65 results on '"Alejo Efeyan"'
Search Results
2. Hepatic nutrient and hormone signaling to mTORC1 instructs the postnatal metabolic zonation of the liver
- Author
-
Ana Belén Plata-Gómez, Lucía de Prado-Rivas, Alba Sanz, Nerea Deleyto-Seldas, Fernando García, Celia de la Calle Arregui, Camila Silva, Eduardo Caleiras, Osvaldo Graña-Castro, Elena Piñeiro-Yáñez, Joseph Krebs, Luis Leiva-Vega, Javier Muñoz, Ajay Jain, Guadalupe Sabio, and Alejo Efeyan
- Subjects
Science - Abstract
Abstract The metabolic functions of the liver are spatially organized in a phenomenon called zonation, linked to the differential exposure of portal and central hepatocytes to nutrient-rich blood. The mTORC1 signaling pathway controls cellular metabolism in response to nutrients and insulin fluctuations. Here we show that simultaneous genetic activation of nutrient and hormone signaling to mTORC1 in hepatocytes results in impaired establishment of postnatal metabolic and zonal identity of hepatocytes. Mutant hepatocytes fail to upregulate postnatally the expression of Frizzled receptors 1 and 8, and show reduced Wnt/β-catenin activation. This defect, alongside diminished paracrine Wnt2 ligand expression by endothelial cells, underlies impaired postnatal maturation. Impaired zonation is recapitulated in a model of constant supply of nutrients by parenteral nutrition to piglets. Our work shows the role of hepatocyte sensing of fluctuations in nutrients and hormones for triggering a latent metabolic zonation program.
- Published
- 2024
- Full Text
- View/download PDF
3. NGFR regulates stromal cell activation in germinal centers
- Author
-
Alberto Hernández-Barranco, Vanesa Santos, Marina S. Mazariegos, Eduardo Caleiras, Laura Nogués, Frédéric Mourcin, Simon Léonard, Christelle Oblet, Steve Genebrier, Delphine Rossille, Alberto Benguría, Alba Sanz, Enrique Vázquez, Ana Dopazo, Alejo Efeyan, Ana Ortega-Molina, Michel Cogne, Karin Tarte, and Héctor Peinado
- Subjects
CP: Immunology ,Biology (General) ,QH301-705.5 - Abstract
Summary: Nerve growth factor receptor (NGFR) is expressed by follicular dendritic cells (FDCs). However, the role of NGFR in the humoral response is not well defined. Here, we study the effect of Ngfr loss on lymph node organization and function, demonstrating that Ngfr depletion leads to spontaneous germinal center (GC) formation and an expansion of the GC B cell compartment. In accordance with this effect, stromal cells are altered in Ngfr−/− mice with a higher frequency of FDCs, characterized by CD21/35, MAdCAM-1, and VCAM-1 overexpression. GCs are located ectopically in Ngfr−/− mice, with lost polarization together with impaired high-affinity antibody production and an increase in circulating autoantibodies. We observe higher levels of autoantibodies in Bcl2 Tg/Ngfr−/− mice, concomitant with a higher incidence of autoimmunity and lower overall survival. Our work shows that NGFR is involved in maintaining GC structure and function, participating in GC activation, antibody production, and immune tolerance.
- Published
- 2024
- Full Text
- View/download PDF
4. Folliculin-interacting protein FNIP2 impacts on overweight and obesity through a polymorphism in a conserved 3′ untranslated region
- Author
-
Lara P. Fernández, Nerea Deleyto-Seldas, Gonzalo Colmenarejo, Alba Sanz, Sonia Wagner, Ana Belén Plata-Gómez, Mónica Gómez-Patiño, Susana Molina, Isabel Espinosa-Salinas, Elena Aguilar-Aguilar, Sagrario Ortega, Osvaldo Graña-Castro, Viviana Loria-Kohen, Pablo J. Fernández-Marcos, Alejo Efeyan, and Ana Ramírez de Molina
- Subjects
Obesity ,Overweight ,Metabolism ,mTOR ,FNIP2 ,Folliculin complex ,Biology (General) ,QH301-705.5 ,Genetics ,QH426-470 - Abstract
Abstract Background Overweight and obesity are defined by an anomalous or excessive fat accumulation that may compromise health. To find single-nucleotide polymorphisms (SNPs) influencing metabolic phenotypes associated with the obesity state, we analyze multiple anthropometric and clinical parameters in a cohort of 790 healthy volunteers and study potential associations with 48 manually curated SNPs, in metabolic genes functionally associated with the mechanistic target of rapamycin (mTOR) pathway. Results We identify and validate rs2291007 within a conserved region in the 3′UTR of folliculin-interacting protein FNIP2 that correlates with multiple leanness parameters. The T-to-C variant represents the major allele in Europeans and disrupts an ancestral target sequence of the miRNA miR-181b-5p, thus resulting in increased FNIP2 mRNA levels in cancer cell lines and in peripheral blood from carriers of the C allele. Because the miRNA binding site is conserved across vertebrates, we engineered the T-to-C substitution in the endogenous Fnip2 allele in mice. Primary cells derived from Fnip2 C/C mice show increased mRNA stability, and more importantly, Fnip2 C/C mice replicate the decreased adiposity and increased leanness observed in human volunteers. Finally, expression levels of FNIP2 in both human samples and mice negatively associate with leanness parameters, and moreover, are the most important contributor in a multifactorial model of body mass index prediction. Conclusions We propose that rs2291007 influences human leanness through an evolutionarily conserved modulation of FNIP2 mRNA levels. Graphical Abstract
- Published
- 2022
- Full Text
- View/download PDF
5. Fatty acids homeostasis during fasting predicts protection from chemotherapy toxicity
- Author
-
Marta Barradas, Adrián Plaza, Gonzalo Colmenarejo, Iolanda Lázaro, Luis Filipe Costa-Machado, Roberto Martín-Hernández, Victor Micó, José Luis López-Aceituno, Jesús Herranz, Cristina Pantoja, Hector Tejero, Alberto Diaz-Ruiz, Fatima Al-Shahrour, Lidia Daimiel, Viviana Loria-Kohen, Ana Ramirez de Molina, Alejo Efeyan, Manuel Serrano, Oscar J. Pozo, Aleix Sala-Vila, and Pablo J. Fernandez-Marcos
- Subjects
Science - Abstract
Fasting has been reported to protect from chemotherapy-associated toxicity. Here, the authors show that fatty acid profiles in erythrocyte membranes and gene expression from peripheral blood mononuclear cells are associated to the fasting-mediated benefits during cancer treatment in mice and patients.
- Published
- 2022
- Full Text
- View/download PDF
6. The metabolic plasticity of B cells
- Author
-
Yurena Vivas-García and Alejo Efeyan
- Subjects
metabolic plasticity ,B-cell activation ,humoral response ,glycolysis ,Oxphos ,anabolism ,Biology (General) ,QH301-705.5 - Abstract
The humoral response requires rapid growth, biosynthetic capacity, proliferation and differentiation of B cells. These processes involve profound B-cell phenotypic transitions that are coupled to drastic changes in metabolism so as to meet the extremely different energetic requirements as B cells switch from resting to an activated, highly proliferative state and to plasma or memory cell fates. Thus, B cells execute a multi-step, energetically dynamic process of profound metabolic rewiring from low ATP production to transient and large increments of energy expenditure that depend on high uptake and consumption of glucose and fatty acids. Such metabolic plasticity is under tight transcriptional and post-transcriptional regulation. Alterations in B-cell metabolism driven by genetic mutations or by extrinsic insults impair B-cell functions and differentiation and may underlie the anomalous behavior of pathological B cells. Herein, we review molecular switches that control B-cell metabolism and fuel utilization, as well as the emerging awareness of the impact of dynamic metabolic adaptations of B cells throughout the different phases of the humoral response.
- Published
- 2022
- Full Text
- View/download PDF
7. Limited survival and impaired hepatic fasting metabolism in mice with constitutive Rag GTPase signaling
- Author
-
Celia de la Calle Arregui, Ana Belén Plata-Gómez, Nerea Deleyto-Seldas, Fernando García, Ana Ortega-Molina, Julio Abril-Garrido, Elena Rodriguez, Ivan Nemazanyy, Laura Tribouillard, Alba de Martino, Eduardo Caleiras, Ramón Campos-Olivas, Francisca Mulero, Mathieu Laplante, Javier Muñoz, Mario Pende, Guadalupe Sabio, David M. Sabatini, and Alejo Efeyan
- Subjects
Science - Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) integrates nutrient and hormonal signals to control metabolism. Here the authors investigate the effects of constitutive nutrient signaling through genetic activation of RagA in adult mice and show that constitutive nutrient signaling regulates the response to feeding-fasting cycles and does not drive liver cancer.
- Published
- 2021
- Full Text
- View/download PDF
8. Protocol for the assessment of mTOR activity in mouse primary hepatocytes
- Author
-
Ana Belén Plata-Gómez, María Crespo, Celia de la Calle Arregui, Lucía de Prado-Rivas, Guadalupe Sabio, and Alejo Efeyan
- Subjects
Cell culture ,Cell isolation ,Cell-based Assays ,Metabolism ,Metabolomics ,Signal Transduction ,Science (General) ,Q1-390 - Abstract
Summary: We present a protocol for measuring the activity of the mechanistic target of rapamycin (mTOR) pathway in ex vivo isolated mouse primary hepatocytes. It can be used as a tool for genetic, pharmacological, metabolomic, and signal transduction procedures. We discuss critical aspects for improving yield, viability, and modulation of the mTOR pathway. This protocol can be adapted to other signaling cascades and is compatible with multiple readouts.For complete details on the use and execution of this protocol, please refer to Ortega-Molina et al. (2021).
- Published
- 2021
- Full Text
- View/download PDF
9. From mouse genetics to targeting the Rag GTPase pathway
- Author
-
Ana Ortega-Molina and Alejo Efeyan
- Subjects
mtor ,rag gtpases ,nutrients ,b cells ,lymphoma ,small molecules ,mice ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The identification of the Rag GTPases initiated the deciphering of the molecular puzzle of nutrient signaling to the mechanistic target of rapamycin (mTOR), and spurred interest in targeting this pathway to combat human disease. Recent mouse genetic studies have provided pathophysiological insight and pointed to potential indications for inhibitors of the Rag GTPase pathway.
- Published
- 2021
- Full Text
- View/download PDF
10. The mTOR–Autophagy Axis and the Control of Metabolism
- Author
-
Nerea Deleyto-Seldas and Alejo Efeyan
- Subjects
autophagy ,mechanistic target of rapamycin ,lysosome ,metabolism ,nutrients ,Biology (General) ,QH301-705.5 - Abstract
The mechanistic target of rapamycin (mTOR), master regulator of cellular metabolism, exists in two distinct complexes: mTOR complex 1 and mTOR complex 2 (mTORC1 and 2). MTORC1 is a master switch for most energetically onerous processes in the cell, driving cell growth and building cellular biomass in instances of nutrient sufficiency, and conversely, allowing autophagic recycling of cellular components upon nutrient limitation. The means by which the mTOR kinase blocks autophagy include direct inhibition of the early steps of the process, and the control of the lysosomal degradative capacity of the cell by inhibiting the transactivation of genes encoding structural, regulatory, and catalytic factors. Upon inhibition of mTOR, autophagic recycling of cellular components results in the reactivation of mTORC1; thus, autophagy lies both downstream and upstream of mTOR. The functional relationship between the mTOR pathway and autophagy involves complex regulatory loops that are significantly deciphered at the cellular level, but incompletely understood at the physiological level. Nevertheless, genetic evidence stemming from the use of engineered strains of mice has provided significant insight into the overlapping and complementary metabolic effects that physiological autophagy and the control of mTOR activity exert during fasting and nutrient overload.
- Published
- 2021
- Full Text
- View/download PDF
11. Inhibition of Rag GTPase signaling in mice suppresses B cell responses and lymphomagenesis with minimal detrimental trade-offs
- Author
-
Ana Ortega-Molina, Cristina Lebrero-Fernández, Alba Sanz, Nerea Deleyto-Seldas, Ana Belén Plata-Gómez, Camino Menéndez, Osvaldo Graña-Castro, Eduardo Caleiras, and Alejo Efeyan
- Subjects
B cell lymphoma ,germinal center ,lymphocytes ,mTOR ,RRAGC ,nutrient signaling ,Biology (General) ,QH301-705.5 - Abstract
Summary: B lymphocytes are exquisitely sensitive to fluctuations in nutrient signaling by the Rag GTPases, and 15% of follicular lymphomas (FLs) harbor activating mutations in RRAGC. Hence, a potential therapeutic approach against malignant B cells is to inhibit Rag GTPase signaling, but because such inhibitors are still to be developed, efficacy and safety remain unknown. We generated knockin mice expressing a hypomorphic variant of RagC (Q119L); RagCQ119L/+ mice are viable and show attenuated nutrient signaling. B lymphocyte activation is cell-intrinsically impaired in RagCQ119L/+ mice, which also show significant suppression of genetically induced lymphomagenesis and autoimmunity. Surprisingly, no overt systemic trade-offs or phenotypic alterations caused by partial suppression of nutrient signaling are seen in other organs, and RagCQ119L/+ mice show normal longevity and normal age-dependent health decline. These results support the efficacy and safety of moderate inhibition of nutrient signaling against pathological B cells.
- Published
- 2021
- Full Text
- View/download PDF
12. Limited role of murine ATM in oncogene-induced senescence and p53-dependent tumor suppression.
- Author
-
Alejo Efeyan, Matilde Murga, Barbara Martinez-Pastor, Ana Ortega-Molina, Rebeca Soria, Manuel Collado, Oscar Fernandez-Capetillo, and Manuel Serrano
- Subjects
Medicine ,Science - Abstract
Recent studies in human fibroblasts have provided a new general paradigm of tumor suppression according to which oncogenic signaling produces DNA damage and this, in turn, results in ATM/p53-dependent cellular senescence. Here, we have tested this model in a variety of murine experimental systems. Overexpression of oncogenic Ras in murine fibroblasts efficiently induced senescence but this occurred in the absence of detectable DNA damage signaling, thus suggesting a fundamental difference between human and murine cells. Moreover, lung adenomas initiated by endogenous levels of oncogenic K-Ras presented abundant senescent cells, but undetectable DNA damage signaling. Accordingly, K-Ras-driven adenomas were also senescent in Atm-null mice, and the tumorigenic progression of these lesions was only modestly accelerated by Atm-deficiency. Finally, we have examined chemically-induced fibrosarcomas, which possess a persistently activated DNA damage response and are highly sensitive to the activity of p53. We found that the absence of Atm favored genomic instability in the resulting tumors, but did not affect the persistent DNA damage response and did not impair p53-dependent tumor suppression. All together, we conclude that oncogene-induced senescence in mice may occur in the absence of a detectable DNA damage response. Regarding murine Atm, our data suggest that it plays a minor role in oncogene-induced senescence or in p53-dependent tumor suppression, being its tumor suppressive activity probably limited to the maintenance of genomic stability.
- Published
- 2009
- Full Text
- View/download PDF
13. Bias in Intracellular Luminescence Thermometry: The Case of the Green Fluorescent Protein
- Author
-
Paloma Rodríguez‐Sevilla, Graham Spicer, Ana Sagrera, Alejandro P. Adam, Alejo Efeyan, Daniel Jaque, Sebastian A. Thompson, and UAM. Departamento de Física de Materiales
- Subjects
intracellular studies ,nucleus ,Física ,fluorescence polarization anisotropy ,luminescence thermometry ,Atomic and Molecular Physics, and Optics ,Electronic, Optical and Magnetic Materials - Abstract
Measurement of intracellular temperature in a fast, accurate, reliable, and remote manner is crucial for the understanding of cellular processes. Nanothermometers based on the green fluorescence protein (GFP) are of special interest because intracellular temperature readouts can be obtained from the analysis of the polarization state of its luminescence. Despite the good results provided by GFP thermometers, the reliability of their intracellular thermal readouts is still a question of debate. Here, light is shed on this issue by introducing cell activity as a relevant bias mechanism that prevents the use of GFP for reliable intranuclear thermal measurements. Experimental evidence that this lack of reliability can affect not only GFP but also other widely used thermometers such as semiconductor nanocrystals is provided. It is discussed how differences observed between calibration curves obtained in presence and absence of cell activity can inform about the presence of bias. The presented results and discussion are aimed to warn the community working in intracellular thermometry and encourage authors to approach the issue in a conscious manner. The performance and reliability of the chosen intracellular thermometers must be judiciously assessed. This is the only way intracellular thermometry can progress and deliver indisputable results, This work was financed by the Spanish Ministerio de Innovación y Ciencias under Project Nos. RTI2018-101050-J-I00, NANONERV PID2019- 106211RB-I00, and EIN2020-112419. Additional funding was provided by the European Union Horizon 2020 FETOpen project NanoTBTech (Grant No. 801305). P.R.-S. is grateful for a Juan de la Cierva – Incorporación scholarship (Grant No. IJC2019-041915-I). A.E. is grateful to Retos Projects Program of the Spanish Ministry of Science, Innovation, and Universities, the Spanish State Research Agency, co-funded by the European Regional Development Fund (A.E. is an EMBO Young Investigator). S.T. is grateful to AECC (Spanish Association Against Cancer) IDEAS21989THOM.
- Published
- 2023
14. A YAP/TAZ-TEAD signalling module links endothelial nutrient acquisition to angiogenic growth
- Author
-
Yu Ting Ong, Jorge Andrade, Max Armbruster, Chenyue Shi, Marco Castro, Ana S. H. Costa, Toshiya Sugino, Guy Eelen, Barbara Zimmermann, Kerstin Wilhelm, Joseph Lim, Shuichi Watanabe, Stefan Guenther, Andre Schneider, Francesca Zanconato, Manuel Kaulich, Duojia Pan, Thomas Braun, Holger Gerhardt, Alejo Efeyan, Peter Carmeliet, Stefano Piccolo, Ana Rita Grosso, Michael Potente, UCIBIO - Applied Molecular Biosciences Unit, and DCV - Departamento de Ciências da Vida
- Subjects
TEA Domain Transcription Factors/metabolism ,Endocrinology, Diabetes and Metabolism ,Endothelial Cells ,TEA Domain Transcription Factors ,YAP-Signaling Proteins ,Nutrients ,Cell Biology ,Endothelial Cells/metabolism ,Mechanistic Target of Rapamycin Complex 1 ,YAP-Signaling Proteins/metabolism ,Mice ,Cardiovascular and Metabolic Diseases ,Acyltransferases/metabolism ,Physiology (medical) ,Trans-Activators ,Internal Medicine ,Animals ,Trans-Activators/metabolism ,Mechanistic Target of Rapamycin Complex 1/metabolism ,Acyltransferases - Abstract
Funding Information: The research in the M.P. laboratory was supported by the Max Planck Society, the European Research Council (ERC) Consolidator Grant EMERGE (no. 773047), the Deutsche Forschungsgemeinschaft (DFG, Project-ID 75732319 – SFB 834), the Leducq Foundation, the European Union’s Horizon 2020 research and innovation programme under the Marie Sklodowska-Curie action (no. 814316), the Excellence Cluster Cardio-Pulmonary Institute (EXC 2026, Project-ID 390649896), the DZHK (German Centre for Cardiovascular Research), the Stiftung Charité and the European Molecular Biology Organization (EMBO) Young Investigator Programme. Work in the H.G. laboratory was supported by the DFG, Project-ID 427826188 – SFB 1444 and Project-ID 437531118 – SFB1470. Research in the Carmeliet laboratory is supported by Methusalem funding by the Flemish government and by an ERC Advanced Research grant (no. EU-ERC269073). This work was performed with assistance from the CSHL Mass Spectrometry Shared Resource, which is supported by a Cancer Centre Support grant (no. 5P30CA045508). Publisher Copyright: © 2022, The Author(s). Angiogenesis, the process by which endothelial cells (ECs) form new blood vessels from existing ones, is intimately linked to the tissue's metabolic milieu and often occurs at nutrient-deficient sites. However, ECs rely on sufficient metabolic resources to support growth and proliferation. How endothelial nutrient acquisition and usage are regulated is unknown. Here we show that these processes are instructed by Yes-associated protein 1 (YAP)/WW domain-containing transcription regulator 1 (WWTR1/TAZ)-transcriptional enhanced associate domain (TEAD): a transcriptional module whose function is highly responsive to changes in the tissue environment. ECs lacking YAP/TAZ or their transcriptional partners, TEAD1, 2 and 4 fail to divide, resulting in stunted vascular growth in mice. Conversely, activation of TAZ, the more abundant paralogue in ECs, boosts proliferation, leading to vascular hyperplasia. We find that YAP/TAZ promote angiogenesis by fuelling nutrient-dependent mTORC1 signalling. By orchestrating the transcription of a repertoire of cell-surface transporters, including the large neutral amino acid transporter SLC7A5, YAP/TAZ-TEAD stimulate the import of amino acids and other essential nutrients, thereby enabling mTORC1 activation. Dissociating mTORC1 from these nutrient inputs-elicited by the loss of Rag GTPases-inhibits mTORC1 activity and prevents YAP/TAZ-dependent vascular growth. Together, these findings define a pivotal role for YAP/TAZ-TEAD in controlling endothelial mTORC1 and illustrate the essentiality of coordinated nutrient fluxes in the vasculature. publishersversion published
- Published
- 2022
- Full Text
- View/download PDF
15. Data from Induction of p53-Dependent Senescence by the MDM2 Antagonist Nutlin-3a in Mouse Cells of Fibroblast Origin
- Author
-
Manuel Serrano, Lyubomir T. Vassilev, Daniel Herranz, Susana Velasco-Miguel, Ana Ortega-Molina, and Alejo Efeyan
- Abstract
Cellular senescence is emerging as an important in vivo anticancer response elicited by multiple stresses, including currently used chemotherapeutic drugs. Nutlin-3a is a recently discovered small-molecule antagonist of the p53-destabilizing protein murine double minute-2 (MDM2) that induces cell cycle arrest and apoptosis in cancer cells with functional p53. Here, we report that nutlin-3a induces cellular senescence in murine primary fibroblasts, oncogenically transformed fibroblasts, and fibrosarcoma cell lines. No evidence of drug-induced apoptosis was observed in any case. Nutlin-induced senescence was strictly dependent on the presence of functional p53 as revealed by the fact that cells lacking p53 were completely insensitive to the drug, whereas cells lacking the tumor suppressor alternative reading frame product of the CDKN2A locus underwent irreversible cell cycle arrest. Interestingly, irreversibility was achieved in neoplastic cells faster than in their corresponding parental primary cells, suggesting that nutlin-3a and oncogenic signaling cooperate in activating p53. Our current results suggest that senescence could be a major cellular outcome of cancer therapy by antagonists of the p53-MDM2 interaction, such as nutlin-3a. [Cancer Res 2007;67(15):7350–7]
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Figure 1 from Induction of p53-Dependent Senescence by the MDM2 Antagonist Nutlin-3a in Mouse Cells of Fibroblast Origin
- Author
-
Manuel Serrano, Lyubomir T. Vassilev, Daniel Herranz, Susana Velasco-Miguel, Ana Ortega-Molina, and Alejo Efeyan
- Abstract
Supplementary Figure 1 from Induction of p53-Dependent Senescence by the MDM2 Antagonist Nutlin-3a in Mouse Cells of Fibroblast Origin
- Published
- 2023
- Full Text
- View/download PDF
17. AMPK knocks at the gate of GATOR
- Author
-
Nerea Deleyto-Seldas and Alejo Efeyan
- Subjects
Physiology (medical) ,Endocrinology, Diabetes and Metabolism ,Internal Medicine ,Cell Biology - Published
- 2023
- Full Text
- View/download PDF
18. 3D bioprinted functional skeletal muscle models have potential applications for studies of muscle wasting in cancer cachexia
- Author
-
Andrea García-Lizarribar, Aranzazu Villasante, Jose Antonio Lopez-Martin, Marta Flandez, M. Carmen Soler-Vázquez, Dolors Serra, Laura Herrero, Ana Sagrera, Alejo Efeyan, and Josep Samitier
- Subjects
Biomaterials ,Biomedical Engineering ,Bioengineering - Published
- 2023
- Full Text
- View/download PDF
19. Inhibition of Rag GTPase signaling in mice suppresses B cell responses and lymphomagenesis with minimal detrimental trade-offs
- Author
-
Alba Sanz, Alejo Efeyan, Ana Belén Plata-Gómez, Eduardo Caleiras, Osvaldo Graña-Castro, Ana Ortega-Molina, Cristina Lebrero-Fernández, Nerea Deleyto-Seldas, and Camino Menéndez
- Subjects
Male ,lymphocytes ,Heterozygote ,Lymphoma ,Carcinogenesis ,QH301-705.5 ,Longevity ,GTPase ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,Autoimmunity ,medicine ,Animals ,Gene Knock-In Techniques ,Biology (General) ,B-cell lymphoma ,PI3K/AKT/mTOR pathway ,B cell ,Monomeric GTP-Binding Proteins ,B-Lymphocytes ,RRAGC ,Cell growth ,Germinal center ,nutrient signaling ,medicine.disease ,Phenotype ,Mice, Mutant Strains ,Immunity, Humoral ,Cell biology ,B cell lymphoma ,medicine.anatomical_structure ,germinal center ,Mutation ,mTOR ,Female ,Signal Transduction - Abstract
SUMMARY B lymphocytes are exquisitely sensitive to fluctuations in nutrient signaling by the Rag GTPases, and 15% of follicular lymphomas (FLs) harbor activating mutations in RRAGC. Hence, a potential therapeutic approach against malignant B cells is to inhibit Rag GTPase signaling, but because such inhibitors are still to be developed, efficacy and safety remain unknown. We generated knockin mice expressing a hypomorphic variant of RagC (Q119L); RagCQ119L/+ mice are viable and show attenuated nutrient signaling. B lymphocyte activation is cell-intrinsically impaired in RagCQ119L/+ mice, which also show significant suppression of genetically induced lymphomagenesis and autoimmunity. Surprisingly, no overt systemic trade-offs or phenotypic alterations caused by partial suppression of nutrient signaling are seen in other organs, and RagCQ119L/+ mice show normal longevity and normal age-dependent health decline. These results support the efficacy and safety of moderate inhibition of nutrient signaling against pathological B cells., Graphical abstract, In brief By generating knockin mice expressing a hypomorphic variant of the RagC GTPase, Ortega-Molina et al. show that partial inhibition of nutrient signaling may be a safe and efficacious approach against pathogenic B cells and B cell lymphomas.
- Published
- 2021
20. Disruption of liver homeostasis and systemic metabolism upon concomitant hepatic activation of growth factor and nutrient signaling
- Author
-
Ana Belén Plata-Gómez, Alejo Efeyan, Eduardo Caleiras, and Celia de la Calle Arregui
- Subjects
Nutrient ,Chemistry ,Growth factor ,medicine.medical_treatment ,Concomitant ,medicine ,Metabolism ,Homeostasis ,Cell biology - Published
- 2021
- Full Text
- View/download PDF
21. Fatty acids homeostasis during fasting predicts protection from chemotherapy toxicity
- Author
-
Marta Barradas, Adrián Plaza, Gonzalo Colmenarejo, Iolanda Lázaro, Luis Filipe Costa-Machado, Roberto Martín-Hernández, Victor Micó, José Luis López-Aceituno, Jesús Herranz, Cristina Pantoja, Hector Tejero, Alberto Diaz-Ruiz, Fatima Al-Shahrour, Lidia Daimiel, Viviana Loria-Kohen, Ana Ramirez de Molina, Alejo Efeyan, Manuel Serrano, Oscar J. Pozo, Aleix Sala-Vila, Pablo J. Fernandez-Marcos, Fundacion Ramon Areces, Asociación Española Contra el Cáncer, Ministerio de Ciencia e Innovación (España), Comunidad de Madrid (España), Instituto de Salud Carlos III, Fundación La Caixa, and Unión Europea. Comisión Europea. European Research Council (ERC)
- Subjects
Multidisciplinary ,Fatty Acids ,General Physics and Astronomy ,General Chemistry ,Fasting ,General Biochemistry, Genetics and Molecular Biology ,Fatty Acids, Monounsaturated ,Oxaliplatin ,Mice ,MicroRNAs ,Doxorubicin ,Leukocytes, Mononuclear ,Chemotherapy ,Animals ,Homeostasis ,Humans ,Insulin ,Fat metabolism ,Biomarkers - Abstract
Fasting exerts beneficial effects in mice and humans, including protection from chemotherapy toxicity. To explore the involved mechanisms, we collect blood from humans and mice before and after 36 or 24 hours of fasting, respectively, and measure lipid composition of erythrocyte membranes, circulating micro RNAs (miRNAs), and RNA expression at peripheral blood mononuclear cells (PBMCs). Fasting coordinately affects the proportion of polyunsaturated versus saturated and monounsaturated fatty acids at the erythrocyte membrane; and reduces the expression of insulin signaling-related genes in PBMCs. When fasted for 24 hours before and 24 hours after administration of oxaliplatin or doxorubicin, mice show a strong protection from toxicity in several tissues. Erythrocyte membrane lipids and PBMC gene expression define two separate groups of individuals that accurately predict a differential protection from chemotherapy toxicity, with important clinical implications. Our results reveal a mechanism of fasting associated with lipid homeostasis, and provide biomarkers of fasting to predict fasting-mediated protection from chemotherapy toxicity. General: We thank Prof. Jose Maria. Ordovas for his kind suggestions; nutritionists Helena Marcos-Pasero, Elena Aguilar-Aguilar and Isabel Espinosa-Salinas for their help with volunteers management; Rosa Serrano for her help with animal experiments; Susana Molina for her advice with PBMC isolation; Luisa Mariscal, Domingo Fernandez, Lola Martinez, Diego Megias, Patricia Gonzalez, Fernando Pelaez, Anabel Sanz, Carolina Pola, Celia de la Calle, Ana Ortega, Ana Sagrera, Jose Miguel Frade, Elena Lopez-Guadamillas, Maribel Munoz, Susana Llanos, Andres Fernandez, Aranzazu Sierra, Andres Lopez, Noemi Haro and Ildefonso Rodriguez for their excellent technical and scientific support. Work at the laboratory of P.J.F.M. is funded by the Ramon Areces Foundation, (CIVP18A3891), Asociacion Espanola contra el Cancer-AECC (SIRTBIO-LABAE18008FERN), a Ramon y Cajal Award from the Spanish Ministry of Science, Innovation and Universities (MICINN) (RYC-2017-22335), RETOS projects Program of MICINN (SAF2017-85766-R) and the Portuguese Foundation for Science and Technology (FCT-MCTES, SFRH/BD/124022/2016). Work at the laboratory of ARM was funded by the MICINN (PID2019-110183RB-C21), Regional Government of Community of Madrid (P2018/BAA-4343-ALIBIRD2020-CM) and the Ramon Areces Foundation. Work at the laboratory of A.D.R. Funded by the Comunidad de Madrid-Talento Grant 2018-T1/BMD-11966 and the MICINN PID-2019-106893RA-100. Work at the laboratory of L.D. is funded by projects from the Health Research Fund (ISCIII FIS PI14/01374 and FISPI17/00508) and from a Manuel de Oya research fellowship from the Beer and Health Foundation. Work at the laboratory of A.E. is funded by a Ramon y Cajal Award from MICINN (RYC-2013-13546) and RETOS projects Program of the MICINN, co-funded by the European Regional Development Fund (ERDF) (SAF2015-67538-R). Work in the laboratory of M.S. was funded by the IRB and by grants from the Spanish Ministry of Economy co-funded by the European Regional Development Fund (ERDF) (SAF2013-48256-R), the European Research Council (ERC-2014-AdG/669622), and the "laCaixa" Foundation. Sí
- Published
- 2021
22. Cyclin D3 drives inertial cell cycling in dark zone germinal center B cells
- Author
-
Alex K. Shalek, Alejo Efeyan, Juhee Pae, Samuel J. Allon, Coraline Mlynarczyk, Jonatan Ersching, Jose Ordovas-Montanes, Ari Melnick, Gabriel D. Victora, Michael Meyer-Hermann, Marta Schips, Tiago B. R. Castro, Luka Mesin, and BRICS, Braunschweiger Zentrum für Systembiologie, Rebenring 56,38106 Braunschweig, Germany.
- Subjects
Male ,0301 basic medicine ,T Follicular Helper Cells ,Immunology ,Cell ,Somatic hypermutation ,Article ,Affinity maturation ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Immunology and Allergy ,Cyclin D3 ,Cells, Cultured ,B cell ,Cell Proliferation ,Cyclin ,Gene Editing ,B-Lymphocytes ,Chimera ,Chemistry ,Cell Cycle ,Germinal center ,Cell cycle ,Germinal Center ,Burkitt Lymphoma ,Cell biology ,Mice, Inbred C57BL ,Leukemia & Lymphoma ,030104 developmental biology ,medicine.anatomical_structure ,Gain of Function Mutation ,030220 oncology & carcinogenesis ,Female ,Somatic Hypermutation, Immunoglobulin ,CRISPR-Cas Systems - Abstract
Cyclin D3 drives a unique form of “inertial” cell cycling responsible for clonal expansion of B cells in the germinal center dark zone., During affinity maturation, germinal center (GC) B cells alternate between proliferation and somatic hypermutation in the dark zone (DZ) and affinity-dependent selection in the light zone (LZ). This anatomical segregation imposes that the vigorous proliferation that allows clonal expansion of positively selected GC B cells takes place ostensibly in the absence of the signals that triggered selection in the LZ, as if by “inertia.” We find that such inertial cycles specifically require the cell cycle regulator cyclin D3. Cyclin D3 dose-dependently controls the extent to which B cells proliferate in the DZ and is essential for effective clonal expansion of GC B cells in response to strong T follicular helper (Tfh) cell help. Introduction into the Ccnd3 gene of a Burkitt lymphoma–associated gain-of-function mutation (T283A) leads to larger GCs with increased DZ proliferation and, in older mice, clonal B cell lymphoproliferation, suggesting that the DZ inertial cell cycle program can be coopted by B cells undergoing malignant transformation., Graphical Abstract
- Published
- 2021
23. Limited survival and impaired hepatic fasting metabolism in mice with constitutive Rag GTPase signaling
- Author
-
Mathieu Laplante, Nerea Deleyto-Seldas, Guadalupe Sabio, Ana Ortega-Molina, Celia de la Calle Arregui, Ivan Nemazanyy, Julio Abril-Garrido, Francisca Mulero, Elena Rodríguez, David M. Sabatini, Alba de Martino, Ana Belén Plata-Gómez, Fernando García, Laura Tribouillard, Ramón Campos-Olivas, Javier Munoz, Alejo Efeyan, Mario Pende, Eduardo Caleiras, Ministerio de Ciencia, Innovación y Universidades (España), Unión Europea. Comisión Europea, Asociación Española Contra el Cáncer, and Canadian Institutes of Health Research
- Subjects
0301 basic medicine ,Proteomics ,LIVER ,Transcription, Genetic ,medicine.medical_treatment ,Science ,General Physics and Astronomy ,Kinases ,mTORC1 ,Biology ,Carbohydrate metabolism ,Mechanistic Target of Rapamycin Complex 1 ,General Biochemistry, Genetics and Molecular Biology ,Article ,Tuberous Sclerosis Complex 1 Protein ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Metabolomics ,Amino acid homeostasis ,REVEALS ,medicine ,Animals ,Homeostasis ,Humans ,PPAR alpha ,Monomeric GTP-Binding Proteins ,MTORC1 ,Sirolimus ,Multidisciplinary ,Growth factor ,Endocrine system and metabolic diseases ,General Chemistry ,Metabolism ,Fasting ,Nutrients ,Cell biology ,MODEL ,Disease Models, Animal ,030104 developmental biology ,Glucose ,Phenotype ,Liver ,030217 neurology & neurosurgery ,Hormone ,Signal Transduction - Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) integrates cellular nutrient signaling and hormonal cues to control metabolism. We have previously shown that constitutive nutrient signaling to mTORC1 by means of genetic activation of RagA (expression of GTP-locked RagA, or RagAGTP) in mice resulted in a fatal energetic crisis at birth. Herein, we rescue neonatal lethality in RagAGTP mice and find morphometric and metabolic alterations that span glucose, lipid, ketone, bile acid and amino acid homeostasis in adults, and a median lifespan of nine months. Proteomic and metabolomic analyses of livers from RagAGTP mice reveal a failed metabolic adaptation to fasting due to a global impairment in PPARα transcriptional program. These metabolic defects are partially recapitulated by restricting activation of RagA to hepatocytes, and revert by pharmacological inhibition of mTORC1. Constitutive hepatic nutrient signaling does not cause hepatocellular damage and carcinomas, unlike genetic activation of growth factor signaling upstream of mTORC1. In summary, RagA signaling dictates dynamic responses to feeding-fasting cycles to tune metabolism so as to match the nutritional state., The mechanistic target of rapamycin complex 1 (mTORC1) integrates nutrient and hormonal signals to control metabolism. Here the authors investigate the effects of constitutive nutrient signaling through genetic activation of RagA in adult mice and show that constitutive nutrient signaling regulates the response to feeding-fasting cycles and does not drive liver cancer.
- Published
- 2021
24. Assessing kinetics and recruitment of DNA repair factors using high content screens
- Author
-
Barbara Martinez-Pastor, Giorgia G. Silveira, Thomas L. Clarke, Dudley Chung, Yuchao Gu, Claudia Cosentino, Lance S. Davidow, Gadea Mata, Sylvana Hassanieh, Jayme Salsman, Alberto Ciccia, Narkhyun Bae, Mark T. Bedford, Diego Megias, Lee L. Rubin, Alejo Efeyan, Graham Dellaire, and Raul Mostoslavsky
- Subjects
Histones ,Kinetics ,Open Reading Frames ,DNA Repair Enzymes ,DNA Repair ,Humans ,Tumor Suppressor p53-Binding Protein 1 ,General Biochemistry, Genetics and Molecular Biology ,Article ,Chromatin ,DNA Damage ,High-Throughput Screening Assays - Abstract
Repair of genetic damage is coordinated in the context of chromatin, so cells dynamically modulate accessibility at DNA breaks for the recruitment of DNA damage response (DDR) factors. The identification of chromatin factors with roles in DDR has mostly relied on loss-of-function screens while lacking robust high-throughput systems to study DNA repair. In this study, we have developed two high-throughput systems that allow the study of DNA repair kinetics and the recruitment of factors to double-strand breaks in a 384-well plate format. Using a customized gain-of-function open-reading frame library ("ChromORFeome" library), we identify chromatin factors with putative roles in the DDR. Among these, we find the PHF20 factor is excluded from DNA breaks, affecting DNA repair by competing with 53BP1 recruitment. Adaptable for genetic perturbations, small-molecule screens, and large-scale analysis of DNA repair, these resources can aid our understanding and manipulation of DNA repair.
- Published
- 2021
25. Cyclin D3 drives inertial cell cycling in dark zone germinal center B cells
- Author
-
Jose Ordovas-Montanes, Ari Melnick, Jonatan Ersching, Michael Meyer-Hermann, Alex K. Shalek, Marta Schips, Tiago B. R. Castro, Alejo Efeyan, Luka Mesin, Gabriel D. Victora, Juhee Pae, Samuel J. Allon, and Coraline Mlynarczyk
- Subjects
Affinity maturation ,medicine.anatomical_structure ,Chemistry ,Cell ,medicine ,Somatic hypermutation ,Germinal center ,Cell cycle ,Cyclin D3 ,B cell ,Cell biology ,Malignant transformation - Abstract
During affinity maturation, germinal center (GC) B cells alternate between proliferation and so-matic hypermutation in the dark zone (DZ) and affinity-dependent selection in the light zone (LZ). This anatomical segregation imposes that the vigorous proliferation that allows clonal expansion of positively-selected GC B cells takes place ostensibly in the absence of the signals that triggered selection in the LZ, as if by “inertia.” We find that such inertial cycles specifically require the cell cycle regulator cyclin D3. Cyclin D3 dose-dependently controls the extent to which B cells proliferate in the DZ and is essential for effective clonal expansion of GC B cells in response to strong T follicular helper (Tfh) cell help. Introduction into the Ccnd3 gene of a Burkitt lymphoma-associated gain-of-function mutation (T283A) leads to larger GCs with increased DZ proliferation and, in older mice, to clonal B cell lymphoproliferation, suggesting that the DZ inertial cell cycle program can be coopted by B cells undergoing malignant transformation.
- Published
- 2020
- Full Text
- View/download PDF
26. Amino acid–insensitive mTORC1 regulation enables nutritional stress resilience in hematopoietic stem cells
- Author
-
Youmna Kfoury, David Alexander Sabatini, Ninib Baryawno, Donna Neuberg, Dongjun Lee, Naema Nayyar, Gabriel D. Victora, David T. Scadden, Francois Mercier, Ani Papazian, Alejo Efeyan, David B. Sykes, and Demetrios Kalaitzidis
- Subjects
0301 basic medicine ,Cellular differentiation ,Population ,Mice, Transgenic ,mTORC1 ,Nutrient sensing ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,03 medical and health sciences ,Stress, Physiological ,Animals ,Homeostasis ,Amino Acids ,Progenitor cell ,education ,Cells, Cultured ,PI3K/AKT/mTOR pathway ,Monomeric GTP-Binding Proteins ,2. Zero hunger ,education.field_of_study ,TOR Serine-Threonine Kinases ,Cell Differentiation ,General Medicine ,Hematopoietic Stem Cells ,Adaptation, Physiological ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Multiprotein Complexes ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Stem cell ,Research Article ,RHEB - Abstract
The mTOR pathway is a critical determinant of cell persistence and growth wherein mTOR complex 1 (mTORC1) mediates a balance between growth factor stimuli and nutrient availability. Amino acids or glucose facilitates mTORC1 activation by inducing RagA GTPase recruitment of mTORC1 to the lysosomal outer surface, enabling activation of mTOR by the Ras homolog Rheb. Thereby, RagA alters mTORC1-driven growth in times of nutrient abundance or scarcity. Here, we have evaluated differential nutrient-sensing dependence through RagA and mTORC1 in hematopoietic progenitors, which dynamically drive mature cell production, and hematopoietic stem cells (HSC), which provide a quiescent cellular reserve. In nutrient-abundant conditions, RagA-deficient HSC were functionally unimpaired and upregulated mTORC1 via nutrient-insensitive mechanisms. RagA was also dispensable for HSC function under nutritional stress conditions. Similarly, hyperactivation of RagA did not affect HSC function. In contrast, RagA deficiency markedly altered progenitor population function and mature cell output. Therefore, RagA is a molecular mechanism that distinguishes the functional attributes of reactive progenitors from a reserve stem cell pool. The indifference of HSC to nutrient sensing through RagA contributes to their molecular resilience to nutritional stress, a characteristic that is relevant to organismal viability in evolution and in modern HSC transplantation approaches.
- Published
- 2017
- Full Text
- View/download PDF
27. Harnessing DNA for nanothermometry
- Author
-
Sebastian Thompson, Hugo Bernard, Alejandro P. Adam, Sylvia Gutierrez-Erlandsson, Graham Spicer, Alejo Efeyan, Ruth Matesanz, Agencia Estatal de Investigación (España), National Institute of General Medical Sciences (US), Ministerio de Economía y Competitividad (España), Spicer, Graham, Gutierrez-Erlandsson, Sylvia, Adam, Alejandro P., Efeyan, A., Thompson, Sebastian, Spicer, Graham [0000-0002-8360-463X], Gutierrez-Erlandsson, Sylvia [0000-0002-5895-868X], Adam, Alejandro P. [https://orcid.org/ 0000-0001-6285-7235, Efeyan, A. [0000-0002-3806-6799], and Thompson, Sebastian [0000-0002-0196-1124]
- Subjects
Hoechst ,General Physics and Astronomy ,Nanoparticle ,Nanotechnology ,01 natural sciences ,Temperature measurement ,General Biochemistry, Genetics and Molecular Biology ,Article ,Nanodiamonds ,010309 optics ,chemistry.chemical_compound ,Thermal information ,0103 physical sciences ,DNA nanotechnology ,General Materials Science ,Fluorescent Dyes ,Temperatures ,Oligonucleotide ,010401 analytical chemistry ,General Engineering ,Temperature ,General Chemistry ,DNA ,Atmospheric temperature range ,Fluorescence ,0104 chemical sciences ,Nanothermometers ,chemistry ,Optical tweezers ,Anisotropy - Abstract
16 p.-4 fig.-1 graph. abst., Temperature measurement at the nano-scale has brought insight to a wide arrayof research interests in modern chemis-try, physics, and biology. These measure-ments have been enabled by the adventof nanothermometers, which relay nano-scale temperature information throughthe analysis of their intrinsic photo-physical behavior. In the past decade, several nanothermometers have beendeveloped including dyes, nanodiamonds, fluorescent proteins, nucleotides,and nanoparticles. However, temperature measurement using intact DNA hasnot yet been achieved. Here, we present a method to study the temperaturesensitivity of the DNA molecule within a physiologic temperature range whencomplexed with fluorescent dye. We theoretically and experimentally reportthe temperature sensitivity of the DNA-Hoechst 33342 complex in differentsizes of double-stranded oligonucleotides and plasmids, showing its potentialuse as a nanothermometer. These findings allow for extending the thermalstudy of DNA to several research fields including DNA nanotechnology, opti-cal tweezers, and DNA nanoparticles., This work was supported by RETOS RTI2018-101050-J-I00 (ST), National Institute of General Medical Sciences of the National Institutes of Health under award numberR01GM124133 (AA), Ramon y Cajal Grant (RYC-2013-13546) and by SAF2015-67538-R (MCIU/AEI/FEDER, UE) (AE)
- Published
- 2020
28. A spotlight on cancer researchers in Spain: new paradigms and disruptive ideas
- Author
-
Julián Pardo, Antonio Antón, Alberto J. Schuhmacher, Manuel Valiente, S. Ramón y Cajal, Ignacio Varela, Laura Soucek, P. M. Duque, Jose Jimeno, Víctor Quesada, María Abad, Héctor Peinado, P. Sancho, Alejo Efeyan, Universidad de Zaragoza, Telefónica, Ibercaja, and Fundación Fero
- Subjects
Biomedical Research ,Carcinogenesis ,Cancer research ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Humans ,Medicine ,Paradigms ,Liquid biopsy ,030304 developmental biology ,0303 health sciences ,Medical education ,business.industry ,Foundation (evidence) ,Cancer ,General Medicine ,Congresses as Topic ,medicine.disease ,Research Personnel ,3. Good health ,Oncology ,Spain ,030220 oncology & carcinogenesis ,Clonal cooperation ,business - Abstract
Over the last years, there have been relevant advances in cancer research and in treatment based on specifc genetic alterations improving survival in some tumors. Nevertheless, progress is still dismal in most carcinomas when are disseminated with metastasis; the survival is less than 30% in most cases. Then, we need to move forward looking for new paradigms which may explain the complex mechanisms involved in carcinogenesis and in new treatment approaches., We thank to the University of Zaragoza for the opportunity to organize this meeting and to Telefonica, Ibercaja and the Fero Foundation for its support.
- Published
- 2020
29. Nutrient mTORC1 signaling underpins regulatory T cell control of immune tolerance
- Author
-
Kristelle J. Capistrano, David M. Sabatini, Briana G. Nixon, Ming O. Li, Chun Chou, Xinxin Wang, Alejo Efeyan, Min Peng, Xian Zhang, Mytrang H. Do, Whitehead Institute for Biomedical Research, and Koch Institute for Integrative Cancer Research at MIT
- Subjects
Male ,Amino Acid Transport Systems ,Regulatory T cell ,Immunology ,Receptors, Antigen, T-Cell ,chemical and pharmacologic phenomena ,mTORC1 ,Nutrient sensing ,Biology ,Mechanistic Target of Rapamycin Complex 1 ,T-Lymphocytes, Regulatory ,Immune tolerance ,Mice ,immune system diseases ,hemic and lymphatic diseases ,medicine ,Immune Tolerance ,Immunology and Allergy ,Animals ,Research Articles ,Monomeric GTP-Binding Proteins ,Cell growth ,TOR Serine-Threonine Kinases ,T-cell receptor ,Brief Definitive Report ,FOXP3 ,hemic and immune systems ,Nutrients ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Female ,Signal transduction ,Signal Transduction - Abstract
Foxp3+ regulatory T (T reg) cells are pivotal regulators of immune tolerance, with T cell receptor (TCR)-driven activated T reg (aT reg) cells playing a central role; yet how TCR signaling propagates to control aT reg cell responses remains poorly understood. Here we show that TCR signaling induces expression of amino acid transporters, and renders amino acid-induced activation of mTORC1 in aT reg cells. T reg cell-specific ablation of the Rag family small GTPases RagA and RagB impairs amino acid-induced mTORC1 signaling, causing defective amino acid anabolism, reduced T reg cell proliferation, and a rampant autoimmune disorder similar in severity to that triggered by T reg cell-specific TCR deficiency. Notably, T reg cells in peripheral tissues, including tumors, are more sensitive to Rag GTPase-dependent nutrient sensing. Ablation of RagA alone impairs T reg cell accumulation in the tumor, resulting in enhanced antitumor immunity. Thus, nutrient mTORC1 signaling is an essential component of TCR-initiated T reg cell reprogramming, and Rag GTPase activities may be titrated to break tumor immune tolerance., Memorial Sloan-Kettering Cancer Center (Support Grant/Core Grant P30CA08748)
- Published
- 2019
30. Abstract IA13: Oncogenic Rag GTPase signaling links cellular nutrients with the FL microenvironment
- Author
-
Cristina Lebrero-Fernández, Nerea Deleyto-Seldas, Alba Sanz, Alejo Efeyan, and Ana Ortega-Molina
- Subjects
Paracrine signalling ,Germinal center ,Endogeny ,General Medicine ,GTPase ,mTORC1 ,Biology ,Signal transduction ,Phenotype ,PI3K/AKT/mTOR pathway ,Cell biology - Abstract
During the humoral response, B cells undergo a sudden anabolic shift that requires high cellular nutrient levels to sustain the subsequent proliferative burst. Follicular lymphoma (FL) originates from B cells that have participated in the humoral response, and 15% of FL samples have selected for point, activating mutations in RRAGC, a member of the Rag GTPase family that controls the activation of the mechanistic target of rapamycin complex 1 downstream of the sensing of cellular nutrients. S74C and T89N, two of the most frequent activating single-amino acid changes in RRAGC, when targeted to the endogenous Rragc locus in mice, confer only a partial insensitivity to nutrient deprivation but strongly exacerbate B-cell responses and accelerate lymphomagenesis. Surprisingly, this moderate increase in nutrient signaling affected the interaction of B cells with the cellular microenvironment, synergizing their effects on mTORC1 activation with paracrine cues from the supportive T-cell microenvironment that activate B cells via the PI3K–Akt–mTORC1 axis. Hence, Rragc mutations sustain induced germinal centers and murine and human FL in the presence of decreased T-cell help. From a therapeutic standpoint, Rragc mutations impose a selective vulnerability to pharmacologic inhibition of mTORC1. Our results support a model in which activating mutations in the nutrient signaling pathway foster lymphomagenesis by corrupting a nutrient-dependent control over paracrine signals from the T-cell microenvironment. While pharmacologic inhibition of mTORC1 with rapamycin yielded exciting preclinical responses in murine lymphomas with activating mutations in Rragc, targeting the nutrient signaling cascade itself, instead of using such allosteric, partial inhibition of mTOR, may constitute a more efficacious intervention. Because nutrient signaling inhibitors are still in development phase, their efficacy and safety remain unproven. Previous genetic approaches to investigate the consequences of inhibition of Rag GTPase signaling relied on deletion of the Rags in mice and led to severe phenotypes and death. Incomplete inhibition of nutrient signaling, an approach that would mirror more closely the effect of small molecules, has not been pursued to support both their efficacy and safety. We have generated knock-in mice endogenously expressing a point-mutant form of RagC (Q119L) that partially suppresses nutrient signaling. RagCQ119L/Q119L mice are not viable, but RagCQ119L/+ mice show minimal phenotypic alterations with partially decreased nutrient signaling. While B-cell development was unaffected, B-cell activation and the humoral response were impaired in RagCQ119L/+ in a B cell-intrinsic manner. When bred to the FL- and autoimmunity-prone strain VavP-Bcl2, RagCQ119L/+ mice were protected against development of both FL and autoimmunity. No obvious systemic tradeoff for the suppression of nutrient signaling seems to occur, because RagCQ119L/+ mice show normal physiology and longevity with a similar age-dependent health decline. Altogether, our work supports the oncogenicity of activating mutations in components of the nutrient signaling pathway, such as RagC, and both the efficacy and safety of a moderate inhibition of nutrient signaling against pathologic B cells without detrimental systemic effects. Citation Format: Ana Ortega-Molina, Cristina Lebrero-Fernández, Nerea Deleyto-Seldas, Alba Sanz, Alejo Efeyan. Oncogenic Rag GTPase signaling links cellular nutrients with the FL microenvironment [abstract]. In: Proceedings of the AACR Virtual Meeting: Advances in Malignant Lymphoma; 2020 Aug 17-19. Philadelphia (PA): AACR; Blood Cancer Discov 2020;1(3_Suppl):Abstract nr IA13.
- Published
- 2020
- Full Text
- View/download PDF
31. Universal guidelines for the conversion of proteins and dyes into functional nanothermometers
- Author
-
Alejo Efeyan, Sebastian Thompson, Alejandro P. Adam, and Graham Spicer
- Subjects
Imagination ,Chemical substance ,Luminescence ,Light ,media_common.quotation_subject ,General Physics and Astronomy ,Nanotechnology ,Biocompatible Materials ,Thermometry ,01 natural sciences ,Temperature measurement ,General Biochemistry, Genetics and Molecular Biology ,Article ,010309 optics ,0103 physical sciences ,Animals ,General Materials Science ,Horses ,Coloring Agents ,media_common ,Protein size ,Fluorescent Dyes ,business.industry ,010401 analytical chemistry ,General Engineering ,Temperature ,Proteins ,General Chemistry ,0104 chemical sciences ,Experimental system ,Anisotropy ,Nanoparticles ,Cattle ,Photonics ,Science, technology and society ,business ,Chickens ,Fluorescence anisotropy - Abstract
In the last decade, technological advances in chemistry and photonics have enabled real-time measurement of temperature at the nanoscale. Nanothermometers, the probes specifically designed to relay these nanoscale temperature changes provide a high degree of temperature, temporal, and spatial resolution and precision. Several different approaches have been proposed, including micro-thermocouples, luminescence, and fluorescence polarization anisotropy-based nanothermometers. Anisotropy-based nanothermometers excel in terms of biocompatibility because they can be built from endogenous proteins conjugated to dyes, minimizing any system perturbation. Moreover, the resulting fluorescent proteins can retain their native structure and activity while performing the temperature measurement, allowing precise temperature recordings from the native environment or during an enzymatic reaction in any given experiment system. To facilitate future use of these nanothermometers in research, here we present a theoretical model that predicts the optimal sensitivity for anisotropy-based thermometers starting with any protein or dye, based on protein size and dye fluorescence lifetime. Using this model, most of the proteins and dyes can be converted to nanothermometers. The utilization of these nanothermometers by a broad spectrum of disciplines within the scientific community will bring new knowledge and understanding that today remains unavailable with current techniques.
- Published
- 2019
32. Corrigendum: mTORC1-dependent AMD1 regulation sustains polyamine metabolism in prostate cancer
- Author
-
Ruzica Bago, Naiara Beraza, Alfredo Caro-Maldonado, Ana Loizaga-Iriarte, Natalia Martín-Martín, Arkaitz Carracedo, Elena Castro, Violeta Serra, Mikel Azkargorta, Diana Cabrera, Michelle Clasquin, Katya Marjon, Phong Quang, Kevin Marks, Inés de Torres, Rosa Farràs, Marco Piva, Ana R. Cortazar, Ludmila Prudkin, Sonia Fernández-Ruiz, José Baselga, Julen Tomás-Cortázar, Josep Tabernero, Amelia Barnett, Sebastiaan M. Van Liempd, Carlos Cordon-Cardo, Miguel Unda, Ianire Astobiza, Rosa Barrio, Juan M. Falcón-Pérez, David Olmos, Javier Munoz, Ajinkya Revandkar, Patricia Zúñiga-García, José Antonio Jiménez, Brendan D. Manning, James D. Sutherland, Mireia Castillo-Martin, Ana M. Aransay, Pilar Sanchez-Mosquera, Leire Arreal, Alejo Efeyan, Antonio Gentilella, Itziar Fernández-Domínguez, Felix Elortza, Paolo Nuciforo, Amaia Zabala-Letona, Amaia Arruabarrena-Aristorena, David Pirman, Isabel Lacasa-Viscasillas, Juan Anguita, Gina Lein, Ylenia Cendon, Stuart Murray, Pilar Ximénez-Embún, Andrea Alimonti, Lorea Valcarcel-Jimenez, Verónica Torrano, George Thomas, Aitziber Ugalde-Olano, Teresa Macarulla, María L. Martínez-Chantar, José M. Mato, and Yinan Zhang
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Multidisciplinary ,Published Erratum ,Biology ,medicine.disease ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Polyamine metabolism - Abstract
Nature 547, 109–113 (2017); doi:10.1038/nature22964 In this Letter, there are errors in Extended Data Figs 5, 8 and 9, and the affiliation of an author. The affiliations for author Violeta Serra should include number 2 (CIBERONC, Instituto de Salud Carlos III, C/ Monforte de Lemos 3-5, Pabellon 11, Planta 0, 28029 Madrid, Spain).
- Published
- 2018
33. mTORC1-dependent AMD1 regulation sustains polyamine metabolism in prostate cancer
- Author
-
Ludmila Prudkin, José Baselga, Amelia Barnett, Leire Arreal, María L. Martínez-Chantar, Alejo Efeyan, Arkaitz Carracedo, Elena Castro, Violeta Serra, Yinan Zhang, David Pirman, Ylenia Cendon, Teresa Macarulla, Patricia Zúñiga-García, Rosa Farràs, José M. Mato, Rosa Barrio, Inés Cristina Torres, Julen Tomás-Cortázar, Juan Anguita, Juan M. Falcón-Pérez, Alfredo Caro-Maldonado, Gina Lein, Josep Tabernero, Ana Loizaga-Iriarte, Amaia Zabala-Letona, James D. Sutherland, Jose Jimenez, David Olmos, Mikel Azkargorta, Naiara Beraza, Carlos Cordon-Cardo, Pilar Sanchez-Mosquera, Ajinkya Revandkar, Paolo Nuciforo, Felix Elortza, Ruzica Bago, Isabel Lacasa-Viscasillas, Stuart Murray, Miguel Unda, Natalia Martín-Martín, Verónica Torrano, Mireia Castillo-Martin, Ana M. Aransay, Itziar Fernández-Domínguez, Antonio Gentilella, Sebastiaan M. Van Liempd, Brendan D. Manning, Aitziber Ugalde-Olano, Pilar Ximénez-Embún, Andrea Alimonti, Lorea Valcarcel-Jimenez, Javier Muñoz, Sonia Fernández-Ruiz, Diana Cabrera, Amaia Arruabarrena-Aristorena, Michelle Clasquin, Katya Marjon, Phong Quang, Marco Piva, Ana R. Cortazar, George Thomas, Kevin R Marks, and Ianire Astobiza
- Subjects
0301 basic medicine ,Male ,Adenosylmethionine Decarboxylase ,S-Adenosylmethionine ,mTORC1 ,Biology ,Oncogenicity ,Mechanistic Target of Rapamycin Complex 1 ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,Mice ,Phosphatidylinositol 3-Kinases ,Downregulation and upregulation ,Protein kinases ,Neoplasms ,medicine ,Polyamines ,Animals ,Humans ,Metabolomics ,Everolimus ,Cell Proliferation ,Multidisciplinary ,Càncer de pròstata ,Cell growth ,Protein Stability ,TOR Serine-Threonine Kinases ,PTEN Phosphohydrolase ,Cancer ,Prostatic Neoplasms ,Adenosylmethionine Decarboxylase/immunology ,Adenosylmethionine Decarboxylase/metabolism ,Enzyme Activation ,Everolimus/therapeutic use ,Multiprotein Complexes/antagonists & inhibitors ,Multiprotein Complexes/metabolism ,PTEN Phosphohydrolase/metabolism ,Phosphatidylinositol 3-Kinases/metabolism ,Polyamines/metabolism ,Prostatic Neoplasms/drug therapy ,Prostatic Neoplasms/metabolism ,Prostatic Neoplasms/pathology ,S-Adenosylmethionine/analogs & derivatives ,S-Adenosylmethionine/metabolism ,TOR Serine-Threonine Kinases/antagonists & inhibitors ,TOR Serine-Threonine Kinases/metabolism ,medicine.disease ,3. Good health ,Proteïnes quinases ,030104 developmental biology ,chemistry ,Biochemistry ,Multiprotein Complexes ,Cancer cell ,Cancer research ,biological phenomena, cell phenomena, and immunity ,Polyamine ,Signal Transduction - Abstract
Users may view, print, copy, and download text and data-mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use: http://www.nature.com/authors/editorial_policies/license.html#terms Activation of the PTEN-PI3K-mTORC1 pathway consolidates metabolic programs that sustain cancer cell growth and proliferation,. Here we show that mTORC1 regulates polyamine dynamics, a metabolic route that is essential for oncogenicity. Through the use of integrative metabolomics in a mouse model and human biopsies of prostate cancer, we identified alterations in tumours impacting on the production of decarboxylated S-adenosylmethionine (dcSAM) and polyamine synthesis. Mechanistically, this metabolic rewiring stems from mTORC1-dependent regulation of S-adenosylmethionine decarboxylase 1 (AMD1) stability. This novel molecular regulation was validated in murine and human cancer specimens. AMD1 was upregulated in prostate cancer specimens with activated mTORC1. Conversely, samples from a clinical trial with the mTORC1 inhibitor everolimus exhibited a predominant decrease in AMD1 immunoreactivity that was associated to a decrease in proliferation, in line with the requirement of dcSAM production for oncogenicity. These findings provide fundamental information about the complex regulatory landscape controlled by mTORC1 to integrate and translate growth signals into an oncogenic metabolic program.
- Published
- 2017
34. RagA, but Not RagB, Is Essential for Embryonic Development and Adult Mice
- Author
-
Steven Chang, Oktay Kirak, Dudley W. Lamming, Angelina M. Bilate, Alejo Efeyan, Lawrence D. Schweitzer, David M. Sabatini, Broad Institute of MIT and Harvard, Efeyan, Alejo, Schweitzer, Lawrence David, Bilate, Angelina M, Chang, Steven H., Lamming, Dudley, and Sabatini, David
- Subjects
Proto-Oncogene Proteins c-akt ,medicine.medical_treatment ,Population ,Mammalian embryology ,Nutrient sensing ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Mice ,Phosphatidylinositol 3-Kinases ,medicine ,Animals ,education ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Monomeric GTP-Binding Proteins ,Mice, Knockout ,education.field_of_study ,TOR Serine-Threonine Kinases ,Growth factor ,Cell Biology ,Embryo, Mammalian ,3. Good health ,Cell biology ,Liver ,Multiprotein Complexes ,Hepatocytes ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Signal Transduction ,Developmental Biology - Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) integrates cues from growth factors and nutrients to control metabolism. In contrast to the growth factor input, genetic disruption of nutrient-dependent activation of mTORC1 in mammals remains unexplored. We engineered mice lacking RagA and RagB genes, which encode the GTPases responsible for mTORC1 activation by nutrients. RagB has limited expression, and its loss shows no effects on mammalian physiology. RagA deficiency leads to E10.5 embryonic death, loss of mTORC1 activity, and severe growth defects. Primary cells derived from these mice exhibit no regulation of mTORC1 by nutrients and maintain high sensitivity to growth factors. Deletion of RagA in adult mice is lethal. Upon RagA loss, a myeloid population expands in peripheral tissues. RagA-specific deletion in liver increases cellular responses to growth factors. These results show the essentiality of nutrient sensing for mTORC1 activity in mice and its suppression of PI3K/Akt signaling., United States. National Institutes of Health (R01 CA129105), United States. National Institutes of Health (R01 CA103866), United States. National Institutes of Health (R01 AI047389), United States. National Institutes of Health (R21 AG042876), American Federation for Aging Research, Starr Foundation, David H. Koch Institute for Integrative Cancer Research at MIT. Frontier Research Program, Ellison Medical Foundation, United States. National Institutes of Health (AG041765), National Cancer Institute (U.S.) (F31CA167872)
- Published
- 2014
- Full Text
- View/download PDF
35. Germinal Center Selection and Affinity Maturation Require Dynamic Regulation of mTORC1 Kinase
- Author
-
Brian C. Grabiner, Johanne T. Jacobsen, David M. Sabatini, Jonatan Ersching, Luka Mesin, Alejo Efeyan, David Dominguez-Sola, Giulia Pasqual, Gabriel D. Victora, Massachusetts Institute of Technology. Department of Biology, and Sabatini, David
- Subjects
0301 basic medicine ,Helper-Inducer ,T-Lymphocytes ,Antibody Affinity ,mTORC1 ,Inbred C57BL ,Transgenic ,Mice ,0302 clinical medicine ,antibody ,Receptors ,Immunology and Allergy ,Clonal Selection, Antigen-Mediated ,Cells, Cultured ,B cell ,B-Lymphocytes ,Cultured ,Kinase ,TOR Serine-Threonine Kinases ,Cell Cycle ,T-Lymphocytes, Helper-Inducer ,Cell cycle ,Antigen-Mediated ,Cell biology ,cell size ,Infectious Diseases ,medicine.anatomical_structure ,Antigen ,030220 oncology & carcinogenesis ,mTOR ,Cytokines ,Cells ,Immunology ,Clonal Selection ,Somatic hypermutation ,Receptors, Antigen, B-Cell ,Mice, Transgenic ,Biology ,Mechanistic Target of Rapamycin Complex 1 ,Affinity maturation ,03 medical and health sciences ,Immunoglobulin ,medicine ,Animals ,cell cycle ,germinal center ,Cell Proliferation ,Germinal Center ,Mice, Inbred C57BL ,Multiprotein Complexes ,Sirolimus ,Somatic Hypermutation, Immunoglobulin ,PI3K/AKT/mTOR pathway ,B-Cell ,Germinal center ,Somatic Hypermutation ,030104 developmental biology ,Cancer research - Abstract
During antibody affinity maturation, germinal center (GC) B cells cycle between affinity-driven selection in the light zone (LZ) and proliferation and somatic hypermutation in the dark zone (DZ). Although selection of GC B cells is triggered by antigen-dependent signals delivered in the LZ, DZ proliferation occurs in the absence of such signals. We show that positive selection triggered by T cell help activates the mechanistic target of rapamycin complex 1 (mTORC1), which promotes the anabolic program that supports DZ proliferation. Blocking mTORC1 prior to growth prevented clonal expansion, whereas blockade after cells reached peak size had little to no effect. Conversely, constitutively active mTORC1 led to DZ enrichment but loss of competitiveness and impaired affinity maturation. Thus, mTORC1 activation is required for fueling B cells prior to DZ proliferation rather than for allowing cell-cycle progression itself and must be regulated dynamically during cyclic re-entry to ensure efficient affinity-based selection. During germinal center selection, signals from Tfh cells in the light zone dictate the extent of B cell proliferation in the dark zone. Ersching et al. (2017) show that Tfh help induces mTORC1 activation in light zone B cells, leading to cell growth that sustains the subsequent dark zone proliferative burst.
- Published
- 2016
36. Regulation of mTORC1 by the Rag GTPases is necessary for neonatal autophagy and survival
- Author
-
Alejo Efeyan, Steven Chang, Iwona Gumper, Rachel L. Wolfson, Roberto Zoncu, Oktay Kirak, David D. Sabatini, Harriet Snitkin, David M. Sabatini, Massachusetts Institute of Technology. Department of Biology, Whitehead Institute for Biomedical Research, Koch Institute for Integrative Cancer Research at MIT, Wolfson, Rachel Laura, and Sabatini, David M.
- Subjects
medicine.medical_specialty ,Multidisciplinary ,GTP' ,Glycogen ,Autophagy ,Endogeny ,mTORC1 ,GTPase ,Biology ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Internal medicine ,medicine ,Glucose homeostasis ,biological phenomena, cell phenomena, and immunity ,Homeostasis - Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) pathway regulates organismal growth in response to many environmental cues, including nutrients and growth factors. Cell-based studies showed that mTORC1 senses amino acids through the RagA–D family of GTPases (also known as RRAGA, B, C and D), but their importance in mammalian physiology is unknown. Here we generate knock-in mice that express a constitutively active form of RagA (RagA[superscript GTP]) from its endogenous promoter. RagA[superscript GTP/GTP] mice develop normally, but fail to survive postnatal day 1. When delivered by Caesarean section, fasted RagA[superscript GTP/GTP] neonates die almost twice as rapidly as wild-type littermates. Within an hour of birth, wild-type neonates strongly inhibit mTORC1, which coincides with profound hypoglycaemia and a decrease in plasma amino-acid concentrations. In contrast, mTORC1 inhibition does not occur in RagA[superscript GTP/GTP] neonates, despite identical reductions in blood nutrient amounts. With prolonged fasting, wild-type neonates recover their plasma glucose concentrations, but RagA[superscript GTP/GTP] mice remain hypoglycaemic until death, despite using glycogen at a faster rate. The glucose homeostasis defect correlates with the inability of fasted RagA[superscript GTP/GTP] neonates to trigger autophagy and produce amino acids for de novo glucose production. Because profound hypoglycaemia does not inhibit mTORC1 in RagA[superscript GTP/GTP] neonates, we considered the possibility that the Rag pathway signals glucose as well as amino-acid sufficiency to mTORC1. Indeed, mTORC1 is resistant to glucose deprivation in RagA[superscript GTP/GTP] fibroblasts, and glucose, like amino acids, controls its recruitment to the lysosomal surface, the site of mTORC1 activation. Thus, the Rag GTPases signal glucose and amino-acid concentrations to mTORC1, and have an unexpectedly key role in neonates in autophagy induction and thus nutrient homeostasis and viability., National Institutes of Health (U.S.) (Grant R01 CA129105), National Institutes of Health (U.S.) (Grant R01 CA103866), National Institutes of Health (U.S.) (Grant R37 AI047389), American Federation for Aging Research, Starr Cancer Consortium, David H. Koch Institute for Integrative Cancer Research at MIT (Frontier Research Program), Ellison Medical Foundation
- Published
- 2012
- Full Text
- View/download PDF
37. DEPTOR Cell-Autonomously Promotes Adipogenesis, and Its Expression Is Associated with Obesity
- Author
-
Zala Prevoršek, Alejo Efeyan, Simon Horvat, Kıvanç Birsoy, David M. Sabatini, Mathieu Laplante, and William T. Festuccia
- Subjects
medicine.medical_specialty ,Physiology ,Adipose Tissue, White ,Immunoblotting ,Quantitative Trait Loci ,Peroxisome proliferator-activated receptor ,Mice, Transgenic ,White adipose tissue ,Mechanistic Target of Rapamycin Complex 1 ,Real-Time Polymerase Chain Reaction ,DEPTOR ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Insulin ,Obesity ,Glucocorticoids ,Molecular Biology ,Protein kinase B ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Adipogenesis ,biology ,TOR Serine-Threonine Kinases ,Intracellular Signaling Peptides and Proteins ,Chromosome Mapping ,Proteins ,food and beverages ,3T3 Cells ,Cell Biology ,Cell biology ,Insulin receptor ,Endocrinology ,chemistry ,Multiprotein Complexes ,030220 oncology & carcinogenesis ,biology.protein - Abstract
SummaryDEP domain-containing mTOR-interacting protein (DEPTOR) inhibits the mechanistic target of rapamycin (mTOR), but its in vivo functions are unknown. Previous work indicates that Deptor is part of the Fob3a quantitative trait locus (QTL) linked to obesity/leanness in mice, with Deptor expression being elevated in white adipose tissue (WAT) of obese animals. This relation is unexpected, considering the positive role of mTOR in adipogenesis. Here, we dissected the Fob3a QTL and show that Deptor is the highest-priority candidate promoting WAT expansion in this model. Consistently, transgenic mice overexpressing DEPTOR accumulate more WAT. Furthermore, in humans, DEPTOR expression in WAT correlates with the degree of obesity. We show that DEPTOR is induced by glucocorticoids during adipogenesis and that its overexpression promotes, while its suppression blocks, adipogenesis. DEPTOR activates the proadipogenic Akt/PKB-PPAR-γ axis by dampening mTORC1-mediated feedback inhibition of insulin signaling. These results establish DEPTOR as a new regulator of adipogenesis.
- Published
- 2012
- Full Text
- View/download PDF
38. Pten Positively Regulates Brown Adipose Function, Energy Expenditure, and Longevity
- Author
-
Gonzalo Gómez-López, Ángela M. Valverde, Alejo Efeyan, M. Mar González-Barroso, Elena Lopez-Guadamillas, Eduardo Rial, Sonia Martinez, Joaquín Pastor, Marta Cañamero, Ana Ortega-Molina, James R. Bischoff, Maribel Muñoz-Martin, Manuel Serrano, Eduardo Romanos, Francisca Mulero, Comunidad de Madrid, Ministerio de Ciencia e Innovación (España), and Instituto de Salud Carlos III
- Subjects
medicine.medical_specialty ,Physiology ,Longevity ,Adipose tissue ,Mice, Transgenic ,Biology ,Calorimetry ,Ion Channels ,Mitochondrial Proteins ,Mice ,Adipose Tissue, Brown ,Internal medicine ,Brown/metabolism ,Brown adipose tissue ,medicine ,Uncoupling protein ,PTEN ,Animals ,Energy Metabolism/genetics/physiology ,Protein kinase B ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Uncoupling Protein 1 ,PRDM16 ,Imidazoles ,PTEN Phosphohydrolase ,Cell Biology ,Thermogenin ,DNA-Binding Proteins ,medicine.anatomical_structure ,Endocrinology ,DNA-Binding Proteins/genetics/metabolism ,Pyrazines ,biology.protein ,Energy Metabolism ,Ion Channels/genetics/metabolism ,Transcription Factors - Abstract
13 páginas, 7 figuras, 7 figuras suplementarias, 7 tablas suplementarias.-- et al., Aging in worms and flies is regulated by the PI3K/Akt/Foxo pathway. Here we extend this paradigm to mammals. Ptentg mice carrying additional genomic copies of Pten are protected from cancer and present a significant extension of life span that is independent of their lower cancer incidence. Interestingly, Ptentg mice have an increased energy expenditure and protection from metabolic pathologies. The brown adipose tissue (BAT) of Ptentg mice is hyperactive and presents high levels of the uncoupling protein Ucp1, which we show is a target of Foxo1. Importantly, a synthetic PI3K inhibitor also increases energy expenditure and hyperactivates the BAT in mice. These effects can be recapitulated in isolated brown adipocytes and, moreover, implants of Ptentg fibroblasts programmed with Prdm16 and Cebpβ form subcutaneous brown adipose pads more efficiently than wild-type fibroblasts. These observations uncover a role of Pten in promoting energy expenditure, thus decreasing nutrient storage and its associated damage, Work in the laboratory of A.M.V. was funded by grant SAF2009-08114 and by the CIBERDEM (ISCIII), and work in the laboratory of E. Rial was funded by grants SAF2010-20256 and Consolider-Ingenio CSD2007-00020. A.O.-M. was recipient of a predoctoral contract of the Regional Government of Madrid. E.L.-G. was recipient of a predoctoral contract from the Spanish Ministry of Education. M.M.G.-B. was supported by the “Ramon y Cajal” program of the Spanish Ministry of Science and Innovation.
- Published
- 2012
- Full Text
- View/download PDF
39. mTORC1 Senses Lysosomal Amino Acids Through an Inside-Out Mechanism That Requires the Vacuolar H + -ATPase
- Author
-
Roberto Zoncu, David M. Sabatini, Alejo Efeyan, Liron Bar-Peled, Shuyu Wang, Yasemin Sancak, Massachusetts Institute of Technology. Department of Biology, Whitehead Institute for Biomedical Research, Koch Institute for Integrative Cancer Research at MIT, Zoncu, Roberto, Bar-Peled, Liron, Efeyan, Alejo, Wang, Shuyu, Sancak, Yasemin, and Sabatini, David M.
- Subjects
Vacuolar Proton-Translocating ATPases ,ATPase ,Guanosine ,GTPase ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Cell Line ,GTP Phosphohydrolases ,chemistry.chemical_compound ,Lysosome ,medicine ,Animals ,Humans ,Amino Acids ,chemistry.chemical_classification ,Multidisciplinary ,biology ,TOR Serine-Threonine Kinases ,Proteins ,Ragulator complex ,Cell biology ,Amino acid ,medicine.anatomical_structure ,Lysosomal lumen ,chemistry ,Biochemistry ,Multiprotein Complexes ,biology.protein ,Drosophila ,RNA Interference ,biological phenomena, cell phenomena, and immunity ,Lysosomes ,Signal Transduction - Abstract
The mTOR complex 1 (mTORC1) protein kinase is a master growth regulator that is stimulated by amino acids. Amino acids activate the Rag guanosine triphosphatases (GTPases), which promote the translocation of mTORC1 to the lysosomal surface, the site of mTORC1 activation. We found that the vacuolar H+–adenosine triphosphatase ATPase (v-ATPase) is necessary for amino acids to activate mTORC1. The v-ATPase engages in extensive amino acid–sensitive interactions with the Ragulator, a scaffolding complex that anchors the Rag GTPases to the lysosome. In a cell-free system, ATP hydrolysis by the v-ATPase was necessary for amino acids to regulate the v-ATPase-Ragulator interaction and promote mTORC1 translocation. Results obtained in vitro and in human cells suggest that amino acid signaling begins within the lysosomal lumen. These results identify the v-ATPase as a component of the mTOR pathway and delineate a lysosome-associated machinery for amino acid sensing., Damon Runyon Cancer Research Foundation, Millennium Pharmaceuticals, Inc., American Lebanese Syrian Associated Charities, Howard Hughes Medical Institute
- Published
- 2011
- Full Text
- View/download PDF
40. Induction of p53-Dependent Senescence by the MDM2 Antagonist Nutlin-3a in Mouse Cells of Fibroblast Origin
- Author
-
Ana Ortega-Molina, Susana Velasco-Miguel, Alejo Efeyan, Lyubomir T. Vassilev, Daniel Herranz, and Manuel Serrano
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Senescence ,Cancer Research ,Cell cycle checkpoint ,Tumor suppressor gene ,Fibrosarcoma ,Piperazines ,Mice ,chemistry.chemical_compound ,Animals ,Cellular Senescence ,Cyclin-Dependent Kinase Inhibitor p16 ,Mice, Knockout ,biology ,Cell Cycle ,Imidazoles ,Proto-Oncogene Proteins c-mdm2 ,Nutlin ,Fibroblasts ,Oncology ,chemistry ,Apoptosis ,Cell culture ,Cancer cell ,Cancer research ,biology.protein ,Mdm2 ,ADP-Ribosylation Factor 1 ,Tumor Suppressor Protein p53 - Abstract
Cellular senescence is emerging as an important in vivo anticancer response elicited by multiple stresses, including currently used chemotherapeutic drugs. Nutlin-3a is a recently discovered small-molecule antagonist of the p53-destabilizing protein murine double minute-2 (MDM2) that induces cell cycle arrest and apoptosis in cancer cells with functional p53. Here, we report that nutlin-3a induces cellular senescence in murine primary fibroblasts, oncogenically transformed fibroblasts, and fibrosarcoma cell lines. No evidence of drug-induced apoptosis was observed in any case. Nutlin-induced senescence was strictly dependent on the presence of functional p53 as revealed by the fact that cells lacking p53 were completely insensitive to the drug, whereas cells lacking the tumor suppressor alternative reading frame product of the CDKN2A locus underwent irreversible cell cycle arrest. Interestingly, irreversibility was achieved in neoplastic cells faster than in their corresponding parental primary cells, suggesting that nutlin-3a and oncogenic signaling cooperate in activating p53. Our current results suggest that senescence could be a major cellular outcome of cancer therapy by antagonists of the p53-MDM2 interaction, such as nutlin-3a. [Cancer Res 2007;67(15):7350–7]
- Published
- 2007
- Full Text
- View/download PDF
41. p53: Guardian of the Genome and Policeman of the Oncogenes
- Author
-
Alejo Efeyan and Manuel Serrano
- Subjects
DNA damage ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,Biology ,DNA-binding protein ,Genome ,Malignant transformation ,Mice ,Tumor Suppressor Protein p14ARF ,medicine ,Animals ,Humans ,Molecular Biology ,Genetics ,Kinase ,Tumor Suppressor Proteins ,Cancer ,Oncogenes ,Cell Biology ,medicine.disease ,DNA-Binding Proteins ,Tumor Suppressor Protein p53 ,Signal transduction ,DNA Damage ,Signal Transduction ,Developmental Biology - Abstract
The process of malignant transformation universally entails genetic damage and oncogenic signaling, two stresses that are signaled to p53 through different genetic pathways. Based on this, it is possible to distinguish two jobs for p53: "guardian of the genome" that consists in sensing and reacting to DNA damage through the ATM/ATR and Chk1/Chk2 kinases, and "policeman of the oncogenes" that, correspondingly, consists in responding to oncogenic signaling through the p53-stabilizing protein ARF. Contrary to expectation, recent genetic evidence in mice indicates that the response of p53 to DNA damage has little or no impact on cancer protection. In contrast, ARF-dependent activation of p53 is critical for p53-mediated tumor suppression. Here, we discuss the mechanistic implications of these observations and their relevance for cancer therapy.
- Published
- 2007
- Full Text
- View/download PDF
42. Recurrent mTORC1-activating RRAGC mutations in follicular lymphoma
- Author
-
Csaba Bödör, Leire Escudero-Ibarz, Andrew Davies, Rachel L. Wolfson, Hilmar Quentmeier, Reiner Siebert, Ahad F. Al Seraihi, Sameena Iqbal, Claude Chelala, Julia Richter, Janet Matthews, Ming-Qing Du, Jessica Okosun, Lucy Wilkins, Andrew Jack, Stephan H. Bernhart, Sharon Barrans, Silvia Montoto, Peter Johnson, Shamzah Araf, T. Andrew Lister, Alejo Efeyan, David M. Sabatini, Jonathan C. Strefford, Maria Calaminici, Andrew Clear, Rebecca Auer, Graham Packham, John G. Gribben, José Afonso Guerra-Assunção, Christopher Mansbridge, Roberto Zoncu, Jude Fitzgibbon, Jun Wang, Brian M. Castellano, Massachusetts Institute of Technology. Department of Biology, Whitehead Institute for Biomedical Research, Koch Institute for Integrative Cancer Research at MIT, Sabatini, David, Wolfson, Rachel Laura, and Efeyan, Alejo
- Subjects
0301 basic medicine ,Molecular Sequence Data ,Follicular lymphoma ,Mechanistic Target of Rapamycin Complex 1 ,Gene mutation ,Biology ,medicine.disease_cause ,Article ,03 medical and health sciences ,Genetics ,medicine ,Animals ,Humans ,Amino Acid Sequence ,B-cell lymphoma ,Lymphoma, Follicular ,Exome ,Monomeric GTP-Binding Proteins ,Mutation ,Sequence Homology, Amino Acid ,TOR Serine-Threonine Kinases ,medicine.disease ,Lymphoma ,030104 developmental biology ,Multiprotein Complexes ,Cancer research ,Clone (B-cell biology) ,Diffuse large B-cell lymphoma - Abstract
Follicular lymphoma is an incurable B cell malignancy characterized by the t(14;18) translocation and mutations affecting the epigenome. Although frequent gene mutations in key signaling pathways, including JAK-STAT, NOTCH and NF-κB, have also been defined, the spectrum of these mutations typically overlaps with that in the closely related diffuse large B cell lymphoma (DLBCL). Using a combination of discovery exome and extended targeted sequencing, we identified recurrent somatic mutations in RRAGC uniquely enriched in patients with follicular lymphoma (17%). More than half of the mutations preferentially co-occurred with mutations in ATP6V1B2 and ATP6AP1, which encode components of the vacuolar H+-ATP ATPase (V-ATPase) known to be necessary for amino acid−induced activation of mTORC1. The RagC variants increased raptor binding while rendering mTORC1 signaling resistant to amino acid deprivation. The activating nature of the RRAGC mutations, their existence in the dominant clone and their stability during disease progression support their potential as an excellent candidate for therapeutic targeting., Experimental Cancer Medicine Centres
- Published
- 2015
43. Genetic dissection of the role of p21Cip1/Waf1 in p53-mediated tumour suppression
- Author
-
Alejo Efeyan, Susana Velasco-Miguel, Manuel Serrano, and Manuel Collado
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Cancer Research ,Tumor suppressor gene ,Cell Cycle ,Cancer ,Apoptosis ,Context (language use) ,Cell cycle ,Biology ,medicine.disease ,medicine.disease_cause ,Mice ,In vivo ,Genetics ,medicine ,Cancer research ,Animals ,Tumor Suppressor Protein p53 ,Fibrosarcoma ,Carcinogenesis ,Molecular Biology - Abstract
Protein p21Cip1/Waf1 is transcriptionally activated by the tumour suppressor p53 and previous studies have shown that p21 plays a role in tumour suppression. However, the involvement of p21 in p53-mediated tumour suppression remains to be directly demonstrated in vivo. Tumour suppression mediated by p53 can be measured by comparing tumour susceptibility in animals carrying two (wild-type mice) or three (super-p53 mice) copies of the p53 gene. We have taken advantage of this genetically defined system to measure p53-mediated cell-cycle arrest, apoptosis and tumorigenesis, in a p21 wild-type and in a p21-null context. The absence of p21 significantly impaired the enhanced p53-mediated cell-cycle arrest characteristic of super-p53 cells, but did not affect the enhanced apoptosis. Importantly, in an experimental model of fibrosarcoma induction, the absence of p21 significantly decreased the tumour suppression benefit of super-p53 mice. We conclude that cell-cycle arrest through p21 plays a significant role in mediating p53-dependent cancer protection.
- Published
- 2006
- Full Text
- View/download PDF
44. Increased gene dosage ofInk4a/Arfresults in cancer resistance and normal aging
- Author
-
Alejo Efeyan, Juana M. Flores, Juan Martín-Caballero, Ander Matheu, Peter Klatt, Cristina Pantoja, Manuel Serrano, and Luis M. Criado
- Subjects
Male ,Heterozygote ,Cell Survival ,9,10-Dimethyl-1,2-benzanthracene ,Transgene ,Gene Dosage ,Mice, Transgenic ,Biology ,Gene dosage ,law.invention ,Mice ,law ,Genetics ,Animals ,Humans ,Genes, Tumor Suppressor ,neoplasms ,Gene ,Cells, Cultured ,Cellular Senescence ,Cyclin-Dependent Kinase Inhibitor p16 ,integumentary system ,Homozygote ,Heterozygote advantage ,Neoplasms, Experimental ,Fibroblasts ,Embryo, Mammalian ,Research Papers ,Phenotype ,Molecular biology ,In vitro ,Mice, Inbred C57BL ,Cell Transformation, Neoplastic ,embryonic structures ,Carcinogens ,Cancer research ,Suppressor ,Female ,biological phenomena, cell phenomena, and immunity ,Cell aging ,Developmental Biology - Abstract
Mammalian genes frequently present allelic variants that differ in their expression levels and that, in the case of tumor suppressor genes, can be of relevance for cancer susceptibility and aging. We report here the characterization of a novel mouse model with increased activity for the Ink4a and Arf tumor suppressors. We have generated a “superInk4a/Arf” mouse strain carrying a transgenic copy of the entireInk4a/Arflocus. Cells derived from superInk4a/Arfmice have increased resistance to in vitro immortalization and oncogenic transformation. Importantly, superInk4a/Arfmice manifest higher resistance to cancer compared to normal, nontransgenic, mice. Finally, superInk4a/Arfmice have normal aging and lifespan. Together, these results indicate that modest increases in the activity of theInk4a/Arftumor suppressor result in a beneficial cancer-resistant phenotype without affecting normal viability or aging.
- Published
- 2004
- Full Text
- View/download PDF
45. Establishment of Two Hormone-responsive Mouse Mammary Carcinoma Cell Lines Derived from a Metastatic Mammary Tumor
- Author
-
Alejo Efeyan, Victoria Teresa Fabris, Alfredo A. Molinolo, Susana Merani, and Claudia Lanari
- Subjects
Genetic Markers ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Antineoplastic Agents, Hormonal ,Blotting, Western ,Mammary gland ,Estrogen receptor ,Mammary Neoplasms, Animal ,Medroxyprogesterone Acetate ,Adenocarcinoma ,Biology ,Mice ,Internal medicine ,Progesterone receptor ,Tumor Cells, Cultured ,medicine ,Animals ,Neoplasm Metastasis ,Receptor ,In Situ Hybridization, Fluorescence ,Mice, Inbred BALB C ,Mammary tumor ,Estradiol ,medicine.disease ,Immunohistochemistry ,Molecular biology ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Endocrinology ,Receptors, Estrogen ,Oncology ,Hormone receptor ,Cell culture ,Female ,Receptors, Progesterone - Abstract
We report the establishment of two mouse mammary cancer cell lines, MC7-2A and MC7-2B obtained from a mouse mammary carcinoma induced by medroxyprogesterone acetate (MPA) and maintained by syngeneic transplantation in BALB/c mice. They are epithelial (express cytokeratins) and express both estrogen receptors alpha (ERalpha) and progesterone receptors (PRs) isoforms A and B (western blots). In vitro, MPA inhibited 3H-thymidine uptake, starting from concentrations as low as 10(-13) M in MC7-2A and 10(10) M in MC7-2B; the antiprogestin RU 486 exerted a stimulatory effect at 10(-14) M in both cell lines; 17-beta-estradiol (E2) also exerted a stimulatory effect starting at 10(-10) M in MC7-2A and at 10(-13) M in MC7-2B. When transplanted in syngeneic mice, both cell lines originated adenocarcinomas that gave rise to lung metastases within 3 months. In in vivo studies, in MC7-2A, the antiprogestin inhibited completely tumor growth, E2 induced a slight although significant ( p < 0.05) stimulatory effect and MPA stimulated tumor growth while MC7-2B cells were unresponsive to all treatments. ER and PR were also expressed in tumors as assessed by immunohistochemistry. Two marker chromosomes were identified by FISH as translocations between chromosomes 4 and 7, and between chromosomes X and 2; the third marker chromosome remains unidentified. All these markers were also present in the parental tumor. A new marker, a centric fusion of chromosomes 2, was acquired in both cell lines. Considering that there are very few murine breast carcinoma responsive cell lines, these cells represent new tools in which the regulatory effect of hormones can be studied.
- Published
- 2004
- Full Text
- View/download PDF
46. Isolation of a stromal cell line from an early passage of a mouse mammary tumor line: A model for stromal parenchymal interactions
- Author
-
Marina Simian, Rocío Soldati, Maria Alicia Gorostiaga, Luisa A. Helguero, Alfredo A. Molinolo, Caroline A. Lamb, Norberto Aníbal Sanjuan, Maria Cecilia Bottino, Alejo Efeyan, Victoria Teresa Fabris, and Claudia Lanari
- Subjects
Genetic Markers ,medicine.medical_specialty ,Cell type ,Stromal cell ,Antineoplastic Agents, Hormonal ,Physiology ,Cellular differentiation ,Clinical Biochemistry ,Cell Culture Techniques ,Estrogen receptor ,Breast Neoplasms ,Medroxyprogesterone Acetate ,Biology ,Transforming Growth Factor beta1 ,Mice ,Hormone Antagonists ,Mammary Glands, Animal ,Transforming Growth Factor beta ,Cell Line, Tumor ,Internal medicine ,medicine ,Animals ,Humans ,Receptor ,Cell Shape ,Mice, Inbred BALB C ,Mammary tumor ,Epidermal Growth Factor ,Estradiol ,Cell growth ,Mesenchymal stem cell ,Cell Differentiation ,Cell Biology ,Fibroblasts ,Coculture Techniques ,Mifepristone ,Endocrinology ,Receptors, Estrogen ,Mutation ,Cancer research ,Keratins ,Female ,Stromal Cells ,Tumor Suppressor Protein p53 ,Receptors, Progesterone ,Neoplasm Transplantation - Abstract
We have developed a murine mammary tumor cell line, MC4-L4, and after 15 passages, a spindle-shaped population became evident. The cuboidal cells, MC4-L4E, cloned by limit dilution, proved to be epithelial tumor cells. When inoculated in syngeneic mice, they gave rise to invasive metastatic carcinomas expressing estrogen and progesterone receptors. These tumors regressed after anti-progestin treatment and stopped growing after 17-beta-estradiol administration. In vitro, they were insensitive to medroxyprogesterone acetate (MPA), 17-beta-estradiol, and EGF and were inhibited by TGFbeta1. They expressed mutated p53 and estrogen receptors alpha; progesterone receptors were undetectable. Cells were polyploid and shared the same four common marker chromosomes present in the parental tumor in addition to an exclusive marker. Spindle-shaped cells, MC4-L4F, were selected by differential attachment and detachment and proved to be non-epithelial non-tumorigenic cells. They were cytokeratin negative, showed mesenchymal features by electron microscopy, differentiated to adipocytes when treated with an adipogenic cocktail, were stimulated by TGFbeta1 and EGF, showed a wild-type p53, and did not exhibit the marker chromosomes of the parental tumor. Although they expressed estrogen receptors alpha, they were insensitive to 17-beta-estradiol in proliferation assays. Co-cultures of both cell types had a synergic effect on progesterone receptors expression and on cell proliferation, being the epithelial cells, the most responsive ones, and 17-beta-estradiol increased cell proliferation only in co-cultures. Cytogenetic studies and data on p53 mutations rule out the possibility of an epithelial mesenchymal transition. Their unique characteristics make them an excellent model to be used in studies of epithelial-stromal interactions in the context of hormone responsiveness in hormone related tumors.
- Published
- 2004
- Full Text
- View/download PDF
47. Nutrient Sensing Mechanisms and Pathways
- Author
-
William C. Comb, David M. Sabatini, Alejo Efeyan, Massachusetts Institute of Technology. Department of Biology, Whitehead Institute for Biomedical Research, Koch Institute for Integrative Cancer Research at MIT, Efeyan, Alejo, Comb, William C., and Sabatini, David M.
- Subjects
Multidisciplinary ,Anabolism ,Autophagy ,Nutrient sensing ,Metabolism ,Carbohydrate metabolism ,Biology ,Lipid Metabolism ,Article ,Glucose ,Biochemistry ,Metabolic Diseases ,Extracellular ,Animals ,Homeostasis ,Humans ,Amino Acids ,Intracellular - Abstract
The ability to sense and respond to fluctuations in environmental nutrient levels is a requisite for life. Nutrient scarcity is a selective pressure that has shaped the evolution of most cellular processes. Different pathways that detect intracellular and extracellular levels of sugars, amino acids, lipids and surrogate metabolites are integrated and coordinated at the organismal level through hormonal signals. During food abundance, nutrient-sensing pathways engage anabolism and storage, whereas scarcity triggers homeostatic mechanisms, such as the mobilization of internal stores through autophagy. Nutrient-sensing pathways are commonly deregulated in human metabolic diseases., National Institutes of Health (U.S.) (Grant R01 CA129105), National Institutes of Health (U.S.) (Grant R01 CA103866), National Institutes of Health (U.S.) (Grant R01 AI047389), National Institutes of Health (U.S.) (Grant R21 AG042876), American Federation for Aging Research, Starr Foundation, David H. Koch Institute for Integrative Cancer Research at MIT (Frontier Research Program), Ellison Medical Foundation, Charles A. King Trust, American Cancer Society (Ellison Medical Foundation Postdoctoral Fellowship PF-13-356-01-TBE)
- Published
- 2015
48. Erratum: Corrigendum: Recurrent mTORC1-activating RRAGC mutations in follicular lymphoma
- Author
-
Christopher Mansbridge, Ahad F. Al Seraihi, Julia Richter, Andrew Jack, David M. Sabatini, Jonathan C. Strefford, Lucy Wilkins, Roberto Zoncu, Csaba Bödör, Rachel L. Wolfson, Hilmar Quentmeier, Shamzah Araf, Janet Matthews, T. Andrew Lister, Alejo Efeyan, Jessica Okosun, Sharon Barrans, Ming-Qing Du, Jude Fitzgibbon, Claude Chelala, José Afonso Guerra-Assunção, Andrew Clear, Maria Calaminici, Rebecca C. Auer, John G. Gribben, Sameena Iqbal, Silvia Montoto, Peter Johnson, Brian M. Castellano, Andrew Davies, Reiner Siebert, Jun Wang, Stephan H. Bernhart, Graham Packham, and Leire Escudero-Ibarz
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Section (typography) ,Follicular lymphoma ,Biology ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,Internal medicine ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Genetics ,medicine ,GeneralLiterature_REFERENCE(e.g.,dictionaries,encyclopedias,glossaries) - Abstract
Nat. Genet.; 10.1038/ng.3473; corrected online 12 January 2016 In the version of this article initially published online, several funding sources were omitted from the Acknowledgments section. The error has been corrected for the print, PDF and HTML versions of this article.
- Published
- 2016
- Full Text
- View/download PDF
49. Amino acids and mTORC1: from lysosomes to disease
- Author
-
Roberto Zoncu, Alejo Efeyan, David M. Sabatini, Massachusetts Institute of Technology. Department of Biology, Whitehead Institute for Biomedical Research, Koch Institute for Integrative Cancer Research at MIT, Efeyan, Alejo, Zoncu, Roberto, and Sabatini, David
- Subjects
mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Article ,Mediator ,Lysosome ,Neoplasms ,medicine ,Diabetes Mellitus ,Humans ,Amino Acids ,Molecular Biology ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,biology ,Catabolism ,Cell growth ,TOR Serine-Threonine Kinases ,Neurodegenerative Diseases ,Cell biology ,medicine.anatomical_structure ,Biochemistry ,Multiprotein Complexes ,biology.protein ,Molecular Medicine ,Lysosomes - Abstract
The mechanistic target of rapamycin (mTOR) kinase controls growth and metabolism, and its deregulation underlies the pathogenesis of many diseases, including cancer, neurodegeneration, and diabetes. mTOR complex 1 (mTORC1) integrates signals arising from nutrients, energy, and growth factors, but how exactly these signals are propagated await to be fully understood. Recent findings have placed the lysosome, a key mediator of cellular catabolism, at the core of mTORC1 regulation by amino acids. A multiprotein complex that includes the Rag GTPases, Ragulator, and the v-ATPase forms an amino acid-sensing machinery on the lysosomal surface that affects the decision between cell growth and catabolism at multiple levels. The involvement of a catabolic organelle in growth signaling may have important implications for our understanding of mTORC1-related pathologies., National Institutes of Health (U.S.) (Grants R01 CA129105, R01 CA103866, and R37 AI047389), American Federation for Aging Research, Starr Foundation, David H. Koch Institute for Integrative Cancer Research at MIT. Frontier Research Program, Ellison Medical Foundation, Jane Coffin Childs Memorial Fund for Medical Research (Fellowship), LAM Foundation, Human Frontier Science Program (Strasbourg, France)
- Published
- 2012
50. A minimally invasive assay for individual assessment of the ATM/CHEK2/p53 pathway activity
- Author
-
Simon Bouffler, Ana Ortega-Molina, Sylwia Kabacik, Christophe Badie, Alejo Efeyan, Paul Finnon, and Manuel Serrano
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Genome instability ,T-Lymphocytes ,Gene Dosage ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,medicine.disease_cause ,Gene dosage ,Li-Fraumeni Syndrome ,Ataxia Telangiectasia ,Mice ,Neoplasms ,Proto-Oncogene Proteins ,Puma ,Report ,medicine ,Animals ,Humans ,Molecular Biology ,CHEK2 ,Regulation of gene expression ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Tumor Suppressor Proteins ,Nuclear Proteins ,Cell Biology ,biology.organism_classification ,medicine.disease ,Molecular biology ,DNA-Binding Proteins ,Gene expression profiling ,Checkpoint Kinase 2 ,Gene Expression Regulation ,Ataxia-telangiectasia ,Cancer research ,Disease Susceptibility ,Tumor Suppressor Protein p53 ,Apoptosis Regulatory Proteins ,Carcinogenesis ,Signal Transduction ,Developmental Biology - Abstract
Ionizing radiation induces DNA Double-Strand Breaks (DSBs), which activate the ATM/CHEK2/p53 pathway leading to cell cycle arrest and apoptosis through transcription of genes including CDKN1A (p21) and BBC3 (PUMA). This pathway prevents genomic instability and tumorigenesis as demonstrated in heritable syndromes [e.g., Ataxia Telangiectasia (AT); Li-Fraumeni syndrome (LFS)]. Here, a simple assay based on gene expression in peripheral blood to measure accurately ATM/CHEK2/p53 pathway activity is described. The expression of p21, Puma and Sesn2 was determined in blood from mice with different gene copy numbers of Atm, Trp53 (p53), Chek2 or Arf and in human blood and mitogen stimulated T-lymphocyte (MSTL) cultures from AT, AT carriers, LFS patients and controls, both before and after ex vivo ionizing irradiation. Mouse Atm/Chek2/p53 activity was highly dependent on the copy number of each gene except Arf. In human MSTL, an AT case, AT carriers and LFS patients showed responses distinct from healthy donors. The relationship between gene copy number and transcriptional induction upon radiation was linear for p21 and Puma and correlated well with cancer incidence in p53 variant mice. This reliable blood test provides an assay to determine ATM/CHEK2/p53 pathway activity and demonstrates the feasibility of assessing the activity of this essential cancer protection pathway in simple assays. These findings may have implications for the individualized prediction of cancer susceptibility.
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.