168 results on '"Fajas, L"'
Search Results
2. Expression of the peroxisome proliferator-activated receptor gamma (PPARgamma) in human atherosclerosis and regulation in macrophages by colony stimulating factors and oxidized low density lipoprotein.
- Author
-
Ricote, M, Huang, J, Fajas, L, Li, A, Welch, J, Najib, J, Witztum, JL, Auwerx, J, Palinski, W, and Glass, CK
- Subjects
Cells ,Cultured ,Foam Cells ,Animals ,Humans ,Mice ,Arteriosclerosis ,Tetradecanoylphorbol Acetate ,Protein Kinase C ,Lipoproteins ,LDL ,Colony-Stimulating Factors ,Receptors ,Cytoplasmic and Nuclear ,Transcription Factors ,Cell Differentiation ,Promoter Regions ,Genetic - Abstract
The peroxisome proliferator-activated receptor gamma (PPARgamma) is a ligand-dependent transcription factor that has been demonstrated to regulate fat cell development and glucose homeostasis. PPARgamma is also expressed in a subset of macrophages and negatively regulates the expression of several proinflammatory genes in response to natural and synthetic ligands. We here demonstrate that PPARgamma is expressed in macrophage foam cells of human atherosclerotic lesions, in a pattern that is highly correlated with that of oxidation-specific epitopes. Oxidized low density lipoprotein (oxLDL) and macrophage colony-stimulating factor, which are known to be present in atherosclerotic lesions, stimulated PPARgamma expression in primary macrophages and monocytic cell lines. PPARgamma mRNA expression was also induced in primary macrophages and THP-1 monocytic leukemia cells by the phorbol ester 12-O-tetradecanoylphorbol 13-acetate (TPA). Inhibition of protein kinase C blocked the induction of PPARgamma expression by TPA, but not by oxLDL, suggesting that more than one signaling pathway regulates PPARgamma expression in macrophages. TPA induced the expression of PPARgamma in RAW 264.7 macrophages by increasing transcription from the PPARgamma1 and PPARgamma3 promoters. In concert, these observations provide insights into the regulation of PPARgamma expression in activated macrophages and raise the possibility that PPARgamma ligands may influence the progression of atherosclerosis.
- Published
- 1998
3. 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
4. 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
5. Metabolism and proliferation share common regulatory pathways in cancer cells
- Author
-
Fritz, V and Fajas, L
- Published
- 2010
- Full Text
- View/download PDF
6. 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
7. Metabolic intervention on lipid synthesis converging pathways abrogates prostate cancer growth
- Author
-
Fritz, V, Benfodda, Z, Henriquet, C, Hure, S, Cristol, J-P, Michel, F, Carbonneau, M-A, Casas, F, and Fajas, L
- Published
- 2013
- Full Text
- View/download PDF
8. Atypical transcriptional regulators and cofactors of PPARγ
- Author
-
Miard, S and Fajas, L
- Published
- 2005
9. 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
10. 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
11. 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
12. 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
13. 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
14. [Emerging key role of cell cycle regulators in cell metabolism]
- Author
-
Lagarrigue, S., Blanchet, E., Annicotte, J. S., Fajas, L., Institut de Génétique Moléculaire de Montpellier (IGMM), and Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM)
- Subjects
[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology - Abstract
The role of cell cycle regulators in the control of cell proliferation has been extensively studied, but independently of these functions in cell proliferation, it now appears that these proteins are also key to the adapted metabolic response of the cells. This has some logic since cell cycle is linked to metabolic control. This review focusses on the involvment of cyclins, cyclin dependent kinases or E2F factor in the control of adipogenesis, glucidic homeostasis, and energy consumption. Murine models in which genes encoding these regulators have been invalidated have been key to unravel these novel functions of cell cycle regulators in cell metabolism. Furthermore, these findings may also have some relevance for metabolic disorders such as obesity or diabetes.
- Published
- 2011
- Full Text
- View/download PDF
15. The Transcriptional Coactivator Peroxisome Proliferator–Activated Receptor (PPAR) Coactivator-1 and the Nuclear Receptor PPAR Control the Expression of Glycerol Kinase and Metabolism Genes Independently of PPAR Activation in Human White Adipocytes
- Author
-
Mazzucotelli, Antoine, Viguerie, A, Tiraby, A, Annicotte, J-S, Klimcakova, E, Lepin, E, Delmar, P, Dejean, S, Tarvernier, G, Lefort, C, Mairal, A, Hidalgo, J, Pineau, Thierry, Fajas, L, Clément, Karine, Langin, Dominique, Inconnu, Département Santé Animale (DEPT SA), and Institut National de la Recherche Agronomique (INRA)
- Subjects
[SDV]Life Sciences [q-bio] ,ComputingMilieux_MISCELLANEOUS - Abstract
International audience
- Published
- 2007
16. PPAR{gamma} Recruits the P-TEFb Complex to Activate Transcription and Promote Adipogenesis
- Author
-
Iankova, I., Petersen, Rk, Annicotte, Jean-Sébastien, Chavey, C., Hansen, Jb, Kratchmarova, I., Sarruf, D., Benkirane, M., Kristiansen, K., Fajas, L., Institut de génétique humaine (IGH), and Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
[SDV.BC.IC]Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,[SDV.BDD]Life Sciences [q-bio]/Development Biology - Published
- 2006
- Full Text
- View/download PDF
17. Downregulation of protein tyrosine phosphatase PTP-BL represses adipogenesis.
- Author
-
Glondu-Lassis, M., Dromard, M., Chavey, C., Puech, C., Fajas, L., Hendriks, W.J.A.J., Freiss, G., Glondu-Lassis, M., Dromard, M., Chavey, C., Puech, C., Fajas, L., Hendriks, W.J.A.J., and Freiss, G.
- Abstract
Contains fulltext : 80913.pdf (publisher's version ) (Closed access), The insulin/insulin-like growth factor 1 (IGF-1) signaling pathway is a major regulator of adipose tissue growth and differentiation. We recently demonstrated that human protein tyrosine phosphatase (PTP) L1, a large cytoplasmic phosphatase also known as PTP-BAS/PTPN13/PTP-1E, is a negative regulator of IGF-1R/IRS-1/Akt pathway in breast cancer cells. This triggered us to investigate the potential role of PTPL1 in adipogenesis. To evaluate the implication of PTP-BL, the mouse orthologue of PTPL1, in adipose tissue biology, we analyzed PTP-BL mRNA expression in adipose tissue in vivo and during proliferation and differentiation of 3T3-L1 pre-adipocytes. To elucidate the role of PTP-BL and of its catalytic activity during adipogenesis we use siRNA techniques in 3T3-L1 pre-adipocytes, and mouse embryonic fibroblasts that lack wildtype PTP-BL and instead express a variant without the PTP domain (Delta P/Delta P MEFs). Here we show that PTP-BL is strongly expressed in white adipose tissue and that PTP-BL transcript and protein levels increase during proliferation and differentiation of 3T3-L1 pre-adipocytes. Strikingly, knockdown of PTP-BL expression in 3T3-L1 adipocytes caused a dramatic decrease in adipogenic gene expression levels (PPAR gamma, aP2) and lipid accumulation but did not interfere with the insulin/Akt pathway. Delta P/Delta P MEFs differentiate into the adipogenic lineage as efficiently as wildtype MEFs. However, when expression of either PTP-BL or PTP-BL Delta P was inhibited a dramatic reduction in the number of MEF-derived adipocytes was observed. These findings demonstrate a key role for PTP-BL in 3T3-L1 and MEF-derived adipocyte differentiation that is independent of its enzymatic activity.
- Published
- 2009
18. Transcriptional control of adipogenesis
- Author
-
Fajas, L, Fruchart, J C, Auwerx, J, Fajas, L, Fruchart, J C, and Auwerx, J
- Abstract
Adipocyte differentiation is coordinatedly regulated by several transcription factors. C/EBP beta, C/EBP delta and ADD-1/SREBP-1 are active early during the differentiation process and induce the expression and/or activity of the peroxisome proliferator activated receptor-gamma (PPAR gamma), the pivotal coordinator of the adipocyte differentiation process. Activated PPAR gamma induces exit from the cell cycle and triggers the expression of adipocyte-specific genes, resulting in increased delivery of energy to the cells. C/EBP alpha, whose expression coincides with the later stages of differentiation, cooperates with PPAR gamma in inducing additional target genes and sustains a high level of PPAR gamma in the mature adipocyte as part of a feedforward loop. Altered activity and/or expression of these transcription factors might underlie the pathogenesis of disorders characterized by increased or decreased adipose tissue depots.
- Published
- 2009
- Full Text
- View/download PDF
19. Peroxisome proliferator-activated receptor-gamma: from adipogenesis to carcinogenesis
- Author
-
Fajas, L, Debril, M B, Auwerx, J, Fajas, L, Debril, M B, and Auwerx, J
- Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear hormone receptors, initially described as molecular targets for synthetic compounds inducing peroxisome proliferation. PPAR-gamma, the best characterized of the PPARs, plays a crucial role in adipogenesis and insulin sensitization. Furthermore, PPAR-gamma has been reported to affect cell proliferation/differentiation pathways in various malignancies. We discuss in the present review recent advances in the understanding of the function of PPAR-gamma in both cell proliferation and adipocyte differentiation.
- Published
- 2009
- Full Text
- View/download PDF
20. PPAR gamma: an essential role in metabolic control
- Author
-
Fajas, L, Debril, M B, Auwerx, J, Fajas, L, Debril, M B, and Auwerx, J
- Abstract
The peroxisome proliferator-activated receptor gamma is a nuclear hormone receptor playing a crucial role in adipogenesis and insulin sensitization. Prostaglandin J2 derivatives and the antidiabetic thiazolidinediones are its respective natural and synthetic ligands. The RXR/PPAR gamma heterodimer has also been reported to have important immunomodulatory activities and its pleiotropic functions suggest wide-ranging medical implications.
- Published
- 2009
21. The pleiotropic functions of peroxisome proliferator-activated receptor gamma
- Author
-
Debril, M B, Renaud, J P, Fajas, L, Auwerx, J, Debril, M B, Renaud, J P, Fajas, L, and Auwerx, J
- Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear hormone receptors, initially described as molecular targets for synthetic compounds that induce peroxisome proliferation. PPARgamma is the best characterized of the PPARs. The heterodimer of PPARgamma with the retinoid X receptor (RXR) plays a crucial role in adipogenesis and insulin sensitization. The RXR/PPARgamma heterodimer furthermore has been reported to have important immunomodulatory activities and to affect cell proliferation/differentiation pathways in various malignancies. PPARgamma is activated by a number of naturally occurring fatty acid derivatives and by several synthetic compounds, including the thiazolidinediones and L-tyrosine-based insulin sensitizers. This review gives an overview of the pleiotropic functions of PPARgamma and discusses the wide-ranging medical implications that modulation of PPARgamma activity might have for various diseases, ranging from obesity and type 2 diabetes to cancer and inflammation.
- Published
- 2009
- Full Text
- View/download PDF
22. Metabolic intervention on lipid synthesis converging pathways abrogates prostate cancer growth
- Author
-
Fritz, V, primary, Benfodda, Z, additional, Henriquet, C, additional, Hure, S, additional, Cristol, J-P, additional, Michel, F, additional, Carbonneau, M-A, additional, Casas, F, additional, and Fajas, L, additional
- Published
- 2012
- Full Text
- View/download PDF
23. Ciglitazone negatively regulates CXCL1 signaling through MITF to suppress melanoma growth
- Author
-
Botton, T, primary, Puissant, A, additional, Cheli, Y, additional, Tomic, T, additional, Giuliano, S, additional, Fajas, L, additional, Deckert, M, additional, Ortonne, J-P, additional, Bertolotto, C, additional, Tartare-Deckert, S, additional, Ballotti, R, additional, and Rocchi, S, additional
- Published
- 2010
- Full Text
- View/download PDF
24. Cyclin A is a mediator of p120E4F-dependent cell cycle arrest in G1
- Author
-
Fajas, L., Paul, C., Vié, A., Estrach, S., Medema, R.H., Blanchard, J.M., Sardet, C., Vignais, M.-L., Fajas, L., Paul, C., Vié, A., Estrach, S., Medema, R.H., Blanchard, J.M., Sardet, C., and Vignais, M.-L.
- Published
- 2001
25. pRB binds to and modulates the transrepressing activity of the E1A-regulated transcription factor p120E4F
- Author
-
Fajas, L., Paul, C., Zugasti, O., Cam, L. Le, Polanowska, J., Fabbrizio, E., Medema, R.H., Vignais, M.-L., Sardet, C., Fajas, L., Paul, C., Zugasti, O., Cam, L. Le, Polanowska, J., Fabbrizio, E., Medema, R.H., Vignais, M.-L., and Sardet, C.
- Published
- 2000
26. p300 interacts with the N- and C-terminal part of PPARgamma2 in a ligand-independent and -dependent manner, respectively.
- Author
-
Gelman, L, Zhou, G, Fajas, L, Raspé, Eric, Fruchart, J C, Auwerx, J, Gelman, L, Zhou, G, Fajas, L, Raspé, Eric, Fruchart, J C, and Auwerx, J
- Abstract
The nuclear peroxisome proliferator-activated receptor gamma (PPARgamma) activates the transcription of multiple genes involved in intra- and extracellular lipid metabolism. Several cofactors are crucial for the stimulation or the silencing of nuclear receptor transcriptional activities. The two homologous cofactors p300 and CREB-binding protein (CBP) have been shown to co-activate the ligand-dependent transcriptional activities of several nuclear receptors as well as the ligand-independent transcriptional activity of the androgen receptor. We show here that the interaction between p300/CBP and PPARgamma is complex and involves multiple domains in each protein. p300/CBP not only bind in a ligand-dependent manner to the DEF region of PPARgamma but also bind directly in a ligand-independent manner to a region in the AB domain localized between residue 31 to 99. In transfection experiments, p300/CBP could thereby enhance the transcriptional activities of both the activating function (AF)-1 and AF-2 domains. p300/CBP displays itself at least two docking sites for PPARgamma located in its N terminus (between residues 1 and 113 for CBP) and in the middle of the protein (between residues 1099 and 1460)., info:eu-repo/semantics/published
- Published
- 1999
27. The organization, promoter analysis, and expression of the human PPARgamma gene.
- Author
-
Fajas, L, Auboeuf, D, Raspé, Eric, Schoonjans, K, Lefebvre, A M, Saladin, R, Najib, J, Laville, M, Fruchart, J C, Deeb, S, Vidal-Puig, A, Flier, J, Briggs, M R, Staels, Bart, Vidal, H, Auwerx, J, Fajas, L, Auboeuf, D, Raspé, Eric, Schoonjans, K, Lefebvre, A M, Saladin, R, Najib, J, Laville, M, Fruchart, J C, Deeb, S, Vidal-Puig, A, Flier, J, Briggs, M R, Staels, Bart, Vidal, H, and Auwerx, J
- Abstract
PPARgamma is a member of the PPAR subfamily of nuclear receptors. In this work, the structure of the human PPARgamma cDNA and gene was determined, and its promoters and tissue-specific expression were functionally characterized. Similar to the mouse, two PPAR isoforms, PPARgamma1 and PPARgamma2, were detected in man. The relative expression of human PPARgamma was studied by a newly developed and sensitive reverse transcriptase-competitive polymerase chain reaction method, which allowed us to distinguish between PPARgamma1 and gamma2 mRNA. In all tissues analyzed, PPARgamma2 was much less abundant than PPARgamma1. Adipose tissue and large intestine have the highest levels of PPARgamma mRNA; kidney, liver, and small intestine have intermediate levels; whereas PPARgamma is barely detectable in muscle. This high level expression of PPARgamma in colon warrants further study in view of the well established role of fatty acid and arachidonic acid derivatives in colonic disease. Similarly as mouse PPARgammas, the human PPARgammas are activated by thiazolidinediones and prostaglandin J and bind with high affinity to a PPRE. The human PPARgamma gene has nine exons and extends over more than 100 kilobases of genomic DNA. Alternate transcription start sites and alternate splicing generate the PPARgamma1 and PPARgamma2 mRNAs, which differ at their 5'-ends. PPARgamma1 is encoded by eight exons, and PPARgamma2 is encoded by seven exons. The 5'-untranslated sequence of PPARgamma1 is comprised of exons A1 and A2, whereas that of PPARgamma2 plus the additional PPARgamma2-specific N-terminal amino acids are encoded by exon B, located between exons A2 and A1. The remaining six exons, termed 1 to 6, are common to the PPARgamma1 and gamma2. Knowledge of the gene structure will allow screening for PPARgamma mutations in humans with metabolic disorders, whereas knowledge of its expression pattern and factors regulating its expression could be of major importance in understanding its biolog, info:eu-repo/semantics/published
- Published
- 1997
28. Peroxisome proliferator-activated receptor-gamma: from adipogenesis to carcinogenesis
- Author
-
Fajas, L, primary, Debril, MB, additional, and Auwerx, J, additional
- Published
- 2001
- Full Text
- View/download PDF
29. Impact of the Peroxisome Proliferator Activated Receptor γ2 Pro12Ala polymorphism on adiposity, lipids and non-insulin-dependent diabetes mellitus
- Author
-
Meirhaeghe, A, primary, Fajas, L, additional, Helbecque, N, additional, Cottel, D, additional, Auwerx, J, additional, Deeb, SS, additional, and Amouyel, P, additional
- Published
- 2000
- Full Text
- View/download PDF
30. Improved LM-PCR Procedure for In Vivo Footprinting Analysis of GC-Rich Promoters
- Author
-
Cam, L. Le, primary, Polanowska, J., additional, Fajas, L., additional, Fabbrizio, E., additional, and Sardet, C., additional
- Published
- 1999
- Full Text
- View/download PDF
31. Ciglitazone negatively regulates CXCL1 signaling through MITF to suppress melanoma growth.
- Author
-
Botton, T., Puissant, A., Cheli, Y., Tomic, T., Giuliano, S., Fajas, L., Deckert, M., Ortonne, J.-P., Bertolotto, C., Tartare-Deckert, S., Ballotti, R., and Rocchi, S.
- Subjects
MELANOMA ,MELANOCYTES ,APOPTOSIS ,GENES ,MICROPHTHALMUS ,CELL lines - Abstract
We have previously demonstrated that the thiazolidinedione ciglitazone inhibited, independently of PPARγ activation, melanoma cell growth. Further investigations now show that ciglitazone effects are mediated through the regulation of secreted factors. Q-PCR screening of several genes involved in melanoma biology reveals that ciglitazone inhibits expression of the CXCL1 chemokine gene. CXCL1 is overexpressed in melanoma and contributes to tumorigenicity. We show that ciglitazone induces a diminution of CXCL1 level in different human melanoma cell lines. This effect is mediated by the downregulation of microphthalmia-associated transcription factor, MITF, the master gene in melanocyte differentiation and involved in melanoma development. Further, recombinant CXCL1 protein is sufficient to abrogate thiazolidinedione effects such as apoptosis induction, whereas extinction of the CXCL1 pathway mimics phenotypic changes observed in response to ciglitazone. Finally, inhibition of human melanoma tumor development in nude mice treated with ciglitazone is associated with a strong decrease in MITF and CXCL1 levels. Our results show that anti-melanoma effects of thiazolidinediones involve an inhibition of the MITF/CXCL1 axis and highlight the key role of this specific pathway in melanoma malignancy. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
32. The transcriptional coactivator peroxisome proliferator activated receptor (PPAR)gamma coactivator-1 alpha and the nuclear receptor PPAR alpha control the expression of glycerol kinase and metabolism genes independently of PPAR gamma activation in human white adipocytes.
- Author
-
Mazzucotelli A, Viguerie N, Tiraby C, Annicotte JS, Mairal A, Klimcakova E, Lepin E, Delmar P, Dejean S, Tavernier G, Lefort C, Hidalgo J, Pineau T, Fajas L, Clément K, and Langin D
- Abstract
OBJECTIVE: The purpose of this work was to determine the pattern of genes regulated by peroxisome proliferator-activated receptor (PPAR) gamma coactivator 1 alpha (PGC-1 alpha) in human adipocytes and the involvement of PPARalpha and PPARgamma in PGC-1 alpha transcriptional action. RESEARCH DESIGN AND METHODS: Primary cultures of human adipocytes were transduced with a PGC-1 alpha adenovirus and treated with PPARgamma and PPARalpha agonists. Variation in gene expression was assessed using pangenomic microarrays and quantitative RT-PCR. To investigate glycerol kinase (GyK), a target of PGC-1 alpha, we measured enzymatic activity and glycerol incorporation into triglycerides. In vivo studies were performed on wild-type and PPARalpha(-/-) mice. The GyK promoter was studied using chromatin immunoprecipitation and promoter reporter gene assays. RESULTS: Among the large number of genes regulated by PGC-1 alpha independently of PPARgamma, new targets involved in metabolism included the gene encoding GyK. The induction of GyK by PGC-1 alpha was observed at the levels of mRNA, enzymatic activity, and glycerol incorporation into triglycerides. PPARalpha was also upregulated by PGC-1 alpha. Its activation led to an increase in GyK expression and activity. PPARalpha was shown to bind and activate the GyK promoter. Experiments in mice confirmed the role of PGC-1 alpha and PPARalpha in the regulation of GyK in vivo. CONCLUSIONS: This work uncovers novel pathways regulated by PGC-1 alpha and reveals that PPARalpha controls gene expression in human white adipocytes. The induction of GyK by PGC-1 alpha and PPARalpha may promote a futile cycle of triglyceride hydrolysis and fatty acid reesterification. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
33. Impact of the Peroxisome Proliferator Activated Receptor γ2Pro12Ala polymorphism on adiposity, lipids and non-insulin-dependent diabetes mellitus.
- Author
-
Meirhaeghe, A, Fajas, L, Helbecque, N, Cottel, D, Auwerx, J, Deeb, S S, and Amouyel, P
- Published
- 2000
- Full Text
- View/download PDF
34. PPARt3 mRNA: a distinct PPARty mRNA subtype transcribed from an independent promoter (FEBS 21031)
- Author
-
Fajas, L., Fruchart, J.-C., and Auwerx, J.
- Published
- 1998
- Full Text
- View/download PDF
35. Peroxisome proliferator-activated receptor gamma and the transcriptional control of adipogenesis and metabolism
- Author
-
Fajas, L., Auwerx, J., Bray, G.A., and Bouchard, C.
36. 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
37. 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
38. 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
39. 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
40. 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
41. 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
42. 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
43. 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
44. 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
45. 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
46. 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
47. 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
48. 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
49. 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
50. 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
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.