63 results on '"Fajas, L"'
Search Results
2. 57P Deciphering the role of E2F transcription factor-1 in glutamine metabolism
- Author
-
Huber, K., primary, Dreos, R., additional, Geller, S., additional, Barquissau, V., additional, Ziegler, D., additional, Tavernari, D., additional, Giralt, A., additional, Gallart-Ayala, H., additional, Ciriello, G., additional, Ivanisevic, J., additional, Pichler, M., additional, and Fajas, L., additional
- Published
- 2023
- Full Text
- View/download PDF
3. The multifaceted role of cell cycle regulators in the coordination of growth and metabolism
- Author
-
Huber, K., Mestres-Arenas, A., Fajas, L., and Leal-Esteban, L.C.
- Subjects
Adaptation, Physiological ,Animals ,Cell Cycle/drug effects ,Cell Cycle/genetics ,Cells, Cultured ,Cyclin-Dependent Kinase Inhibitor Proteins/genetics ,Cyclin-Dependent Kinase Inhibitor Proteins/metabolism ,Cyclin-Dependent Kinases/antagonists & inhibitors ,Cyclin-Dependent Kinases/genetics ,Cyclin-Dependent Kinases/metabolism ,Cyclins/genetics ,Cyclins/metabolism ,E2F Transcription Factors/genetics ,E2F Transcription Factors/metabolism ,Endosomes/drug effects ,Endosomes/metabolism ,Gene Expression Regulation ,Humans ,Lysosomes/drug effects ,Lysosomes/metabolism ,Mitochondria/drug effects ,Mitochondria/metabolism ,Nutritive Value/physiology ,Protein Kinase Inhibitors/pharmacology ,Retinoblastoma Protein/genetics ,Retinoblastoma Protein/metabolism ,Saccharomyces cerevisiae/drug effects ,Saccharomyces cerevisiae/growth & development ,Saccharomyces cerevisiae/metabolism ,Signal Transduction ,cell cycle regulators ,cell growth ,endolysosomes ,metabolism ,mitochondria - Abstract
Adapting to changes in nutrient availability and environmental conditions is a fundamental property of cells. This adaptation requires a multi-directional coordination between metabolism, growth, and the cell cycle regulators (consisting of the family of cyclin-dependent kinases (CDKs), their regulatory subunits known as cyclins, CDK inhibitors, the retinoblastoma family members, and the E2F transcription factors). Deciphering the mechanisms accountable for this coordination is crucial for understanding various patho-physiological processes. While it is well established that metabolism and growth affect cell division, this review will focus on recent observations that demonstrate how cell cycle regulators coordinate metabolism, cell cycle progression, and growth. We will discuss how the cell cycle regulators directly regulate metabolic enzymes and pathways and summarize their involvement in the endolysosomal pathway and in the functions and dynamics of mitochondria.
- Published
- 2021
4. Cdkn2a deficiency promotes adipose tissue browning
- Author
-
Rabhi, N., Hannou, S.A., Gromada, X., Salas, E., Yao, X., Oger, F., Carney, C., Lopez-Mejia, I.C., Durand, E., Rabearivelo, I., Bonnefond, A., Caron, E., Fajas, L., Dani, C., Froguel, P., and Annicotte, J.S.
- Subjects
Genome-wide association study ,lcsh:Internal medicine ,Adipogenesis ,cdkn2a ,Induced Pluripotent Stem Cells ,Adipocytes, Brown/cytology ,Adipocytes, Brown/metabolism ,Animals ,Cells, Cultured ,Cyclin-Dependent Kinase Inhibitor p16/genetics ,Cyclin-Dependent Kinase Inhibitor p16/metabolism ,Gene Regulatory Networks ,Glucose/metabolism ,Humans ,Induced Pluripotent Stem Cells/cytology ,Induced Pluripotent Stem Cells/metabolism ,Mice ,Obesity/metabolism ,Thermogenesis ,Adipose tissue browning ,Energy expenditure ,Insulin sensitivity ,Obesity ,Type 2 diabetes ,stomatognathic diseases ,Adipocytes, Brown ,Glucose ,Original Article ,lcsh:RC31-1245 ,neoplasms ,Cyclin-Dependent Kinase Inhibitor p16 - Abstract
Objectives Genome-wide association studies have reported that DNA polymorphisms at the CDKN2A locus modulate fasting glucose in human and contribute to type 2 diabetes (T2D) risk. Yet the causal relationship between this gene and defective energy homeostasis remains elusive. Here we sought to understand the contribution of Cdkn2a to metabolic homeostasis. Methods We first analyzed glucose and energy homeostasis from Cdkn2a-deficient mice subjected to normal or high fat diets. Subsequently Cdkn2a-deficient primary adipose cells and human-induced pluripotent stem differentiated into adipocytes were further characterized for their capacity to promote browning of adipose tissue. Finally CDKN2A levels were studied in adipocytes from lean and obese patients. Results We report that Cdkn2a deficiency protects mice against high fat diet-induced obesity, increases energy expenditure and modulates adaptive thermogenesis, in addition to improving insulin sensitivity. Disruption of Cdkn2a associates with increased expression of brown-like/beige fat markers in inguinal adipose tissue and enhances respiration in primary adipose cells. Kinase activity profiling and RNA-sequencing analysis of primary adipose cells further demonstrate that Cdkn2a modulates gene networks involved in energy production and lipid metabolism, through the activation of the Protein Kinase A (PKA), PKG, PPARGC1A and PRDM16 signaling pathways, key regulators of adipocyte beiging. Importantly, CDKN2A expression is increased in adipocytes from obese compared to lean subjects. Moreover silencing CDKN2A expression during human-induced pluripotent stem cells adipogenic differentiation promoted UCP1 expression. Conclusion Our results offer novel insight into brown/beige adipocyte functions, which has recently emerged as an attractive therapeutic strategy for obesity and T2D. Modulating Cdkn2a-regulated signaling cascades may be of interest for the treatment of metabolic disorders., Highlights • Cdkn2a deficiency protects mice against high fat diet-induced obesity. • Cdkn2a modulates brown-like/beige fat gene networks involved in energy production and lipid metabolism. • Increased CDKN2A expression in human obese adipocytes. • Increased UCP1 levels in adipocytes differentiated from CDKN2A-silenced hiPS cells.
- Published
- 2018
5. Cancer: Linking Powerhouses to Suicidal Bags
- Author
-
Martinez-Carreres, L., Nasrallah, A., and Fajas, L.
- Subjects
AMP-activated protein kinase ,Rab7 ,cancer ,lysosomal membrane permeabilization ,lysosomes ,mammalian target of rapamycin ,mitochondria ,mitophagy - Abstract
Membrane-bound organelles are integrated into cellular networks and work together for a common goal: regulating cell metabolism, cell signaling pathways, cell fate, cellular maintenance, and pathogen defense. Many of these interactions are well established, but little is known about the interplay between mitochondria and lysosomes, and their deregulation in cancer. The present review focuses on the common signaling pathways of both organelles, as well as the processes in which they both physically interact, their changes under pathological conditions, and the impact on targeting those organelles for treating cancer.
- Published
- 2017
6. E2F1, a Novel Regulator of Metabolism
- Author
-
Denechaud, P.D., Fajas, L., and Giralt, A.
- Subjects
endocrine system ,E2F1 ,cancer metabolism ,cell cycle regulators ,metabolic diseases ,obesity ,biological phenomena, cell phenomena, and immunity - Abstract
In the past years, several lines of evidence have shown that cell cycle regulatory proteins also can modulate metabolic processes. The transcription factor E2F1 is a central player involved in cell cycle progression, DNA-damage response, and apoptosis. Its crucial role in the control of cell fate has been extensively studied and reviewed before; however, here, we focus on the participation of E2F1 in the regulation of metabolism. We summarize recent findings about the cell cycle-independent roles of E2F1 in various tissues that contribute to global metabolic homeostasis and highlight that E2F1 activity is increased during obesity. Finally, coming back to the pivotal role of E2F1 in cancer development, we discuss how E2F1 links cell cycle progression with different metabolic adaptations required for cell growth and survival.
- Published
- 2017
7. CDK4, a new metabolic sensor that antagonizes AMPK
- Author
-
Fajas, L., primary and Lopez-Mejia, I. C., additional
- Published
- 2018
- Full Text
- View/download PDF
8. Role of cell cycle regulators in adipose tissue and whole body energy homeostasis
- Author
-
Lopez-Mejia, I. C., primary, Castillo-Armengol, J., additional, Lagarrigue, S., additional, and Fajas, L., additional
- Published
- 2017
- Full Text
- View/download PDF
9. CDK4 is an essential insulin effector in adipocytes
- Author
-
Universitat Rovira i Virgili, Lagarrigue S; Lopez-Mejia IC; Denechaud PD; Escoté X; Castillo-Armengol J; Jimenez V; Chavey C; Giralt A; Lai Q; Zhang L; Martinez-Carreres L; Delacuisine B; Annicotte JS; Blanchet E; Huré S; Abella A; Tinahones FJ; Vendrell J; Dubus P; Bosch F; Kahn CR; Fajas L, Universitat Rovira i Virgili, and Lagarrigue S; Lopez-Mejia IC; Denechaud PD; Escoté X; Castillo-Armengol J; Jimenez V; Chavey C; Giralt A; Lai Q; Zhang L; Martinez-Carreres L; Delacuisine B; Annicotte JS; Blanchet E; Huré S; Abella A; Tinahones FJ; Vendrell J; Dubus P; Bosch F; Kahn CR; Fajas L
- Abstract
Insulin resistance is a fundamental pathogenic factor that characterizes various metabolic disorders, including obesity and type 2 diabetes. Adipose tissue contributes to the development of obesity-related insulin resistance through increased release of fatty acids, altered adipokine secretion, and/or macrophage infiltration and cytokine release. Here, we aimed to analyze the participation of the cyclin-dependent kinase 4 (CDK4) in adipose tissue biology. We determined that white adipose tissue (WAT) from CDK4-deficient mice exhibits impaired lipogenesis and increased lipolysis. Conversely, lipolysis was decreased and lipogenesis was increased in mice expressing a mutant hyperactive form of CDK4 (CDK4R24C). A global kinome analysis of CDK4-deficient mice following insulin stimulation revealed that insulin signaling is impaired in these animals. We determined that insulin activates the CCND3-CDK4 complex, which in turn phosphorylates insulin receptor substrate 2 (IRS2) at serine 388, thereby creating a positive feedback loop that maintains adipocyte insulin signaling. Furthermore, we found that CCND3 expression and IRS2 serine 388 phosphorylation are increased in human obese subjects. Together, our results demonstrate that CDK4 is a major regulator of insulin signaling in WAT.
- Published
- 2016
10. O63 Le facteur de transcription E2F1 contrôle la glycolyse et la lipogenèse de novo et contribue au développement de la stéatose hépatique
- Author
-
Denechaud, P.-D., primary, Lopez-Mejia, I., additional, Blanchet, E., additional, Nicolay, B., additional, Dyson, N., additional, Annicotte, J.-S., additional, and Fajas, L., additional
- Published
- 2015
- Full Text
- View/download PDF
11. BCL2L13 at endoplasmic reticulum-mitochondria contact sites regulates calcium homeostasis to maintain skeletal muscle function.
- Author
-
Grepper D, Tabasso C, Zanou N, Aguettaz AKF, Castro-Sepulveda M, Ziegler DV, Lagarrigue S, Arribat Y, Martinotti A, Ebrahimi A, Daraspe J, Fajas L, and Amati F
- Abstract
The physical connection between mitochondria and endoplasmic reticulum (ER) is an essential signaling hub to ensure organelle and cellular functions. In skeletal muscle, ER-mitochondria calcium (Ca
2+ ) signaling is crucial to maintain cellular homeostasis during physical activity. High expression of BCL2L13, a member of the BCL-2 family, was suggested as an adaptive response in endurance-trained human subjects. In adult zebrafish, we found that the loss of Bcl2l13 impairs skeletal muscle structure and function. Ca2+ signaling is altered in Bcl2l13 knockout animals and mitochondrial complexes activity is decreased. Organelle fractioning in mammalian cells shows BCL2L13 at mitochondria, ER, and mitochondria-associated membranes. ER-mitochondria contact sites number is not modified by BCL2L13 modulation, but knockdown of BCL2L13 in C2C12 cells changes cytosolic Ca2+ release and mitochondrial Ca2+ uptake. This suggests that BCL2L13 interaction with mitochondria and ER, and its role in Ca2+ signaling, contributes to proper skeletal muscle function., Competing Interests: The authors have no competing interests., (© 2024 The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
12. CDK4 Phosphorylates AMPKα2 to Inhibit Its Activity and Repress Fatty Acid Oxidation.
- Author
-
Lopez-Mejia IC, Lagarrigue S, Giralt A, Martinez-Carreres L, Martín J, Ortega S, Zanou N, Denechaud PD, Castillo-Armengol J, Chavey C, Orpinell M, Delacuisine B, Nasrallah A, Collodet C, Zhang L, Viollet B, Hardie DG, and Fajas L
- Published
- 2024
- Full Text
- View/download PDF
13. E2F transcription factor-1 modulates expression of glutamine metabolic genes in mouse embryonic fibroblasts and uterine sarcoma cells.
- Author
-
Huber K, Giralt A, Dreos R, Michenthaler H, Geller S, Barquissau V, Ziegler DV, Tavernari D, Gallart-Ayala H, Krajina K, Jonas K, Ciriello G, Ivanisevic J, Prokesch A, Pichler M, and Fajas L
- Subjects
- Animals, Mice, Female, Humans, Sarcoma genetics, Sarcoma metabolism, Sarcoma pathology, Mice, Knockout, Cell Line, Tumor, Cell Proliferation, Promoter Regions, Genetic, E2F1 Transcription Factor metabolism, E2F1 Transcription Factor genetics, Glutamine metabolism, Uterine Neoplasms genetics, Uterine Neoplasms metabolism, Uterine Neoplasms pathology, Fibroblasts metabolism, Gene Expression Regulation, Neoplastic
- Abstract
Metabolic reprogramming is considered as a hallmark of cancer and is clinically exploited as a novel target for therapy. The E2F transcription factor-1 (E2F1) regulates various cellular processes, including proliferative and metabolic pathways, and acts, depending on the cellular and molecular context, as an oncogene or tumor suppressor. The latter is evident by the observation that E2f1-knockout mice develop spontaneous tumors, including uterine sarcomas. This dual role warrants a detailed investigation of how E2F1 loss impacts metabolic pathways related to cancer progression. Our data indicate that E2F1 binds to the promoter of several glutamine metabolism-related genes. Interestingly, the expression of genes in the glutamine metabolic pathway were increased in mouse embryonic fibroblasts (MEFs) lacking E2F1. In addition, we confirm that E2f1
-/- MEFs are more efficient in metabolizing glutamine and producing glutamine-derived precursors for proliferation. Mechanistically, we observe a co-occupancy of E2F1 and MYC on glutamine metabolic promoters, increased MYC binding after E2F1 depletion and that silencing of MYC decreased the expression of glutamine-related genes in E2f1-/- MEFs. Analyses of transcriptomic profiles in 29 different human cancers identified uterine sarcoma that showed a negative correlation between E2F1 and glutamine metabolic genes. CRISPR/Cas9 knockout of E2F1 in the uterine sarcoma cell line SK-UT-1 confirmed elevated glutamine metabolic gene expression, increased proliferation and increased MYC binding to glutamine-related promoters upon E2F1 loss. Together, our data suggest a crucial role of E2F1 in energy metabolism and metabolic adaptation in uterine sarcoma cells., Competing Interests: Declaration of competing interest The authors declare no competing interests, except A.G., who is an employee of Société des Produits Nestlé S.A., (Copyright © 2024 Université de Lausanne. Published by Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
14. Beyond cell cycle regulation: The pleiotropic function of CDK4 in cancer.
- Author
-
Ziegler DV, Parashar K, and Fajas L
- Subjects
- Humans, Cyclin-Dependent Kinase 4 genetics, Cyclin-Dependent Kinase 4 metabolism, Cyclin D metabolism, Phosphorylation, Cell Cycle genetics, Cell Cycle Proteins metabolism, Tumor Microenvironment, Cyclins genetics, Cyclins metabolism, Neoplasms genetics
- Abstract
CDK4, along with its regulatory subunit, cyclin D, drives the transition from G1 to S phase, during which DNA replication and metabolic activation occur. In this canonical pathway, CDK4 is essentially a transcriptional regulator that acts through phosphorylation of retinoblastoma protein (RB) and subsequent activation of the transcription factor E2F, ultimately triggering the expression of genes involved in DNA synthesis and cell cycle progression to S phase. In this review, we focus on the newly reported functions of CDK4, which go beyond direct regulation of the cell cycle. In particular, we describe the extranuclear roles of CDK4, including its roles in the regulation of metabolism, cell fate, cell dynamics and the tumor microenvironment. We describe direct phosphorylation targets of CDK4 and decipher how CDK4 influences these physiological processes in the context of cancer., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
15. β-Cell-Specific E2f1 Deficiency Impairs Glucose Homeostasis, β-Cell Identity, and Insulin Secretion.
- Author
-
Oger F, Bourouh C, Friano ME, Courty E, Rolland L, Gromada X, Moreno M, Carney C, Rabhi N, Durand E, Amanzougarene S, Berberian L, Derhourhi M, Blanc E, Hannou SA, Denechaud PD, Benfodda Z, Meffre P, Fajas L, Kerr-Conte J, Pattou F, Froguel P, Pourcet B, Bonnefond A, Collombat P, and Annicotte JS
- Subjects
- Animals, Humans, Mice, Chromatin metabolism, Glucose metabolism, Histones metabolism, Homeostasis genetics, Insulin metabolism, Insulin Secretion, Mice, Knockout, Diabetes Mellitus, Type 2 genetics, Diabetes Mellitus, Type 2 metabolism, Insulin-Secreting Cells metabolism
- Abstract
The loss of pancreatic β-cell identity has emerged as an important feature of type 2 diabetes development, but the molecular mechanisms are still elusive. Here, we explore the cell-autonomous role of the cell-cycle regulator and transcription factor E2F1 in the maintenance of β-cell identity, insulin secretion, and glucose homeostasis. We show that the β-cell-specific loss of E2f1 function in mice triggers glucose intolerance associated with defective insulin secretion, altered endocrine cell mass, downregulation of many β-cell genes, and concomitant increase of non-β-cell markers. Mechanistically, epigenomic profiling of the promoters of these non-β-cell upregulated genes identified an enrichment of bivalent H3K4me3/H3K27me3 or H3K27me3 marks. Conversely, promoters of downregulated genes were enriched in active chromatin H3K4me3 and H3K27ac histone marks. We find that specific E2f1 transcriptional, cistromic, and epigenomic signatures are associated with these β-cell dysfunctions, with E2F1 directly regulating several β-cell genes at the chromatin level. Finally, the pharmacological inhibition of E2F transcriptional activity in human islets also impairs insulin secretion and the expression of β-cell identity genes. Our data suggest that E2F1 is critical for maintaining β-cell identity and function through sustained control of β-cell and non-β-cell transcriptional programs., Article Highlights: β-Cell-specific E2f1 deficiency in mice impairs glucose tolerance. Loss of E2f1 function alters the ratio of α- to β-cells but does not trigger β-cell conversion into α-cells. Pharmacological inhibition of E2F activity inhibits glucose-stimulated insulin secretion and alters β- and α-cell gene expression in human islets. E2F1 maintains β-cell function and identity through control of transcriptomic and epigenetic programs., (© 2023 by the American Diabetes Association.)
- Published
- 2023
- Full Text
- View/download PDF
16. CDK4 is an essential insulin effector in adipocytes.
- Author
-
Lagarrigue S, Lopez-Mejia IC, Denechaud PD, Escoté X, Castillo-Armengol J, Jimenez V, Chavey C, Giralt A, Lai Q, Zhang L, Martinez-Carreres L, Delacuisine B, Annicotte JS, Blanchet E, Huré S, Abella A, Tinahones FJ, Vendrell J, Dubus P, Bosch F, Kahn CR, and Fajas L
- Published
- 2023
- Full Text
- View/download PDF
17. Hepatic lipid overload triggers biliary epithelial cell activation via E2Fs.
- Author
-
Yildiz E, El Alam G, Perino A, Jalil A, Denechaud PD, Huber K, Fajas L, Auwerx J, Sorrentino G, and Schoonjans K
- Subjects
- Animals, Mice, Liver metabolism, Epithelial Cells metabolism, Cell Division, Lipids, Non-alcoholic Fatty Liver Disease metabolism
- Abstract
During severe or chronic hepatic injury, biliary epithelial cells (BECs) undergo rapid activation into proliferating progenitors, a crucial step required to establish a regenerative process known as ductular reaction (DR). While DR is a hallmark of chronic liver diseases, including advanced stages of non-alcoholic fatty liver disease (NAFLD), the early events underlying BEC activation are largely unknown. Here, we demonstrate that BECs readily accumulate lipids during high-fat diet feeding in mice and upon fatty acid treatment in BEC-derived organoids. Lipid overload induces metabolic rewiring to support the conversion of adult cholangiocytes into reactive BECs. Mechanistically, we found that lipid overload activates the E2F transcription factors in BECs, which drive cell cycle progression while promoting glycolytic metabolism. These findings demonstrate that fat overload is sufficient to reprogram BECs into progenitor cells in the early stages of NAFLD and provide new insights into the mechanistic basis of this process, revealing unexpected connections between lipid metabolism, stemness, and regeneration., Competing Interests: EY, GE, AP, AJ, PD, KH, LF, JA, GS, KS No competing interests declared, (© 2023, Yildiz et al.)
- Published
- 2023
- Full Text
- View/download PDF
18. Oxidative stress-induced FAK activation contributes to uterine serous carcinoma aggressiveness.
- Author
-
Lopez-Mejia IC, Pijuan J, Navaridas R, Santacana M, Gatius S, Velasco A, Castellà G, Panosa A, Cabiscol E, Pinyol M, Coll L, Bonifaci N, Peña LP, Vidal A, Villanueva A, Gari E, Llobet-Navàs D, Fajas L, Matias-Guiu X, and Yeramian A
- Subjects
- Humans, Cell Line, Tumor, Cell Movement, Oxidative Stress, Phosphorylation, Reactive Oxygen Species, Tyrosine metabolism, Animals, Antioxidants metabolism, Cystadenocarcinoma, Serous drug therapy, Cystadenocarcinoma, Serous pathology, Focal Adhesion Kinase 1 metabolism
- Abstract
Uterine serous carcinoma (USC) is an aggressive form of endometrial cancer (EC), characterized by its high propensity for metastases. In fact, while endometrioid endometrial carcinoma (EEC), which accounts for 85% of EC, presents a good prognosis, USC is the most frequently fatal. Herein, we used for the first time a peptide-based tyrosine-kinase-activity profiling approach to quantify the changes in tyrosine kinase activation between USC and EEC. Among the tyrosine kinases highly activated in USC, we identified focal adhesion kinase (FAK). We conducted mechanistic studies using cellular models. In a USC cell line, targeting FAK either by inhibitors PF-573228 and defactinib (VS-6063) or by gene silencing limits 3D cell growth and reduces cell migration. Moreover, results from our studies suggest that oxidative stress is increased in USC tumors compared to EEC ones. Reactive oxygen species (ROS) induce tyrosine phosphorylation of FAK and a concomitant tyrosine phosphorylation of paxillin, a mediator of FAK signal transduction. Mechanistically, by tracking hundreds of individual cells per condition, we show that ROS increased cell distance and migration velocity, highlighting the role of ROS-FAK-PAX signaling in cell migration. Both defactinib and ROS scavenger N-acetylcysteine (NAC) revert this effect, pointing toward ROS as potential culprits for the increase in USC cell motility. A proof of concept of the role of FAK in controlling cell growth was obtained in in vivo experiments using cancer-tissue-originated spheroids (CTOS) and a patient-derived orthotopic xenograft model (orthoxenograft/PDOX). Defactinib reduces cell proliferation and protein oxidation, supporting a pro-tumoral antioxidant role of FAK, whereas antioxidant NAC reverts FAK inhibitor effects. Overall, our data points to ROS-mediated FAK activation in USC as being responsible for the poor prognosis of this tumor type and emphasize the potential of FAK inhibition for USC treatment., (© 2022 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2023
- Full Text
- View/download PDF
19. CDK4 is an essential insulin effector in adipocytes.
- Author
-
Lagarrigue S, Lopez-Mejia IC, Denechaud PD, Escoté X, Castillo-Armengol J, Jimenez V, Chavey C, Giralt A, Lai Q, Zhang L, Martinez-Carreres L, Delacuisine B, Annicotte JS, Blanchet E, Huré S, Abella A, Tinahones FJ, Vendrell J, Dubus P, Bosch F, Kahn CR, and Fajas L
- Published
- 2022
- Full Text
- View/download PDF
20. Sex-Biased Control of Inflammation and Metabolism by a Mitochondrial Nod-Like Receptor.
- Author
-
Snäkä T, Bekkar A, Desponds C, Prével F, Claudinot S, Isorce N, Teixeira F, Grasset C, Xenarios I, Lopez-Mejia IC, Fajas L, and Fasel N
- Subjects
- Animals, Female, Hormones metabolism, Inflammation metabolism, Male, Mice, Mitochondria metabolism, Mitochondrial Proteins metabolism, NLR Proteins metabolism
- Abstract
Mitochondria regulate steroid hormone synthesis, and in turn sex hormones regulate mitochondrial function for maintaining cellular homeostasis and controlling inflammation. This crosstalk can explain sex differences observed in several pathologies such as in metabolic or inflammatory disorders. Nod-like receptor X1 (NLRX1) is a mitochondria-associated innate receptor that could modulate metabolic functions and attenuates inflammatory responses. Here, we showed that in an infectious model with the human protozoan parasite, Leishmania guyanensis , NLRX1 attenuated inflammation in females but not in male mice. Analysis of infected female and male bone marrow derived macrophages showed both sex- and genotype-specific differences in both inflammatory and metabolic profiles with increased type I interferon production, mitochondrial respiration, and glycolytic rate in Nlrx1 -deficient female BMDMs in comparison to wild-type cells, while no differences were observed between males. Transcriptomics of female and male BMDMs revealed an altered steroid hormone signaling in Nlrx1 -deficient cells, and a "masculinization" of Nlrx1 -deficient female BMDMs. Thus, our findings suggest that NLRX1 prevents uncontrolled inflammation and metabolism in females and therefore may contribute to the sex differences observed in infectious and inflammatory diseases., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Snäkä, Bekkar, Desponds, Prével, Claudinot, Isorce, Teixeira, Grasset, Xenarios, Lopez-Mejia, Fajas and Fasel.)
- Published
- 2022
- Full Text
- View/download PDF
21. Glucose Starvation or Pyruvate Dehydrogenase Activation Induce a Broad, ERK5-Mediated, Metabolic Remodeling Leading to Fatty Acid Oxidation.
- Author
-
Khan AUH, Salehi H, Alexia C, Valdivielso JM, Bozic M, Lopez-Mejia IC, Fajas L, Gerbal-Chaloin S, Daujat-Chavanieu M, Gitenay D, and Villalba M
- Subjects
- Fatty Acids metabolism, Oxidation-Reduction, Oxidoreductases metabolism, Pyruvates, Glucose metabolism, Mitogen-Activated Protein Kinase 7 metabolism
- Abstract
Cells have metabolic flexibility that allows them to adapt to changes in substrate availability. Two highly relevant metabolites are glucose and fatty acids (FA), and hence, glycolysis and fatty acid oxidation (FAO) are key metabolic pathways leading to energy production. Both pathways affect each other, and in the absence of one substrate, metabolic flexibility allows cells to maintain sufficient energy production. Here, we show that glucose starvation or sustained pyruvate dehydrogenase (PDH) activation by dichloroacetate (DCA) induce large genetic remodeling to propel FAO. The extracellular signal-regulated kinase 5 (ERK5) is a key effector of this multistep metabolic remodeling. First, there is an increase in the lipid transport by expression of low-density lipoprotein receptor-related proteins (LRP), e.g., CD36, LRP1 and others. Second, an increase in the expression of members of the acyl-CoA synthetase long-chain (ACSL) family activates FA. Finally, the expression of the enzymes that catalyze the initial step in each cycle of FAO, i.e., the acyl-CoA dehydrogenases (ACADs), is induced. All of these pathways lead to enhanced cellular FAO. In summary, we show here that different families of enzymes, which are essential to perform FAO, are regulated by the signaling pathway, i.e., MEK5/ERK5, which transduces changes from the environment to genetic adaptations.
- Published
- 2022
- Full Text
- View/download PDF
22. The Intricate Interplay between Cell Cycle Regulators and Autophagy in Cancer.
- Author
-
Ziegler DV, Huber K, and Fajas L
- Abstract
In the past decade, cell cycle regulators have extended their canonical role in cell cycle progression to the regulation of various cellular processes, including cellular metabolism. The regulation of metabolism is intimately connected with the function of autophagy, a catabolic process that promotes the efficient recycling of endogenous components from both extrinsic stress, e.g., nutrient deprivation, and intrinsic sub-lethal damage. Mediating cellular homeostasis and cytoprotection, autophagy is found to be dysregulated in numerous pathophysiological contexts, such as cancer. As an adaptative advantage, the upregulation of autophagy allows tumor cells to integrate stress signals, escaping multiple cell death mechanisms. Nevertheless, the precise role of autophagy during tumor development and progression remains highly context-dependent. Recently, multiple articles has suggested the importance of various cell cycle regulators in the modulation of autophagic processes. Here, we review the current clues indicating that cell-cycle regulators, including cyclin-dependent kinase inhibitors (CKIs), cyclin-dependent kinases (CDKs), and E2F transcription factors, are intrinsically linked to the regulation of autophagy. As an increasing number of studies highlight the importance of autophagy in cancer progression, we finally evoke new perspectives in therapeutic avenues that may include both cell cycle inhibitors and autophagy modulators to synergize antitumor efficacy.
- Published
- 2021
- Full Text
- View/download PDF
23. The multifunctional protein E4F1 links P53 to lipid metabolism in adipocytes.
- Author
-
Lacroix M, Linares LK, Rueda-Rincon N, Bloch K, Di Michele M, De Blasio C, Fau C, Gayte L, Blanchet E, Mairal A, Derua R, Cardona F, Beuzelin D, Annicotte JS, Pirot N, Torro A, Tinahones FJ, Bernex F, Bertrand-Michel J, Langin D, Fajas L, Swinnen JV, and Le Cam L
- Subjects
- Adipocytes pathology, Adipose Tissue pathology, Adult, Aged, Animals, Body Mass Index, Fatty Acids, Monounsaturated metabolism, Female, Gene Expression Regulation, Humans, Insulin Resistance, Lipid Metabolism genetics, Male, Mice, Mice, Knockout, Middle Aged, Obesity metabolism, Obesity pathology, Repressor Proteins deficiency, Repressor Proteins metabolism, Signal Transduction, Stearoyl-CoA Desaturase metabolism, Tumor Suppressor Protein p53 metabolism, Ubiquitin-Protein Ligases deficiency, Ubiquitin-Protein Ligases metabolism, Adipocytes metabolism, Adipose Tissue metabolism, Obesity genetics, Repressor Proteins genetics, Stearoyl-CoA Desaturase genetics, Tumor Suppressor Protein p53 genetics, Ubiquitin-Protein Ligases genetics
- Abstract
Growing evidence supports the importance of the p53 tumor suppressor in metabolism but the mechanisms underlying p53-mediated control of metabolism remain poorly understood. Here, we identify the multifunctional E4F1 protein as a key regulator of p53 metabolic functions in adipocytes. While E4F1 expression is upregulated during obesity, E4f1 inactivation in mouse adipose tissue results in a lean phenotype associated with insulin resistance and protection against induced obesity. Adipocytes lacking E4F1 activate a p53-dependent transcriptional program involved in lipid metabolism. The direct interaction between E4F1 and p53 and their co-recruitment to the Steaoryl-CoA Desaturase-1 locus play an important role to regulate monounsaturated fatty acids synthesis in adipocytes. Consistent with the role of this E4F1-p53-Steaoryl-CoA Desaturase-1 axis in adipocytes, p53 inactivation or diet complementation with oleate partly restore adiposity and improve insulin sensitivity in E4F1-deficient mice. Altogether, our findings identify a crosstalk between E4F1 and p53 in the control of lipid metabolism in adipocytes that is relevant to obesity and insulin resistance., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
24. Enforced PGC-1α expression promotes CD8 T cell fitness, memory formation and antitumor immunity.
- Author
-
Dumauthioz N, Tschumi B, Wenes M, Marti B, Wang H, Franco F, Li W, Lopez-Mejia IC, Fajas L, Ho PC, Donda A, Romero P, and Zhang L
- Subjects
- Animals, CD8-Positive T-Lymphocytes metabolism, Mice, Mitochondria metabolism, Organelle Biogenesis, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha metabolism, Vaccines, Subunit, Cancer Vaccines
- Abstract
Memory CD8 T cells can provide long-term protection against tumors, which depends on their enhanced proliferative capacity, self-renewal and unique metabolic rewiring to sustain cellular fitness. Specifically, memory CD8 T cells engage oxidative phosphorylation and fatty acid oxidation to fulfill their metabolic demands. In contrast, tumor-infiltrating lymphocytes (TILs) display severe metabolic defects, which may underlie their functional decline. Here, we show that overexpression of proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), the master regulator of mitochondrial biogenesis (MB), favors CD8 T cell central memory formation rather than resident memory generation. PGC-1α-overexpressing CD8 T cells persist and mediate more robust recall responses to bacterial infection or peptide vaccination. Importantly, CD8 T cells with enhanced PGC-1α expression provide stronger antitumor immunity in a mouse melanoma model. Moreover, TILs overexpressing PGC-1α maintain higher mitochondrial activity and improved expansion when rechallenged in a tumor-free host. Altogether, our findings indicate that enforcing mitochondrial biogenesis promotes CD8 T cell memory formation, metabolic fitness, and antitumor immunity in vivo.
- Published
- 2021
- Full Text
- View/download PDF
25. The multifaceted role of cell cycle regulators in the coordination of growth and metabolism.
- Author
-
Huber K, Mestres-Arenas A, Fajas L, and Leal-Esteban LC
- Subjects
- Adaptation, Physiological, Animals, Cell Cycle drug effects, Cells, Cultured, Cyclin-Dependent Kinase Inhibitor Proteins metabolism, Cyclin-Dependent Kinases antagonists & inhibitors, Cyclin-Dependent Kinases metabolism, Cyclins metabolism, E2F Transcription Factors genetics, E2F Transcription Factors metabolism, Endosomes drug effects, Endosomes metabolism, Gene Expression Regulation, Humans, Lysosomes drug effects, Lysosomes metabolism, Mitochondria drug effects, Mitochondria metabolism, Protein Kinase Inhibitors pharmacology, Retinoblastoma Protein genetics, Retinoblastoma Protein metabolism, Saccharomyces cerevisiae drug effects, Saccharomyces cerevisiae growth & development, Saccharomyces cerevisiae metabolism, Signal Transduction, Cell Cycle genetics, Cyclin-Dependent Kinase Inhibitor Proteins genetics, Cyclin-Dependent Kinases genetics, Cyclins genetics, Nutritive Value physiology
- Abstract
Adapting to changes in nutrient availability and environmental conditions is a fundamental property of cells. This adaptation requires a multi-directional coordination between metabolism, growth, and the cell cycle regulators (consisting of the family of cyclin-dependent kinases (CDKs), their regulatory subunits known as cyclins, CDK inhibitors, the retinoblastoma family members, and the E2F transcription factors). Deciphering the mechanisms accountable for this coordination is crucial for understanding various patho-physiological processes. While it is well established that metabolism and growth affect cell division, this review will focus on recent observations that demonstrate how cell cycle regulators coordinate metabolism, cell cycle progression, and growth. We will discuss how the cell cycle regulators directly regulate metabolic enzymes and pathways and summarize their involvement in the endolysosomal pathway and in the functions and dynamics of mitochondria., (© 2020 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2021
- Full Text
- View/download PDF
26. PamgeneAnalyzeR: open and reproducible pipeline for kinase profiling.
- Author
-
Bekkar A, Nasrallah A, Guex N, Fajas L, Xenarios I, and Lopez-Mejia IC
- Subjects
- Microarray Analysis, Phosphorylation, Protein Processing, Post-Translational, Software, Protein Kinases genetics, Protein Kinases metabolism, Proteome
- Abstract
Protein phosphorylation--catalyzed by protein kinases-is the most common post-translational modification. It increases the functional diversity of the proteome and influences various aspects of normal physiology and can be altered in disease states. High throughput profiling of kinases is becoming an essential experimental approach to investigate their activity and this can be achieved using technologies such as PamChip® arrays provided by PamGene for kinase activity measurement. Here, we present 'pamgeneAnalyzeR', an R package developed as an alternative to the manual steps necessary to extract the data from PamChip® peptide microarrays images in a reproducible and robust manner. The extracted data can be directly used for downstream analysis., Availability and Implementation: PamgeneAnalyzeR is implemented in R and can be obtained from https://github.com/amelbek/pamgeneAnalyzeR., (© The Author(s) 2020. Published by Oxford University Press.)
- Published
- 2020
- Full Text
- View/download PDF
27. Tumor regression and resistance mechanisms upon CDK4 and RAF1 inactivation in KRAS/P53 mutant lung adenocarcinomas.
- Author
-
Esteban-Burgos L, Wang H, Nieto P, Zheng J, Blanco-Aparicio C, Varela C, Gómez-López G, Fernández-García F, Sanclemente M, Guerra C, Drosten M, Galán J, Caleiras E, Martínez-Torrecuadrada J, Fajas L, Peng SB, Santamaría D, Musteanu M, and Barbacid M
- Subjects
- Adenocarcinoma of Lung drug therapy, Adenocarcinoma of Lung metabolism, Adenocarcinoma of Lung pathology, Animals, Antineoplastic Agents administration & dosage, Cell Line, Tumor, Cyclin-Dependent Kinase 4 metabolism, Disease Progression, Drug Resistance, Neoplasm, Gene Silencing, Humans, Lung Neoplasms drug therapy, Lung Neoplasms metabolism, Lung Neoplasms pathology, Mice, Mice, Inbred C57BL, Mutation, Proto-Oncogene Proteins c-raf metabolism, Proto-Oncogene Proteins p21(ras) metabolism, Tumor Suppressor Protein p53 genetics, Adenocarcinoma of Lung genetics, Cyclin-Dependent Kinase 4 genetics, Lung Neoplasms genetics, Proto-Oncogene Proteins c-raf genetics, Proto-Oncogene Proteins p21(ras) genetics, Tumor Suppressor Protein p53 metabolism
- Abstract
KRAS mutant lung adenocarcinomas remain intractable for targeted therapies. Genetic interrogation of KRAS downstream effectors, including the MAPK pathway and the interphase CDKs, identified CDK4 and RAF1 as the only targets whose genetic inactivation induces therapeutic responses without causing unacceptable toxicities. Concomitant CDK4 inactivation and RAF1 ablation prevented tumor progression and induced complete regression in 25% of KRAS/p53-driven advanced lung tumors, yet a significant percentage of those tumors that underwent partial regression retained a population of CDK4/RAF1-resistant cells. Characterization of these cells revealed two independent resistance mechanisms implicating hypermethylation of several tumor suppressors and increased PI3K activity. Importantly, these CDK4/RAF1-resistant cells can be pharmacologically controlled. These studies open the door to new therapeutic strategies to treat KRAS mutant lung cancer, including resistant tumors., Competing Interests: Competing interest statement: S.-B.P. is an employee at the Oncology Research at Eli Lilly and Company. M.B. reports a research contract with Oncology Research, Eli Lilly and Company.
- Published
- 2020
- Full Text
- View/download PDF
28. Hypothalamic CDK4 regulates thermogenesis by modulating sympathetic innervation of adipose tissues.
- Author
-
Castillo-Armengol J, Barquissau V, Geller S, Ji H, Severi I, Venema W, Fenandez EA, Moret C, Huber K, Leal-Esteban LC, Nasrallah A, Martinez-Carreres L, Niederhäuser G, Seoane-Collazo P, Lagarrigue S, López M, Giordano A, Croizier S, Thorens B, Lopez-Mejia IC, and Fajas L
- Subjects
- Adipocytes, Brown, Adipose Tissue, Brown, Animals, Hypothalamus, Mice, Adipose Tissue, White, Thermogenesis genetics
- Abstract
This study investigated the role of CDK4 in the oxidative metabolism of brown adipose tissue (BAT). BAT from Cdk4
-/- mice exhibited fewer lipids and increased mitochondrial volume and expression of canonical thermogenic genes, rendering these mice more resistant to cold exposure. Interestingly, these effects were not BAT cell-autonomous but rather driven by increased sympathetic innervation. In particular, the ventromedial hypothalamus (VMH) is known to modulate BAT activation via the sympathetic nervous system. We thus examined the effects of VMH neuron-specific Cdk4 deletion. These mice display increased sympathetic innervation and enhanced cold tolerance, similar to Cdk4-/- mice, in addition to browning of scWAT. Overall, we provide evidence showing that CDK4 modulates thermogenesis by regulating sympathetic innervation of adipose tissue depots through hypothalamic nuclei, including the VMH. This demonstrates that CDK4 not only negatively regulates oxidative pathways, but also modulates the central regulation of metabolism through its action in the brain., (© 2020 The Authors. Published under the terms of the CC BY 4.0 license.)- Published
- 2020
- Full Text
- View/download PDF
29. CDK7 Mediates the Beta-Adrenergic Signaling in Thermogenic Brown and White Adipose Tissues.
- Author
-
Ji H, Chen Y, Castillo-Armengol J, Dreos R, Moret C, Niederhäuser G, Delacuisine B, Lopez-Mejia IC, Denechaud PD, and Fajas L
- Abstract
Cyclin-dependent kinases (CDKs) are emerging regulators of adipose tissue metabolism. Here we aimed to explore the role of CDK7 in thermogenic fat. We found that CDK7 brown adipose tissue (BAT)-specific knockout mice (Cdk7
bKO ) have decreased BAT mass and impaired β3-adrenergic signaling and develop hypothermia upon cold exposure. We found that loss of CDK7 in BAT disrupts the induction of thermogenic genes in response to cold. However, Cdk7bKO mice do not show systemic metabolic dysfunction. Increased expression of genes of the creatine metabolism compensates for the heat generation in the BAT of Cdk7bKO mice in response to cold. Finally, we show that CDK7 is required for beta 3-adrenergic agonist-induced browning of white adipose tissue (WAT). Indeed, Cdk7 ablation in all adipose tissues (Cdk7aKO ) has impaired browning in WAT. Together, our results demonstrate that CDK7 is an important mediator of beta-adrenergic signaling in thermogenic brown and beige fat., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
30. Cell cycle regulators in cancer cell metabolism.
- Author
-
Leal-Esteban LC and Fajas L
- Subjects
- Animals, Carcinogenesis drug effects, Carcinogenesis genetics, Carcinogenesis pathology, Cell Cycle drug effects, Cell Cycle genetics, Cell Line, Tumor, Cyclin-Dependent Kinase Inhibitor Proteins genetics, Cyclin-Dependent Kinases antagonists & inhibitors, Cyclin-Dependent Kinases genetics, Cyclins genetics, Disease Models, Animal, Disease Progression, Energy Metabolism drug effects, Energy Metabolism physiology, Gene Expression Regulation, Neoplastic drug effects, Humans, Mitochondria drug effects, Mitochondria metabolism, Mutation, Neoplasms drug therapy, Neoplasms genetics, Protein Kinase Inhibitors pharmacology, Cyclin-Dependent Kinase Inhibitor Proteins metabolism, Cyclin-Dependent Kinases metabolism, Cyclins metabolism, Neoplasms pathology, Protein Kinase Inhibitors therapeutic use
- Abstract
Cancer proliferation and progression involves altered metabolic pathways as a result of continuous demand for energy and nutrients. In the last years, cell cycle regulators have been involved in the control of metabolic processes, such as glucose and insulin pathways and lipid synthesis, in addition to their canonical function controlling cell cycle progression. Here we describe recent data demonstrating the role of cell cycle regulators in the metabolic control especially in studies performed in cancer models. Moreover, we discuss the importance of these findings in the context of current cancer therapies to provide an overview of the relevance of targeting metabolism using inhibitors of the cell cycle regulation., Competing Interests: Declaration of competing interest The authors have declared that no competing interests exist., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
31. CDK4 Regulates Lysosomal Function and mTORC1 Activation to Promote Cancer Cell Survival.
- Author
-
Martínez-Carreres L, Puyal J, Leal-Esteban LC, Orpinell M, Castillo-Armengol J, Giralt A, Dergai O, Moret C, Barquissau V, Nasrallah A, Pabois A, Zhang L, Romero P, Lopez-Mejia IC, and Fajas L
- Subjects
- Adenylate Kinase metabolism, Aminopyridines pharmacology, Aminopyridines therapeutic use, Animals, Autophagosomes physiology, Autophagy physiology, Benzimidazoles pharmacology, Benzimidazoles therapeutic use, Biphenyl Compounds, Cell Line, Tumor, Cellular Senescence physiology, Cyclin-Dependent Kinase 4 antagonists & inhibitors, Cyclin-Dependent Kinase 4 genetics, Drug Synergism, Female, Gene Knockout Techniques, Humans, Insulin physiology, Lysosomes ultrastructure, Mice, Mice, Inbred NOD, Molecular Targeted Therapy, Phosphorylation, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Protein Processing, Post-Translational, Protein Transport, Proto-Oncogene Proteins metabolism, Pyrones pharmacology, Pyrones therapeutic use, Recombinant Fusion Proteins metabolism, Signal Transduction physiology, Thiophenes pharmacology, Thiophenes therapeutic use, Tumor Suppressor Proteins metabolism, Xenograft Model Antitumor Assays, Cyclin-Dependent Kinase 4 physiology, Lysosomes physiology, Mechanistic Target of Rapamycin Complex 1 metabolism, Neoplasm Proteins physiology
- Abstract
Cyclin-dependent kinase 4 (CDK4) is well-known for its role in regulating the cell cycle, however, its role in cancer metabolism, especially mTOR signaling, is undefined. In this study, we established a connection between CDK4 and lysosomes, an emerging metabolic organelle crucial for mTORC1 activation. On the one hand, CDK4 phosphorylated the tumor suppressor folliculin (FLCN), regulating mTORC1 recruitment to the lysosomal surface in response to amino acids. On the other hand, CDK4 directly regulated lysosomal function and was essential for lysosomal degradation, ultimately regulating mTORC1 activity. Pharmacologic inhibition or genetic inactivation of CDK4, other than retaining FLCN at the lysosomal surface, led to the accumulation of undigested material inside lysosomes, which impaired the autophagic flux and induced cancer cell senescence in vitro and in xenograft models. Importantly, the use of CDK4 inhibitors in therapy is known to cause senescence but not cell death. To overcome this phenomenon and based on our findings, we increased the autophagic flux in cancer cells by using an AMPK activator in combination with a CDK4 inhibitor. The cotreatment induced autophagy (AMPK activation) and impaired lysosomal function (CDK4 inhibition), resulting in cell death and tumor regression. Altogether, we uncovered a previously unknown role for CDK4 in lysosomal biology and propose a novel therapeutic strategy to target cancer cells. SIGNIFICANCE: These findings uncover a novel function of CDK4 in lysosomal biology, which promotes cancer progression by activating mTORC1; targeting this function offers a new therapeutic strategy for cancer treatment., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
32. Inter-organ communication: a gatekeeper for metabolic health.
- Author
-
Castillo-Armengol J, Fajas L, and Lopez-Mejia IC
- Subjects
- Animals, Diabetes Mellitus, Type 2 metabolism, Energy Metabolism genetics, Homeostasis genetics, Humans, Models, Biological, Energy Metabolism physiology, Homeostasis physiology, Obesity metabolism
- Abstract
Multidirectional interactions between metabolic organs in the periphery and the central nervous system have evolved concomitantly with multicellular organisms to maintain whole-body energy homeostasis and ensure the organism's adaptation to external cues. These interactions are altered in pathological conditions such as obesity and type 2 diabetes. Bioactive peptides and proteins, such as hormones and cytokines, produced by both peripheral organs and the central nervous system, are key messengers in this inter-organ communication. Despite the early discovery of the first hormones more than 100 years ago, recent studies taking advantage of novel technologies have shed light on the multiple ways used by cells in the body to communicate and maintain energy balance. This review briefly summarizes well-established concepts and focuses on recent advances describing how specific proteins and peptides mediate the crosstalk between gut, brain, and other peripheral metabolic organs in order to maintain energy homeostasis. Additionally, this review outlines how the improved knowledge about these inter-organ networks is helping us to redefine therapeutic strategies in an effort to promote healthy living and fight metabolic disorders and other diseases., (© 2019 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2019
- Full Text
- View/download PDF
33. Human adipose tissue H3K4me3 histone mark in adipogenic, lipid metabolism and inflammatory genes is positively associated with BMI and HOMA-IR.
- Author
-
Castellano-Castillo D, Denechaud PD, Fajas L, Moreno-Indias I, Oliva-Olivera W, Tinahones F, Queipo-Ortuño MI, and Cardona F
- Subjects
- Adipogenesis, Adipose Tissue metabolism, Adult, Chromatin genetics, DNA Methylation, Epigenesis, Genetic, Female, Humans, Inflammation Mediators metabolism, Male, Middle Aged, Promoter Regions, Genetic, Adipose Tissue pathology, Body Mass Index, Gene Expression Regulation, Histones genetics, Inflammation genetics, Insulin Resistance, Lipid Metabolism genetics
- Abstract
Introduction: Adipose tissue is considered an important metabolic tissue, in charge of energy storage as well as being able to act in systemic homeostasis and inflammation. Epigenetics involves a series of factors that are important for gene regulation or for chromatin structure, mostly DNA methylation and histone-tail modifications, which can be modified by environmental conditions (nutrition, lifestyle, smoking…). Since metabolic diseases like obesity and diabetes are closely related to lifestyle and nutrition, epigenetic deregulation could play an important role in the onset of these diseases and vice versa. However, little is known about histone marks in human adipose tissue. In a previous work, we developed a protocol for chromatin immunoprecipitation (ChIP) of frozen human adipose tissue. By using this method, this study investigates, for the first time, the H3K4 trimethylation (H3K4me3) mark (open chromatin) on the promoter of several factors involved in adipogenesis, lipid metabolism and inflammation in visceral adipose tissue (VAT) from human subjects with different degrees of body mass index (BMI) and metabolic disease., Methodology: VAT was collected and frozen at -80°C. 100 mg VAT samples were fixed in 0.5% formaldehyde and homogenized. After sonication, the sheared chromatin was immune-precipitated with an anti-H3K4me3 antibody linked to magnetic beads and purified. H3K4me3 enrichment was analyzed by qPCR for LEP, LPL, SREBF2, SCD1, PPARG, IL6, TNF and E2F1 promoters. mRNA extraction on the same samples was performed to quantify gene expression of these genes., Results: H3K4me3 was enriched at the promoter of E2F1, LPL, SREBF2, SCD1, PPARG and IL6 in lean normoglycemic compared to morbid obese subjects with prediabetes. Accordingly H3K4me3 mark enrichment at E2F1, LPL, SREBF2, SCD1, PPARG and IL6 promoters was positively correlated with the BMI and the HOMA-IR. Regression analysis showed a strong relationship between the BMI with H3K4me3 at the promoter of E2F1 and LPL, and with mRNA levels of LEP and SCD. In the case of HOMA-IR, the regression analysis showed associations with H3K4me3 enrichment at the promoter of SCD1 and IL6, and with the mRNA of LEP and SCD1. Moreover H3K4me3 at the E2F1 promoter was positively associated to E2F1 mRNA levels., Conclusions: H3K4me3 enrichment in the promoter of LEP, LPL, SREBF2, SCD1, PPARG, IL6, TNF and E2F1 is directly associated with increasing BMI and metabolic deterioration. The H3k4me3 mark could be regulating E3F1 mRNA levels in adipose tissue, while no associations between the promoter enrichment of this mark and mRNA levels existed for the other genes studied., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
34. β-Klotho deficiency shifts the gut-liver bile acid axis and induces hepatic alterations in mice.
- Author
-
Somm E, Henry H, Bruce SJ, Bonnet N, Montandon SA, Niederländer NJ, Messina A, Aeby S, Rosikiewicz M, Fajas L, Sempoux C, Ferrari SL, Greub G, and Pitteloud N
- Subjects
- Adiposity genetics, Animals, Energy Metabolism genetics, Fibroblast Growth Factors metabolism, Gluconeogenesis physiology, Ketone Bodies blood, Klotho Proteins, Membrane Proteins genetics, Mice, Mice, Knockout, Signal Transduction physiology, Bile Acids and Salts metabolism, Body Weight physiology, Gastrointestinal Tract metabolism, Liver metabolism, Liver Cirrhosis metabolism, Membrane Proteins metabolism
- Abstract
β-Klotho (encoded by Klb) is an obligate coreceptor, mediating both fibroblast growth factor (FGF)15 and FGF21 signaling. Klb
-/- mice are refractory to metabolic FGF15 and FGF21 action and exhibit derepressed (increased) bile acid (BA) synthesis. Here, we deeply phenotyped male Klb-/- mice on a pure C57BL/6J genetic background, fed a chow diet focusing on metabolic aspects. This aims to better understand the physiological consequences of concomitant FGF15 and FGF21 signaling deficiency, in particular on the gut-liver axis. Klb-/- mice present permanent growth restriction independent of adiposity and energy balance. Klb-/- mice also exhibit few changes in carbohydrate metabolism, combining normal gluco-tolerance, insulin sensitivity, and fasting response with increased gluconeogenic capacity and decreased glycogen mobilization. Livers of Klb-/- mice reveal pathologic features, including a proinflammatory status and initiation of fibrosis. These defects are associated to a massive shift in BA composition in the enterohepatic system and blood circulation featured by a large excess of microbiota-derived deoxycholic acid, classically known for its genotoxicity in the gastrointestinal tract. In conclusion, β-Klotho is a gatekeeper of hepatic integrity through direct action (mediating FGF21 anti-inflammatory signaling) and indirect mechanisms (mediating FGF15 signaling that maintains BA level and composition).- Published
- 2018
- Full Text
- View/download PDF
35. Dietary Fiber Confers Protection against Flu by Shaping Ly6c - Patrolling Monocyte Hematopoiesis and CD8 + T Cell Metabolism.
- Author
-
Trompette A, Gollwitzer ES, Pattaroni C, Lopez-Mejia IC, Riva E, Pernot J, Ubags N, Fajas L, Nicod LP, and Marsland BJ
- Subjects
- Adaptive Immunity drug effects, Adaptive Immunity immunology, Animals, CD8-Positive T-Lymphocytes metabolism, Dietary Fiber administration & dosage, Fatty Acids, Volatile immunology, Fatty Acids, Volatile metabolism, Hematopoiesis drug effects, Humans, Immunity, Innate drug effects, Immunity, Innate immunology, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Monocytes drug effects, Monocytes metabolism, Protective Agents administration & dosage, Protective Agents pharmacology, Antigens, Ly immunology, CD8-Positive T-Lymphocytes immunology, Dietary Fiber pharmacology, Hematopoiesis immunology, Monocytes immunology, Orthomyxoviridae Infections immunology
- Abstract
Dietary fiber protects against chronic inflammatory diseases by dampening immune responses through short-chain fatty acids (SCFAs). Here we examined the effect of dietary fiber in viral infection, where the anti-inflammatory properties of SCFAs in principle could prevent protective immunity. Instead, we found that fermentable dietary fiber increased survival of influenza-infected mice through two complementary mechanisms. High-fiber diet (HFD)-fed mice exhibited altered bone marrow hematopoiesis, characterized by enhanced generation of Ly6c
- patrolling monocytes, which led to increased numbers of alternatively activated macrophages with a limited capacity to produce the chemokine CXCL1 in the airways. Blunted CXCL1 production reduced neutrophil recruitment to the airways, thus limiting tissue immunopathology during infection. In parallel, diet-derived SCFAs boosted CD8+ T cell effector function by enhancing cellular metabolism. Hence, dietary fermentable fiber and SCFAs set an immune equilibrium, balancing innate and adaptive immunity so as to promote the resolution of influenza infection while preventing immune-associated pathology., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
36. E2F1 promotes hepatic gluconeogenesis and contributes to hyperglycemia during diabetes.
- Author
-
Giralt A, Denechaud PD, Lopez-Mejia IC, Delacuisine B, Blanchet E, Bonner C, Pattou F, Annicotte JS, and Fajas L
- Subjects
- Animals, Cells, Cultured, E2F1 Transcription Factor genetics, Hep G2 Cells, Humans, Liver metabolism, Mice, Mice, Inbred C57BL, Signal Transduction, Diabetes Mellitus, Type 2 metabolism, E2F1 Transcription Factor metabolism, Gluconeogenesis, Hyperglycemia metabolism
- Abstract
Objective: Aberrant hepatic glucose production contributes to the development of hyperglycemia and is a hallmark of type 2 diabetes. In a recent study, we showed that the transcription factor E2F1, a component of the cell cycle machinery, contributes to hepatic steatosis through the transcriptional regulation of key lipogenic enzymes. Here, we investigate if E2F1 contributes to hyperglycemia by regulating hepatic gluconeogenesis., Methods: We use different genetic models to investigate if E2F1 regulates gluconeogenesis in primary hepatocytes and in vivo. We study the impact of depleting E2F1 or inhibiting E2F1 activity in diabetic mouse models to evaluate if this transcription factor contributes to hyperglycemia during insulin resistance. We analyze E2F1 mRNA levels in the livers of human diabetic patients to assess the relevance of E2F1 in human pathophysiology., Results: Lack of E2F1 impaired gluconeogenesis in primary hepatocytes. Conversely, E2F1 overexpression increased glucose production in hepatocytes and in mice. Several genetic models showed that the canonical CDK4-RB1-E2F1 pathway is directly involved in this regulation. E2F1 mRNA levels were increased in the livers from human diabetic patients and correlated with the expression of the gluconeogenic enzyme Pck1. Genetic invalidation or pharmacological inhibition of E2F1 improved glucose homeostasis in diabetic mouse models., Conclusions: Our study unveils that the transcription factor E2F1 contributes to mammalian glucose homeostasis by directly controlling hepatic gluconeogenesis. Together with our previous finding that E2F1 promotes hepatic steatosis, the data presented here show that E2F1 contributes to both hyperlipidemia and hyperglycemia in diabetes, suggesting that specifically targeting E2F1 in the liver could be an interesting strategy for therapies against type 2 diabetes., (Copyright © 2018 The Authors. Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
37. Chromatin immunoprecipitation improvements for the processing of small frozen pieces of adipose tissue.
- Author
-
Castellano-Castillo D, Denechaud PD, Moreno-Indias I, Tinahones F, Fajas L, Queipo-Ortuño MI, and Cardona F
- Subjects
- Adult, Female, Freezing, Genome, Human, Humans, Male, Middle Aged, Proteome, Real-Time Polymerase Chain Reaction, Adipose Tissue metabolism, Chromatin Immunoprecipitation
- Abstract
Chromatin immunoprecipitation (ChIP) has gained importance to identify links between the genome and the proteome. Adipose tissue has emerged as an active tissue, which secretes a wide range of molecules that have been related to metabolic and obesity-related disorders, such as diabetes, cardiovascular failure, metabolic syndrome, or cancer. In turn, epigenetics has raised the importance in discerning the possible relationship between metabolic disorders, lifestyle and environment. However, ChIP application in human adipose tissue is limited by several factors, such as sample size, frozen sample availability, high lipid content and cellular composition of the tissue. Here, we optimize the standard protocol of ChIP for small pieces of frozen human adipose tissue. In addition, we test ChIP for the histone mark H3K4m3, which is related to active promoters, and validate the performance of the ChIP by analyzing gene promoters for factors usually studied in adipose tissue using qPCR. Our improvements result in a higher performance in chromatin shearing and DNA recovery of adipocytes from the tissue, which may be useful for ChIP-qPCR or ChIP-seq analysis.
- Published
- 2018
- Full Text
- View/download PDF
38. Editorial: Metabolic Adaptation to Cell Growth and Proliferation in Normal and Pathological Conditions.
- Author
-
Giralt A and Fajas L
- Published
- 2017
- Full Text
- View/download PDF
39. E2F1, a Novel Regulator of Metabolism.
- Author
-
Denechaud PD, Fajas L, and Giralt A
- Abstract
In the past years, several lines of evidence have shown that cell cycle regulatory proteins also can modulate metabolic processes. The transcription factor E2F1 is a central player involved in cell cycle progression, DNA-damage response, and apoptosis. Its crucial role in the control of cell fate has been extensively studied and reviewed before; however, here, we focus on the participation of E2F1 in the regulation of metabolism. We summarize recent findings about the cell cycle-independent roles of E2F1 in various tissues that contribute to global metabolic homeostasis and highlight that E2F1 activity is increased during obesity. Finally, coming back to the pivotal role of E2F1 in cancer development, we discuss how E2F1 links cell cycle progression with different metabolic adaptations required for cell growth and survival.
- Published
- 2017
- Full Text
- View/download PDF
40. CDK4 Phosphorylates AMPKα2 to Inhibit Its Activity and Repress Fatty Acid Oxidation.
- Author
-
Lopez-Mejia IC, Lagarrigue S, Giralt A, Martinez-Carreres L, Zanou N, Denechaud PD, Castillo-Armengol J, Chavey C, Orpinell M, Delacuisine B, Nasrallah A, Collodet C, Zhang L, Viollet B, Hardie DG, and Fajas L
- Subjects
- AMP-Activated Protein Kinases genetics, Animals, Cyclin-Dependent Kinase 4 genetics, Embryo, Mammalian metabolism, Fatty Acids genetics, Fibroblasts metabolism, Mice, Mice, Knockout, Mutation, Oxidation-Reduction, AMP-Activated Protein Kinases metabolism, Cyclin-Dependent Kinase 4 metabolism, Fatty Acids metabolism, Muscle, Skeletal metabolism, Physical Conditioning, Animal
- Abstract
The roles of CDK4 in the cell cycle have been extensively studied, but less is known about the mechanisms underlying the metabolic regulation by CDK4. Here, we report that CDK4 promotes anaerobic glycolysis and represses fatty acid oxidation in mouse embryonic fibroblasts (MEFs) by targeting the AMP-activated protein kinase (AMPK). We also show that fatty acid oxidation (FAO) is specifically induced by AMPK complexes containing the α2 subunit. Moreover, we report that CDK4 represses FAO through direct phosphorylation and inhibition of AMPKα2. The expression of non-phosphorylatable AMPKα2 mutants, or the use of a CDK4 inhibitor, increased FAO rates in MEFs and myotubes. In addition, Cdk4
-/- mice have increased oxidative metabolism and exercise capacity. Inhibition of CDK4 mimicked these alterations in normal mice, but not when skeletal muscle was AMPK deficient. This novel mechanism explains how CDK4 promotes anabolism by blocking catabolic processes (FAO) that are activated by AMPK., (Crown Copyright © 2017. Published by Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
41. The PDK1 Inhibitor Dichloroacetate Controls Cholesterol Homeostasis Through the ERK5/MEF2 Pathway.
- Author
-
Khan AUH, Allende-Vega N, Gitenay D, Gerbal-Chaloin S, Gondeau C, Vo DN, Belkahla S, Orecchioni S, Talarico G, Bertolini F, Bozic M, Valdivielso JM, Bejjani F, Jariel I, Lopez-Mejia IC, Fajas L, Lecellier CH, Hernandez J, Daujat M, and Villalba M
- Subjects
- Animals, Cell Line, Tumor, Cell Survival drug effects, Hepatocytes drug effects, Hepatocytes metabolism, Homeostasis drug effects, Mice, Pyruvate Dehydrogenase Acetyl-Transferring Kinase, Reactive Oxygen Species metabolism, Receptors, LDL genetics, Receptors, LDL metabolism, Cholesterol metabolism, Dichloroacetic Acid pharmacology, Lipid Metabolism drug effects, MEF2 Transcription Factors metabolism, Mitogen-Activated Protein Kinase 7 metabolism, Protein Serine-Threonine Kinases antagonists & inhibitors, Signal Transduction drug effects
- Abstract
Controlling cholesterol levels is a major challenge in human health, since hypercholesterolemia can lead to serious cardiovascular disease. Drugs that target carbohydrate metabolism can also modify lipid metabolism and hence cholesterol plasma levels. In this sense, dichloroacetate (DCA), a pyruvate dehydrogenase kinase (PDK) inhibitor, augments usage of the glycolysis-produced pyruvate in the mitochondria increasing oxidative phosphorylation (OXPHOS). In several animal models, DCA decreases plasma cholesterol and triglycerides. Thus, DCA was used in the 70 s to treat diabetes mellitus, hyperlipoproteinemia and hypercholesterolemia with satisfactory results. However, the mechanism of action remained unknown and we describe it here. DCA increases LDLR mRNA and protein levels as well as LDL intake in several cell lines, primary human hepatocytes and two different mouse models. This effect is mediated by transcriptional activation as evidenced by H3 acetylation on lysine 27 on the LDLR promoter. DCA induces expression of the MAPK ERK5 that turns on the transcription factor MEF2. Inhibition of this ERK5/MEF2 pathway by genetic or pharmacological means decreases LDLR expression and LDL intake. In summary, our results indicate that DCA, by inducing OXPHOS, promotes ERK5/MEF2 activation leading to LDLR expression. The ERK5/MEF2 pathway offers an interesting pharmacological target for drug development.
- Published
- 2017
- Full Text
- View/download PDF
42. Cancer: Linking Powerhouses to Suicidal Bags.
- Author
-
Martinez-Carreres L, Nasrallah A, and Fajas L
- Abstract
Membrane-bound organelles are integrated into cellular networks and work together for a common goal: regulating cell metabolism, cell signaling pathways, cell fate, cellular maintenance, and pathogen defense. Many of these interactions are well established, but little is known about the interplay between mitochondria and lysosomes, and their deregulation in cancer. The present review focuses on the common signaling pathways of both organelles, as well as the processes in which they both physically interact, their changes under pathological conditions, and the impact on targeting those organelles for treating cancer.
- Published
- 2017
- Full Text
- View/download PDF
43. E2F1 inhibits circulating cholesterol clearance by regulating Pcsk9 expression in the liver.
- Author
-
Lai Q, Giralt A, Le May C, Zhang L, Cariou B, Denechaud PD, and Fajas L
- Abstract
Cholesterol accumulation in the liver is an early event in nonalcoholic fatty liver disease (NAFLD). Here, we demonstrate that E2F1 plays a crucial role in maintaining cellular cholesterol homeostasis by regulating cholesterol uptake via proprotein convertase subtilisin/kexin 9 (PCSK9), an enzyme that promotes low-density lipoprotein receptor (LDLR) degradation upon activation. E2f1-/- mice display reduced total plasma cholesterol levels and increased cholesterol content in the liver. In this study, we show that E2f1 deletion in cellular and mouse models leads to a marked decrease in Pcsk9 expression and an increase in LDLR expression. In addition to the upregulation of LDLR, we report that E2f1-/- hepatocytes exhibit increased LDL uptake. ChIP-Seq and PCSK9 promoter reporter experiments confirmed that E2F1 binds to and transactivates the PCSK9 promoter. Interestingly, E2f1-/- mice fed a high-cholesterol diet (HCD) display a fatty liver phenotype and liver fibrosis, which is reversed by reexpression of PCSK9 in the liver. Collectively, these data indicate that E2F1 regulates cholesterol uptake and that the loss of E2F1 leads to abnormal cholesterol accumulation in the liver and the development of fibrosis in response to an HCD.
- Published
- 2017
- Full Text
- View/download PDF
44. β-Klotho deficiency protects against obesity through a crosstalk between liver, microbiota, and brown adipose tissue.
- Author
-
Somm E, Henry H, Bruce SJ, Aeby S, Rosikiewicz M, Sykiotis GP, Asrih M, Jornayvaz FR, Denechaud PD, Albrecht U, Mohammadi M, Dwyer A, Acierno JS Jr, Schoonjans K, Fajas L, Greub G, and Pitteloud N
- Abstract
β-Klotho (encoded by Klb) is the obligate coreceptor mediating FGF21 and FGF15/19 signaling. Klb-/- mice are refractory to beneficial action of pharmacological FGF21 treatment including stimulation of glucose utilization and thermogenesis. Here, we investigated the energy homeostasis in Klb-/- mice on high-fat diet in order to better understand the consequences of abrogating both endogenous FGF15/19 and FGF21 signaling during caloric overload. Surprisingly, Klb-/- mice are resistant to diet-induced obesity (DIO) owing to enhanced energy expenditure and BAT activity. Klb-/- mice exhibited not only an increase but also a shift in bile acid (BA) composition featured by activation of the classical (neutral) BA synthesis pathway at the expense of the alternative (acidic) pathway. High hepatic production of cholic acid (CA) results in a large excess of microbiota-derived deoxycholic acid (DCA). DCA is specifically responsible for activating the TGR5 receptor that stimulates BAT thermogenic activity. In fact, combined gene deletion of Klb and Tgr5 or antibiotic treatment abrogating bacterial conversion of CA into DCA both abolish DIO resistance in Klb-/- mice. These results suggested that DIO resistance in Klb-/- mice is caused by high levels of DCA, signaling through the TGR5 receptor. These data also demonstrated that gut microbiota can regulate host thermogenesis via conversion of primary into secondary BA. Pharmacologic or nutritional approaches to selectively modulate BA composition may be a promising target for treating metabolic disorders.
- Published
- 2017
- Full Text
- View/download PDF
45. Growth factor receptor binding protein 14 inhibition triggers insulin-induced mouse hepatocyte proliferation and is associated with hepatocellular carcinoma.
- Author
-
Morzyglod L, Caüzac M, Popineau L, Denechaud PD, Fajas L, Ragazzon B, Fauveau V, Planchais J, Vasseur-Cognet M, Fartoux L, Scatton O, Rosmorduc O, Guilmeau S, Postic C, Desdouets C, Desbois-Mouthon C, and Burnol AF
- Subjects
- Animals, Carcinoma, Hepatocellular epidemiology, Carcinoma, Hepatocellular metabolism, Cell Line, Tumor, Cell Proliferation genetics, Cells, Cultured, Diabetes Mellitus, Type 2 epidemiology, Diabetes Mellitus, Type 2 metabolism, Disease Models, Animal, Down-Regulation, Hepatocytes cytology, Hepatocytes metabolism, Humans, Liver Neoplasms epidemiology, Liver Neoplasms metabolism, Male, Mice, Mice, Inbred C57BL, Random Allocation, Sensitivity and Specificity, Adaptor Proteins, Signal Transducing genetics, Carcinoma, Hepatocellular physiopathology, Diabetes Mellitus, Type 2 physiopathology, Liver Neoplasms physiopathology, Receptor, Insulin metabolism
- Abstract
Metabolic diseases such as obesity and type 2 diabetes are recognized as independent risk factors for hepatocellular carcinoma (HCC). Hyperinsulinemia, a hallmark of these pathologies, is suspected to be involved in HCC development. The molecular adapter growth factor receptor binding protein 14 (Grb14) is an inhibitor of insulin receptor catalytic activity, highly expressed in the liver. To study its involvement in hepatocyte proliferation, we specifically inhibited its liver expression using a short hairpin RNA strategy in mice. Enhanced insulin signaling upon Grb14 inhibition was accompanied by a transient induction of S-phase entrance by quiescent hepatocytes, indicating that Grb14 is a potent repressor of cell division. The proliferation of Grb14-deficient hepatocytes was cell-autonomous as it was also observed in primary cell cultures. Combined Grb14 down-regulation and insulin signaling blockade using pharmacological approaches as well as genetic mouse models demonstrated that Grb14 inhibition-mediated hepatocyte division involved insulin receptor activation and was mediated by the mechanistic target of rapamycin complex 1-S6K pathway and the transcription factor E2F1. In order to determine a potential dysregulation in GRB14 gene expression in human pathophysiology, a collection of 85 human HCCs was investigated. This revealed a highly significant and frequent decrease in GRB14 expression in hepatic tumors when compared to adjacent nontumoral parenchyma, with 60% of the tumors exhibiting a reduced Grb14 mRNA level., Conclusion: Our study establishes Grb14 as a physiological repressor of insulin mitogenic action in the liver and further supports that dysregulation of insulin signaling is associated with HCC. (Hepatology 2017;65:1352-1368)., (© 2016 by the American Association for the Study of Liver Diseases.)
- Published
- 2017
- Full Text
- View/download PDF
46. [Cell cycle regulators CDK4 and E2F1 control glucose and lipid homeostasis].
- Author
-
Denechaud PD, Lopez-Mejia IC, and Fajas L
- Subjects
- Adipose Tissue metabolism, Fatty Liver, Glycolysis physiology, Humans, Insulin metabolism, Lipogenesis physiology, Liver metabolism, Obesity metabolism, Signal Transduction physiology, Cyclin-Dependent Kinase 4 physiology, E2F1 Transcription Factor physiology, Glucose metabolism, Homeostasis, Lipid Metabolism physiology
- Published
- 2016
- Full Text
- View/download PDF
47. KAT2B Is Required for Pancreatic Beta Cell Adaptation to Metabolic Stress by Controlling the Unfolded Protein Response.
- Author
-
Rabhi N, Denechaud PD, Gromada X, Hannou SA, Zhang H, Rashid T, Salas E, Durand E, Sand O, Bonnefond A, Yengo L, Chavey C, Bonner C, Kerr-Conte J, Abderrahmani A, Auwerx J, Fajas L, Froguel P, and Annicotte JS
- Subjects
- Adaptation, Physiological, Animals, Cell Line, Endoplasmic Reticulum metabolism, Humans, Insulin Secretion, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, RNA Interference, RNA, Small Interfering, Signal Transduction, Stress, Physiological, Transcription Factors metabolism, Unfolded Protein Response genetics, p300-CBP Transcription Factors deficiency, Endoplasmic Reticulum Stress physiology, Glucose Intolerance genetics, Insulin metabolism, Insulin-Secreting Cells metabolism, Unfolded Protein Response physiology, p300-CBP Transcription Factors genetics
- Abstract
The endoplasmic reticulum (ER) unfolded protein response (UPR(er)) pathway plays an important role in helping pancreatic β cells to adapt their cellular responses to environmental cues and metabolic stress. Although altered UPR(er) gene expression appears in rodent and human type 2 diabetic (T2D) islets, the underlying molecular mechanisms remain unknown. We show here that germline and β cell-specific disruption of the lysine acetyltransferase 2B (Kat2b) gene in mice leads to impaired insulin secretion and glucose intolerance. Genome-wide analysis of Kat2b-regulated genes and functional assays reveal a critical role for Kat2b in maintaining UPR(er) gene expression and subsequent β cell function. Importantly, Kat2b expression is decreased in mouse and human diabetic β cells and correlates with UPR(er) gene expression in normal human islets. In conclusion, Kat2b is a crucial transcriptional regulator for adaptive β cell function during metabolic stress by controlling UPR(er) and represents a promising target for T2D prevention and treatment., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
48. Retinoblastoma Protein Knockdown Favors Oxidative Metabolism and Glucose and Fatty Acid Disposal in Muscle Cells.
- Author
-
Petrov PD, Ribot J, López-Mejía IC, Fajas L, Palou A, and Bonet ML
- Subjects
- Animals, Gene Knockdown Techniques methods, Insulin pharmacology, Mice, Muscle Development drug effects, Muscle Development physiology, Muscle, Skeletal drug effects, Oxidation-Reduction drug effects, Retinoblastoma Protein deficiency, Fatty Acids metabolism, Glucose metabolism, Muscle Fibers, Skeletal metabolism, Muscle, Skeletal metabolism, Retinoblastoma Protein metabolism
- Abstract
Deficiency in the retinoblastoma protein (Rb) favors leanness and a healthy metabolic profile in mice largely attributed to activation of oxidative metabolism in white and brown adipose tissues. Less is known about Rb modulation of skeletal muscle metabolism. This was studied here by transiently knocking down Rb expression in differentiated C2C12 myotubes using small interfering RNAs. Compared with control cells transfected with non-targeting RNAs, myotubes silenced for Rb (by 80-90%) had increased expression of genes related to fatty acid uptake and oxidation such as Cd36 and Cpt1b (by 61% and 42%, respectively), increased Mitofusin 2 protein content (∼2.5-fold increase), increased mitochondrial to nuclear DNA ratio (by 48%), increased oxygen consumption (by 65%) and decreased intracellular lipid accumulation. Rb silenced myotubes also displayed up-regulated levels of glucose transporter type 4 expression (∼5-fold increase), increased basal glucose uptake, and enhanced insulin-induced Akt phosphorylation. Interestingly, exercise in mice led to increased Rb phosphorylation (inactivation) in skeletal muscle as evidenced by immunohistochemistry analysis. In conclusion, the silencing of Rb enhances mitochondrial oxidative metabolism and fatty acid and glucose disposal in skeletal myotubes, and changes in Rb status may contribute to muscle physiological adaptation to exercise., (© 2015 Wiley Periodicals, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
49. Mammalian Target of Rapamycin Complex 2 Controls CD8 T Cell Memory Differentiation in a Foxo1-Dependent Manner.
- Author
-
Zhang L, Tschumi BO, Lopez-Mejia IC, Oberle SG, Meyer M, Samson G, Rüegg MA, Hall MN, Fajas L, Zehn D, Mach JP, Donda A, and Romero P
- Subjects
- Animals, Carrier Proteins metabolism, Cell Nucleus metabolism, Forkhead Box Protein O1, Interleukin-2 biosynthesis, Mechanistic Target of Rapamycin Complex 2, Mice, Inbred C57BL, Mice, Knockout, Rapamycin-Insensitive Companion of mTOR Protein, T-Box Domain Proteins metabolism, Transcription, Genetic, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology, Cell Differentiation genetics, Forkhead Transcription Factors metabolism, Immunologic Memory genetics, Multiprotein Complexes metabolism, TOR Serine-Threonine Kinases metabolism
- Abstract
Upon infection, antigen-specific naive CD8 T cells are activated and differentiate into short-lived effector cells (SLECs) and memory precursor cells (MPECs). The underlying signaling pathways remain largely unresolved. We show that Rictor, the core component of mammalian target of rapamycin complex 2 (mTORC2), regulates SLEC and MPEC commitment. Rictor deficiency favors memory formation and increases IL-2 secretion capacity without dampening effector functions. Moreover, mTORC2-deficient memory T cells mount more potent recall responses. Enhanced memory formation in the absence of mTORC2 was associated with Eomes and Tcf-1 upregulation, repression of T-bet, enhanced mitochondrial spare respiratory capacity, and fatty acid oxidation. This transcriptional and metabolic reprogramming is mainly driven by nuclear stabilization of Foxo1. Silencing of Foxo1 reversed the increased MPEC differentiation and IL-2 production and led to an impaired recall response of Rictor KO memory T cells. Therefore, mTORC2 is a critical regulator of CD8 T cell differentiation and may be an important target for immunotherapy interventions., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
50. Modulation of mTOR Signalling Triggers the Formation of Stem Cell-like Memory T Cells.
- Author
-
Scholz G, Jandus C, Zhang L, Grandclément C, Lopez-Mejia IC, Soneson C, Delorenzi M, Fajas L, Held W, Dormond O, and Romero P
- Subjects
- Animals, CD4-Positive T-Lymphocytes immunology, Cells, Cultured, Female, Humans, Lymphopoiesis, Mechanistic Target of Rapamycin Complex 1, Mice, Multiprotein Complexes antagonists & inhibitors, Precursor Cells, T-Lymphoid immunology, Pyrimidines pharmacology, Pyrroles pharmacology, TOR Serine-Threonine Kinases antagonists & inhibitors, CD4-Positive T-Lymphocytes cytology, Immunologic Memory, Multiprotein Complexes metabolism, Precursor Cells, T-Lymphoid cytology, Signal Transduction, TOR Serine-Threonine Kinases metabolism
- Abstract
Robust, long-lasting immune responses are elicited by memory T cells that possess properties of stem cells, enabling them to persist long-term and to permanently replenish the effector pools. Thus, stem cell-like memory T (TSCM) cells are of key therapeutic value and efforts are underway to characterize TSCM cells and to identify means for their targeted induction. Here, we show that inhibition of mechanistic/mammalian Target of Rapamycin (mTOR) complex 1 (mTORC1) by rapamycin or the Wnt-β-catenin signalling activator TWS119 in activated human naive T cells leads to the induction of TSCM cells. We show that these compounds switch T cell metabolism to fatty acid oxidation as favoured metabolic programme for TSCM cell generation. Of note, pharmacologically induced TSCM cells possess superior functional features as a long-term repopulation capacity after adoptive transfer. Furthermore, we provide insights into the transcriptome of TSCM cells. Our data identify a mechanism of pharmacological mTORC1 inhibitors, allowing us to confer stemness to human naive T cells which may be significantly relevant for the design of innovative T cell-based cancer immunotherapies.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.