Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Anal Chem ; 92(18): 12101-12110, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32790291

RESUMEN

Chemoresistance, i.e., tumor insensitivity to chemotherapy, shortens life expectancy of cancer patients. Despite the availability of new treatment options, initial systemic regimens for solid tumors are dominated by a set of standard chemotherapy drugs, and alternative therapies are used only when a patient has demonstrated chemoresistance clinically. Chemoresistance predictors use laboratory parameters measured on tissue samples to predict the patient's response to chemotherapy and help to avoid application of chemotherapy to chemoresistant patients. Despite thousands of publications on putative chemoresistance predictors, there are only about a dozen predictors that are sufficiently accurate for precision oncology. One of the major reasons for inaccuracy of predictors is inaccuracy of analytical methods utilized to measure their laboratory parameters: an inaccurate method leads to an inaccurate predictor. The goal of this study was to identify analytical challenges in chemoresistance-predictor development and suggest ways to overcome them. Here we describe principles of chemoresistance predictor development via correlating a clinical parameter, which manifests disease state, with a laboratory parameter. We further classify predictors based on the nature of laboratory parameters and analyze advantages and limitations of different predictors using the reliability of analytical methods utilized for measuring laboratory parameters as a criterion. Our eventual focus is on predictors with known mechanisms of reactions involved in drug resistance (drug extrusion, drug degradation, and DNA damage repair) and using rate constants of these reactions to establish accurate and robust laboratory parameters. Many aspects and conclusions of our analysis are applicable to all types of disease biomarkers built upon the correlation of clinical and laboratory parameters.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Antineoplásicos/análisis , Resistencia a Antineoplásicos/genética , Humanos , Neoplasias/diagnóstico , Neoplasias/genética
2.
Anal Chem ; 92(13): 9348-9355, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32522000

RESUMEN

Cytometry of Reaction Rate Constant (CRRC) uses time-lapse fluorescence microscopy to measure a rate constant of a catalytic reaction in individual cells and, thus, facilitate accurate size determination for cell subpopulations with distinct efficiencies of this reaction. Reliable CRRC requires uniform exposure of cells to the reaction substrate followed by their uniform imaging, which in turn, requires that a tissue sample be disintegrated into a suspension of dispersed cells, and these cells settle on the support surface before being analyzed by CRRC. We call such cells "dispersed-settled" to distinguish them from cells cultured as a monolayer. Studies of the dispersed-settled cells can be tissue-relevant only if the cells maintain their 3D tissue state during the multi-hour CRRC procedure. Here, we propose an approach for assessing tissue relevance of the CRRC-based analysis of the dispersed-settled cells. Our approach utilizes cultured multicellular spheroids as a 3D cell model and cultured cell monolayers as a 2D cell model. The CRRC results of the dispersed-settled cells derived from spheroids are compared to those of spheroids and monolayers in order to find if the dispersed-settled cells are representative of the spheroids. To demonstrate its practical use, we applied this approach to a cellular reaction of multidrug resistance (MDR) transport, which was followed by extrusion of a fluorescent substrate from the cells. The approach proved to be reliable and revealed long-term maintenance of MDR transport in the dispersed-settled cells obtained from cultured ovarian cancer spheroids. Accordingly, CRRC can be used to determine accurately the size of a cell subpopulation with an elevated level of MDR transport in tumor samples, which makes CRRC a suitable method for the development of MDR-based predictors of chemoresistance. The proposed spheroid-based approach for validation of CRRC is applicable to other types of cellular reactions and, thus, will be an indispensable tool for transforming CRRC from an experimental technique into a practical analytical tool.


Asunto(s)
Microscopía Fluorescente/métodos , Esferoides Celulares/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Fluoresceína/química , Humanos , Cinética , Esferoides Celulares/citología , Esferoides Celulares/patología , Imagen de Lapso de Tiempo
3.
Anal Chem ; 91(6): 4186-4194, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30829484

RESUMEN

Robust and accurate analysis of cell-population heterogeneity is challenging but required in many areas of biology and medicine. In particular, it is pivotal to the development of reliable cancer biomarkers. Here, we prove that cytometry of reaction rate constant (CRRC) can facilitate such analysis when the kinetic mechanism of a reaction associated with the heterogeneity is known. In CRRC, the cells are loaded with a reaction substrate, and its conversion into a product is followed by time-lapse fluorescence microscopy at the single-cell level. A reaction rate constant is determined for every cell, and a kinetic histogram "number of cells versus the rate constant" is used to determine quantitative parameters of reaction-based cell-population heterogeneity. Such parameters include, for example, the number and sizes of subpopulations. In this work, we applied CRRC to a reaction of substrate extrusion from cells by ATP-binding cassette (ABC) transporters. This reaction is viewed as a potential basis for predictive biomarkers of chemoresistance in cancer. CRRC proved to be robust (insensitive to variations in experimental settings) and accurate for finding quantitative parameters of cell-population heterogeneity. In contrast, a typical nonkinetic analysis, performed on the same data sets, proved to be both nonrobust and inaccurate. Our results suggest that CRRC can potentially facilitate the development of reliable cancer biomarkers on the basis of quantitative parameters of cell-population heterogeneity. A plausible implementation scenario of CRRC-based development, validation, and clinical use of a predictor of ovarian cancer chemoresistance to its frontline therapy is presented.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Citometría de Flujo/métodos , Microscopía Fluorescente/métodos , Neoplasias Ováricas/patología , Resistencia a Múltiples Medicamentos , Femenino , Humanos , Neoplasias Ováricas/metabolismo , Células Tumorales Cultivadas
4.
J Cell Biochem ; 118(1): 154-162, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27283126

RESUMEN

In functional cytometric studies, cultured cells are exposed to effectors (e.g., drugs), and the heterogeneity of cell responses are studied using cytometry techniques (e.g., image cytometry). Such studies are difficult to perform on 3D cell cultures. A solution is to disperse 3D clusters and transfer the cells to the 2D state before applying effectors and using cytometry. This approach requires that the lifetime of the 3D phenotype be longer than the duration of the experiment. Here we studied the dynamics of phenotype transformation from 3D to 2D and searched for means of slowing this transformation down in dispersed spheroids of MCF7 cells. We found three functional biomarkers of the 3D phenotype in MCF7 cell spheroids that are absent in the 2D cell culture: (i) the presence of a subpopulation with an elevated drug-expelling capacity; (ii) the presence of a subpopulation with an elevated cytoprotective capacity; and (iii) the accumulation of cells in the G1 phase of the cell cycle. Monitoring these biomarkers in cells transferred from the 3D state to the 2D state revealed their gradual extinction. We found that the combined application of an elevated cell density and thiol-containing medium supplements increased the lifetime of the 3D phenotype by several fold to as long as 96 h. Our results suggest that extending the lifetime of the 3D phenotype in the cells transferred from the 3D state to the 2D state can facilitate detailed functional cytometric studies, such as measurements of population heterogeneity of cytotoxicity, chemosensitivity, and radiosensitivity. J. Cell. Biochem. 118: 154-162, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Fase G1/fisiología , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Técnicas de Cultivo de Célula/métodos , Femenino , Humanos , Células MCF-7
5.
J Cell Biochem ; 117(1): 59-65, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26054050

RESUMEN

Inhibition of metabolic features which distinguish cancer cells from their non-malignant counterparts is a promising approach to cancer treatment. Energy support for drug extrusion in multidrug resistance (MDR) is a potential target for metabolic inhibition. Two major sources of ATP-based metabolic energy are partial (glycolysis) and complete (mitochondrial oxidative phosphorylation) oxidation of metabolic fuels. In cancer cells, the balance between them tends to be shifted toward glycolysis; this shift is considered to be characteristic of the cancer metabolic phenotype. Numerous earlier studies, conducted with cells cultured in a monolayer (2-D model), suggested inhibition of glycolytic ATP production as an efficient tool to suppress MDR in cancer cells. Yet, more recent work challenged the appropriateness of the 2-D model for such studies and suggested that a more clinically relevant approach would utilize a more advanced cellular model such as a 3-D model. Here, we show that the transition from the 2-D model (cultured monolayer) to a 3-D model (cultured spheroids) introduces essential changes into the concept of energetic suppression of MDR. The 3-D cell organization leads to the formation of a discrete cell subpopulation (not formed in the 2-D model) with elevated MDR transport capacity. This subpopulation has a specific metabolic phenotype (mixed glycolytic/oxidative MDR support) different from that of cells cultured in the 2-D model. Finally, the shift to the oxidative phenotype becomes greater when the spheroids are grown under conditions of lactic acidosis that are typical for solid tumors. The potential clinical significance of these findings is discussed.


Asunto(s)
Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Adenosina Trifosfato/metabolismo , Supervivencia Celular/fisiología , Citometría de Flujo , Humanos , Células MCF-7 , Modelos Biológicos
6.
Anal Chem ; 85(5): 2578-81, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23394374

RESUMEN

Phenotypically uniform cell populations may contain subpopulations with different activities of enzymes, membrane transporters, and other functional units which can be characterized kinetically. Here we propose the first approach to the discovery of such subpopulations; we term it single-cell approach to discover subpopulations or SCADS for short. SCADS combines microscopy, single-cell kinetic analysis, and population/cluster analysis to discover a functionally distinct subpopulation of cells. In this proof-of-principle work, we used SCADS to search for subpopulations with distinct kinetic patterns of membrane transport in bulk tumor cells (BTCs) and tumor-initiating cells (TICs). We used two classical Michaelis parameters, Vmax and KM, to kinetically characterize the rate of transport. We found that the BTC population was homogeneous with respect to membrane transport. When analyzing TICs, we discovered three main functionally distinct subpopulations: (i) cells with a high rate of transport (high Vmax), (ii) cells with high-affinity transporters (low KM), and (iii) cells with activity and affinity similar to those in BTCs (low Vmax and high KM).


Asunto(s)
Análisis de la Célula Individual/métodos , Transporte Biológico , Cinética , Neoplasias/patología , Fenotipo
7.
PLoS One ; 18(7): e0282990, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37399195

RESUMEN

Cytometry of Reaction Rate Constant (CRRC) is a method for studying cell-population heterogeneity using time-lapse fluorescence microscopy, which allows one to follow reaction kinetics in individual cells. The current and only CRRC workflow utilizes a single fluorescence image to manually identify cell contours which are then used to determine fluorescence intensity of individual cells in the entire time-stack of images. This workflow is only reliable if cells maintain their positions during the time-lapse measurements. If the cells move, the original cell contours become unsuitable for evaluating intracellular fluorescence and the CRRC experiment will be inaccurate. The requirement of invariant cell positions during a prolonged imaging is impossible to satisfy for motile cells. Here we report a CRRC workflow developed to be applicable to motile cells. The new workflow combines fluorescence microscopy with transmitted-light microscopy and utilizes a new automated tool for cell identification and tracking. A transmitted-light image is taken right before every fluorescence image to determine cell contours, and cell contours are tracked through the time-stack of transmitted-light images to account for cell movement. Each unique contour is used to determine fluorescence intensity of cells in the associated fluorescence image. Next, time dependencies of the intracellular fluorescence intensities are used to determine each cell's rate constant and construct a kinetic histogram "number of cells vs rate constant." The new workflow's robustness to cell movement was confirmed experimentally by conducting a CRRC study of cross-membrane transport in motile cells. The new workflow makes CRRC applicable to a wide range of cell types and eliminates the influence of cell motility on the accuracy of results. Additionally, the workflow could potentially monitor kinetics of varying biological processes at the single-cell level for sizable cell populations. Although our workflow was designed ad hoc for CRRC, this cell-segmentation/cell-tracking strategy also represents an entry-level, user-friendly option for a variety of biological assays (i.e., migration, proliferation assays, etc.). Importantly, no prior knowledge of informatics (i.e., training a model for deep learning) is required.


Asunto(s)
Rastreo Celular , Procesamiento de Imagen Asistido por Computador , Movimiento Celular , Rastreo Celular/métodos , Microscopía Fluorescente/métodos , Procesamiento de Imagen Asistido por Computador/métodos
8.
Anal Chem ; 83(16): 6132-4, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21770449

RESUMEN

Multidrug resistance (MDR) driven by active efflux of drugs from the cells is one of the major obstacles in chemotherapies. Understanding the nature of MDR and designing more efficient chemotherapies requires the comparison of the efflux rate between different subpopulations of cells. Here we propose a single-cell-kinetics approach for such a comparison. In essence, the entire cell population is loaded with a suitable fluorescent substrate for MDR-associated membrane transporters. The kinetics of substrate efflux from individual cells is followed by time-lapse fluorescence microscopy and analyzed at the single-cell level. Microscopy is also used to assign cells to different subpopulations based on differences in morphology or level of staining by molecular probes. The kinetic parameters obtained for individual cells are then averaged for different cell subpopulations and the mean values of these parameters are finally compared between subpopulations. To test our single-cell-kinetics approach, we studied MDR-related efflux for two subpopulations of cultured breast cancer cells: cells in 2N and 4N phases of the cell cycle. The assignment of cells to 2N and 4N subpopulations was done by fluorescent DNA staining after the completion of efflux. By using the single-cell-kinetics approach, we were able to prove for the first time that the rates of MDR-related efflux differ in 2N and 4N phases of the cell cycle. We foresee that this approach will be an important tool in studies of MDR and in designing combination chemotherapies.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Análisis de la Célula Individual/métodos , Antineoplásicos/metabolismo , Transporte Biológico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Ciclo Celular , Línea Celular Tumoral , Resistencia a Múltiples Medicamentos , Femenino , Fluoresceína/análisis , Humanos , Cinética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Ploidias
9.
Proc Natl Acad Sci U S A ; 105(33): 12057-62, 2008 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-18701716

RESUMEN

In pancreatic beta-cells, uncoupling protein 2 (UCP2) influences mitochondrial oxidative phosphorylation and insulin secretion. Here, we show that alpha-cells express significantly higher levels of UCP2 than do beta-cells. Greater mitochondrial UCP2-related uncoupling was observed in alpha-cells compared with beta-cells and was accompanied by a lower oxidative phosphorylation efficiency (ATP/O). Conversely, reducing UCP2 activity in alpha-cells was associated with higher mitochondrial membrane potential generated by glucose oxidation and with increased ATP synthesis, indicating more efficient metabolic coupling. In vitro, the suppression of UCP2 activity led to reduced glucagon secretion in response to low glucose; however, in vivo, fasting glucagon levels were normal in UCP2(-/-) mice. In addition to its effects on secretion, UCP2 played a cytoprotective role in islets, with UCP2(-/-) alpha-cells being more sensitive to specific death stimuli. In summary, we demonstrate a direct role for UCP2 in maintaining alpha-cell function at the level of glucose metabolism, glucagon secretion, and cytoprotection.


Asunto(s)
Células Secretoras de Glucagón/metabolismo , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Adenosina Trifosfato/biosíntesis , Animales , Línea Celular , Supervivencia Celular , Glucagón/metabolismo , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Canales Iónicos/deficiencia , Canales Iónicos/genética , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Noqueados , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , ARN Interferente Pequeño/genética , Proteína Desacopladora 2
10.
Mol Clin Oncol ; 15(2): 161, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34295468

RESUMEN

A primary reason for chemotherapy failure is chemoresistance, which is driven by various mechanisms. Multi-drug resistance (MDR) is one such mechanism that is responsible for drug extrusion from the intracellular space. MDR can be intrinsic and thus, may pre-exist the first application of chemotherapy. However, MDR may also be acquired during tumor exposure to chemotherapeutic agents. To understand whether cell clustering can influence intrinsic and acquired MDR, the present study assessed cultured monolayers (representing individual cells) and spheroids (representing clusters) formed by cisplatin-naïve (intrinsic MDR) and cisplatin-exposed (acquired MDR) lines of ovarian cancer A2780 cells by determining the cytometry of reaction rate constant (CRRC). MDR efflux was characterized using accurate and robust cell number vs. MDR efflux rate constant (k MDR) histograms. Both cisplatin-naïve and cisplatin-exposed monolayer cells presented unimodal histograms; the histogram of cisplatin-exposed cells was shifted towards a higher k MDR value suggesting greater MDR activity. Spheroids of cisplatin-naïve cells presented a bimodal histogram indicating the presence of two subpopulations with different MDR activity. In contrast, spheroids of cisplatin-exposed cells presented a unimodal histogram qualitatively similar to that of the monolayers of cisplatin-exposed cells but with a moderate shift towards greater MDR activity. A flow-cytometry assessment of multidrug resistance-associated protein 1 transporter levels in monolayers and dissociated spheroids revealed distributions similar to those of k MDR, thus, suggesting a plausible molecular mechanism for the observed differences in MDR activity. The observed greater effect of cell clustering on intrinsic rather than in acquired MDR can help guide the development of new therapeutic strategies targeting clusters of circulating tumor cells.

11.
Diabetes ; 53 Suppl 1: S136-42, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14749279

RESUMEN

Stressors such as chronic hyperglycemia or hyperlipidemia may lead to insufficient insulin secretion in susceptible individuals, contributing to type 2 diabetes. The molecules mediating this effect are just beginning to be identified. Uncoupling protein (UCP)-2 may be one such negative modulator of insulin secretion. Accumulating evidence shows that beta-cell UCP2 expression is upregulated by glucolipotoxic conditions and that increased activity of UCP2 decreases insulin secretion. Mitochondrial superoxide has been identified as a posttranslational regulator of UCP2 activity in islets; thus, UCP2 may provide protection to beta-cells at one level while simultaneously having detrimental effects on insulin secretion. Interestingly, the latter appears to be the dominant outcome, because UCP2 knockout mice display an increased beta-cell mass and retained insulin secretion capacity in the face of glucolipotoxicity.


Asunto(s)
Islotes Pancreáticos/fisiología , Proteínas de Transporte de Membrana/fisiología , Proteínas Mitocondriales/fisiología , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatología , Regulación de la Expresión Génica/fisiología , Humanos , Insulina/genética , Insulina/metabolismo , Secreción de Insulina , Canales Iónicos , Fenómenos Fisiológicos de la Nutrición , Especies Reactivas de Oxígeno/metabolismo , Proteína Desacopladora 2
12.
Diabetes ; 53(1): 129-40, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14693707

RESUMEN

Oligonucleotide microarrays were used to define oleic acid (OA)-regulated gene expression and proteomic technology to screen protein kinases in MIN6 insulinoma cells. The effects of oxidative stress caused by OA and potential protective effects of N-acetyl-L-cysteine (NAC), a scavenger of reactive oxygen species (ROS), on global gene expression and beta-cell function were investigated. Long-term exposure of MIN6 cells to OA led to a threefold increase in basal insulin secretion, a 50% decrease in insulin content, an inhibition of glucose-stimulated insulin secretion (GSIS), and a twofold increase in the level of ROS. The addition of NAC normalized both the OA-induced insulin content and ROS elevation, but it failed to restore GSIS. Microarray studies and subsequent quantitative PCR analysis showed that OA consistently regulated the expression of 45 genes involved in metabolism, cell growth, signal transduction, transcription, and protein processing. The addition of NAC largely normalized the expression of the OA-regulated genes involved in cell growth and differentiation but not other functions. A protein kinase screen showed that OA regulated the expression and/or phosphorylation levels of kinases involved in stress-response mitogen-activated protein kinase, phosphatidylinositol 3-kinase, and cell cycle control pathways. Importantly, these findings indicate that chronic OA exposure can impair beta-cell function through ROS-dependent and -independent mechanisms.


Asunto(s)
Perfilación de la Expresión Génica , Islotes Pancreáticos/fisiología , Estrés Oxidativo/fisiología , Proteínas Quinasas/genética , Especies Reactivas de Oxígeno/farmacología , Acetilcisteína/farmacología , Animales , Línea Celular , Enzimas/efectos de los fármacos , Enzimas/genética , Islotes Pancreáticos/efectos de los fármacos , Ácido Ocadaico/farmacología , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo/efectos de los fármacos , Procesamiento Postranscripcional del ARN/efectos de los fármacos
13.
Diabetes ; 51(11): 3211-9, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12401712

RESUMEN

Uncoupling protein 2 (UCP2) may act as an important regulator of insulin secretion. In this study, beta-cell function in UCP2-deficient mice was examined after a 45% high-fat diet (HFD) to assess its role during the development of diet-induced type 2 diabetes. HFD-fed UCP2 (-/-) mice have lower fasting blood glucose and elevated insulin levels when compared with wild-type (WT) mice. UCP2 (-/-) mice also have enhanced beta-cell glucose sensitivity compared with WT mice after HFD, a result that is due in part to the deterioration of glucose responsiveness in WT mice. HFD-fed UCP2 (-/-) mice have increased insulin secretory capacity as a result of increased pancreatic beta-cell mass and insulin content per islet. Islets from WT mice exposed to 0.5 mmol/l palmitate for 48 h have significantly reduced mitochondrial membrane potential, ATP concentrations, and glucose responsiveness compared with UCP2 (-/-) islets, suggesting that elevated UCP2 in WT mice increases proton leak and decreases mitochondrial ATP production. Highly increased carnitine palmitoyl transferase-1 gene expression in UCP2 (-/-) mice is suggestive of enhanced fatty acid oxidizing capacity, particularly after HFD stress. These results further establish UCP2 as a component in glucose sensing and suggest a possible new aspect of UCP2 function during the progression of type 2 diabetes.


Asunto(s)
Grasas de la Dieta/farmacología , Insulina/metabolismo , Proteínas de Transporte de Membrana , Proteínas Mitocondriales , Animales , Secuencia de Bases , Glucemia/metabolismo , Cartilla de ADN , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Ácidos Grasos no Esterificados/sangre , Glucagón/sangre , Insulina/sangre , Secreción de Insulina , Canales Iónicos , Ratones , Ratones Noqueados , Proteínas/genética , Proteínas/fisiología , Triglicéridos/sangre , Proteína Desacopladora 2 , Aumento de Peso
14.
PLoS One ; 8(11): e79222, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24223908

RESUMEN

Multidrug resistance (MDR) driven by ABC (ATP binding cassette) membrane transporters is one of the major causes of treatment failure in human malignancy. MDR capacity is thought to be unevenly distributed among tumor cells, with higher capacity residing in tumor-initiating cells (TIC) (though opposite finding are occasionally reported). Functional evidence for enhanced MDR of TICs was previously provided using a "side population" assay. This assay estimates MDR capacity by a single parameter - cell's ability to retain fluorescent MDR substrate, so that cells with high MDR capacity ("side population") demonstrate low substrate retention. In the present work MDR in TICs was investigated in greater detail using a kinetic approach, which monitors MDR efflux from single cells. Analysis of kinetic traces obtained allowed for the estimation of both the velocity (V max) and affinity (K M) of MDR transport in single cells. In this way it was shown that activation of MDR in TICs occurs in two ways: through the increase of V max in one fraction of cells, and through decrease of K M in another fraction. In addition, kinetic data showed that heterogeneity of MDR parameters in TICs significantly exceeds that of bulk cells. Potential consequences of these findings for chemotherapy are discussed.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Células Madre Neoplásicas/metabolismo , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Animales , Transporte Biológico/efectos de los fármacos , División Celular , Línea Celular Tumoral , Citometría de Flujo , Fluoresceína/metabolismo , Fase G1 , Fase G2 , Gliburida/farmacología , Humanos , Receptores de Hialuranos/metabolismo , Cinética , Células MCF-7 , Microscopía Fluorescente , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Esferoides Celulares/metabolismo
15.
PLoS One ; 7(7): e41368, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22848474

RESUMEN

Multidrug resistance driven by ABC membrane transporters is one of the major reasons for treatment failure in human malignancy. Some limited evidence has previously been reported on the cell cycle dependence of ABC transporter expression. However, it has never been demonstrated that the functional activity of these transporters correlates with the cell cycle position. Here, we studied the rate of intrinsic ABC transport in different phases of the cell cycle in cultured MCF-7 breast cancer cells. The rate was characterized in terms of the efflux kinetics from cells loaded with an ABC transporter substrate. As averaging the kinetics over a cell population could lead to errors, we studied kinetics of ABC transport at the single-cell level. We found that the rate of ABC transport in MCF-7 cells could be described by Michaelis-Menten kinetics with two classical parameters, V(max) and K(M). Each of these parameters showed similar unimodal distributions with different positions of maxima for cell subpopulations in the 2c and 4c states. Compared to the 2c cells, the 4c cells exhibited greater V(max) values, indicating a higher activity of transport. They also exhibited a greater V(max)/K(M) ratio, indicating a higher efficiency of transport. Our findings suggest that cell cycle-related modulation of MDR may need to be taken into account when designing chemotherapy regimens which include cytostatic agents.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Neoplasias de la Mama/metabolismo , Ciclo Celular , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Citostáticos/farmacología , Citostáticos/uso terapéutico , Femenino , Humanos
16.
J Med Chem ; 55(7): 3546-52, 2012 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-22471443

RESUMEN

The Escherichia coli (E. coli) AlkB protein and its functional human homologues belong to a subfamily of 2-oxoglutarate (2OG) dependent oxygenases (2OG oxygenases for simplicity) that enable the repair of cytotoxic methylation damage in nucleic acids and that catalyze t-RNA oxidations. DNA alkylation is a major mechanism of action for cytotoxic anticancer drugs. Thus, the inhibition of oxidative demethylation, catalyzed by these enzymes, has the potential to improve the efficacy of chemotherapies. Here we report that oligonucleotide aptamers constitute a new class of potent inhibitors of 2OG oxygenases. DNA aptamers can selectively bind to AlkB, with nanomolar affinity, and efficiently inhibit catalysis. The mechanism of inhibition was studied by capillary electrophoresis (CE) with laser-induced fluorescence (LIF) detection. Inhibition constants of the aptamers were determined and shown to correlate well with K(d) values. The results of kinetic analyses imply that the aptamers bind AlkB away from the active site. Our findings should stimulate the development of oligonucleotide aptamers for human homologues of AlkB and further their study as potential enhancers of chemotherapy efficiency.


Asunto(s)
Aptámeros de Nucleótidos/química , ADN/química , Proteínas de Escherichia coli/antagonistas & inhibidores , Ácidos Cetoglutáricos/metabolismo , Oxigenasas de Función Mixta/antagonistas & inhibidores , Aminoácidos Dicarboxílicos/química , Biocatálisis , Metilación de ADN , Pruebas de Enzimas , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Compuestos Ferrosos/química , Humanos , Cinética , Oxigenasas de Función Mixta/química , Oxigenasas de Función Mixta/metabolismo , Unión Proteica , Relación Estructura-Actividad
17.
Diabetes ; 60(11): 2710-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21984579

RESUMEN

OBJECTIVE: The role of uncoupling protein 2 (UCP2) in pancreatic ß-cells is highly debated, partly because of the broad tissue distribution of UCP2 and thus limitations of whole-body UCP2 knockout mouse models. To investigate the function of UCP2 in the ß-cell, ß-cell-specific UCP2 knockout mice (UCP2BKO) were generated and characterized. RESEARCH DESIGN AND METHODS: UCP2BKO mice were generated by crossing loxUCP2 mice with mice expressing rat insulin promoter-driven Cre recombinase. Several in vitro and in vivo parameters were measured, including respiration rate, mitochondrial membrane potential, islet ATP content, reactive oxygen species (ROS) levels, glucose-stimulated insulin secretion (GSIS), glucagon secretion, glucose and insulin tolerance, and plasma hormone levels. RESULTS: UCP2BKO ß-cells displayed mildly increased glucose-induced mitochondrial membrane hyperpolarization but unchanged rates of uncoupled respiration and islet ATP content. UCP2BKO islets had elevated intracellular ROS levels that associated with enhanced GSIS. Surprisingly, UCP2BKO mice were glucose-intolerant, showing greater α-cell area, higher islet glucagon content, and aberrant ROS-dependent glucagon secretion under high glucose conditions. CONCLUSIONS: Using a novel ß-cell-specific UCP2KO mouse model, we have shed light on UCP2 function in primary ß-cells. UCP2 does not behave as a classical metabolic uncoupler in the ß-cell, but has a more prominent role in the regulation of intracellular ROS levels that contribute to GSIS amplification. In addition, ß-cell UCP2 contributes to the regulation of intraislet ROS signals that mediate changes in α-cell morphology and glucagon secretion.


Asunto(s)
Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Canales Iónicos/fisiología , Proteínas Mitocondriales/fisiología , Especies Reactivas de Oxígeno/metabolismo , Animales , Genes Reporteros , Células Secretoras de Glucagón/patología , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Humanos , Hiperglucemia/metabolismo , Insulina/genética , Secreción de Insulina , Células Secretoras de Insulina/patología , Canales Iónicos/genética , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Masculino , Potencial de la Membrana Mitocondrial , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/genética , Especificidad de Órganos , Regiones Promotoras Genéticas , Ratas , Técnicas de Cultivo de Tejidos , Proteína Desacopladora 2
18.
Diabetes ; 59(2): 448-59, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19903739

RESUMEN

OBJECTIVE The inability of pancreatic beta-cells to appropriately respond to glucose and secrete insulin are primary defects associated with beta-cell failure in type 2 diabetes. Mitochondrial dysfunction has been implicated as a key factor in the development of type 2 diabetes; however, a link between mitochondrial dysfunction and defective insulin secretion is unclear. RESEARCH DESIGN AND METHODS We investigated the changes in islet mitochondrial function and morphology during progression from insulin resistance (3 weeks old), immediately before hyperglycemia (5 weeks old), and after diabetes onset (10 weeks old) in transgenic MKR mice compared with controls. The molecular and protein changes at 10 weeks were determined using microarray and iTRAQ proteomic screens. RESULTS At 3 weeks, MKR mice were hyperinsulinemic but normoglycemic and beta-cells showed negligible mitochondrial or morphological changes. At 5 weeks, MKR islets displayed abrogated hyperpolarization of mitochondrial membrane potential (DeltaPsi(m)), reduced mitochondrial Ca(2+) uptake, slightly enlarged mitochondria, and reduced glucose-stimulated insulin secretion. By 10 weeks, MKR mice were hyperglycemic and hyperinsulinemic and beta-cells contained swollen mitochondria with disordered cristae. beta-Cells displayed impaired stimulus-secretion coupling including reduced hyperpolarization of DeltaPsi(m), impaired Ca(2+)-signaling, and reduced glucose-stimulated ATP/ADP and insulin release. Furthermore, decreased cytochrome c oxidase-dependent oxygen consumption and signs of oxidative stress were observed in diabetic islets. Protein profiling of diabetic islets revealed that 36 mitochondrial proteins were differentially expressed, including inner membrane proteins of the electron transport chain. CONCLUSIONS We provide novel evidence for a critical role of defective mitochondrial oxidative phosphorylation and morphology in the pathology of insulin resistance-induced beta-cell failure.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/fisiología , Mitocondrias/patología , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Glucemia/metabolismo , Calcio/metabolismo , ADN Mitocondrial/genética , Diabetes Mellitus Tipo 2/genética , Modelos Animales de Enfermedad , Humanos , Hiperglucemia/complicaciones , Hiperglucemia/fisiopatología , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Secreción de Insulina , Células Secretoras de Insulina/patología , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos , Ratones Transgénicos , Fosforilación Oxidativa , Estrés Oxidativo/fisiología , Proteínas/genética , ARN Ribosómico/genética , ARN Ribosómico/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Diabetes ; 58(9): 2070-83, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19542200

RESUMEN

OBJECTIVE: Zinc ions are essential for the formation of hexameric insulin and hormone crystallization. A nonsynonymous single nucleotide polymorphism rs13266634 in the SLC30A8 gene, encoding the secretory granule zinc transporter ZnT8, is associated with type 2 diabetes. We describe the effects of deleting the ZnT8 gene in mice and explore the action of the at-risk allele. RESEARCH DESIGN AND METHODS: Slc30a8 null mice were generated and backcrossed at least twice onto a C57BL/6J background. Glucose and insulin tolerance were measured by intraperitoneal injection or euglycemic clamp, respectively. Insulin secretion, electrophysiology, imaging, and the generation of adenoviruses encoding the low- (W325) or elevated- (R325) risk ZnT8 alleles were undertaken using standard protocols. RESULTS: ZnT8(-/-) mice displayed age-, sex-, and diet-dependent abnormalities in glucose tolerance, insulin secretion, and body weight. Islets isolated from null mice had reduced granule zinc content and showed age-dependent changes in granule morphology, with markedly fewer dense cores but more rod-like crystals. Glucose-stimulated insulin secretion, granule fusion, and insulin crystal dissolution, assessed by total internal reflection fluorescence microscopy, were unchanged or enhanced in ZnT8(-/-) islets. Insulin processing was normal. Molecular modeling revealed that residue-325 was located at the interface between ZnT8 monomers. Correspondingly, the R325 variant displayed lower apparent Zn(2+) transport activity than W325 ZnT8 by fluorescence-based assay. CONCLUSIONS: ZnT8 is required for normal insulin crystallization and insulin release in vivo but not, remarkably, in vitro. Defects in the former processes in carriers of the R allele may increase type 2 diabetes risks.


Asunto(s)
Glucemia/metabolismo , Proteínas de Transporte de Catión/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Insulina/metabolismo , Zinc/metabolismo , Animales , Proteínas de Transporte de Catión/genética , Gránulos Citoplasmáticos/metabolismo , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/genética , Exocitosis/fisiología , Femenino , Expresión Génica/fisiología , Células HeLa , Homeostasis/fisiología , Humanos , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Polimorfismo Genético , Factores de Riesgo , Transportador 8 de Zinc
20.
J Biol Chem ; 283(12): 7936-48, 2008 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-18162472

RESUMEN

Involvement of the mitochondrial permeability transition (MPT) pore in early stages of lipotoxic stress in the pancreatic beta-cell lines MIN6 and INS-1 was the focus of this study. Both long term (indirect) and acute (direct) effects of fatty acid (FA) application on beta-cell susceptibility to Ca(2+)-induced MPT induction were examined using both permeabilized and intact beta-cells. Long term exposure to moderate (i.e. below cytotoxic) levels of the saturated FA palmitate sensitized beta-cell mitochondria to MPT induced by Ca(2+). Long term exposure to palmitate was significantly a more efficient inducer of MPT than the unsaturated FA oleate, although upon acute application both caused similar MPT activation. Application of antioxidants, inhibitors of the ceramide pathway, or modifiers of membrane fluidity did not protect beta-cell mitochondria from FA exposure. However, significant protection was provided by co-application of the unsaturated FA oleate in a phosphatidylinositol 3-kinase-dependent manner. Characterization of MPT pore opening in response to moderate palmitate treatment revealed the opening of a unique form of MPT in beta-cells as it encompassed features of both low and high conductance MPT states. Specifically, this MPT showed solute selectivity, characteristic of a low conductance MPT; however, it affected mitochondrial respiration and membrane potential in a way typical of a high conductance MPT. Activation of the full-size/high conductance form of MPT required application of high levels of FA that reduced growth and initiated apoptosis. These findings suggest that in the beta-cell, MPTs can act as both initiators of cell death and as versatile modulators of cell metabolism, depending on the mode of the MPT pore induced.


Asunto(s)
Permeabilidad de la Membrana Celular/efectos de los fármacos , Inhibidores Enzimáticos/toxicidad , Células Secretoras de Insulina/metabolismo , Mitocondrias/metabolismo , Ácido Palmítico/toxicidad , Animales , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Línea Celular , Células Secretoras de Insulina/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/patología , Ácido Oléico/metabolismo , Ácido Oléico/toxicidad , Consumo de Oxígeno/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA