Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38815134

RESUMEN

The non-physiological nutrient levels found in traditional culture media have been shown to affect numerous aspects of cancer cell physiology, including how cells respond to certain therapeutic agents. Here, we comprehensively evaluated how physiological nutrient levels impact therapeutic response by performing drug screening in human plasma-like medium (HPLM). We observed dramatic nutrient-dependent changes in sensitivity to a variety of FDA-approved and clinically trialed compounds including rigosertib, an experimental cancer therapeutic that has recently failed in phase 3 clinical trials. Mechanistically, we found that the ability of rigosertib to destabilize microtubules is strongly inhibited by the purine metabolism end product uric acid, which is uniquely abundant in humans relative to traditional in vitro and in vivo cancer models. These results demonstrate the broad and dramatic effects nutrient levels can have on drug response, and how incorporation of human-specific physiological nutrient media might help to identify compounds whose efficacy could be impacted in humans.

2.
bioRxiv ; 2023 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-37873453

RESUMEN

The non-essential amino acid serine is a critical nutrient for cancer cells due to its diverse biosynthetic functions. While some tumors can synthesize serine de novo, others are auxotrophic for serine and therefore reliant on the uptake of exogenous serine. Importantly, however, the transporter(s) that mediate serine uptake in cancer cells are not known. Here, we characterize the amino acid transporter ASCT2 (coded for by the gene SLC1A5) as the primary serine transporter in cancer cells. ASCT2 is well-known as a glutamine transporter in cancer, and our work demonstrates that serine and glutamine compete for uptake through ASCT2. We further show that ASCT2-mediated serine uptake is essential for purine nucleotide biosynthesis and that ERα promotes serine uptake by directly activating SLC1A5 transcription. Together, our work defines an additional important role for ASCT2 as a serine transporter in cancer and evaluates ASCT2 as a potential therapeutic target in serine metabolism.

3.
bioRxiv ; 2023 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-37546939

RESUMEN

The non-physiological nutrient levels found in traditional culture media have been shown to affect numerous aspects of cancer cell physiology, including how cells respond to certain therapeutic agents. Here, we comprehensively evaluated how physiological nutrient levels impact therapeutic response by performing drug screening in human plasma-like medium (HPLM). We observed dramatic nutrient-dependent changes in sensitivity to a variety of FDA-approved and clinically trialed compounds, including rigosertib, an experimental cancer therapeutic that has recently failed in phase 3 clinical trials. Mechanistically, we found that the ability of rigosertib to destabilize microtubules is strongly inhibited by the purine metabolism waste product uric acid, which is uniquely abundant in humans relative to traditional in vitro and in vivo cancer models. Structural modelling studies suggest that uric acid interacts with the tubulin-rigosertib complex and may act as an uncompetitive inhibitor of rigosertib. These results offer a possible explanation for the failure of rigosertib in clinical trials and demonstrate the utility of physiological media to achieve in vitro results that better represent human therapeutic responses.

5.
Oncogene ; 41(8): 1190-1202, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35067686

RESUMEN

New strategies are needed to predict and overcome metastatic progression and therapy resistance in prostate cancer. One potential clinical target is the stem cell transcription factor SOX2, which has a critical role in prostate development and cancer. We thus investigated the impact of SOX2 expression on patient outcomes and its function within prostate cancer cells. Analyses of SOX2 expression among a case-control cohort of 1028 annotated tumor specimens demonstrated that SOX2 expression confers a more rapid time to metastasis and decreased patient survival after biochemical recurrence. SOX2 ChIP-Seq analyses revealed SOX2-binding sites within prostate cancer cells which differ significantly from canonical embryonic SOX2 gene targets, and prostate-specific SOX2 gene targets are associated with multiple oncogenic pathways. Interestingly, phenotypic and gene expression analyses after CRISPR-mediated deletion of SOX2 in castration-resistant prostate cancer cells, as well as ectopic SOX2 expression in androgen-sensitive prostate cancer cells, demonstrated that SOX2 promotes changes in multiple metabolic pathways and metabolites. SOX2 expression in prostate cancer cell lines confers increased glycolysis and glycolytic capacity, as well as increased basal and maximal oxidative respiration and increased spare respiratory capacity. Further, SOX2 expression was associated with increased quantities of mitochondria, and metabolomic analyses revealed SOX2-associated changes in the metabolism of purines, pyrimidines, amino acids and sugars, and the pentose phosphate pathway. Analyses of SOX2 gene targets with central functions metabolism (CERK, ECHS1, HS6SDT1, LPCAT4, PFKP, SLC16A3, SLC46A1, and TST) document significant expression correlation with SOX2 among RNA-Seq datasets derived from patient tumors and metastases. These data support a key role for SOX2 in metabolic reprogramming of prostate cancer cells and reveal new mechanisms to understand how SOX2 enables metastatic progression, lineage plasticity, and therapy resistance. Further, our data suggest clinical opportunities to exploit SOX2 as a biomarker for staging and imaging, as well as a potential pharmacologic target.


Asunto(s)
Factores de Transcripción SOXB1
6.
Cell Rep ; 38(3): 110278, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35045283

RESUMEN

A major challenge of targeting metabolism for cancer therapy is pathway redundancy, in which multiple sources of critical nutrients can limit the effectiveness of some metabolism-targeted therapies. Here, we analyze lineage-dependent gene expression in human breast tumors to identify differences in metabolic gene expression that may limit pathway redundancy and create therapeutic vulnerabilities. We find that the serine synthesis pathway gene PSAT1 is the most depleted metabolic gene in luminal breast tumors relative to basal tumors. Low PSAT1 prevents de novo serine biosynthesis and sensitizes luminal breast cancer cells to serine and glycine starvation in vitro and in vivo. This PSAT1 expression disparity preexists in the putative cells of origin of basal and luminal tumors and is due to luminal-specific hypermethylation of the PSAT1 gene. Our data demonstrate that luminal breast tumors are auxotrophic for serine and may be uniquely sensitive to therapies targeting serine availability.


Asunto(s)
Neoplasias de la Mama/metabolismo , Serina/metabolismo , Transaminasas/metabolismo , Neoplasias de la Mama/patología , Femenino , Humanos
7.
FEBS J ; 288(19): 5629-5649, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33811729

RESUMEN

Many metabolic phenotypes in cancer cells are also characteristic of proliferating nontransformed mammalian cells, and attempts to distinguish between phenotypes resulting from oncogenic perturbation from those associated with increased proliferation are limited. Here, we examined the extent to which metabolic changes corresponding to oncogenic KRAS expression differed from those corresponding to epidermal growth factor (EGF)-driven proliferation in human mammary epithelial cells (HMECs). Removal of EGF from culture medium reduced growth rates and glucose/glutamine consumption in control HMECs despite limited changes in respiration and fatty acid synthesis, while the relative contribution of branched-chain amino acids to the TCA cycle and lipogenesis increased in the near-quiescent conditions. Most metabolic phenotypes measured in HMECs expressing mutant KRAS were similar to those observed in EGF-stimulated control HMECs that were growing at comparable rates. However, glucose and glutamine consumption as well as lactate and glutamate production were lower in KRAS-expressing cells cultured in media without added EGF, and these changes correlated with reduced sensitivity to GLUT1 inhibitor and phenformin treatment. Our results demonstrate the strong dependence of metabolic behavior on growth rate and provide a model to distinguish the metabolic influences of oncogenic mutations and nononcogenic growth.


Asunto(s)
Neoplasias de la Mama/genética , Carcinogénesis/genética , Factor de Crecimiento Epidérmico/genética , Transportador de Glucosa de Tipo 1/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Mama/crecimiento & desarrollo , Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/antagonistas & inhibidores , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Humanos , Ácido Láctico/metabolismo , Glándulas Mamarias Humanas/crecimiento & desarrollo , Glándulas Mamarias Humanas/patología , Células Tumorales Cultivadas
8.
Cancer Metab ; 8: 6, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32549981

RESUMEN

BACKGROUND: d-3-phosphoglycerate dehydrogenase (PHGDH), which encodes the first enzyme in serine biosynthesis, is overexpressed in human cancers and has been proposed as a drug target. However, whether PHGDH is critical for the proliferation or homeostasis of tissues following the postnatal period is unknown. METHODS: To study PHGDH inhibition in adult animals, we developed a knock-in mouse model harboring a PHGDH shRNA under the control of a doxycycline-inducible promoter. With this model, PHGDH depletion can be globally induced in adult animals, while sparing the brain due to poor doxycycline delivery. RESULTS: We found that PHGDH depletion is well tolerated, and no overt phenotypes were observed in multiple highly proliferative cell compartments. Further, despite detectable knockdown and impaired serine synthesis, liver and pancreatic functions were normal. Interestingly, diminished PHGDH expression reduced liver serine and ceramide levels without increasing the levels of deoxysphingolipids. Further, liver triacylglycerol profiles were altered, with an accumulation of longer chain, polyunsaturated tails upon PHGDH knockdown. CONCLUSIONS: These results suggest that dietary serine is adequate to support the function of healthy, adult murine tissues, but PHGDH-derived serine supports liver ceramide synthesis and sustains general lipid homeostasis.

9.
Cancers (Basel) ; 11(5)2019 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-31096630

RESUMEN

Far beyond simply being 11 of the 20 amino acids needed for protein synthesis, non-essential amino acids play numerous important roles in tumor metabolism. These diverse functions include providing precursors for the biosynthesis of macromolecules, controlling redox status and antioxidant systems, and serving as substrates for post-translational and epigenetic modifications. This functional diversity has sparked great interest in targeting non-essential amino acid metabolism for cancer therapy and has motivated the development of several therapies that are either already used in the clinic or are currently in clinical trials. In this review, we will discuss the important roles that each of the 11 non-essential amino acids play in cancer, how their metabolic pathways are linked, and how researchers are working to overcome the unique challenges of targeting non-essential amino acid metabolism for cancer therapy.

10.
Cell Metab ; 29(5): 1166-1181.e6, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30799286

RESUMEN

Cells are subjected to oxidative stress during the initiation and progression of tumors, and this imposes selective pressure for cancer cells to adapt mechanisms to tolerate these conditions. Here, we examined the dependency of cancer cells on glutathione (GSH), the most abundant cellular antioxidant. While cancer cell lines displayed a broad range of sensitivities to inhibition of GSH synthesis, the majority were resistant to GSH depletion. To identify cellular pathways required for this resistance, we carried out genetic and pharmacologic screens. Both approaches revealed that inhibition of deubiquitinating enzymes (DUBs) sensitizes cancer cells to GSH depletion. Inhibition of GSH synthesis, in combination with DUB inhibition, led to an accumulation of polyubiquitinated proteins, induction of proteotoxic stress, and cell death. These results indicate that depletion of GSH renders cancer cells dependent on DUB activity to maintain protein homeostasis and cell viability and reveal a potentially exploitable vulnerability for cancer therapy.


Asunto(s)
Antioxidantes/metabolismo , Supervivencia Celular/efectos de los fármacos , Enzimas Desubicuitinizantes/metabolismo , Glutatión/metabolismo , Proteostasis/efectos de los fármacos , Células A549 , Aminopiridinas/farmacología , Animales , Butionina Sulfoximina/farmacología , Dominio Catalítico/efectos de los fármacos , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Femenino , Glutamato-Cisteína Ligasa/antagonistas & inhibidores , Glutamato-Cisteína Ligasa/química , Glutamato-Cisteína Ligasa/metabolismo , Humanos , Células MCF-7 , Glándulas Mamarias Animales/citología , Glándulas Mamarias Humanas/citología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Organoides/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Tiocianatos/farmacología , Carga Tumoral/efectos de los fármacos , Proteínas Ubiquitinadas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Mol Cell ; 69(1): 1-2, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29304329

RESUMEN

Tumor cell metabolism can be altered to support specific pathological functions or to adapt to environmental stresses. In this issue of Molecular Cell, Jin et al. (2018) identify induction of glutamate dehydrogenase as a critical metabolic adaptation in matrix-detached cancer cells that is required for metastasis of LKB1-deficient lung tumors.


Asunto(s)
Anoicis , Glutamato Deshidrogenasa , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina , Proteínas de Unión al ADN , Humanos , Neoplasias Pulmonares , Proteínas Serina-Treonina Quinasas , Transducción de Señal
12.
Elife ; 62017 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-29239720

RESUMEN

Cells use multiple feedback controls to regulate metabolism in response to nutrient and signaling inputs. However, feedback creates the potential for unstable network responses. We examined how concentrations of key metabolites and signaling pathways interact to maintain homeostasis in proliferating human cells, using fluorescent reporters for AMPK activity, Akt activity, and cytosolic NADH/NAD+ redox. Across various conditions, including glycolytic or mitochondrial inhibition or cell proliferation, we observed distinct patterns of AMPK activity, including both stable adaptation and highly dynamic behaviors such as periodic oscillations and irregular fluctuations that indicate a failure to reach a steady state. Fluctuations in AMPK activity, Akt activity, and cytosolic NADH/NAD+ redox state were temporally linked in individual cells adapting to metabolic perturbations. By monitoring single-cell dynamics in each of these contexts, we identified PI3K/Akt regulation of glycolysis as a multifaceted modulator of single-cell metabolic dynamics that is required to maintain metabolic stability in proliferating cells.


Asunto(s)
Metabolismo Energético , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Glucólisis , Fosfatidilinositol 3-Quinasas/análisis , Proteínas Proto-Oncogénicas c-akt/análisis , Proteínas Quinasas Activadas por AMP/análisis , Línea Celular , Proliferación Celular , Humanos , NAD/análisis
13.
Proc Natl Acad Sci U S A ; 114(52): E11276-E11284, 2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29229826

RESUMEN

Large, multidimensional cancer datasets provide a resource that can be mined to identify candidate therapeutic targets for specific subgroups of tumors. Here, we analyzed human breast cancer data to identify transcriptional programs associated with tumors bearing specific genetic driver alterations. Using an unbiased approach, we identified thousands of genes whose expression was enriched in tumors with specific genetic alterations. However, expression of the vast majority of these genes was not enriched if associations were analyzed within individual breast tumor molecular subtypes, across multiple tumor types, or after gene expression was normalized to account for differences in proliferation or tumor lineage. Together with linear modeling results, these findings suggest that most transcriptional programs associated with specific genetic alterations in oncogenes and tumor suppressors are highly context-dependent and are predominantly linked to differences in proliferation programs between distinct breast cancer subtypes. We demonstrate that such proliferation-dependent gene expression dominates tumor transcriptional programs relative to matched normal tissues. However, we also identified a relatively small group of cancer-associated genes that are both proliferation- and lineage-independent. A subset of these genes are attractive candidate targets for combination therapy because they are essential in breast cancer cell lines, druggable, enriched in stem-like breast cancer cells, and resistant to chemotherapy-induced down-regulation.


Asunto(s)
Neoplasias de la Mama , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias , Modelos Biológicos , Células Madre Neoplásicas/metabolismo , Transcripción Genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Femenino , Humanos
14.
Nat Cell Biol ; 19(5): 414-415, 2017 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-28446818

RESUMEN

Dysfunctional cells are eliminated from epithelial monolayers by a process known as cell extrusion to maintain tissue homeostasis. Normal epithelial cells are now shown to induce the extrusion of oncogene-transformed cells by inducing metabolic changes in the oncogene-expressing cells through PDK4-mediated inhibition of PDH and mitochondrial metabolism.


Asunto(s)
Carcinogénesis , Oncogenes , Línea Celular , Células Epiteliales , Homeostasis , Humanos
15.
Nat Commun ; 8: 13989, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-28071763

RESUMEN

Extracellular matrix adhesion is required for normal epithelial cell survival, nutrient uptake and metabolism. This requirement can be overcome by oncogene activation. Interestingly, inhibition of PI3K/mTOR leads to apoptosis of matrix-detached, but not matrix-attached cancer cells, suggesting that matrix-attached cells use alternate mechanisms to maintain nutrient supplies. Here we demonstrate that under conditions of dietary restriction or growth factor starvation, where PI3K/mTOR signalling is decreased, matrix-attached human mammary epithelial cells upregulate and internalize ß4-integrin along with its matrix substrate, laminin. Endocytosed laminin localizes to lysosomes, results in increased intracellular levels of essential amino acids and enhanced mTORC1 signalling, preventing cell death. Moreover, we show that starved human fibroblasts secrete matrix proteins that maintain the growth of starved mammary epithelial cells contingent upon epithelial cell ß4-integrin expression. Our study identifies a crosstalk between stromal fibroblasts and epithelial cells under starvation that could be exploited therapeutically to target tumours resistant to PI3K/mTOR inhibition.


Asunto(s)
Células Epiteliales/fisiología , Matriz Extracelular/metabolismo , Integrina beta4/metabolismo , Laminina/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Línea Celular , Supervivencia Celular/fisiología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Femenino , Fibroblastos/metabolismo , Humanos , Integrina beta4/genética , Laminina/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Endogámicos , Fosfatidilinositol 3-Quinasas/metabolismo , Inanición
16.
Cancer Res ; 76(24): 7168-7180, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27913436

RESUMEN

Aberrant activation of the PI3K/mTOR pathway is a common feature of many cancers and an attractive target for therapy, but resistance inevitably evolves as is the case for any cancer cell-targeted therapy. In animal tumor models, chronic inhibition of PI3K/mTOR initially inhibits tumor growth, but over time, tumor cells escape inhibition. In this study, we identified a context-dependent mechanism of escape whereby tumor cells upregulated the proto-oncogene transcriptional regulators c-MYC and YAP1. This mechanism was dependent on both constitutive ERK activity as well as inhibition of the stress kinase p38. Inhibition of p38 relieved proliferation arrest and allowed upregulation of MYC and YAP through stabilization of CREB. These data provide new insights into cellular signaling mechanisms that influence resistance to PI3K/mTOR inhibitors. Furthermore, they suggest that therapies that inactivate YAP or MYC or augment p38 activity could enhance the efficacy of PI3K/mTOR inhibitors. Cancer Res; 76(24); 7168-80. ©2016 AACR.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Resistencia a Antineoplásicos/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Neoplasias Experimentales/patología , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Técnica del Anticuerpo Fluorescente , Xenoinjertos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos NOD , Microscopía Confocal , Neoplasias Experimentales/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Proto-Oncogenes Mas , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Factores de Transcripción , Proteínas Señalizadoras YAP
17.
Clin Cancer Res ; 22(24): 6039-6050, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27330058

RESUMEN

PURPOSE: To provide further insight into the role of proliferation and other cellular processes in chemosensitivity and resistance, we evaluated the association of a diverse set of gene expression signatures with response to neoadjuvant chemotherapy (NAC) in breast cancer. EXPERIMENTAL DESIGN: Expression data from primary breast cancer biopsies for 1,419 patients in 17 studies prior to NAC were identified and aggregated using common normalization procedures. Clinicopathologic characteristics, including response to NAC, were collected. Scores for 125 previously published breast cancer-related gene expression signatures were calculated for each tumor. RESULTS: Within each receptor-based subgroup or PAM50 subtype, breast tumors with high proliferation signature scores were significantly more likely to achieve pathologic complete response to NAC. To distinguish "proliferation-associated" from "proliferation-independent" signatures, we used correlation and linear modeling approaches. Most signatures associated with response to NAC were proliferation associated: 90.5% (38/42) in ER+/HER2- and 63.3% (38/60) in triple-negative breast cancer (TNBC). Proliferation-independent signatures predictive of response to NAC in ER+/HER2- breast cancer were related to immune activity, while those in TNBC comprised a diverse set of signatures, including immune, DNA damage, signaling pathways (PI3K, AKT, Ras, and EGFR), and "stemness" phenotypes. CONCLUSIONS: Proliferation differences account for the vast majority of predictive capacity of gene expression signatures in neoadjuvant chemosensitivity for ER+/HER2- breast cancers and, to a lesser extent, TNBCs. Immune activation signatures are proliferation-independent predictors of pathologic complete response in ER+/HER2- breast cancers. In TNBCs, significant proliferation-independent signatures include gene sets that represent a diverse set of cellular processes. Clin Cancer Res; 22(24); 6039-50. ©2016 AACR.


Asunto(s)
Antineoplásicos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Mama/efectos de los fármacos , Mama/patología , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Persona de Mediana Edad , Terapia Neoadyuvante/métodos , Estadificación de Neoplasias/métodos , Fosfatidilinositol 3-Quinasas/genética , Pronóstico , Receptor ErbB-2/genética , Receptores de Estrógenos/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
18.
Cell Res ; 26(7): 757-8, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27216116

RESUMEN

Detachment from extracellular matrix causes metabolic defects that transformed cells must overcome in order to survive and proliferate outside of their normal niche. A recent report from Jiang et al. published in Nature describes how cancer cells grown in suspension utilize reductive carboxylation of glutamine to transfer reducing power from the cytosol to mitochondria to detoxify reactive oxygen species and promote anchorage-independent growth and survival.


Asunto(s)
Mitocondrias , Estrés Fisiológico , Adaptación Psicológica , Homeostasis , Oxidación-Reducción , Especies Reactivas de Oxígeno
19.
Cell Metab ; 23(5): 867-80, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27133130

RESUMEN

Mammary epithelial cells transition between periods of proliferation and quiescence during development, menstrual cycles, and pregnancy, and as a result of oncogenic transformation. Utilizing an organotypic 3D tissue culture model coupled with quantitative metabolomics and proteomics, we identified significant differences in glutamate utilization between proliferating and quiescent cells. Relative to quiescent cells, proliferating cells catabolized more glutamate via transaminases to couple non-essential amino acid (NEAA) synthesis to α-ketoglutarate generation and tricarboxylic acid (TCA) cycle anaplerosis. As cells transitioned to quiescence, glutamine consumption and transaminase expression were reduced, while glutamate dehydrogenase (GLUD) was induced, leading to decreased NEAA synthesis. Highly proliferative human tumors display high transaminase and low GLUD expression, suggesting that proliferating cancer cells couple glutamine consumption to NEAA synthesis to promote biosynthesis. These findings describe a competitive and partially redundant relationship between transaminases and GLUD, and they reveal how coupling of glutamate-derived carbon and nitrogen metabolism can be regulated to support cell proliferation.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/metabolismo , Ácido Glutámico/metabolismo , Glándulas Mamarias Humanas/citología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Proliferación Celular , Células Cultivadas , Femenino , Glutamato Deshidrogenasa/metabolismo , Humanos , Metabolómica , Modelos Biológicos , Isótopos de Nitrógeno , Fosfatidilinositol 3-Quinasas/metabolismo , Proteómica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Transaminasas/metabolismo
20.
Genes Dev ; 30(7): 870, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27036968

RESUMEN

In the above-mentioned article, it has come to the authors' attention that, during the preparation of Figure 5C and Supplemental Figure S2C for the final version of this article, the authors unintentionally assembled incorrect tubulin immunoblots due to similarities in the markings or names, such as FLT3 versus FT, between two similar experiments. The amended versions of these figures are shown below. Neither the quantitative determinations nor the conclusions of this article are altered. The authors apologize for these errors.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...