Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Oncol Rep ; 37(1): 617-621, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27878300

RESUMEN

As part of the efforts to drug repurposing, some HIV drugs have recently been identified to exert anticancer effects. Selected nucleoside analogues of nucleosidic reverse-transcriptase inhibitors (NRTIs) have been shown to interfere with RNA transcription of HI viruses as well as with the replication of DNA in cancer cells. Non-nucleosidic reverse transcriptase inhibitors (NNRTIs) are believed to have less effects on human DNA replication and, thus, on cancer cell proliferation. Assessment of the effect of the NNRTI efavirenz in human cancer cells, however, revealed a high sensitivity of leukemia cells to this agent at pharmacologically relevant concentrations of less than 10 µg/ml. Cell death induced by efavirenz was caused by apoptosis, as shown by FACScan analysis (Annexin binding) and western blot analysis (cleavage of caspases and PARP). Western blot analyses also revealed a pronounced activation and phosphorylation of the DNA damage marker proteins p53, chk2 and H2AX, indicating DNA replication and genomic integrity as primary targets of efavirenz in leukemia cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzoxazinas/farmacología , Daño del ADN/efectos de los fármacos , Leucemia/patología , Alquinos , Apoptosis/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Ciclopropanos , Células HL-60 , Humanos , Células Jurkat , Leucemia/genética , Leucemia/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Células Tumorales Cultivadas , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
2.
Eur J Cell Biol ; 95(10): 389-400, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27567537

RESUMEN

Functional protein homeostasis is essential for the maintenance of normal cellular physiology, cell growth, and cell survival. Proteasome inhibition in cancer cells can disturb protein homeostasis in such a way that synthetic proteasome inhibitors like bortezomib may selectively kill myeloma cells. Solid cancer cells appear to respond less to bortezomib which may in part be due to a rescue mechanism of the unfolded protein response/endoplasmic reticulum stress mechanism which leads to a temporary shutdown of protein biosynthesis at the translational level. Here we show that proteasome inhibition by bortezomib may also interfere with general protein biosynthesis already at the stage of nucleolar ribosome biogenesis. Ultrastructural analysis revealed not only that bortezomib induces conspicuous changes in cytoplasmic morphology but also pronounced morphological changes of the nucleolar ultrastructure, associated with an accumulation of the transcription factor ATF4 at nucleolar sites. Stress-induced intra-nucleolar ATF4 accumulation was observed in cancer cells in a dose and time dependent manner and ultrastructural studies revealed that ATF4 is preferentially localized inside the dense fibrillar and granular component of nucleoli. Furthermore, bortezomib affected not only the number of nucleoli, but also the volume and distribution of nucleolar components. The localization of ATF4 in the granular component of nucleoli together with its association with nascent RNA transcripts in cells undergoing proteotoxic cell stress could suggest a new function for ATF4 in cell stress management.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Bortezomib/farmacología , ARN Ribosómico/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Nucléolo Celular/efectos de los fármacos , Nucléolo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Células HeLa , Humanos , Células MCF-7 , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Inhibidores de Proteasoma/farmacología , Transducción de Señal/efectos de los fármacos , Estrés Fisiológico/fisiología
3.
Onco Targets Ther ; 9: 2535-45, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27175087

RESUMEN

To improve the prognosis of glioblastoma, we developed an adjuvant treatment directed to a neglected aspect of glioblastoma growth, the contribution of nonmalignant monocyte lineage cells (MLCs) (monocyte, macrophage, microglia, dendritic cells) that infiltrated a main tumor mass. These nonmalignant cells contribute to glioblastoma growth and tumor homeostasis. MLCs comprise of approximately 10%-30% of glioblastoma by volume. After integration into the tumor mass, these become polarized toward an M2 immunosuppressive, pro-angiogenic phenotype that promotes continued tumor growth. Glioblastoma cells initiate and promote this process by synthesizing 13 kDa MCP-1 that attracts circulating monocytes to the tumor. Infiltrating monocytes, after polarizing toward an M2 phenotype, synthesize more MCP-1, forming an amplification loop. Three noncytotoxic drugs, an antibiotic - minocycline, an antihypertensive drug - telmisartan, and a bisphosphonate - zoledronic acid, have ancillary attributes of MCP-1 synthesis inhibition and could be re-purposed, singly or in combination, to inhibit or reverse MLC-mediated immunosuppression, angiogenesis, and other growth-enhancing aspects. Minocycline, telmisartan, and zoledronic acid - the MTZ Regimen - have low-toxicity profiles and could be added to standard radiotherapy and temozolomide. Re-purposing older drugs has advantages of established safety and low drug cost. Four core observations support this approach: 1) malignant glioblastoma cells require a reciprocal trophic relationship with nonmalignant macrophages or microglia to thrive; 2) glioblastoma cells secrete MCP-1 to start the cycle, attracting MLCs, which subsequently also secrete MCP-1 perpetuating the recruitment cycle; 3) increasing cytokine levels in the tumor environment generate further immunosuppression and tumor growth; and 4) MTZ regimen may impede MCP-1-driven processes, thereby interfering with glioblastoma growth.

4.
Cardiovasc Toxicol ; 16(1): 90-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25666561

RESUMEN

The non-nucleoside reverse transcriptase inhibitor efavirenz is a widely prescribed antiretroviral drug used in combined antiretroviral therapy. Despite being an essential and life-saving medication, the required lifelong use of HIV drugs has been associated with a variety of adverse effects, including disturbances in lipid metabolism and increased cardiovascular risk. Efavirenz belongs to those HIV drugs for which cardiovascular and endothelial dysfunctions have been reported. It is here shown that elevated concentrations of efavirenz can inhibit endothelial meshwork formation on extracellular matrix gels by normal and immortalized human umbilical vein cells. This inhibition was associated with an increase in oxidative stress markers, endoplasmic reticulum (ER) stress markers, and autophagy. Induction of ER stress occurred at pharmacologically relevant concentrations of efavirenz and resulted in reduced proliferation and cell viability of endothelial cells, which worsened in the presence of elevated efavirenz concentrations. In combination with the HIV protease inhibitor nelfinavir, both oxidative stress and ER stress became elevated in endothelial cells. These data indicate that pharmacologically relevant concentrations of efavirenz can impair cell viability of endothelial cells and that these effects may be aggravated by either elevated concentrations of efavirenz or by a combined use of efavirenz with other oxidative stress-inducing medications.


Asunto(s)
Fármacos Anti-VIH/toxicidad , Autofagia/efectos de los fármacos , Benzoxazinas/toxicidad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Estrés Oxidativo/efectos de los fármacos , Inhibidores de la Transcriptasa Inversa/toxicidad , Alquinos , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclopropanos , Inhibidores de la Proteasa del VIH/toxicidad , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Microtúbulos/efectos de los fármacos , Microtúbulos/patología , Nelfinavir/toxicidad
5.
Front Oncol ; 5: 47, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25759792

RESUMEN

The application of cytostatic drugs targeting the high proliferation rates of cancer cells is currently the most commonly used treatment option in cancer chemotherapy. However, severe side effects and resistance mechanisms may occur as a result of such treatment, possibly limiting the therapeutic efficacy of these agents. In recent years, several therapeutic strategies have been developed that aim at targeting not the genomic integrity and replication machinery of cancer cells but instead their protein homeostasis. During malignant transformation, the cancer cell proteome develops vast aberrations in the expression of mutated proteins, oncoproteins, drug- and apoptosis-resistance proteins, etc. A complex network of protein quality-control mechanisms, including chaperoning by heat shock proteins (HSPs), not only is essential for maintaining the extravagant proteomic lifestyle of cancer cells but also represents an ideal cancer-specific target to be tackled. Furthermore, the high rate of protein synthesis and turnover in certain types of cancer cells can be specifically directed by interfering with the proteasomal and autophagosomal protein recycling and degradation machinery, as evidenced by the clinical application of proteasome inhibitors. Since proteins with loss of their native conformation are prone to unspecific aggregations and have proved to be detrimental to normal cellular function, specific induction of misfolded proteins by HSP inhibitors, proteasome inhibitors, hyperthermia, or inducers of endoplasmic reticulum stress represents a new method of cancer cell killing exploitable for therapeutic purposes. This review describes drugs - approved, repurposed, or under investigation - that can be used to accumulate misfolded proteins in cancer cells, and particularly focuses on the molecular aspects that lead to the cytotoxicity of misfolded proteins in cancer cells.

6.
Reprod Toxicol ; 50: 122-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25461911

RESUMEN

Combined antiretroviral therapy has proven efficacy in decreasing vertical HIV transmission. However, endoplasmic reticulum stress is a known side effect of HIV protease inhibitors. We investigated endoplasmic reticulum stress in placentas of HIV-infected and uninfected mothers by PCR-based splicing analysis of the specific endoplasmic reticulum stress marker XBP1 in post-delivery placental samples of uninfected mothers and in HIV-infected mothers taking antiretroviral therapy. No elevated XBP1 splicing could be detected in placentas of uninfected mothers and most of the mothers receiving combined anti-retroviral therapy. However, markedly elevated XBP1 splicing was found in the placentas of three individuals on combined antiviral therapy, all receiving lopinavir or atazanavir. In vitro experiments confirmed induction of endoplasmic reticulum stress by lopinavir and atazanavir in trophoblast-derived cell lines. Since endoplasmic reticulum stress occurred in selective patients only, individual differences in susceptibility of HIV-infected mothers to protease inhibitor induced endoplasmic reticulum stress can be postulated.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , Inhibidores de la Proteasa del VIH/efectos adversos , Placenta/metabolismo , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Adulto , Autofagia , Células Cultivadas , Proteínas de Unión al ADN/genética , Femenino , Humanos , Embarazo , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/genética , Proteína 1 de Unión a la X-Box
7.
Cancer Metastasis Rev ; 33(4): 943-51, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25319202

RESUMEN

The family of metastasis-associated (MTA) genes is a small group of transcriptional co-regulators which are involved in various physiological functions, ranging from lymphopoietic cell differentiation to the development and maintenance of epithelial cell adhesions. By recruiting histone-modifying enzymes to specific promoter sequences, MTA proteins can function both as transcriptional repressors and activators of a number of cancer-relevant proteins, including Snail, E-cadherin, signal transducer and activator of transcriptions (STATs), and the estrogen receptor. Their involvement in the epithelial-mesenchymal transition process and regulatory interactions with estrogen receptor activity has made MTA proteins highly interesting research candidates, especially in the field of hormone-sensitive breast cancer and malignancies of the female reproductive tract. This review focuses on the current knowledge about the function and regulation of MTA1 and MTA3 proteins in gynecological cancer, including ovarian, endometrial, and cervical tumors.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Neoplasias de los Genitales Femeninos/genética , Histona Desacetilasas/genética , Proteínas de Neoplasias/genética , Proteínas Represoras/genética , Diferenciación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Neoplasias de los Genitales Femeninos/patología , Histona Desacetilasas/biosíntesis , Humanos , Proteínas de Neoplasias/biosíntesis , Receptores de Estrógenos/metabolismo , Proteínas Represoras/biosíntesis , Transactivadores
8.
Gynecol Oncol ; 135(2): 342-8, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25178996

RESUMEN

OBJECTIVE: Therapeutic options are limited for patients with advanced cervical cancer, and more effective drugs with favorable side-effect profiles are needed. We developed a nucleoside analogue duplex drug (5-FdU-ECyd), in which the DNA synthesis inhibitor 5-fluorodeoxyuridine is coupled to the RNA synthesis inhibitor 3'-C-ethinylcytidine. We therefore aimed to test its efficacy in cervical carcinoma cells in vitro and to establish its mechanism of action. METHODS: The cytotoxic effects of 5-FdU-ECyd on cervical cancer cells were assessed using the MTT assay, clonality assays, FACScan analysis, and its effect on cancer cell spheroids. Mechanisms of cell death were analyzed by Western blotting for apoptosis and autophagy pathways and mitochondrial membrane potential. RESULTS: HeLa, CaSki, SiHa, and Me180 cervical cancer cells were highly sensitive to 5-FdU-ECyd in both 2- and 3-dimensional cancer models. The cell death induced by 5-FdU-ECyd was associated with characteristic morphological and biochemical signs of apoptosis, including nuclear chromatin condensation and fragmentation, PARP cleavage, and a breakdown in mitochondrial membrane potential. 5-FdU-ECyd treatment led to an early S-phase arrest and drastically reduced expression of the anti-apoptosis protein Mcl-1 and increased signaling via the JNK and p38 MAPK pathways. CONCLUSIONS: 5-FdU-ECyd is highly cytotoxic in cervical cancer cells and exploits apoptosis pathways that might be specific to cancer, but not normal cells. 5-FdU-ECyd might represent a new chemotherapeutic option for patients with advanced or treatment refractory cervical cancer.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas , Puntos de Control del Ciclo Celular/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Oligonucleótidos/farmacología , Neoplasias del Cuello Uterino , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Células HeLa , Humanos
9.
Cell Cycle ; 13(10): 1513-4, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24759086
10.
Arch Gynecol Obstet ; 290(1): 149-54, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24554348

RESUMEN

PURPOSE: Due to very unspecific symptoms ovarian cancer often is diagnosed only at a late stage of the disease. Thus, morbidity and mortality of the patients are high. Even the established tumor marker CA12-5 shows only low specificity, rising the need for alternative biomarkers capable of detecting early stages of ovarian cancer. We analyzed the expression of the tumor suppressor candidate gene LDOC1 (leucine zipper downregulated in cancer 1) as a potential early biomarker in ovarian cancer cell lines. METHODS: A total of seven ovarian cancer cell lines were analyzed by RT-PCR (reverse transcriptase polymerase chain reaction) and real-time PCR for expression of LDOC1. Verification of promoter methylation was performed using methylation-specific primers on bisulfite-modified genomic DNA. RESULTS: Three out of seven ovarian cancer cell lines showed a complete loss of LDOC1 gene expression. LDOC1 silencing was caused neither by gene deletion nor gene rearrangements, but by methylation and subsequent inactivation of the concerned promoter as proofed by methylation specific primers. Similarly, promoter methylation could be inhibited by adding AdC (5-aza-2'-deoxycytidine), an inhibitor of DNA methyltransferases. As a result, a reactivation of the LDOC1 gene was seen. CONCLUSIONS: The tumor suppressor gene LDOC1 in ovarian cancer cell lines is downregulated by promoter methylation and thus may serve as an early biomarker. Further investigation will show if detection of methylated LDOC1 in peripheral blood has both adequate sensitivity and specificity for a timely non-invasive detection of ovarian cancer.


Asunto(s)
Metilación de ADN , Metilasas de Modificación del ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Ováricas/genética , Adulto , Anciano , Anciano de 80 o más Años , Azacitidina/análogos & derivados , Biomarcadores de Tumor/genética , Línea Celular , Metilasas de Modificación del ADN/antagonistas & inhibidores , Metilasas de Modificación del ADN/genética , Cartilla de ADN/genética , Decitabina , Inhibidores Enzimáticos , Femenino , Silenciador del Gen , Genes Supresores de Tumor/efectos de los fármacos , Humanos , Persona de Mediana Edad , Neoplasias Ováricas/metabolismo , Regiones Promotoras Genéticas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Oncoscience ; 1(1): 21-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25593981

RESUMEN

A valuable strategy to develop new therapeutic options for a variety of diseases has been the identification of new targets and applications for already approved drugs, the so-called drug repositioning. Recurrent ovarian cancer is a nearly incurable malignancy for which new and effective treatments are urgently needed. The alcohol-deterring drug disulfiram has been shown to cause preferential cell death in a variety of cancer cells. In this study, it is shown that disulfiram mediates effective cell death in ovarian cancer cells by promoting a pro-oxidative intracellular environment in a copper-dependent mechanism. Within few hours of application, disulfiram caused irreversible cell damage associated with pronounced induction of the inducible heat shock proteins HSP70, HSP40, and HSP32. The small heat shock protein HSP27 was found to be covalently dimerized via oxidized disulfide bonds and precipitated in para-nuclear protein aggregates. Simultaneous inhibition of the cellular thioredoxin system by auranofin further enhanced the cytotoxic effect of disulfiram. These data indeed indicate that the combination of two approved drugs, the anti-alcoholic disulfiram and the anti-rheumatic auranofin, may be of interest for the treatment of recurrent and genotoxic drug-resistant ovarian cancer by inducing a proteotoxic cell death mechanism.

12.
Exp Cell Res ; 320(2): 281-9, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24280420

RESUMEN

Tetracyclines have long been used as valuable broad-spectrum antibiotics. The high antibacterial activity of tetracyclines, combined with their good tolerability, has led to their widespread use in treating various infectious diseases. However, similar to other antibiotics, tetracyclines are also known for their adverse effects on different human tissues, including hepatic steatosis. We observed that tetracyclines, including doxycycline and minocycline, caused enhanced expression of the liver chalone inhibin ßE in HepG2 cells, mediated by the cell stress-regulated transcription factor ATF4. ATF4 and its target genes ATF3, CHOP, and inhibin ßE are involved in cell cycle control, cell survival, cell metabolism, and modulation of cytokine expression. Furthermore, we observed that long term tetracycline incubation also caused inhibition of the mTOR complex, a central regulator of cell metabolism, further contributing to the observed cell-cycle arrest and autophagy in doxycycline- and minocycline-treated cell lines. ATF4 activation and mTOR inhibition link two crucial regulators of the cellular stress response and cell metabolism to the effects of tetracyclines on eukaryotic cell metabolism, and may help to understand the antibiotic-independent influence of these drugs on human tissues. Since the observed effects of tetracyclines on human cells were also found to be dependent on the magnesium ion concentrations supplied, the data further indicate the importance of magnesium supplementation to reduce or prevent side effects of long term treatment with tetracyclines.


Asunto(s)
Factor de Transcripción Activador 4/agonistas , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Tetraciclinas/farmacología , Factor de Transcripción Activador 4/metabolismo , Células CACO-2 , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células HeLa , Células Hep G2 , Humanos , Subunidades beta de Inhibinas/genética , Subunidades beta de Inhibinas/metabolismo , Estrés Fisiológico/fisiología , Activación Transcripcional/efectos de los fármacos
13.
J Appl Toxicol ; 34(10): 1109-13, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24155182

RESUMEN

Clinically effective anti-cancer drugs have to tread a narrow line between selective cytotoxicity on tumor cells and tolerable adverse effects against healthy tissues. This causes the failure of many potential cancer drugs in advanced clinical trials, hence signifying the importance of a comprehensive initial estimate of the cytotoxicity of prospective anti-cancer drugs in preclinical studies. In this study, the cytotoxicity of borrelidin, a macrolide antibiotic with a high cytotoxic selectivity for proliferating endothelial cells and leukemia cells, was tested on malignant and non-malignant breast cells. Highly metastatic breast cancer cell lines (MDA-MB-231 and MDA-MB-435) showed promising results and exhibited good sensitivity to borrelidin at low nanomolar concentrations, but borrelidin was cytotoxic to a non-malignant breast epithelial cell line (MCF10A) as well. Furthermore, although a high sensitivity of endothelial cells (human umbilical vein endothelial cells; HUVEC) and individual leukemia cell lines (Jurkat and IM9) to borrelidin was confirmed in this study, another leukemia cell line (HL60) and an immortalized endothelial cell line (EA.hy926) displayed a significantly decreased sensitivity. Reduced sensitivity to borrelidin was associated with elevated bcl-2 expression in these cell lines. In conclusion, the results presented show that borrelidin displays high and selective cytotoxicity against subgroups of cancer cells and endothelial cells, but, owing to its non-specific toxicity to non-malignant cells, its clinical application might be restricted because of likely adverse effects and limited efficacy in bcl2-overexpressing cancer cells.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Mama/metabolismo , Células Epiteliales/metabolismo , Genes bcl-2/genética , Leucemia/tratamiento farmacológico , Leucemia/genética , Adenosina Trifosfato/metabolismo , Western Blotting , Mama/efectos de los fármacos , Línea Celular Tumoral , Células Epiteliales/efectos de los fármacos , Alcoholes Grasos/farmacología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Sales de Tetrazolio , Tiazoles
14.
Cancer Invest ; 31(9): 571-7, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24125169

RESUMEN

Cervical cancer lacks reliable prognostic factors for both progression and chemotherapeutic responsiveness. The expression of the LDOC1 tumor suppressor candidate was therefore investigated. In four of six cervical cancer cell lines tested, expression of LDOC1 was silenced. Downregulation of LDOC1 could also be shown in biopsies of cervical cancer specimens. PCR-based promoter methylation analysis revealed a significant association between promoter methylation and the loss of LDOC1 expression, which could be reverted by DNA methyltransferase inhibitors. This indicates that silencing of LDOC1 is a frequent event in cervical cancer and may be of interest as a molecular marker in cervical cancer.


Asunto(s)
Metilación de ADN , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Proteínas Supresoras de Tumor/genética , Neoplasias del Cuello Uterino/genética , Biopsia , Muerte Celular , Metilasas de Modificación del ADN/antagonistas & inhibidores , Metilasas de Modificación del ADN/metabolismo , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Células HeLa , Humanos , Proteínas Nucleares/metabolismo , Transfección , Proteínas Supresoras de Tumor/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología
15.
Mol Oncol ; 7(6): 1012-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23916134

RESUMEN

Endoplasmic reticulum (ER) stress and autophagy are two basic cell survival mechanisms often occurring in concert. Extensive ER stress in cancer cells deliberately induced by chemotherapeutic drugs may lead to growth arrest and cell death. However, the link between ER stress and autophagy is not well understood. In this study, the treatment of cancer cells with ER stress-inducing drug nelfinavir resulted in the expression of endogenous mTOR inhibitor sestrin-2 (SESN2). Upregulation of SESN2 expression was associated with expression of ER stress markers ATF4, ATF3, and CHOP. SESN2 upregulation also occurred in cells treated with the proteasome inhibitor bortezomib. Ectopic expression of ATF4, but not of ATF3 or CHOP, caused transcriptional upregulation of SESN2 expression, indicating expressional regulation of SESN2 by ATF4. Transient overexpression of ectopic SESN2 resulted in mTOR inhibition and autophagy, confirming a link between ER stress, SESN2 upregulation, and mTOR inhibition. Accordingly, cancer cells treated with the ER stress-inducing agent nelfinavir showed reduced mTOR activity and associated increases in the expression levels of ATF4 and SESN2. These results show that ATF4-regulated SESN2 expression presents a new link between ER stress and mTOR inhibition and autophagy. mTOR inhibition by nelfinavir, which is currently in clinical trials for cancer patients, may also explain its observed ability to induce autophagy, growth arrest, and radiosensitization in cancer cells.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de la Proteasa del VIH/farmacología , Nelfinavir/farmacología , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Proteínas Nucleares/biosíntesis , Pirazinas/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Factor de Transcripción Activador 4/genética , Autofagia/efectos de los fármacos , Autofagia/genética , Bortezomib , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Células HeLa , Humanos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/genética , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
16.
Oncotarget ; 4(4): 502-30, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23594434

RESUMEN

To improve prognosis in recurrent glioblastoma we developed a treatment protocol based on a combination of drugs not traditionally thought of as cytotoxic chemotherapy agents but that have a robust history of being well-tolerated and are already marketed and used for other non-cancer indications. Focus was on adding drugs which met these criteria: a) were pharmacologically well characterized, b) had low likelihood of adding to patient side effect burden, c) had evidence for interfering with a recognized, well-characterized growth promoting element of glioblastoma, and d) were coordinated, as an ensemble had reasonable likelihood of concerted activity against key biological features of glioblastoma growth. We found nine drugs meeting these criteria and propose adding them to continuous low dose temozolomide, a currently accepted treatment for relapsed glioblastoma, in patients with recurrent disease after primary treatment with the Stupp Protocol. The nine adjuvant drug regimen, Coordinated Undermining of Survival Paths, CUSP9, then are aprepitant, artesunate, auranofin, captopril, copper gluconate, disulfiram, ketoconazole, nelfinavir, sertraline, to be added to continuous low dose temozolomide. We discuss each drug in turn and the specific rationale for use- how each drug is expected to retard glioblastoma growth and undermine glioblastoma's compensatory mechanisms engaged during temozolomide treatment. The risks of pharmacological interactions and why we believe this drug mix will increase both quality of life and overall survival are reviewed.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Aprepitant , Artemisininas/administración & dosificación , Auranofina/administración & dosificación , Captopril/administración & dosificación , Dacarbazina/administración & dosificación , Dacarbazina/análogos & derivados , Disulfiram/administración & dosificación , Gluconatos/administración & dosificación , Humanos , Cetoconazol/administración & dosificación , Morfolinas/administración & dosificación , Nelfinavir/administración & dosificación , Sertralina/administración & dosificación , Succinatos/administración & dosificación , Temozolomida
17.
Anticancer Agents Med Chem ; 13(7): 1025-31, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23272907

RESUMEN

Quercetin is an abundant micronutrient in our daily diet. Several beneficial health effects are associated with the dietary uptake of this bioflavonoid, including alleviating effects on chronic inflammation and atherosclerosis. A variety of in vitro data indicate a possible use of quercetin for cancer treatment purposes through its interaction with multiple cancer-related pathways. Among these, recent data reveal that quercetin can inhibit mTOR activity in cancer cells. Inhibition of the mTOR signaling pathway by quercetin has directly been described and can further be deduced from its interference with PI3K-dependent Akt stimulation, AMP-dependent protein kinase activation and hamartin upregulation. The ability of quercetin to interfere with both mTOR activity and activation of the PI3K/Akt signaling pathway gives quercetin the advantage to function as a dual-specific mTOR/PI3K inhibitor. The mTOR complex, often hyperactivated in cancer, is a crucial regulator of homeostasis controlling essential pathways leading to cell growth, protein biosynthesis and autophagy. The ability of quercetin to inhibit mTOR activity by multiple pathways makes this otherwise safe bioflavonoid an interesting tool for the treatment of cancers and other diseases associated with mTOR deregulation.


Asunto(s)
Antioxidantes/farmacología , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Quercetina/farmacología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Animales , Humanos , Neoplasias/metabolismo , Neoplasias/prevención & control , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo
20.
Gynecol Oncol ; 128(2): 383-90, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23107612

RESUMEN

OBJECTIVE: The proteasome inhibitor bortezomib is currently being tested in clinical trials against refractory cervical cancer. However, high doses of bortezomib are associated with adverse effects, which may lead to treatment abrogation or to the use of lower, ineffective doses. We investigated combination drug treatments that could enhance the efficacy of low bortezomib concentrations on cervical cancer cells. METHODS: The cervical cancer cell lines CaSki, HeLa and SW756 were treated with various combinations of bortezomib and eeyarestatin. Treated cells were analyzed for cell viability by clonal assays and the MTT assay, and for expression of pro-apoptotic proteins and cell stress markers by immunofluorescence, immunoblots and RT-PCR analysis. RESULTS: Cotreatment of bortezomib with eeyarestatin markedly enhanced cell death in cervical cancer cells, allowing reduction of the bortezomib concentration necessary for efficient cell death to as low as 5 ng/ml. Combination of bortezomib with eeyarestatin resulted in a massive induction of the endoplasmic reticulum stress reaction, small and large heat shock protein activation, autophagy, and upregulation of pro-apoptotic CHOP. CONCLUSION: Eeyarestatin is a small molecule recently shown to cause endoplasmic reticulum stress by inhibiting the endoplasmic reticulum-associated degradation pathway, which directs misfolded cytotoxic proteins to proteasomal degradation. Concomitant inhibition of both pathways markedly enhances the efficacy of bortezomib against cervical cancer cells and thus may be applied to reduce the bortezomib dosage required for efficient cervical cancer treatment.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Factor de Transcripción CHOP/biosíntesis , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/metabolismo , Ácidos Borónicos/administración & dosificación , Bortezomib , Sinergismo Farmacológico , Femenino , Células HeLa , Humanos , Hidrazonas/administración & dosificación , Hidroxiurea/administración & dosificación , Hidroxiurea/análogos & derivados , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Pirazinas/administración & dosificación , Ubiquitina/metabolismo , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...