Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Front Oncol ; 14: 1375905, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38807762

RESUMEN

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that is widely recognized to play important, but complex, modulatory roles in a variety of tumor types. In this review, we comprehensively summarize the increasingly controversial role of AhR as a tumor regulator and the mechanisms by which it alters tumor progression based on the cancer cell type. Finally, we discuss new and emerging strategies to therapeutically modulate AhR, focusing on novel agents that hold promise in current human clinical trials as well as existing FDA-approved drugs that could potentially be repurposed for cancer therapy.

2.
Sci Rep ; 14(1): 5731, 2024 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-38459088

RESUMEN

Triple-negative breast cancer (TNBC) is one of the most aggressive types of cancer. Despite decades of intense investigation, treatment options remain limited, and rapid recurrence with distant metastases remains a significant challenge. Cancer cell-intrinsic production of cytokines such as type I interferons (IFN-I) is a known potent modulator of response to therapy in many cancers, including TNBC, and can influence therapeutic outcome. Here, we report that, in TNBC systems, the aryl hydrocarbon receptor (AhR) suppresses IFN-I expression via inhibition of STImulator of Interferon Genes (STING), a key mediator of interferon production. Intratumoral STING activity is essential in mediating the efficacy of PARP inhibitors (PARPi) which are used in the treatment of cancers harboring BRCA1 deficiency. We find that, in TNBC cells, PARPi treatment activates AhR in a BRCA1 deficiency-dependent manner, thus suggesting the presence of a negative feedback loop aimed at modulating PARPi efficacy. Importantly, our results indicate that the combined inhibition of PARP and AhR is superior in elevating IFN-I expression as compared to PARPi-alone. Thus, AhR inhibition may allow for enhanced IFN-I production upon PARPi in BRCA1-deficient breast cancers, most of which are of TNBC origin, and may represent a therapeutically viable strategy to enhance PARPi efficacy.


Asunto(s)
Interferón Tipo I , Neoplasias de la Mama Triple Negativas , Humanos , Proteína BRCA2/genética , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Receptores de Hidrocarburo de Aril/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
3.
iScience ; 26(11): 108303, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-38026169

RESUMEN

Neuroblastoma is the most common extracranial solid tumor in children. MYCN amplification is detected in almost half of high-risk cases and is associated with poorly differentiated tumors, poor patient prognosis and poor response to therapy, including retinoids. We identify the aryl hydrocarbon receptor (AhR) as a transcription factor promoting the growth and suppressing the differentiation of MYCN-amplified neuroblastoma. A neuroblastoma specific AhR transcriptional signature reveals an inverse correlation of AhR activity with patients' outcome, suggesting AhR activity is critical for disease progression. AhR modulates chromatin structures, reducing accessibility to regions responsive to retinoic acid. Genetic and pharmacological inhibition of AhR results in induction of differentiation. Importantly, AhR antagonism with clofazimine synergizes with retinoic acid in inducing differentiation both in vitro and in vivo. Thus, we propose AhR as a target for MYCN-amplified neuroblastoma and that its antagonism, combined with current standard-of-care, may result in a more durable response in patients.

4.
Trends Mol Med ; 28(9): 758-769, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35718686

RESUMEN

Genetic or pharmacological inhibition of enzymes involved in GTP biosynthesis has substantial biological effects, underlining the need to better understand the function of GTP levels in regulation of cellular processes and the significance of targeting GTP biosynthesis enzymes for therapeutic intervention. Our current understanding of spatiotemporal regulation of GTP metabolism and its role in physiological and pathological cellular processes is far from complete. Novel methodologies such as genetically encoded sensors of free GTP offered insights into intracellular distribution and function of GTP molecules. In the current Review, we provide analysis of recent discoveries in the field of GTP metabolism and evaluate the key enzymes as molecular targets.


Asunto(s)
Guanosina Trifosfato , Humanos , Fenotipo
5.
Cell Chem Biol ; 29(6): 970-984.e6, 2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35148834

RESUMEN

Signal transduction pathways post-translationally regulating nucleotide metabolism remain largely unknown. Guanosine monophosphate reductase (GMPR) is a nucleotide metabolism enzyme that decreases GTP pools by converting GMP to IMP. We observed that phosphorylation of GMPR at Tyr267 is critical for its activity and found that this phosphorylation by ephrin receptor tyrosine kinase EPHA4 decreases GTP pools in cell protrusions and levels of GTP-bound RAC1. EPHs possess oncogenic and tumor-suppressor activities, although the mechanisms underlying switches between these two modes are poorly understood. We demonstrated that GMPR plays a key role in EPHA4-mediated RAC1 suppression. This supersedes GMPR-independent activation of RAC1 by EPHA4, resulting in a negative overall effect on melanoma cell invasion and tumorigenicity. Accordingly, EPHA4 levels increase during melanoma progression and inversely correlate with GMPR levels in individual melanoma tumors. Therefore, phosphorylation of GMPR at Tyr267 is a metabolic signal transduction switch controlling GTP biosynthesis and transformed phenotypes.


Asunto(s)
Melanoma , Receptor EphA4/metabolismo , GMP-Reductasa/genética , GMP-Reductasa/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Melanoma/metabolismo , Nucleótidos/metabolismo , Fosforilación
6.
Methods Mol Biol ; 2394: 163-169, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35094327

RESUMEN

Changes in intracellular GTP levels, even incremental ones, profoundly affect the activity of several GTP-binding proteins ultimately resulting in alteration of several basal cellular phenotypes including cell motility, invasion, and tumorigenesis. However, until recently, no tools were available for GTP quantification in live cells. Therefore, in the current chapter, we describe the methodology for the quantitative assessment of spatiotemporal changes in GTP levels in the cells using genetically encoded fluorescent ratiometric GTP sensors termed GEVALs for GTP evaluators.


Asunto(s)
Colorantes Fluorescentes , Proteína de Unión al GTP rhoA , Movimiento Celular/fisiología , Colorantes Fluorescentes/química , Guanosina Trifosfato/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
8.
Nat Commun ; 12(1): 6091, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34667203

RESUMEN

Physiological changes in GTP levels in live cells have never been considered a regulatory step of RAC1 activation because intracellular GTP concentration (determined by chromatography or mass spectrometry) was shown to be substantially higher than the in vitro RAC1 GTP dissociation constant (RAC1-GTP Kd). Here, by combining genetically encoded GTP biosensors and a RAC1 activity biosensor, we demonstrated that GTP levels fluctuating around RAC1-GTP Kd correlated with changes in RAC1 activity in live cells. Furthermore, RAC1 co-localized in protrusions of invading cells with several guanylate metabolism enzymes, including rate-limiting inosine monophosphate dehydrogenase 2 (IMPDH2), which was partially due to direct RAC1-IMPDH2 interaction. Substitution of endogenous IMPDH2 with IMPDH2 mutants incapable of binding RAC1 did not affect total intracellular GTP levels but suppressed RAC1 activity. Targeting IMPDH2 away from the plasma membrane did not alter total intracellular GTP pools but decreased GTP levels in cell protrusions, RAC1 activity, and cell invasion. These data provide a mechanism of regulation of RAC1 activity by local GTP pools in live cells.


Asunto(s)
Guanosina Trifosfato/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Membrana Celular/metabolismo , Movimiento Celular , Guanosina Trifosfato/química , Células HEK293 , Humanos , IMP Deshidrogenasa/genética , IMP Deshidrogenasa/metabolismo , Cinética , Unión Proteica , Proteína de Unión al GTP rac1/química , Proteína de Unión al GTP rac1/genética
9.
Blood Adv ; 5(7): 1933-1946, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33821992

RESUMEN

Resistance to the proteasome inhibitor bortezomib (BTZ) represents a major obstacle in the treatment of multiple myeloma (MM). The contribution of lipid metabolism in the resistance of MM cells to BTZ is mostly unknown. Here we report that levels of fatty acid elongase 6 (ELOVL6) were lower in MM cells from BTZ-nonresponsive vs BTZ-responsive patients and in cultured MM cells selected for BTZ resistance compared with parental counterparts. Accordingly, depletion of ELOVL6 in parental MM cells suppressed BTZ-induced endoplasmic reticulum (ER) stress and cytotoxicity, whereas restoration of ELOVL6 levels in BTZ-resistant MM cells sensitized them to BTZ in tissue culture settings and, as xenografts, in a plasmacytoma mouse model. Furthermore, for the first time, we identified changes in the BTZ-induced lipidome between parental and BTZ-resistant MM cell lines underlying a functional difference in their response to BTZ. We demonstrated that restoration of ELOVL6 levels in BTZ-resistant MM cells resensitized them to BTZ largely via upregulation of ELOVL6-dependent ceramide species, which was a prerequisite for BTZ-induced ER stress and cell death in these cells. Our data characterize ELOVL6 as a major clinically relevant regulator of MM cell resistance to BTZ, which can emerge from the impaired ability of these cells to alter ceramide composition in response to BTZ.


Asunto(s)
Mieloma Múltiple , Animales , Bortezomib/farmacología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Elongasas de Ácidos Grasos , Humanos , Ratones , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética
10.
Nat Commun ; 11(1): 52, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31911608

RESUMEN

Prostatic luminal epithelial cells secrete high levels of acetylated polyamines into the prostatic lumen, sensitizing them to perturbations of connected metabolic pathways. Enhanced flux is driven by spermidine/spermine N1-acetyltransferase (SSAT) activity, which acetylates polyamines leading to their secretion and drives biosynthetic demand. The methionine salvage pathway recycles one-carbon units lost to polyamine biosynthesis to the methionine cycle to overcome stress. Prostate cancer (CaP) relies on methylthioadenosine phosphorylase (MTAP), the rate-limiting enzyme, to relieve strain. Here, we show that inhibition of MTAP alongside SSAT upregulation is synergistic in androgen sensitive and castration recurrent CaP models in vitro and in vivo. The combination treatment increases apoptosis in radical prostatectomy ex vivo explant samples. This unique high metabolic flux through polyamine biosynthesis and connected one carbon metabolism in CaP creates a metabolic dependency. Enhancing this flux while simultaneously targeting this dependency in prostate cancer results in an effective therapeutic approach potentially translatable to the clinic.


Asunto(s)
Metionina/metabolismo , Poliaminas/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Adenina/administración & dosificación , Adenina/análogos & derivados , Animales , Apoptosis , Línea Celular Tumoral , Quimioterapia Combinada , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Purina-Nucleósido Fosforilasa/genética , Purina-Nucleósido Fosforilasa/metabolismo , Pirrolidinas/administración & dosificación , Terapia Recuperativa , Espermina/administración & dosificación , Espermina/análogos & derivados , Espermina/metabolismo
11.
Oncogene ; 38(19): 3585-3597, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30664687

RESUMEN

Although antioxidants promote melanoma metastasis, the role of reactive oxygen species (ROS) in other stages of melanoma progression is controversial. Moreover, genes regulating ROS have not been functionally characterized throughout the entire tumor progression in mouse models of cancer. To address this question, we crossed mice-bearing knock-out of Klf9, an ubiquitous transcriptional regulator of oxidative stress, with two conditional melanocytic mouse models: BrafCA mice, where BrafV600E causes premalignant melanocytic hyperplasia, and BrafCA/Pten-/- mice, where BrafV600E and loss of Pten induce primary melanomas and metastases. Klf9 deficiency inhibited premalignant melanocytic hyperplasia in BrafCA mice but did not affect formation and growth of BrafCA/Pten-/- primary melanomas. It also, as expected, promoted BrafCA/Pten-/- metastasis. Treatment with antioxidant N-acetyl cysteine phenocopied loss of Klf9 including suppression of melanocytic hyperplasia. We were interested in a different role of Klf9 in regulation of cell proliferation in BrafCA and BrafCA/Pten-/- melanocytic cells. Mechanistically, we demonstrated that BRAFV600E signaling transcriptionally upregulated KLF9 and that KLF9-dependent ROS were required for full-scale activation of ERK1/2 and induction of cell proliferation by BRAFV600E. PTEN depletion in BRAFV600E-melanocytes did not further activate ERK1/2 and cell proliferation, but rendered these phenotypes insensitive to KLF9 and ROS. Our data identified an essential role of KLF9-dependent ROS in BRAFV600E signaling in premalignant melanocytes, offered an explanation to variable role of ROS in premalignant and transformed melanocytic cells and suggested a novel mechanism for suppression of premalignant growth by topical antioxidants.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Melanoma/patología , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Cutáneas/patología , Acetilcisteína/efectos adversos , Adulto , Anciano , Anciano de 80 o más Años , Animales , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Melanocitos/efectos de los fármacos , Melanocitos/metabolismo , Melanocitos/patología , Melanoma/genética , Melanoma/metabolismo , Melanoma Experimental/inducido químicamente , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones Noqueados , Persona de Mediana Edad , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/metabolismo
12.
Cell Rep ; 25(1): 212-223.e4, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30282030

RESUMEN

Transcription factor XBP1s, activated by endoplasmic reticulum (ER) stress in a dose-dependent manner, plays a central role in adaptive unfolded protein response (UPR) via direct activation of multiple genes controlling protein refolding. Here, we report that elevation of ER stress above a critical threshold causes accumulation of XBP1s protein sufficient for binding to the promoter and activation of a gene encoding a transcription factor KLF9. In comparison to other XBP1s targets, KLF9 promoter contains an evolutionary conserved lower-affinity binding site that requires higher amounts of XBP1s for activation. In turn, KLF9 induces expression of two regulators of ER calcium storage, TMEM38B and ITPR1, facilitating additional calcium release from ER, exacerbation of ER stress, and cell death. Accordingly, Klf9 deficiency attenuates tunicamycin-induced ER stress in mouse liver. These data reveal a role for XBP1s in cytotoxic UPR and provide insights into mechanisms of life-or-death decisions in cells under ER stress.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Respuesta de Proteína Desplegada/fisiología , Proteína 1 de Unión a la X-Box/metabolismo , Animales , Estrés del Retículo Endoplásmico , Femenino , Células HCT116 , Células HEK293 , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba , Proteína 1 de Unión a la X-Box/genética
13.
J Clin Invest ; 128(10): 4682-4696, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30198908

RESUMEN

Polyamine inhibition for cancer therapy is, conceptually, an attractive approach but has yet to meet success in the clinical setting. The aryl hydrocarbon receptor (AHR) is the central transcriptional regulator of the xenobiotic response. Our study revealed that AHR also positively regulates intracellular polyamine production via direct transcriptional activation of 2 genes, ODC1 and AZIN1, which are involved in polyamine biosynthesis and control, respectively. In patients with multiple myeloma (MM), AHR levels were inversely correlated with survival, suggesting that AHR inhibition may be beneficial for the treatment of this disease. We identified clofazimine (CLF), an FDA-approved anti-leprosy drug, as a potent AHR antagonist and a suppressor of polyamine biosynthesis. Experiments in a transgenic model of MM (Vk*Myc mice) and in immunocompromised mice bearing MM cell xenografts revealed high efficacy of CLF comparable to that of bortezomib, a first-in-class proteasome inhibitor used for the treatment of MM. This study identifies a previously unrecognized regulatory axis between AHR and polyamine metabolism and reveals CLF as an inhibitor of AHR and a potentially clinically relevant anti-MM agent.


Asunto(s)
Poliaminas Biogénicas/biosíntesis , Clofazimina/farmacología , Mieloma Múltiple , Proteínas de Neoplasias , Neoplasias Experimentales , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Células HEK293 , Humanos , Ratones , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo
14.
Cell Death Differ ; 25(6): 1040-1049, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29463842

RESUMEN

Oncogenic transcription factor FOXQ1 has been implicated in promotion of multiple transformed phenotypes in carcinoma cells. Recently, we have characterized FOXQ1 as a melanoma tumor suppressor that acts via repression of N-cadherin gene, and invasion and metastasis. Here we report that FOXQ1 induces differentiation in normal and transformed melanocytic cells at least partially via direct transcriptional activation of MITF gene, melanocytic lineage-specific regulator of differentiation. Importantly, we demonstrate that pigmentation induced in cultured melanocytic cells and in mice by activation of cAMP/CREB1 pathway depends in large part on FOXQ1. Moreover, our data reveal that FOXQ1 acts as a critical mediator of BRAFV600E-dependent regulation of MITF levels, thus providing a novel link between two major signal transduction pathways controlling MITF and differentiation in melanocytic cells.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Melanocitos/metabolismo , Melanoma/metabolismo , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Animales , Línea Celular Tumoral , Factores de Transcripción Forkhead/genética , Melanocitos/patología , Melanoma/genética , Melanoma/patología , Ratones , Ratones Noqueados , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología
15.
Nat Methods ; 14(10): 1003-1009, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28869758

RESUMEN

GTP is a major regulator of multiple cellular processes, but tools for quantitative evaluation of GTP levels in live cells have not been available. We report the development and characterization of genetically encoded GTP sensors, which we constructed by inserting a circularly permuted yellow fluorescent protein (cpYFP) into a region of the bacterial G protein FeoB that undergoes a GTP-driven conformational change. GTP binding to these sensors results in a ratiometric change in their fluorescence, thereby providing an internally normalized response to changes in GTP levels while minimally perturbing those levels. Mutations introduced into FeoB to alter its affinity for GTP created a series of sensors with a wide dynamic range. Critically, in mammalian cells the sensors showed consistent changes in ratiometric signal upon depletion or restoration of GTP pools. We show that these GTP evaluators (GEVALs) are suitable for detection of spatiotemporal changes in GTP levels in living cells and for high-throughput screening of molecules that modulate GTP levels.


Asunto(s)
Proteínas Bacterianas/metabolismo , Técnicas Biosensibles , Guanosina Trifosfato/metabolismo , Proteínas Luminiscentes/metabolismo , Animales , Proteínas Bacterianas/genética , Línea Celular Tumoral , Guanosina Trifosfato/genética , Humanos , Concentración de Iones de Hidrógeno , Proteínas Luminiscentes/genética , Mutación
16.
Cell Rep ; 20(12): 2820-2832, 2017 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-28930679

RESUMEN

Lineage-specific regulation of tumor progression by the same transcription factor is understudied. We find that levels of the FOXQ1 transcription factor, an oncogene in carcinomas, are decreased during melanoma progression. Moreover, in contrast to carcinomas, FOXQ1 suppresses epithelial-to-mesenchymal transition, invasion, and metastasis in melanoma cells. We find that these lineage-specific functions of FOXQ1 largely depend on its ability to activate (in carcinomas) or repress (in melanoma) transcription of the N-cadherin gene (CDH2). We demonstrate that FOXQ1 interacts with nuclear ß-catenin and TLE proteins, and the ß-catenin/TLE ratio, which is higher in carcinoma than melanoma cells, determines the effect of FOXQ1 on CDH2 transcription. Accordingly, other FOXQ1-dependent phenotypes can be manipulated by altering nuclear ß-catenin or TLE proteins levels. Our data identify FOXQ1 as a melanoma suppressor and establish a mechanism underlying its inverse lineage-specific transcriptional regulation of transformed phenotypes.


Asunto(s)
Factores de Transcripción Forkhead/genética , Melanoma/genética , Melanoma/patología , Oncogenes , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Transformación Celular Neoplásica/patología , Progresión de la Enfermedad , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Ratones SCID , Factor de Transcripción Asociado a Microftalmía/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Fenotipo , beta Catenina/metabolismo
17.
Methods Mol Biol ; 1534: 1-15, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27812863

RESUMEN

Activation of oncogenic signaling paradoxically results in the permanent withdrawal from cell cycle and induction of senescence (oncogene-induced senescence (OIS)). OIS is a fail-safe mechanism used by the cells to prevent uncontrolled tumor growth, and, as such, it is considered as the first barrier against cancer. In order to progress, tumor cells thus need to first overcome the senescent phenotype. Despite the increasing attention gained by OIS in the past 20 years, this field is still rather young due to continuous emergence of novel pathways and processes involved in OIS. Among the many factors contributing to incomplete understanding of OIS are the lack of unequivocal markers for senescence and the complexity of the phenotypes revealed by senescent cells in vivo and in vitro. OIS has been shown to play major roles at both the cellular and organismal levels in biological processes ranging from embryonic development to barrier to cancer progression. Here we will briefly outline major advances in methodologies that are being utilized for induction, identification, and characterization of molecular processes in cells undergoing oncogene-induced senescence. The full description of such methodologies is provided in the corresponding chapters of the book.


Asunto(s)
Senescencia Celular/fisiología , Animales , Biomarcadores , Susceptibilidad a Enfermedades , Metabolismo Energético , Regulación de la Expresión Génica , Humanos , Oncogenes , Transducción de Señal , beta-Galactosidasa/metabolismo
18.
Mol Cell Oncol ; 3(1)2016 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-26989767

RESUMEN

Enzymes involved in de novo production of guanosine triphosphate (GTP) have been recently revealed as integral components of melanoma progression through modulation of the activity of small GTPases. Here, we discuss the biology and therapeutic implications of these findings.

19.
Mol Cell ; 53(6): 916-928, 2014 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-24613345

RESUMEN

Reactive oxygen species (ROS) activate NF-E2-related transcription factor 2 (Nrf2), a key transcriptional regulator driving antioxidant gene expression and protection from oxidant injury. Here, we report that in response to elevation of intracellular ROS above a critical threshold, Nrf2 stimulates expression of transcription Kruppel-like factor 9 (Klf9), resulting in further Klf9-dependent increases in ROS and subsequent cell death. We demonstrated that Klf9 independently causes increased ROS levels in various types of cultured cells and in mouse tissues and is required for pathogenesis of bleomycin-induced pulmonary fibrosis in mice. Mechanistically, Klf9 binds to the promoters and alters the expression of several genes involved in the metabolism of ROS, including suppression of thioredoxin reductase 2, an enzyme participating in ROS clearance. Our data reveal an Nrf2-dependent feedforward regulation of ROS and identify Klf9 as a ubiquitous regulator of oxidative stress and lung injury.


Asunto(s)
Regulación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/genética , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo , Fibrosis Pulmonar/genética , Animales , Sitios de Unión , Bleomicina , Línea Celular Tumoral , Genes Reporteros , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Pulmón/metabolismo , Pulmón/patología , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Células 3T3 NIH , Regiones Promotoras Genéticas , Unión Proteica , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Especies Reactivas de Oxígeno , Transducción de Señal
20.
Cell Rep ; 5(2): 493-507, 2013 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-24139804

RESUMEN

Melanoma is one of the most aggressive types of human cancers, and the mechanisms underlying melanoma invasive phenotype are not completely understood. Here, we report that expression of guanosine monophosphate reductase (GMPR), an enzyme involved in de novo biosynthesis of purine nucleotides, was downregulated in the invasive stages of human melanoma. Loss- and gain-of-function experiments revealed that GMPR downregulates the amounts of several GTP-bound (active) Rho-GTPases and suppresses the ability of melanoma cells to form invadopodia, degrade extracellular matrix, invade in vitro, and grow as tumor xenografts in vivo. Mechanistically, we demonstrated that GMPR partially depletes intracellular GTP pools. Pharmacological inhibition of de novo GTP biosynthesis suppressed whereas addition of exogenous guanosine increased invasion of melanoma cells as well as cells from other cancer types. Our data identify GMPR as a melanoma invasion suppressor and establish a link between guanosine metabolism and Rho-GTPase-dependent melanoma cell invasion.


Asunto(s)
GMP-Reductasa/metabolismo , Melanoma/enzimología , Nucleósidos de Purina/biosíntesis , Animales , Línea Celular Tumoral , Movimiento Celular , Matriz Extracelular/metabolismo , GMP-Reductasa/antagonistas & inhibidores , GMP-Reductasa/genética , Guanosina Trifosfato/metabolismo , Células HCT116 , Humanos , IMP Deshidrogenasa/metabolismo , Melanoma/metabolismo , Melanoma/patología , Ratones , Fenotipo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Trasplante Heterólogo , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA