Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Haematologica ; 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38934082

RESUMEN

The treatment of blast phase chronic myeloid leukemia (bpCML) remains a challenge due at least in part to drug resistance of leukemia stem cells (LSCs). Recent clinical evidence suggests that the BCL-2 inhibitor venetoclax in combination with ABL-targeting tyrosine kinase inhibitors (TKIs) can eradicate bpCML LSCs. In this report, we employed preclinical models of bpCML to investigate the efficacy and underlying mechanism of LSC-targeting with venetoclax/TKI combinations. Transcriptional analysis of LSCs exposed to venetoclax and dasatinib revealed upregulation of genes involved in lysosomal biology, in particular lysosomal acid lipase A (LIPA), a regulator of free fatty acids. Metabolomic analysis confirmed increased levels of free fatty acids in response to venetoclax/dasatinib. Pre-treatment of leukemia cells with bafilomycin, a specific lysosome inhibitor, or genetic perturbation of LIPA, resulted in increased sensitivity of leukemia cells toward venetoclax/dasatinib, implicating LIPA in treatment resistance. Importantly, venetoclax/dasatinib treatment does not affect normal stem cell function, suggestive of a leukemia-specific response. These results demonstrate that venetoclax/dasatinib is an LSCselective regimen in bpCML and that disrupting LIPA and fatty acid transport enhances venetoclax/dasatinib response in targeting LSCs, providing a rationale for exploring lysosomal disruption as an adjunct therapeutic strategy to prolong disease remission.

2.
Dev Cell ; 59(3): 384-399.e5, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38198890

RESUMEN

Different types of cells uptake fatty acids in response to different stimuli or physiological conditions; however, little is known about context-specific regulation of fatty acid uptake. Here, we show that muscle injury induces fatty acid uptake in muscle stem cells (MuSCs) to promote their proliferation and muscle regeneration. In humans and mice, fatty acids are mobilized after muscle injury. Through CD36, fatty acids function as both fuels and growth signals to promote MuSC proliferation. Mechanistically, injury triggers the translocation of CD36 in MuSCs, which relies on dynamic palmitoylation of STX11. Palmitoylation facilitates the formation of STX11/SNAP23/VAMP4 SANRE complex, which stimulates the fusion of CD36- and STX11-containing vesicles. Restricting fatty acid supply, blocking fatty acid uptake, or inhibiting STX11 palmitoylation attenuates muscle regeneration in mice. Our studies have identified a critical role of fatty acids in muscle regeneration and shed light on context-specific regulation of fatty acid sensing and uptake.


Asunto(s)
Ácidos Grasos , Lipoilación , Músculo Esquelético , Proteínas Qa-SNARE , Regeneración , Animales , Humanos , Ratones , Transporte Biológico , Antígenos CD36/metabolismo , Membrana Celular/metabolismo , Ácidos Grasos/metabolismo , Músculo Esquelético/lesiones , Músculo Esquelético/fisiología , Proteínas Qa-SNARE/metabolismo
3.
Nat Commun ; 15(1): 133, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38168040

RESUMEN

Adipocytes are the primary sites for fatty acid storage, but the synthesis rate of fatty acids is very low. The physiological significance of this phenomenon remains unclear. Here, we show that surplus fatty acid synthesis in adipocytes induces necroptosis and lipodystrophy. Transcriptional activation of FASN elevates fatty acid synthesis, but decreases NADPH level and increases ROS production, which ultimately leads to adipocyte necroptosis. We identify MED20, a subunit of the Mediator complex, as a negative regulator of FASN transcription. Adipocyte-specific male Med20 knockout mice progressively develop lipodystrophy, which is reversed by scavenging ROS. Further, in a murine model of HIV-associated lipodystrophy and a human patient with acquired lipodystrophy, ROS neutralization significantly improves metabolic disorders, indicating a causal role of ROS in disease onset. Our study well explains the low fatty acid synthesis rate in adipocytes, and sheds light on the management of acquired lipodystrophy.


Asunto(s)
Adipocitos , Lipodistrofia , Masculino , Ratones , Humanos , Animales , Especies Reactivas de Oxígeno/metabolismo , Adipocitos/metabolismo , Lipodistrofia/genética , Lipodistrofia/metabolismo , Ácidos Grasos/metabolismo , Estrés Oxidativo , Ratones Noqueados
4.
Adv Mater ; 35(1): e2209015, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36245327

RESUMEN

Solar desalination is one of the most promising strategies to address the global freshwater shortage crisis. However, the residual salt accumulated on the top surface of solar evaporators severely reduces light absorption and steam evaporation efficiency, thus impeding the further industrialization of this technology. Herein, a metal-phenolic network (MPN)-engineered 3D evaporator composed of photothermal superhydrophilic/superhydrophobic sponges and side-twining hydrophilic threads for efficient desalination with directional salt crystallization and zero liquid discharge is reported. The MPN coatings afford the engineering of alternating photothermal superhydrophilic/superhydrophobic sponges with high heating efficiency and defined vapor escape channels, while the side-twining threads induce site-selective salt crystallization. The 3D evaporator exhibits a high and stable indoor desalination rate (≈2.3 kg m-2  h-1 ) of concentrated seawater (20 wt%) under simulated sun irradiation for over 21 days without the need for salt crystallization inhibitors. This direct desalination is also achieved in outdoor field operations with a production rate of clean water up to ≈1.82 kg m-2  h-1 from concentrated seawater (10 wt%). Together with the high affinity and multiple functions of MPNs, this work is expected to facilitate the rational design of solar desalination devices and boost the research translation of MPN materials in broader applications.

5.
Blood ; 139(4): 584-596, 2022 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-34525179

RESUMEN

Acute myeloid leukemia (AML) is characterized by the presence of leukemia stem cells (LSCs), and failure to fully eradicate this population contributes to disease persistence/relapse. Prior studies have characterized metabolic vulnerabilities of LSCs, which demonstrate preferential reliance on oxidative phosphorylation (OXPHOS) for energy metabolism and survival. In the present study, using both genetic and pharmacologic strategies in primary human AML specimens, we show that signal transducer and activator of transcription 3 (STAT3) mediates OXPHOS in LSCs. STAT3 regulates AML-specific expression of MYC, which in turn controls transcription of the neutral amino acid transporter gene SLC1A5. We show that genetic inhibition of MYC or SLC1A5 acts to phenocopy the impairment of OXPHOS observed with STAT3 inhibition, thereby establishing this axis as a regulatory mechanism linking STAT3 to energy metabolism. Inhibition of SLC1A5 reduces intracellular levels of glutamine, glutathione, and multiple tricarboxylic acid (TCA) cycle metabolites, leading to reduced TCA cycle activity and inhibition of OXPHOS. Based on these findings, we used a novel small molecule STAT3 inhibitor, which binds STAT3 and disrupts STAT3-DNA, to evaluate the biological role of STAT3. We show that STAT3 inhibition selectively leads to cell death in AML stem and progenitor cells derived from newly diagnosed patients and patients who have experienced relapse while sparing normal hematopoietic cells. Together, these findings establish a STAT3-mediated mechanism that controls energy metabolism and survival in primitive AML cells.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/metabolismo , Leucemia Mieloide Aguda/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Supervivencia Celular , Humanos , Células Madre Neoplásicas/citología , Fosforilación Oxidativa , Células Tumorales Cultivadas
6.
Cancer Discov ; 11(2): 500-519, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33028621

RESUMEN

Due to the disseminated nature of leukemia, malignant cells are exposed to many different tissue microenvironments, including a variety of extramedullary sites. In the present study, we demonstrate that leukemic cells residing in the liver display unique biological properties and also contribute to systemic changes that influence physiologic responses to chemotherapy. Specifically, the liver microenvironment induces metabolic adaptations via upregulating expression of endothelial lipase in leukemia cells, which not only stimulates tumor cell proliferation through polyunsaturated fatty acid-mediated pathways, but also promotes survival by stabilizing antiapoptotic proteins. Additionally, hepatic infiltration and tissue damage caused by malignant cells induces release of liver-derived enzymes capable of degrading chemotherapy drugs, an event that further protects leukemia cells from conventional therapies. Together, these studies demonstrate a unique role for liver in modulating the pathogenesis of leukemic disease and suggest that the hepatic microenvironment may protect leukemia cells from chemotherapeutic challenge. SIGNIFICANCE: The studies presented herein demonstrate that the liver provides a microenvironment in which leukemia cells acquire unique metabolic properties. The adaptations that occur in the liver confer increased resistance to chemotherapy. Therefore, we propose that therapies designed to overcome liver-specific metabolic changes will yield improved outcomes for patients with leukemia.This article is highlighted in the In This Issue feature, p. 211.


Asunto(s)
Leucemia/metabolismo , Lipasa/metabolismo , Hígado/metabolismo , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Microambiente Tumoral
7.
Cell Stem Cell ; 27(5): 748-764.e4, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-32822582

RESUMEN

We previously demonstrated that leukemia stem cells (LSCs) in de novo acute myeloid leukemia (AML) patients are selectively reliant on amino acid metabolism and that treatment with the combination of venetoclax and azacitidine (ven/aza) inhibits amino acid metabolism, leading to cell death. In contrast, ven/aza fails to eradicate LSCs in relapsed/refractory (R/R) patients, suggesting altered metabolic properties. Detailed metabolomic analysis revealed elevated nicotinamide metabolism in relapsed LSCs, which activates both amino acid metabolism and fatty acid oxidation to drive OXPHOS, thereby providing a means for LSCs to circumvent the cytotoxic effects of ven/aza therapy. Genetic and pharmacological inhibition of nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in nicotinamide metabolism, demonstrated selective eradication of R/R LSCs while sparing normal hematopoietic stem/progenitor cells. Altogether, these findings demonstrate that elevated nicotinamide metabolism is both the mechanistic basis for ven/aza resistance and a metabolic vulnerability of R/R LSCs.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes , Leucemia Mieloide Aguda , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Células Madre Neoplásicas , Niacinamida/farmacología , Células Madre , Sulfonamidas
8.
Cancer Discov ; 10(4): 536-551, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31974170

RESUMEN

Venetoclax-based therapy can induce responses in approximately 70% of older previously untreated patients with acute myeloid leukemia (AML). However, up-front resistance as well as relapse following initial response demonstrates the need for a deeper understanding of resistance mechanisms. In the present study, we report that responses to venetoclax +azacitidine in patients with AML correlate closely with developmental stage, where phenotypically primitive AML is sensitive, but monocytic AML is more resistant. Mechanistically, resistant monocytic AML has a distinct transcriptomic profile, loses expression of venetoclax target BCL2, and relies on MCL1 to mediate oxidative phosphorylation and survival. This differential sensitivity drives a selective process in patients which favors the outgrowth of monocytic subpopulations at relapse. Based on these findings, we conclude that resistance to venetoclax + azacitidine can arise due to biological properties intrinsic to monocytic differentiation. We propose that optimal AML therapies should be designed so as to independently target AML subclones that may arise at differing stages of pathogenesis. SIGNIFICANCE: Identifying characteristics of patients who respond poorly to venetoclax-based therapy and devising alternative therapeutic strategies for such patients are important topics in AML. We show that venetoclax resistance can arise due to intrinsic molecular/metabolic properties of monocytic AML cells and that such properties can potentially be targeted with alternative strategies.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Sulfonamidas/uso terapéutico , Anciano , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Humanos , Sulfonamidas/farmacología
10.
Nat Cancer ; 1(12): 1176-1187, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33884374

RESUMEN

Venetoclax with azacitidine (ven/aza) has emerged as a promising regimen for acute myeloid leukemia (AML), with a high percentage of clinical remissions in newly diagnosed patients. However, approximately 30% of newly diagnosed and the majority of relapsed patients do not achieve remission with ven/aza. We previously reported that ven/aza efficacy is based on eradication of AML stem cells through a mechanism involving inhibition of amino acid metabolism, a process which is required in primitive AML cells to drive oxidative phosphorylation. Herein we demonstrate that resistance to ven/aza occurs via up-regulation of fatty acid oxidation (FAO), which occurs due to RAS pathway mutations, or as a compensatory adaptation in relapsed disease. Utilization of FAO obviates the need for amino acid metabolism, thereby rendering ven/aza ineffective. Pharmacological inhibition of FAO restores sensitivity to ven/aza in drug resistant AML cells. We propose inhibition of FAO as a therapeutic strategy to address ven/aza resistance.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Leucemia Mieloide Aguda , Aminoácidos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Compuestos Bicíclicos Heterocíclicos con Puentes , Ácidos Grasos/uso terapéutico , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Células Madre , Sulfonamidas
11.
Cell Rep ; 27(1): 238-254.e6, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30943405

RESUMEN

The NADPH-dependent oxidase NOX2 is an important effector of immune cell function, and its activity has been linked to oncogenic signaling. Here, we describe a role for NOX2 in leukemia-initiating stem cell populations (LSCs). In a murine model of leukemia, suppression of NOX2 impaired core metabolism, attenuated disease development, and depleted functionally defined LSCs. Transcriptional analysis of purified LSCs revealed that deficiency of NOX2 collapses the self-renewal program and activates inflammatory and myeloid-differentiation-associated programs. Downstream of NOX2, we identified the forkhead transcription factor FOXC1 as a mediator of the phenotype. Notably, suppression of NOX2 or FOXC1 led to marked differentiation of leukemic blasts. In xenotransplantation models of primary human myeloid leukemia, suppression of either NOX2 or FOXC1 significantly attenuated disease development. Collectively, these findings position NOX2 as a critical regulator of malignant hematopoiesis and highlight the clinical potential of inhibiting NOX2 as a means to target LSCs.


Asunto(s)
Autorrenovación de las Células , Leucemia/sangre , Leucopoyesis , Células Progenitoras Mieloides/metabolismo , NADPH Oxidasa 2/metabolismo , Animales , Células Cultivadas , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Células HEK293 , Humanos , Leucemia/genética , Leucemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/patología , NADPH Oxidasa 2/genética
13.
Cancer Cell ; 34(5): 724-740.e4, 2018 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-30423294

RESUMEN

In this study we interrogated the metabolome of human acute myeloid leukemia (AML) stem cells to elucidate properties relevant to therapeutic intervention. We demonstrate that amino acid uptake, steady-state levels, and catabolism are all elevated in the leukemia stem cell (LSC) population. Furthermore, LSCs isolated from de novo AML patients are uniquely reliant on amino acid metabolism for oxidative phosphorylation and survival. Pharmacological inhibition of amino acid metabolism reduces oxidative phosphorylation and induces cell death. In contrast, LSCs obtained from relapsed AML patients are not reliant on amino acid metabolism due to their ability to compensate through increased fatty acid metabolism. These findings indicate that clinically relevant eradication of LSCs can be achieved with drugs that target LSC metabolic vulnerabilities.


Asunto(s)
Aminoácidos/metabolismo , Ácidos Grasos/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Azacitidina/farmacología , Transporte Biológico/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular Tumoral , Femenino , Glucólisis/fisiología , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Metabolismo de los Lípidos/efectos de los fármacos , Metaboloma/fisiología , Ratones , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Sulfonamidas/farmacología
14.
Cancer Cell ; 34(4): 659-673.e6, 2018 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-30270124

RESUMEN

From an organismal perspective, cancer cell populations can be considered analogous to parasites that compete with the host for essential systemic resources such as glucose. Here, we employed leukemia models and human leukemia samples to document a form of adaptive homeostasis, where malignant cells alter systemic physiology through impairment of both host insulin sensitivity and insulin secretion to provide tumors with increased glucose. Mechanistically, tumor cells induce high-level production of IGFBP1 from adipose tissue to mediate insulin sensitivity. Further, leukemia-induced gut dysbiosis, serotonin loss, and incretin inactivation combine to suppress insulin secretion. Importantly, attenuated disease progression and prolonged survival are achieved through disruption of the leukemia-induced adaptive homeostasis. Our studies provide a paradigm for systemic management of leukemic disease.


Asunto(s)
Glucosa/metabolismo , Homeostasis/fisiología , Resistencia a la Insulina/fisiología , Leucemia/metabolismo , Animales , Dieta Alta en Grasa , Humanos , Insulina/biosíntesis , Ratones
15.
Cell Stem Cell ; 19(1): 23-37, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27374788

RESUMEN

Adipose tissue (AT) has previously been identified as an extra-medullary reservoir for normal hematopoietic stem cells (HSCs) and may promote tumor development. Here, we show that a subpopulation of leukemic stem cells (LSCs) can utilize gonadal adipose tissue (GAT) as a niche to support their metabolism and evade chemotherapy. In a mouse model of blast crisis chronic myeloid leukemia (CML), adipose-resident LSCs exhibit a pro-inflammatory phenotype and induce lipolysis in GAT. GAT lipolysis fuels fatty acid oxidation in LSCs, especially within a subpopulation expressing the fatty acid transporter CD36. CD36(+) LSCs have unique metabolic properties, are strikingly enriched in AT, and are protected from chemotherapy by the GAT microenvironment. CD36 also marks a fraction of human blast crisis CML and acute myeloid leukemia (AML) cells with similar biological properties. These findings suggest striking interplay between leukemic cells and AT to create a unique microenvironment that supports the metabolic demands and survival of a distinct LSC subpopulation.


Asunto(s)
Adaptación Fisiológica , Tejido Adiposo/patología , Antineoplásicos/farmacología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Animales , Antineoplásicos/uso terapéutico , Crisis Blástica/tratamiento farmacológico , Crisis Blástica/patología , Antígenos CD36/metabolismo , Citoprotección/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Ácidos Grasos/metabolismo , Gónadas/patología , Humanos , Inflamación/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Lipólisis/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Neoplásicas/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Carga Tumoral/efectos de los fármacos
16.
J Biol Chem ; 288(47): 33542-33558, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24089526

RESUMEN

The development of strategies to eradicate primary human acute myelogenous leukemia (AML) cells is a major challenge to the leukemia research field. In particular, primitive leukemia cells, often termed leukemia stem cells, are typically refractory to many forms of therapy. To investigate improved strategies for targeting of human AML cells we compared the molecular mechanisms regulating oxidative state in primitive (CD34(+)) leukemic versus normal specimens. Our data indicate that CD34(+) AML cells have elevated expression of multiple glutathione pathway regulatory proteins, presumably as a mechanism to compensate for increased oxidative stress in leukemic cells. Consistent with this observation, CD34(+) AML cells have lower levels of reduced glutathione and increased levels of oxidized glutathione compared with normal CD34(+) cells. These findings led us to hypothesize that AML cells will be hypersensitive to inhibition of glutathione metabolism. To test this premise, we identified compounds such as parthenolide (PTL) or piperlongumine that induce almost complete glutathione depletion and severe cell death in CD34(+) AML cells. Importantly, these compounds only induce limited and transient glutathione depletion as well as significantly less toxicity in normal CD34(+) cells. We further determined that PTL perturbs glutathione homeostasis by a multifactorial mechanism, which includes inhibiting key glutathione metabolic enzymes (GCLC and GPX1), as well as direct depletion of glutathione. These findings demonstrate that primitive leukemia cells are uniquely sensitive to agents that target aberrant glutathione metabolism, an intrinsic property of primary human AML cells.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Dioxolanos/farmacología , Glutatión/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Sesquiterpenos/farmacología , Antígenos CD34 , Femenino , Glutamato-Cisteína Ligasa/antagonistas & inhibidores , Glutamato-Cisteína Ligasa/metabolismo , Glutatión/antagonistas & inhibidores , Glutatión Peroxidasa/antagonistas & inhibidores , Glutatión Peroxidasa/metabolismo , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , Oxidación-Reducción/efectos de los fármacos , Células Tumorales Cultivadas , Glutatión Peroxidasa GPX1
17.
Chin J Cancer ; 31(11): 519-31, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23114090

RESUMEN

A large amount of nicotinamide adenine dinucleotide phosphate (NADPH) is required for fatty acid synthesis and maintenance of the redox state in cancer cells. Malic enzyme 1(ME1)-dependent NADPH production is one of the three pathways that contribute to the formation of the cytosolic NADPH pool. ME1 is generally considered to be overexpressed in cancer cells to meet the high demand for increased de novo fatty acid synthesis. In the present study, we found that glucose induced higher ME1 activity and that repressing ME1 had a profound impact on glucose metabolism of nasopharyngeal carcinoma(NPC) cells. High incorporation of glucose and an enhancement of the pentose phosphate pathway were observed in ME1-repressed cells. However, there were no obvious changes in the other two pathways for glucose metabolism: glycolysis and oxidative phosphorylation. Interestingly, NADPH was decreased under low-glucose condition in ME1-repressed cells relative to wild-type cells, whereas no significant difference was observed under high-glucose condition. ME1-repressed cells had significantly decreased tolerance to low-glucose condition. Moreover, NADPH produced by ME1 was not only important for fatty acid synthesis but also essential for maintenance of the intracellular redox state and the protection of cells from oxidative stress. Furthermore, diminished migration and invasion were observed in ME1-repressed cells due to a reduced level of Snail protein. Collectively, these results suggest an essential role for ME1 in the production of cytosolic NADPH and maintenance of migratory and invasive abilities of NPC cells.


Asunto(s)
Movimiento Celular , Glucosa/metabolismo , Malato Deshidrogenasa/metabolismo , NADP/metabolismo , Neoplasias Nasofaríngeas , Oxidación-Reducción , Carcinoma , Línea Celular Tumoral , Supervivencia Celular , Glucólisis , Humanos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Invasividad Neoplásica , Fosforilación Oxidativa , Vía de Pentosa Fosfato , Proteínas Proto-Oncogénicas c-akt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...