Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
FASEB J ; 34(9): 12024-12039, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32692445

RESUMEN

Macropinocytosis supports the metabolic requirement of RAS-transformed pancreatic ductal adenocarcinoma cells (PDACs). However, regulators of RAS-transformation (activation) that lead to macropinocytosis have not been identified. Herein, we report that UBAP2 (ubiquitin-binding associated protein 2), regulates the activation of KRAS and macropinocytosis in pancreatic cancer. We demonstrate that UBAP2 is highly expressed in both pancreatic cancer cell lines and tumor tissues of PDAC patients. The expression of UBAP2 is associated with poor overall survival in several cancers, including PDAC. Silencing UBAP2 decreases the levels of activated KRAS, and inhibits macropinocytosis, and tumor growth in vivo. Using a UBAP2-deletion construct, we demonstrate that the UBA-domain of UBAP2 is critical for the regulation of macropinocytosis and maintaining the levels of activated KRAS. In addition, UBAP2 regulates RAS downstream signaling and helps maintain RAS in the GTP-bound form. However, the exact mechanism by which UBAP2 regulates KRAS activation is unknown and needs further investigation. Thus, UBAP2 may be exploited as a potential therapeutic target to inhibit macropinocytosis and tumor growth in activated KRAS-driven cancers.


Asunto(s)
Proteínas Portadoras/metabolismo , Neoplasias Pancreáticas/metabolismo , Pinocitosis , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Portadoras/genética , Línea Celular Tumoral , Activación Enzimática , Silenciador del Gen , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Dominios Proteicos , Proteínas Proto-Oncogénicas p21(ras)/genética
4.
Nat Commun ; 8: 14634, 2017 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-28530221

RESUMEN

Cancer cells actively promote aerobic glycolysis to sustain their metabolic requirements through mechanisms not always clear. Here, we demonstrate that the gatekeeper of mitochondrial Ca2+ uptake, Mitochondrial Calcium Uptake 1 (MICU1/CBARA1) drives aerobic glycolysis in ovarian cancer. We show that MICU1 is overexpressed in a panel of ovarian cancer cell lines and that MICU1 overexpression correlates with poor overall survival (OS). Silencing MICU1 in vitro increases oxygen consumption, decreases lactate production, inhibits clonal growth, migration and invasion of ovarian cancer cells, whereas silencing in vivo inhibits tumour growth, increases cisplatin efficacy and OS. Mechanistically, silencing MICU1 activates pyruvate dehydrogenase (PDH) by stimulating the PDPhosphatase-phosphoPDH-PDH axis. Forced-expression of MICU1 in normal cells phenocopies the metabolic aberrations of malignant cells. Consistent with the in vitro and in vivo findings we observe a significant correlation between MICU1 and pPDH (inactive form of PDH) expression with poor prognosis. Thus, MICU1 could serve as an important therapeutic target to normalize metabolic aberrations responsible for poor prognosis in ovarian cancer.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Transporte de Catión/metabolismo , Resistencia a Antineoplásicos , Glucólisis , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Neoplasias Ováricas/metabolismo , Animales , Antineoplásicos/uso terapéutico , Apoptosis , Calcio/metabolismo , Línea Celular Tumoral , Cisplatino/uso terapéutico , Femenino , Humanos , Ratones Desnudos , Análisis por Micromatrices , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/mortalidad , Fosforilación Oxidativa , Fenotipo , Complejo Piruvato Deshidrogenasa/metabolismo
5.
ACS Nano ; 10(12): 10636-10651, 2016 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-27758098

RESUMEN

Altered tumor microenvironment (TME) arising from a bidirectional crosstalk between the pancreatic cancer cells (PCCs) and the pancreatic stellate cells (PSCs) is implicated in the dismal prognosis in pancreatic ductal adenocarcinoma (PDAC), yet effective strategies to disrupt the crosstalk is lacking. Here, we demonstrate that gold nanoparticles (AuNPs) inhibit proliferation and migration of both PCCs and PSCs by disrupting the bidirectional communication via alteration of the cell secretome. Analyzing the key proteins identified from a functional network of AuNP-altered secretome in PCCs and PSCs, we demonstrate that AuNPs impair secretions of major hub node proteins in both cell types and transform activated PSCs toward a lipid-rich quiescent phenotype. By reducing activation of PSCs, AuNPs inhibit matrix deposition, enhance angiogenesis, and inhibit tumor growth in an orthotopic co-implantation model in vivo. Auto- and heteroregulations of secretory growth factors/cytokines are disrupted by AuNPs resulting in reprogramming of the TME. By utilizing a kinase dead mutant of IRE1-α, we demonstrate that AuNPs alter the cellular secretome through the ER-stress-regulated IRE1-dependent decay pathway (RIDD) and identify endostatin and matrix metalloproteinase 9 as putative RIDD targets. Thus, AuNPs could potentially be utilized as a tool to effectively interrogate bidirectional communications in the tumor microenvironment, reprogram it, and inhibit tumor growth by its therapeutic function.


Asunto(s)
Carcinoma Ductal Pancreático/terapia , Oro , Nanopartículas del Metal , Neoplasias Pancreáticas/terapia , Microambiente Tumoral , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pancreáticas/metabolismo , Células Estrelladas Pancreáticas
6.
Autophagy ; 12(4): 659-70, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27050456

RESUMEN

The clonal self-renewal property conferred by BMI1 is instrumental in maintenance of not only normal stem cells but also cancer-initiating cells from several different malignancies that represent a major challenge to chemotherapy. Realizing the immense pathological significance, PTC-209, a small molecule inhibitor of BMI1 transcription has recently been described. While targeting BMI1 in various systems significantly decreases clonal growth, the mechanisms differ, are context-dependent, and somewhat unclear. We report here that genetic or pharmacological inhibition of BMI1 significantly impacts clonal growth without altering CDKN2A/INK4/ARF or CCNG2 and induces autophagy in ovarian cancer (OvCa) cells through ATP depletion. While autophagy can promote survival or induce cell death, targeting BMI1 engages the PINK1-PARK2-dependent mitochondrial pathway and induces a novel mode of nonapoptotic, necroptosis-mediated cell death. In OvCa, necroptosis is potentiated by activation of the RIPK1-RIPK3 complex that phosphorylates its downstream substrate, MLKL. Importantly, genetic or pharmacological inhibitors of autophagy or RIPK3 rescue clonal growth in BMI1 depleted cells. Thus, we have established a novel molecular link between BMI1, clonal growth, autophagy and necroptosis. In chemoresistant OvCa where apoptotic pathways are frequently impaired, necroptotic cell death modalities provide an important alternate strategy that leverage overexpression of BMI1.


Asunto(s)
Apoptosis , Autofagia , Complejo Represivo Polycomb 1/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Caspasas/metabolismo , Línea Celular Tumoral , Compuestos Heterocíclicos con 2 Anillos/farmacología , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Biológicos , Necrosis , Complejo Represivo Polycomb 1/metabolismo , Tiazoles/farmacología
7.
FASEB J ; 30(1): 441-56, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26405298

RESUMEN

Deficiencies of the human cystathionine ß-synthase (CBS) enzyme are characterized by a plethora of vascular disorders and hyperhomocysteinemia. However, several clinical trials demonstrated that despite reduction in homocysteine levels, disease outcome remained unaffected, thus the mechanism of endothelial dysfunction is poorly defined. Here, we show that the loss of CBS function in endothelial cells (ECs) leads to a significant down-regulation of cellular hydrogen sulfide (H2S) by 50% and of glutathione (GSH) by 40%. Silencing CBS in ECs compromised phenotypic and signaling responses to the VEGF that were potentiated by decreased transcription of VEGF receptor (VEGFR)-2 and neuropilin (NRP)-1, the primary receptors regulating endothelial function. Transcriptional down-regulation of VEGFR-2 and NRP-1 was mediated by a lack in stability of the transcription factor specificity protein 1 (Sp1), which is a sulfhydration target of H2S at residues Cys68 and Cys755. Reinstating H2S but not GSH in CBS-silenced ECs restored Sp1 levels and its binding to the VEGFR-2 promoter and VEGFR-2, NRP-1 expression, VEGF-dependent proliferation, and migration phenotypes. Thus, our study emphasizes the importance of CBS-mediated protein S-sulfhydration in maintaining vascular health and function.-Saha, S., Chakraborty, P. K., Xiong, X., Dwivedi, S. K. D., Mustafi, S. B., Leigh, N. R., Ramchandran, R., Mukherjee, P., Bhattacharya, R. Cystathionine ß-synthase regulates endothelial function via protein S-sulfhydration.


Asunto(s)
Cistationina betasintasa/metabolismo , Endotelio Vascular/metabolismo , Sulfuro de Hidrógeno/metabolismo , Movimiento Celular , Proliferación Celular , Cistationina betasintasa/genética , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Glutatión/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Neuropilinas/genética , Neuropilinas/metabolismo , Sistemas de Mensajero Secundario , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
8.
Oncotarget ; 6(35): 37367-84, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26452259

RESUMEN

Elevated lipid metabolism is implicated in poor survival in ovarian cancer (OC) and other cancers; however, current lipogenesis-targeting strategies lack cancer cell specificity. Here, we identify a novel role of cystathionine beta-synthase (CBS), a sulphur amino acid metabolizing enzyme highly expressed in several ovarian cancer cell lines, in driving deregulated lipid metabolism in OC. We examined the role of CBS in regulation of triglycerides, cholesterol and lipogenic enzymes via the lipogenic transcription factors SREBP1 and SREBP2. CBS silencing attenuated the expression of number of key enzymes involved in lipid synthesis (FASN and ACC1). Additionally CBS abrogates lipid uptake in OC cells. Gene silencing of CBS or SREBPs abrogated cellular migration and invasion in OC, while ectopic expression of SREBPs can rescue phenotypic effects of CBS silencing by restoring cell migration and invasion. Mechanistically, CBS represses SREBP1 and SREBP2 at the transcription levels by modulating the transcription factor Sp1. We further established the roles of both CBS and SREBPs in regulating ovarian tumor growth in vivo. In orthotopic tumor models, CBS or SREBP silencing resulted in reduced tumor cells proliferation, blood vessels formation and lipid content. Hence, cancer-selective disruption of the lipid metabolism pathway is possible by targeting CBS and, at least for OC, promises a profound benefit.


Asunto(s)
Colesterol/metabolismo , Cistationina betasintasa/metabolismo , Lipogénesis , Neoplasias Ováricas/enzimología , Triglicéridos/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Cistationina betasintasa/genética , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genotipo , Xenoinjertos , Humanos , Ratones Desnudos , Invasividad Neoplásica , Neovascularización Fisiológica , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Fenotipo , Interferencia de ARN , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Transcripción Genética , Transfección
9.
Oncotarget ; 5(15): 6453-65, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25071019

RESUMEN

Recently we reported that gold nanoparticles (AuNPs) inhibit ovarian tumor growth and metastasis in mice by reversing epithelial-mesenchymal transition (EMT). Since EMT is known to confer drug resistance to cancer cells, we wanted to investigate whether anti-EMT property of AuNP could be utilized to sensitize ovarian cancer cells to cisplatin. Herein, we report that AuNPs prevent cisplatin-induced acquired chemoresistance and stemness in ovarian cancer cells and sensitize them to cisplatin. AuNPs inhibit cisplatin induced EMT, decrease the side population cells and key stem cell markers such as ALDH1, CD44, CD133, Sox2, MDR1 and ABCG2 in ovarian cancer cells. Mechanistically, AuNPs prevent cisplatin-induced activation of Akt and NF-kB signaling axis in ovarian cancer cells that are critical for EMT, stem cell maintenance and drug resistance. In vivo, AuNPs sensitize orthotopically implanted ovarian tumor to a low dose of cisplatin and significantly inhibit tumor growth via facilitated delivery of both AuNP and cisplatin. These findings suggest that by depleting stem cell pools and inhibiting key molecular pathways gold nanoparticles sensitize ovarian cancer cells to cisplatin and may be used in combination to inhibit tumor growth and metastasis in ovarian cancer.


Asunto(s)
Cisplatino/farmacología , Oro/administración & dosificación , Nanopartículas del Metal/administración & dosificación , Neoplasias Ováricas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Regulación hacia Abajo , Sinergismo Farmacológico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Desnudos , Nanopartículas , Neoplasias Ováricas/patología , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Bioconjug Chem ; 25(6): 1078-90, 2014 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-24831101

RESUMEN

Molecular identification of protein molecules surrounding nanoparticles (NPs) may provide useful information that influences NP clearance, biodistribution, and toxicity. Hence, nanoproteomics provides specific information about the environment that NPs interact with and can therefore report on the changes in protein distribution that occurs during tumorigenesis. Therefore, we hypothesized that characterization and identification of protein molecules that interact with 20 nm AuNPs from cancer and noncancer cells may provide mechanistic insights into the biology of tumor growth and metastasis and identify new therapeutic targets in ovarian cancer. Hence, in the present study, we systematically examined the interaction of the protein molecules with 20 nm AuNPs from cancer and noncancerous cell lysates. Time-resolved proteomic profiles of NP-protein complexes demonstrated electrostatic interaction to be the governing factor in the initial time-points which are dominated by further stabilization interaction at longer time-points as determined by ultraviolet-visible spectroscopy (UV-vis), dynamic light scattering (DLS), ζ-potential measurements, transmission electron microscopy (TEM), and tandem mass spectrometry (MS/MS). Reduction in size, charge, and number of bound proteins were observed as the protein-NP complex stabilized over time. Interestingly, proteins related to mRNA processing were overwhelmingly represented on the NP-protein complex at all times. More importantly, comparative proteomic analyses revealed enrichment of a number of cancer-specific proteins on the AuNP surface. Network analyses of these proteins highlighted important hub nodes that could potentially be targeted for maximal therapeutic advantage in the treatment of ovarian cancer. The importance of this methodology and the biological significance of the network proteins were validated by a functional study of three hubs that exhibited variable connectivity, namely, PPA1, SMNDC1, and PI15. Western blot analysis revealed overexpression of these proteins in ovarian cancer cells when compared to normal cells. Silencing of PPA1, SMNDC1, and PI15 by the siRNA approach significantly inhibited proliferation of ovarian cancer cells and the effect correlated with the connectivity pattern obtained from our network analyses.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/uso terapéutico , Oro/química , Nanopartículas del Metal/química , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Neoplasias Ováricas/tratamiento farmacológico , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Proliferación Celular/efectos de los fármacos , Biología Computacional , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Oro/efectos adversos , Oro/farmacocinética , Oro/uso terapéutico , Humanos , Nanopartículas del Metal/efectos adversos , Nanopartículas del Metal/uso terapéutico , Modelos Moleculares , Neoplasias Ováricas/patología , Tamaño de la Partícula , Proteómica , Relación Estructura-Actividad , Propiedades de Superficie , Células Tumorales Cultivadas
11.
PLoS One ; 8(11): e79167, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24236104

RESUMEN

BACKGROUND: Epithelial ovarian cancer is the leading cause of gynecologic cancer deaths. Most patients respond initially to platinum-based chemotherapy after surgical debulking, however relapse is very common and ultimately platinum resistance emerges. Understanding the mechanism of tumor growth, metastasis and drug resistant relapse will profoundly impact the therapeutic management of ovarian cancer. METHODS/PRINCIPAL FINDINGS: Using patient tissue microarray (TMA), in vitro and in vivo studies we report a role of of cystathionine-beta-synthase (CBS), a sulfur metabolism enzyme in ovarian carcinoma. We report here that the expression of cystathionine-beta-synthase (CBS), a sulfur metabolism enzyme, is common in primary serous ovarian carcinoma. The in vitro effects of CBS silencing can be reversed by exogenous supplementation with the GSH and H2S producing chemical Na2S. Silencing CBS in a cisplatin resistant orthotopic model in vivo by nanoliposomal delivery of CBS siRNA inhibits tumor growth, reduces nodule formation and sensitizes ovarian cancer cells to cisplatin. The effects were further corroborated by immunohistochemistry that demonstrates a reduction of H&E, Ki-67 and CD31 positive cells in si-RNA treated as compared to scrambled-RNA treated animals. Furthermore, CBS also regulates bioenergetics of ovarian cancer cells by regulating mitochondrial ROS production, oxygen consumption and ATP generation. This study reports an important role of CBS in promoting ovarian tumor growth and maintaining drug resistant phenotype by controlling cellular redox behavior and regulating mitochondrial bioenergetics. CONCLUSION: The present investigation highlights CBS as a potential therapeutic target in relapsed and platinum resistant ovarian cancer.


Asunto(s)
Cistationina betasintasa/metabolismo , Resistencia a Antineoplásicos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Adenosina Trifosfato/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antioxidantes/metabolismo , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Cisplatino/farmacología , Cistationina betasintasa/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Inmunohistoquímica , Ratones , Persona de Mediana Edad , Mitocondrias/metabolismo , Clasificación del Tumor , Estadificación de Neoplasias , Neoplasias Ováricas/genética , Fenotipo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Adulto Joven
12.
Philos Trans A Math Phys Eng Sci ; 371(1995): 20120190, 2013 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-23776297

RESUMEN

Iron(III) complexes [FeL(B)] (1-4) of a tetradentate phenolate-based ligand (H3L) and biotin-conjugated dipyridophenazine bases (B), viz. 7-aminodipyrido [3,2-a:2',3'-c]-phenazine (dppza in 1), (N-dipyrido[3,2-a:2',3'-c]-phenazino)amidobiotin (dppzNB in 2), dipyrido [3,2-a:2',3'-c]-phenazine-11-carboxylic acid (dppzc in 3) and 2-((2-biotinamido)ethyl) amido-dipyrido[3,2-a:2',3'-c]-phenazine (dppzCB in 4) are prepared, characterized and their interaction with streptavidin and DNA and their photocytotoxicity and cellular uptake in various cells studied. The high-spin iron(III) complexes display Fe(III)/Fe(II) redox couple near -0.7 V versus saturated calomel electrode in dimethyl sulfoxide-0.1 M tetrabutylammonium perchlorate. The complexes show non-specific interaction with DNA as determined from the binding studies. Complexes with appended biotin moiety show similar binding to streptavidin as that of free biotin, suggesting biotin conjugation to dppz does not cause any loss in its binding affinity to streptavidin. The photocytotoxicity of the complexes is tested in HepG2, HeLa and HEK293 cell lines. Complex 2 shows higher photocytotoxicity in HepG2 cells than in HeLa or HEK293, forming reactive oxygen species. This effect is attributed to the presence of overexpressed sodium-dependent multi-vitamin transporters in HepG2 cells. Microscopic studies in HepG2 cells show internalization of the biotin complexes 2 and 4 essentially occurring by receptor-mediated endocytosis, which is similar to that of native biotin and biotin fluorescein isothiocyanate conjugate.


Asunto(s)
Biotina/farmacocinética , Hierro/uso terapéutico , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/uso terapéutico , Células Hep G2 , Humanos , Luz , Neoplasias Experimentales/patología , Resultado del Tratamiento
13.
Proc Natl Acad Sci U S A ; 110(17): 6700-5, 2013 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-23569259

RESUMEN

Although biomedical applications of nanotechnology, which typically involve functionalized nanoparticles, have taken significant strides, biological characterization of unmodified nanoparticles remains underinvestigated. Herein we demonstrate that unmodified gold nanoparticles (AuNPs) inhibit the proliferation of cancer cells in a size- and concentration-dependent manner by abrogating MAPK-signaling. In addition, these AuNPs reverse epithelial-mesenchymal transition (EMT) in cancer cells by reducing secretion of a number of proteins involved in EMT, up-regulating E-Cadherin, and down-regulating Snail, N-Cadherin, and Vimentin. Inhibition of MAPK signaling and reversal of EMT upon AuNP treatment inhibits tumor growth and metastasis in two separate orthotopic models of ovarian cancer. Western blot analyses of tumor tissues reveal up-regulation of E-Cadherin and down-regulation of Snail and phospho-MAPK, confirming the reversal of EMT and inhibition of MAPK signaling upon AuNP treatment. The ability of a single self-therapeutic nanoparticle to abrogate signaling cascades of multiple growth factors is distinctive and purports possible medical applications as potential antitumor and antimetastatic agent.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Oro/uso terapéutico , Sistema de Señalización de MAP Quinasas/fisiología , Nanopartículas/uso terapéutico , Nanotecnología/métodos , Metástasis de la Neoplasia/prevención & control , Neoplasias Ováricas/tratamiento farmacológico , Western Blotting , Cadherinas/metabolismo , Proliferación Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Oro/química , Oro/farmacología , Humanos , Inmunohistoquímica , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microscopía Confocal , Microscopía Electrónica de Transmisión , Nanopartículas/química , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo , Vimentina/metabolismo
14.
J Biol Chem ; 288(24): 17610-8, 2013 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-23615904

RESUMEN

Nanoparticles provide a potent tool for targeting and understanding disease mechanisms. In this regard, cancer cells are surprisingly resistant to the expected toxic effects of positively charged gold nanoparticles ((+)AuNPs). Our investigations led to the identification of MICU1, regulator of mitochondrial calcium uniporter, as a key molecule conferring cancer cells with resistance to (+)AuNPs. The increase in cytosolic [Ca(2+)]cyto in malignant cells induced by (+)AuNPs is counteracted by MICU1, preventing cell death. Pharmacological or siRNA-mediated inhibition of mitochondrial Ca(+2) entry leads to endoplasmic reticulum stress and sensitizes cancer cells to (+)AuNP-induced cytotoxicity. Silencing MICU1 decreases Bcl-2 expression and increases caspase-3 activity and cytosolic cytochrome c levels, thus initiating the mitochondrial pathway for apoptosis: effects further enhanced by (+)AuNPs. This study highlights the potential of nanomaterials as a tool to broaden our understanding of cellular processes, establishes MICU1 as a novel regulator of the machinery in cancer cells that prevents apoptosis, and emphasizes the need to synergize nanoparticle design with understanding of mitochondrial machinery for enhancing targeted cellular toxicity.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Unión al Calcio/metabolismo , Proteínas de Transporte de Catión/metabolismo , Oro/farmacología , Nanopartículas del Metal/química , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Transporte de Catión/genética , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/genética , Neoplasias Ováricas
15.
J Inorg Biochem ; 116: 77-87, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23022691

RESUMEN

Iron(II) complexes [Fe(L)(2)](2+) as perchlorate (1-3) and chloride (1a-3a) salts, where L is 4'-phenyl-2,2':6',2″-terpyridine (phtpy in 1, 1a), 4'-(9-anthracenyl)-2,2':6',2″-terpyridine (antpy in 2, 2a) and 4'-(1-pyrenyl)-2,2':6',2″-terpyridine (pytpy in 3, 3a), were prepared and their photocytotoxicity studied. The diamagnetic complexes 1-3 having an FeN(6) core showed an Fe(III)-Fe(II) redox couple near 1.0 V vs. saturated calomel electrode in MeCN-0.1 M tetrabutylammonium perchlorate. Complexes 2 and 3, in addition, displayed a quasi-reversible ligand-based redox process near 0.0 V. The redox and spectral properties are rationalized from the theoretical studies. The complexes bind to DNA in a partial intercalative mode. The pytpy complex efficiently photo-cleaves DNA in green light via superoxide and hydroxyl radical formation. The antpy and pytpy complexes exhibited a remarkable photocytotoxic effect in HeLa cancer cells (IC(50), ~9 µM) in visible light (400-700nm), while remaining essentially nontoxic in dark (IC(50), ~90 µM). Formation of reactive oxygen species (ROS) inside the HeLa cells was evidenced from the fluorescence enhancement of dichlorofluorescein upon treatment with the pytpy complex followed by photo-exposure. The antpy and pytpy complexes were used for cellular imaging. Confocal imaging and dual staining study using propidium iodide (PI) showed nuclear localization of the complexes.


Asunto(s)
Núcleo Celular/química , Compuestos Ferrosos/química , Piridinas/química , División del ADN , Electroquímica , Compuestos Ferrosos/toxicidad , Células HeLa , Humanos , Modelos Moleculares , Piridinas/toxicidad , Especies Reactivas de Oxígeno/metabolismo
16.
Inorg Chem ; 50(7): 2975-87, 2011 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-21391682

RESUMEN

Iron(III) complexes [FeL(B)] (1-5) of a tetradentate trianionic phenolate-based ligand (L) and modified dipyridophenazine bases (B), namely, dipyrido-6,7,8,9-tetrahydrophenazine (dpqC in 1), dipyrido[3,2-a:2',3'-c]phenazine-2-carboxylic acid (dppzc in 2), dipyrido[3,2-a:2',3'-c]phenazine-11-sulfonic acid (dppzs in 3), 7-aminodipyrido[3,2-a:2',3'-c]phenazine (dppza in 4) and benzo[i]dipyrido[3,2-a:2',3'-c]phenazine (dppn in 5), have been synthesized and their photocytotoxic properties studied along with their dipyridophenazine analogue (6). The complexes have a five electron paramagnetic iron(III) center, and the Fe(III)/Fe(II) redox couple appears at about -0.69 V versus SCE in DMF-0.1 M TBAP. The physicochemical data also suggest that the complexes possess similar structural features as that of its parent complex [FeL(dppz)] with FeO3N3 coordination in a distorted octahedral geometry. The DNA-complex and protein-complex interaction studies have revealed that the complexes interact favorably with the biomolecules, the degree of which depends on the nature of the substituents present on the dipyridophenazine ring. Photocleavage of pUC19 DNA by the complexes has been studied using visible light of 476, 530, and 647 nm wavelengths. Mechanistic investigations with inhibitors show formation of HO(•) radicals via a photoredox pathway. Photocytotoxicity study of the complexes in HeLa cells has shown that the dppn complex (5) is highly active in causing cell death in visible light with sub micromolar IC(50) value. The effect of substitutions and the planarity of the phenazine moiety on the cellular uptake are quantified by determining the total cellular iron content using the inductively coupled plasma-optical emission spectrometry (ICP-OES) technique. The cellular uptake increases marginally with an increase in the hydrophobicity of the dipyridophenazine ligands whereas complex 3 with dppzs shows very high uptake. Insights into the cell death mechanism by the dppn complex 5, obtained through DAPI nuclear staining in HeLa cells, reveal a rapid programmed cell death mechanism following photoactivation of complex 5 with visible light. The effect of substituent on the DNA photocleavage activity of the complexes has been rationalized from the theoretical studies.


Asunto(s)
Compuestos Férricos/farmacología , Fenazinas/química , Fármacos Fotosensibilizantes/farmacología , Muerte Celular/efectos de los fármacos , ADN/efectos de los fármacos , División del ADN , Compuestos Férricos/síntesis química , Compuestos Férricos/química , Células HeLa , Humanos , Ligandos , Estructura Molecular , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/química , Teoría Cuántica , Estereoisomerismo , Relación Estructura-Actividad
17.
Dalton Trans ; 40(6): 1233-42, 2011 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-21186388

RESUMEN

Cobalt(II) complexes of terpyridine bases [Co(L)2](ClO4)2 (1-3), where L is 4'-phenyl-2,2':6',2''-terpyridine (ph-tpy in 1), 4'-(9-anthracenyl)-2,2':6',2''-terpyridine (an-tpy in 2) and 4'-(1- pyrenyl)-2,2':6',2''-terpyridine (py-tpy in 3), are prepared and their photo-induced DNA and protein cleavage activity and photocytotoxic property in HeLa cells studied. The 1 : 2 electrolytic and three-electron paramagnetic complexes show a visible band near 550 nm in DMF-Tris-HCl buffer. The complexes 1-3 show emission spectral bands at 355, 421 and 454 nm, respectively, when excited at 287, 368 and 335 nm. The quantum yield values for 1-3 in DMF-H2O (2 : 1 v/v) are 0.025, 0.060 and 0.28, respectively. The complexes are redox active in DMF-0.1 M TBAP. The Co(III)-Co(II) and Co(II)-Co(I) couples appear as quasi-reversible cyclic voltammetric responses near 0.2 and -0.7 V vs. SCE, respectively. Complexes 2 and 3 are avid binders to calf thymus DNA giving K(b) value of ∼106 M⁻¹. The complexes show chemical nuclease activity. Complexes 2 and 3 exhibit oxidative cleavage of pUC19 DNA in UV-A and visible light. The DNA photocleavage reaction of 3 at 365 nm shows formation of singlet oxygen and hydroxyl radical species, while only hydroxyl radical formation is evidenced in visible light. Complexes 2 and 3 show non-specific photo-induced bovine serum albumin protein cleavage activity at 365 nm. The an-tpy and py-tpy complexes exhibit significant photocytotoxicity in HeLa cervical cancer cells on exposure to visible light giving IC50 values of 24.2 and 7.6 µM, respectively. Live cell imaging study shows accumulation of the complexes in the cytosol of HeLa cancer cells.


Asunto(s)
Cobalto/química , Complejos de Coordinación/química , Luz , Compuestos Organometálicos/química , Fármacos Fotosensibilizantes/química , Piridinas/química , Animales , Bovinos , Complejos de Coordinación/metabolismo , Complejos de Coordinación/toxicidad , División del ADN , Células HeLa , Humanos , Ligandos , Microscopía Fluorescente , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/toxicidad , Oxidación-Reducción , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/toxicidad , Teoría Cuántica , Albúmina Sérica Bovina/metabolismo
18.
Metallomics ; 2(11): 754-65, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21072367

RESUMEN

Ternary cobalt(III) complexes [CoL(B)] (1-3) of a trianionic tetradentate phenolate-based ligand (L) and phenanthroline bases (B), viz. 1,10-phenanthroline (phen in 1), dipyridoquinoxaline (dpq in 2) and dipyridophenazine (dppz in 3) are synthesized, characterized from X-ray crystallographic, analytical and spectral techniques, and their utility in photodynamic therapy (PDT) of thyroid diseases caused by TSH receptor dysfunction is probed. The complexes display a visible spectral band within the PDT spectral window at ~690 nm. Photodynamic potential was estimated through DNA cleavage activity of the dpq and dppz complexes in UV-A light of 365 nm and red light of 676 nm. The reactions proceed via the hydroxyl radical pathway. The complexes retain their DNA photocleavage activity in red light under anaerobic conditions, a situation normally prevails in hypoxic tumor core. Investigation into the photocytotoxic potential of these complexes showed that the dppz complex 3 is approximately 4-fold more active in the HEK293 cells expressing human thyrotropin receptor (HEK293-hTSHR) than in the parental cell line and has an insignificant effect on an unrelated human cervical carcinoma cell line (HeLa). Photoexcitation of complex 3 in HEK293-hTSHR cells leads to damage hTSHR as evidenced from the decrease in cAMP formation both in absence and presence of hTSH and decrease in the TSHR immunofluorescence with a concomitant cytoplasmic translocation of the membrane protein, cadherin. The involvement of hTSHR is evidenced from the ability of complex 3 to bind to the extracellular domain of hTSHR (hTSHR-ECD) with a K(d) value of 81 nM and from the photocleavage of hTSHR-ECD.


Asunto(s)
Células/efectos de los fármacos , Células/efectos de la radiación , Cobalto/farmacología , Fenazinas/farmacología , Fotoquimioterapia , Receptores de Tirotropina/metabolismo , Rayos Ultravioleta , Células HEK293 , Células HeLa , Humanos , Modelos Moleculares , Compuestos Organometálicos/farmacología , Piridinas/farmacología
19.
Dalton Trans ; 39(7): 1807-16, 2010 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-20449426

RESUMEN

Cobalt(iii) complexes [Co(pnt)(B)(2)](NO(3))(2) (1-3) of pyridine-2-thiol (pnt) and phenanthroline bases (B), viz. 1,10-phenanthroline (phen in 1), dipyrido[3,2-d:2',3'-f]quinoxaline (dpq in ) and dipyrido[3,2-a:2',3'-c]phenazine (dppz in 3), have been prepared, characterized and their photo-induced anaerobic DNA cleavage activity studied. The crystal structure of 1a as mixed ClO(4)(-) and PF(6)(-) salt of 1 shows a Co(III)N(5)S coordination geometry in which the pnt and phen showed N,S- and N,N-donor binding modes, respectively. The complexes exhibit Co(iii)/Co(ii) redox couple near -0.3 V (vs. SCE) in 20% DMF-Tris-HCl buffer having 0.1 M TBAP. The complexes show binding propensity to calf thymus DNA giving K(b) values within 2.2 x 10(4)-7.3 x 10(5) M(-1). Thermal melting and viscosity data suggest DNA surface and/or groove binding of the complexes. The complexes show significant anaerobic DNA cleavage activity in red light under argon atmosphere possibly involving sulfide anion radical or thiyl radical species. The DNA cleavage reaction under aerobic medium in red light is found to involve both singlet oxygen and hydroxyl radical pathways. The dppz complex shows non-specific BSA and lysozyme protein cleavage activity in UV-A light of 365 nm via both hydroxyl and singlet oxygen pathways. The dppz complex exhibits photocytotoxicity in HeLa cervical cancer cells giving IC(50) values of 767 nM and 19.38 microM in UV-A light of 365 nm and in the dark, respectively. A significant reduction of the dark toxicity of the dppz base (IC(50) = 8.34 microM in dark) is observed on binding to the cobalt(iii) center.


Asunto(s)
ADN/efectos de los fármacos , Luz , Compuestos Organometálicos/farmacología , Fenantrolinas/química , Animales , Bovinos , Cristalografía por Rayos X , ADN/química , División del ADN , Modelos Moleculares , Compuestos Organometálicos/química , Fotoquímica
20.
Inorg Chem ; 49(9): 4036-45, 2010 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-20380391

RESUMEN

Lanthanide complexes of formulation [La(B)(2)(NO(3))(3)] (1-3) and [Gd(B)(2)(NO(3))(3)] (4-6), where B is a N,N-donor phenanthroline base, namely, 1,10-phenanthroline (phen in 1, 4), dipyrido[3,2-d:2',3'-f]quinoxaline (dpq in 2, 5) and dipyrido[3,2-a:2',3'-c]phenazine (dppz in 3, 6), have been prepared, characterized from physicochemical data, and their photoinduced DNA and protein cleavage activity studied. The photocytotoxicity of the dppz complexes 3 and 6 has been studied using HeLa cancer cells. The complexes exhibit ligand centered bands in the UV region. The dppz complexes show the lowest energy band at 380 nm in N,N-dimethylformamide (DMF). The La(III) complexes are diamagnetic. The Gd(III) complexes (4-6) have magnetic moments that correspond to seven unpaired electrons. The complexes are 1:1 electrolytic in aqueous DMF. The dpq and dppz complexes in DMF show ligand-based reductions. The complexes display moderate binding propensity to calf thymus DNA giving binding constant values in the range of 5.7 x 10(4)-5.8 x 10(5) M(-1) with a relative order: 3, 6 (dppz) > 2, 5 (dpq) > 1, 4 (phen). The binding data suggest DNA surface and/or groove binding nature of the complexes. The complexes do not show any hydrolytic cleavage of plasmid supercoiled pUC19 DNA. The dpq and dppz complexes efficiently cleave SC DNA to its nicked circular form on exposure to UV-A light of 365 nm at nanomolar complex concentration. Mechanistic studies reveal the involvement of singlet oxygen ((1)O(2)) and hydroxyl radical (HO*) as the cleavage active species. The complexes show binding propensity to bovine serum albumin (BSA) protein giving K(BSA) values of approximately 10(5) M(-1). The dppz complexes 3 and 6 show BSA protein cleavage activity in UV-A light of 365 nm. The dppz complexes 3 and 6 exhibit significant photocytotoxicity in HeLa cells giving respective IC(50) values of 341 nM and 573 nM in UV-A light of 365 nm for an exposure time of 15 min (IC(50) > 100 microM in dark for both the complexes). Control experiments show significant dark and phototoxicity of the dppz base alone (IC(50) = 413 nM in light with 4 h incubation in dark and 11.6 microM in dark with 24 h incubation). A significant decrease in the dark toxicity of the dppz base is observed on binding to the lanthanide ions while retaining similar phototoxicity.


Asunto(s)
División del ADN/efectos de la radiación , ADN/química , Gadolinio/química , Lantano/química , Compuestos Organometálicos/farmacología , Fenantrolinas/química , Rayos Ultravioleta , Animales , Bovinos , Supervivencia Celular/efectos de los fármacos , Células HeLa , Humanos , Estructura Molecular , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/química , Fotoquímica , Albúmina Sérica Bovina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...