER(+)PR(+) breast cancer cell lines. We have developed models to induce and quantitate CK5(+)ER(-)PR(-) cells, using CK5 promoter-driven luciferase (Fluc) or green fluorescent protein (GFP) reporters stably transduced into T47D breast cancer cells (CK5Pro-GFP or CK5Pro-Luc). We validated the CK5Pro-GFP-T47D model for high-content screening in 96-well microplates and performed a pilot screen using a focused library of 280 compounds from the National Institutes of Health clinical collection. Four hits were obtained that significantly abrogated the progestin-induced CK5(+) cell population, three of which were members of the retinoid family. Hence, this approach will be useful in discovering small molecules that could potentially be developed as combination therapies, preventing the acquisition of a drug-resistant subpopulation.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Descoberta de Drogas/métodos , Resistencia a Medicamentos Antineoplásicos , Células-Tronco Neoplásicas/efeitos dos fármacos , Retinoides/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Acitretina/farmacologia , Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Receptores de Hialuronatos/metabolismo , Isotretinoína/farmacologia , Queratina-5/metabolismo , Luciferases/genética , Luciferases/metabolismo , Miconazol/farmacologia , Células-Tronco Neoplásicas/metabolismo , Progesterona/farmacologia , Progestinas/farmacologia , Regiões Promotoras Genéticas , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Tretinoína/farmacologia
11.
Expert Opin Drug Discov ; 7(9): 819-30, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22788761

RESUMO

INTRODUCTION: For the past 30 years 2D-cell-based assay models have dominated preclinical cancer drug discovery efforts. 2D-cell-based models fail to predict in vivo efficacy, contributing to a lower success rate and higher cost required to translate an investigational new drug to clinical approval. Technological advances in 3D-cell culture models bridge the gap between 2D and in vivo models to improve upon the current success rates of cancer drug discovery. AREAS COVERED: This review focuses on the multicellular tumor spheroid (MCTS), particularly how this model can be utilized for HTS drug discovery. We discuss the current technologies for uniform culture of MCTS suitable for HTS and detection methods utilized for assay development and drug screening. EXPERT OPINION: Substantial hurdles remain before we reach the ultimate goal of robust HTS of large compound libraries with MCTS models. Specifically, we can group these challenges into three categories: MCTS growth, data collection, and data analysis. The MCTS model should be utilized with fluorescent readouts and high-content imaging with a systems biology approach to model human tumors in vitro. Such models will be more predictive of in vivo efficacy, improving on the current success rates of cancer drug discovery from bench to bedside.


Assuntos
Antineoplásicos/farmacologia , Descoberta de Drogas/métodos , Neoplasias/tratamento farmacológico , Esferoides Celulares/efeitos dos fármacos , Animais , Desenho de Fármacos , Descoberta de Drogas/tendências , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Modelos Biológicos , Células Tumorais Cultivadas
12.
Int J Cancer ; 131(2): 357-66, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21834075

RESUMO

Resistance of carcinoma cells to anoikis, apoptosis that is normally induced by detachment of nonmalignant epithelial cells from the extracellular matrix, is thought to be critical for carcinoma progression. Molecular mechanisms that control anoikis of nonmalignant and cancer cells are understood poorly. In an effort to understand them we found that detachment of nonmalignant intestinal epithelial cells triggers upregulation of Chk2, a pro-apoptotic protein kinase that has never been implicated in anoikis and has been thought to kill cells mainly under the conditions compromising genome integrity. We found that enforced downregulation of Chk2 protects intestinal epithelial cells from anoikis. Chk2 can kill cells by stabilizing p53 tumor suppressor protein or via p53-independent mechanisms, and we established that Chk2-mediated anoikis of intestinal epithelial cells is p53-independent. We further found that, unlike nonmalignant intestinal epithelial cells whose anoikis is triggered by detachment-induced Chk2 upregulation, intestinal epithelial cells carrying oncogenic ras, a known inhibitor of anoikis, remain anoikis-resistant in response to enforced Chk2 upregulation. By contrast, drugs, such as topoisomerase I inhibitors, that can kill cells via Chk2-indpendent mechanisms, efficiently triggered anoikis of ras-transformed cells. Thus, oncogenic ras can prevent Chk2 from triggering anoikis even when levels of this protein kinase are elevated in cancer cells, and the use of therapeutic agents that kill cells in a Chk-2-independent, rather than Chk-2-dependent, manner could represent an efficient strategy for overcoming ras-induced anoikis resistance of these cells. We conclude that Chk-2 is an important novel component of anoikis-promoting machinery of intestinal epithelial cells.


Assuntos
Anoikis , Células Epiteliais/citologia , Mucosa Intestinal/citologia , Proteína Oncogênica p21(ras)/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Anoikis/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular , Transformação Celular Neoplásica , Quinase do Ponto de Checagem 2 , Matriz Extracelular/metabolismo , Camundongos , Proteína Oncogênica p21(ras)/genética , Interferência de RNA , RNA Interferente Pequeno , Ratos , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Mar Drugs ; 9(11): 2397-2408, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22163192

RESUMO

Type IIα DNA topoisomerase (TopoIIα) is among the most important clinical drug targets for the treatment of cancer. Recently, the DNA repair protein Metnase was shown to enhance TopoIIα activity and increase resistance to TopoIIα poisons. Using in vitro DNA decatenation assays we show that neoamphimedine potently inhibits TopoIIα-dependent DNA decatenation in the presence of Metnase. Cell proliferation assays demonstrate that neoamphimedine can inhibit Metnase-enhanced cell growth with an IC(50) of 0.5 µM. Additionally, we find that the apparent K(m) of TopoIIα for ATP increases linearly with higher concentrations of neoamphimedine, indicating ATP-competitive inhibition, which is substantiated by molecular modeling. These findings support the continued development of neoamphimedine as an anticancer agent, particularly in solid tumors that over-express Metnase.


Assuntos
Acridinas/farmacologia , Trifosfato de Adenosina/metabolismo , Antígenos de Neoplasias/efeitos dos fármacos , DNA Topoisomerases Tipo II/efeitos dos fármacos , Proteínas de Ligação a DNA/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/metabolismo , Acridinas/administração & dosagem , Antígenos de Neoplasias/metabolismo , Proliferação de Células/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Técnicas In Vitro , Concentração Inibidora 50 , Modelos Moleculares
14.
J Biol Chem ; 286(45): 38894-903, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21903589

RESUMO

Resistance of carcinoma cells to anoikis, apoptosis that is normally induced by loss of cell-to-extracellular matrix adhesion, is thought to be essential for the ability of these cells to form primary tumors, invade adjacent tissues, and metastasize to distant organs. Current knowledge about the mechanisms by which cancer cells evade anoikis is far from complete. In an effort to understand these mechanisms, we found that ras, a major oncogene, down-regulates protease caspase-2 (which initiates certain steps of the cellular apoptotic program) in malignant human and rat intestinal epithelial cells. This down-regulation could be reversed by inhibition of a protein kinase Mek, a mediator of Ras signaling. We also found that enforced down-regulation of caspase-2 in nonmalignant intestinal epithelial cells by RNA interference protected them from anoikis. Furthermore, the reversal of the effect of Ras on caspase-2 achieved by the expression of exogenous caspase-2 in detached ras-transformed intestinal epithelial cells promoted well established apoptotic events, such as the release of the pro-apoptotic mitochondrial factors cytochrome c and HtrA2/Omi into the cytoplasm of these cells, significantly enhanced their anoikis susceptibility, and blocked their long term growth in the absence of adhesion to the extracellular matrix. Finally, the blockade of the effect of Ras on caspase-2 substantially suppressed growth of tumors formed by the ras-transformed cells in mice. We conclude that ras-induced down-regulation of caspase-2 represents a novel mechanism by which oncogenic Ras protects malignant intestinal epithelial cells from anoikis, promotes their anchorage-independent growth, and allows them to form tumors in vivo.


Assuntos
Caspase 2/biossíntese , Transformação Celular Neoplásica/metabolismo , Cisteína Endopeptidases/biossíntese , Regulação para Baixo , Células Epiteliais/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Mucosa Intestinal/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas ras/metabolismo , Animais , Anoikis/genética , Caspase 2/genética , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Cisteína Endopeptidases/genética , Citocromos c/genética , Citocromos c/metabolismo , Citoplasma/genética , Citoplasma/metabolismo , Citoplasma/patologia , Células Epiteliais/patologia , Serina Peptidase 2 de Requerimento de Alta Temperatura A , Humanos , Mucosa Intestinal/patologia , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Proteínas de Neoplasias/genética , Ratos , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Transdução de Sinais/genética , Proteínas ras/genética
15.
BMC Bioinformatics ; 11: 252, 2010 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-20470414

RESUMO

BACKGROUND: The enumeration of tetrameric and other sequence motifs that are positively or negatively correlated with in vivo antisense DNA effects has been a useful addition to the arsenal of information needed to predict effective targets for antisense DNA control of gene expression. Such retrospective information derived from in vivo cellular experiments characterizes aspects of the sequence dependence of antisense inhibition that are not predicted by nearest-neighbor (NN) thermodynamic parameters derived from in vitro experiments. However, quantitation of the antisense contributions of motifs is problematic, since individual motifs are not isolated from the effects of neighboring nucleotides, and motifs may be overlapping. These problems are circumvented by a next-nearest-neighbor (NNN) analysis of antisense DNA effects in which the overlapping nature of nearest-neighbors is taken into account. RESULTS: Next-nearest-neighbor triplet combinations of nucleotides are the simplest that include overlapping sequence effects and therefore can encompass interactions beyond those of nearest neighbors. We used singular value decomposition (SVD) to fit experimental data from our laboratory in which phosphorothioate-modified antisense DNAs (S-DNAs) 20 nucleotides long were used to inhibit cellular protein expression in 112 experiments involving four gene targets and two cell lines. Data were fitted using a NNN model, neglecting end effects, to derive NNN inhibition parameters that could be combined to give parameters for a set of 49 sequences that represents the inhibitory effects of all possible overlapping triplet interactions in the cellular targets of these antisense S-DNAs. We also show that parameters to describe subsets of the data, such as the mRNAs being targeted and the cell lines used, can be included in such a derivation. While NNN triplet parameters provided an adequate model to fit our data, NN doublet parameters did not. CONCLUSIONS: The methodology presented illustrates how NNN antisense inhibitory information can be derived from in vivo cellular experiments. Subsequent calculations of the antisense inhibitory parameters for any mRNA target sequence automatically take into account the effects of all possible overlapping combinations of nearest-neighbors in the sequence. This procedure is more robust than the tallying of tetrameric motifs that have positive or negative antisense effects. The specific parameters derived in this work are limited in their applicability by the relatively small database of experiments that was used in their derivation.


Assuntos
DNA Antissenso/química , Sequência de Bases , Células Cultivadas , Bases de Dados de Ácidos Nucleicos , Proteínas/química , Termodinâmica
16.
J Biol Chem ; 285(8): 5438-49, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-19778902

RESUMO

Detachment of non-malignant epithelial cells from the extracellular matrix causes their growth arrest and, ultimately, death. By contrast, cells composing carcinomas, cancers of epithelial origin, can survive and proliferate without being attached to the extracellular matrix. These properties of tumor cells represent hallmarks of malignant transformation and are critical for cancer progression. Previously we identified several mechanisms by which ras, a major oncogene, blocks detachment-induced apoptosis of intestinal epithelial cells, but mechanisms by which Ras promotes proliferation of those cells that remain viable following detachment are unknown. We show here that detachment of non-malignant intestinal epithelial cells promotes formation of autophagosomes, vacuole-like structures that mediate autophagy (a process of cellular self-cannibalization), and that oncogenic ras prevents this autophagosome formation. We also found that ras activates a GTPase RhoA, that RhoA promotes activation of a protease calpain, and that calpain triggers degradation of Beclin-1, a critical mediator of autophagy, in these cells. The reversal of the effect of ras on Beclin-1 (achieved by expression of exogenous Beclin-1) promoted autophagosome formation following cell detachment, significantly reduced the fraction of detached cells in the S phase of the cell cycle and their rate of proliferation without affecting their viability. Furthermore, RNA interference-induced Beclin-1 down-regulation in non-malignant intestinal epithelial cells prevented detachment-dependent reduction of the fraction of these cells in the S phase of the cell cycle. Thus, ras oncogene promotes proliferation of those malignant intestinal epithelial cells that remain viable following detachment via a distinct novel mechanism that involves Ras-induced down-regulation of Beclin-1.


Assuntos
Proteínas Reguladoras de Apoptose/biossíntese , Autofagia , Transformação Celular Neoplásica/metabolismo , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Neoplasias Intestinais/metabolismo , Proteínas de Membrana/biossíntese , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas Reguladoras de Apoptose/genética , Proteína Beclina-1 , Transformação Celular Neoplásica/genética , Regulação para Baixo/genética , Humanos , Neoplasias Intestinais/genética , Proteínas de Membrana/genética , Fagossomos/genética , Fagossomos/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Fase S/genética , Células Tumorais Cultivadas , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
17.
Autophagy ; 5(8): 1166-79, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19901551

RESUMO

Disruption of tumor blood supply causes tumor hypoxia. Hypoxia can induce cell death, but cancer cells that remain viable in the absence of oxygen often possess an increased survival potential, and tumors formed by these cells tend to grow particularly aggressively. Thus, developing approaches aimed at increasing the susceptibility of malignant cells to hypoxia-induced death represents a potentially important avenue for cancer treatment. Molecular mechanisms that control the survival of cancer cells under hypoxia are not well understood. In an effort to understand them we found that hypoxia downregulates Beclin 1, a mediator of autophagy, in malignant intestinal epithelial cells. The reversal of this downregulation promoted autophagosome accumulation, enhanced the activation of a pro-apoptotic protease caspase-9 and subsequent caspase-9-dependent activation of two other pro-apoptotic proteases caspases 3 and 7 in these cells. Furthermore, the reversal of hypoxia-induced downregulation of Beclin 1-stimulated caspase-9-dependent apoptosis of the indicated cells under hypoxia. Interestingly, we found that Beclin 1-dependent caspase-9 activation in hypoxic cells was not associated with an increased release of cytochrome c from the mitochondria to the cytoplasm (such release represents a frequently occurring mechanism for caspase-9 activation). We also observed that Beclin 1-dependent apoptosis of hypoxic malignant cells was independent of FADD, a mediator of death receptor signaling. We conclude that hypoxia triggers a feedback mechanism that delays apoptosis of oxygen-deprived malignant intestinal epithelial cells and is driven by hypoxia-induced Beclin 1 downregulation. Thus, approaches aimed at the disruption of this mechanism can be expected to enhance the susceptibility of such cells to hypoxia-induced apoptosis.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose , Autofagia , Regulação para Baixo , Células Epiteliais/patologia , Intestinos/patologia , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Proteína Beclina-1 , Camptotecina/análogos & derivados , Camptotecina/farmacologia , Caspases/metabolismo , Hipóxia Celular/efeitos dos fármacos , Citocromos c/metabolismo , Regulação para Baixo/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Proteína de Domínio de Morte Associada a Fas/metabolismo , Serina Peptidase 2 de Requerimento de Alta Temperatura A , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Irinotecano , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Proteínas Mitocondriais/metabolismo , Oxigênio/metabolismo , Ratos , Serina Endopeptidases/metabolismo , Vacúolos/efeitos dos fármacos , Vacúolos/metabolismo
18.
J Biol Chem ; 284(4): 2012-22, 2009 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-18957423

RESUMO

Resistance of solid tumor cells to anoikis, apoptosis induced by cell detachment from the extracellular matrix, is thought to be critical for the ability of these cells to grow anchorage independently within three-dimensional tumor masses and from metastases. Beta-catenin, a major oncoprotein, can inhibit anoikis of cancer cells via unknown mechanisms. In an effort to identify these mechanisms we found that beta-catenin blocks anoikis of malignant kidney and intestinal epithelial cells and promotes their anchorage-independent growth by down-regulating death-associated protein kinase-2 (DAPk-2), a pro-apoptotic protein whose cellular functions have so far remained unexplored. We found that beta-catenin-induced down-regulation of DAPk-2 requires the presence of the transcription factor Tcf-4, a known mediator of beta-catenin signaling. We also observed that DAPk-2 contributes to the execution of anoikis of the non-malignant epithelial cells. Thus, beta-catenin-induced down-regulation of DAPk-2 represents a novel signaling mechanism by which beta-catenin promotes the survival of malignant epithelial cells following their detachment from the ECM and enables these cells to grow in an anchorage-independent manner.


Assuntos
Anoikis , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Neoplasias do Colo/metabolismo , Regulação para Baixo , Células Epiteliais/metabolismo , Neoplasias Renais/metabolismo , beta Catenina/metabolismo , Proteínas Reguladoras de Apoptose/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Linhagem Celular , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas Quinases Associadas com Morte Celular , Células Epiteliais/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Renais/genética , Neoplasias Renais/patologia , Mutação/genética , Ligação Proteica , RNA Interferente Pequeno/genética , Fator de Transcrição 4 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica/genética , beta Catenina/genética
19.
Nucleic Acids Res ; 32(6): 2008-16, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15064360

RESUMO

Antisense oligodeoxynucleotides (ODNs) have biological activity in treating various forms of cancer. The antisense effects of two types of 20mer ODNs, phosphorothioate-modified ODNs (S-ODNs) and S-ODNs with 12 2'-O-methyl groups (Me-S-ODNs), targeted to sites 109 and 277 of bcl-2 mRNA, were compared. Both types were at least as effective as G3139 (Genta, Inc.) in reducing the level of Bcl-2 protein in T24 cells following a 4 h transfection at a dose of 0.1 micro M. Circular dichroism spectra showed that both types formed A-form duplexes with the complementary RNA, and the melting temperatures were in the order of Me-S-ODN.RNA > normal DNA.RNA > S-ODN.RNA. In comparison with the S-ODN, the Me-S-ODN had reduced toxic growth inhibitory effects, was less prone to bind the DNA-binding domain A of human replication protein A, and was as resistant to serum nucleases. Neither type of oligomer induced apoptosis, according to a PARP-cleavage assay. Hybrids formed with Me-S-ODN sequences were less sensitive to RNase H degradation than those formed with S-ODN sequences. Despite this latter disadvantage, the addition of 2'-O-methyl groups to a phosphorothioate-modified ODN is advantageous because of increased stability of binding and reduced non-specific effects.


Assuntos
Oligonucleotídeos Antissenso/química , Oligonucleotídeos Antissenso/farmacologia , Tionucleotídeos/química , Apoptose , Divisão Celular , Linhagem Celular Tumoral , Dicroísmo Circular , DNA/biossíntese , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Humanos , Metilação , Oligonucleotídeos Antissenso/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA/química , RNA Mensageiro/metabolismo , Proteína de Replicação A , Ribonuclease H/metabolismo , Temperatura
20.
Kisaengchunghak Chapchi ; 23(1): 58-72, 1985 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12888687

RESUMO

A histopathological study was carried out on the duodenum of mice and rats experimentally infected by F. seoulensis. Each mouse was infected wit 500 metacercariae and killed after 1, 2, 3 days, 1 and 2 weeks from infection. Each rat was given 1,000 metacercariae and was examined after 1, 2, and 4 weeks from infection. The duodenal tissue sections of mice and rats were stained with hematoxylin and eosin, and PAS stained for the rats of 1 week group. The pathological findings are summarized as below. There were no differences in mucosal findings between the mice and the rats, adn between the location of duodenum, 1 and 5 cm distal to the pylorus. Each worm embraced a villus exclusively with its foliate forebody which was inserted into the intervillous spaces. The fluke pinched villous epithelia using its oral and ventral suckers. The tribocytic organ destroyed the villous epithelia deeply up to the stroma after 3 days from infection. Apparent villous changes were observed in the mice after 3 days from infection. Villous changes were shortening, widening, blunting or fusion. The villous stroma showed edema, microscopic hemorrhage, capillary congestion, dilatation of lymphatics and inflammatory cell infiltration. The cells were lymphocytes, plasma cells, eosinophils and giant cells. Rarely submucosal and transmural inflammation was encountered.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...