Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Genes Dev ; 34(7-8): 544-559, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32079653

ABSTRACT

Excessive reactive oxygen species (ROS) can cause oxidative stress and consequently cell injury contributing to a wide range of diseases. Addressing the critical gaps in our understanding of the adaptive molecular events downstream ROS provocation holds promise for the identification of druggable metabolic vulnerabilities. Here, we unveil a direct molecular link between the activity of two estrogen-related receptor (ERR) isoforms and the control of glutamine utilization and glutathione antioxidant production. ERRα down-regulation restricts glutamine entry into the TCA cycle, while ERRγ up-regulation promotes glutamine-driven glutathione production. Notably, we identify increased ERRγ expression/activation as a hallmark of oxidative stress triggered by mitochondrial disruption or chemotherapy. Enhanced tumor antioxidant capacity is an underlying feature of human breast cancer (BCa) patients that respond poorly to treatment. We demonstrate that pharmacological inhibition of ERRγ with the selective inverse agonist GSK5182 increases antitumor efficacy of the chemotherapeutic paclitaxel on poor outcome BCa tumor organoids. Our findings thus underscore the ERRs as novel redox sensors and effectors of a ROS defense program and highlight the potential therapeutic advantage of exploiting ERRγ inhibitors for the treatment of BCa and other diseases where oxidative stress plays a central role.


Subject(s)
Breast Neoplasms/physiopathology , Drug Resistance, Neoplasm/drug effects , Oxidative Stress , Reactive Oxygen Species/metabolism , Receptors, Estrogen/metabolism , Signal Transduction/physiology , Animals , Antineoplastic Agents/pharmacology , Biosensing Techniques , Breast Neoplasms/drug therapy , Female , Gene Expression Regulation, Neoplastic/drug effects , Glutamine/metabolism , Glutathione/metabolism , Humans , Mice , Oxidative Stress/drug effects , Oxidative Stress/physiology , Paclitaxel/pharmacology , Receptors, Estrogen/genetics , Rotenone/pharmacology , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , ERRalpha Estrogen-Related Receptor
2.
Proc Natl Acad Sci U S A ; 119(35): e2121251119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994670

ABSTRACT

GCN2 (general control nonderepressible 2) is a serine/threonine-protein kinase that controls messenger RNA translation in response to amino acid availability and ribosome stalling. Here, we show that GCN2 controls erythrocyte clearance and iron recycling during stress. Our data highlight the importance of liver macrophages as the primary cell type mediating these effects. During different stress conditions, such as hemolysis, amino acid deficiency or hypoxia, GCN2 knockout (GCN2-/-) mice displayed resistance to anemia compared with wild-type (GCN2+/+) mice. GCN2-/- liver macrophages exhibited defective erythrophagocytosis and lysosome maturation. Molecular analysis of GCN2-/- cells demonstrated that the ATF4-NRF2 pathway is a critical downstream mediator of GCN2 in regulating red blood cell clearance and iron recycling.


Subject(s)
Amino Acids , Erythrocytes , Iron , Liver , Macrophages , Protein Serine-Threonine Kinases , Activating Transcription Factor 4/metabolism , Amino Acids/deficiency , Amino Acids/metabolism , Anemia/metabolism , Animals , Cytophagocytosis , Erythrocytes/metabolism , Gene Deletion , Hemolysis , Hypoxia/metabolism , Iron/metabolism , Liver/cytology , Lysosomes/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Stress, Physiological
3.
Genes Dev ; 31(12): 1228-1242, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28724614

ABSTRACT

Androgen receptor (AR) signaling reprograms cellular metabolism to support prostate cancer (PCa) growth and survival. Another key regulator of cellular metabolism is mTOR, a kinase found in diverse protein complexes and cellular localizations, including the nucleus. However, whether nuclear mTOR plays a role in PCa progression and participates in direct transcriptional cross-talk with the AR is unknown. Here, via the intersection of gene expression, genomic, and metabolic studies, we reveal the existence of a nuclear mTOR-AR transcriptional axis integral to the metabolic rewiring of PCa cells. Androgens reprogram mTOR-chromatin associations in an AR-dependent manner in which activation of mTOR-dependent metabolic gene networks is essential for androgen-induced aerobic glycolysis and mitochondrial respiration. In models of castration-resistant PCa cells, mTOR was capable of transcriptionally regulating metabolic gene programs in the absence of androgens, highlighting a potential novel castration resistance mechanism to sustain cell metabolism even without a functional AR. Remarkably, we demonstrate that increased mTOR nuclear localization is indicative of poor prognosis in patients, with the highest levels detected in castration-resistant PCa tumors and metastases. Identification of a functional mTOR targeted multigene signature robustly discriminates between normal prostate tissues, primary tumors, and hormone refractory metastatic samples but is also predictive of cancer recurrence. This study thus underscores a paradigm shift from AR to nuclear mTOR as being the master transcriptional regulator of metabolism in PCa.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/physiopathology , Receptors, Androgen/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Androgens/metabolism , Cell Nucleus/metabolism , DNA/metabolism , Disease Progression , Humans , Male , Protein Binding , TOR Serine-Threonine Kinases/genetics , Transcription, Genetic
4.
J Biol Chem ; 298(9): 102277, 2022 09.
Article in English | MEDLINE | ID: mdl-35863436

ABSTRACT

La-related protein 1 (LARP1) has been identified as a key translational inhibitor of terminal oligopyrimidine (TOP) mRNAs downstream of the nutrient sensing protein kinase complex, mTORC1. LARP1 exerts this inhibitory effect on TOP mRNA translation by binding to the mRNA cap and the adjacent 5'TOP motif, resulting in the displacement of the cap-binding protein eIF4E from TOP mRNAs. However, the involvement of additional signaling pathway in regulating LARP1-mediated inhibition of TOP mRNA translation is largely unexplored. In the present study, we identify a second nutrient sensing kinase GCN2 that converges on LARP1 to control TOP mRNA translation. Using chromatin-immunoprecipitation followed by massive parallel sequencing (ChIP-seq) analysis of activating transcription factor 4 (ATF4), an effector of GCN2 in nutrient stress conditions, in WT and GCN2 KO mouse embryonic fibroblasts, we determined that LARP1 is a GCN2-dependent transcriptional target of ATF4. Moreover, we identified GCN1, a GCN2 activator, participates in a complex with LARP1 on stalled ribosomes, suggesting a role for GCN1 in LARP1-mediated translation inhibition in response to ribosome stalling. Therefore, our data suggest that the GCN2 pathway controls LARP1 activity via two mechanisms: ATF4-dependent transcriptional induction of LARP1 mRNA and GCN1-mediated recruitment of LARP1 to stalled ribosomes.


Subject(s)
Amino Acids , Protein Biosynthesis , Protein Serine-Threonine Kinases , RNA 5' Terminal Oligopyrimidine Sequence , RNA, Messenger , RNA-Binding Proteins , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Amino Acids/metabolism , Animals , Cell Culture Techniques , Chromatin Immunoprecipitation , Eukaryotic Initiation Factor-4E/metabolism , Fibroblasts , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
5.
Genes Dev ; 24(6): 537-42, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20194433

ABSTRACT

Estrogen-related receptor alpha (ERRalpha) and proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha) play central roles in the transcriptional control of energy homeostasis, but little is known about factors regulating their activity. Here we identified the homeobox protein prospero-related homeobox 1 (Prox1) as one such factor. Prox1 interacts with ERRalpha and PGC-1alpha, occupies promoters of metabolic genes on a genome-wide scale, and inhibits the activity of the ERRalpha/PGC-1alpha complex. DNA motif analysis suggests that Prox1 interacts with the genome through tethering to ERRalpha and other factors. Importantly, ablation of Prox1 and ERRalpha have opposite effects on the respiratory capacity of liver cells, revealing an unexpected role for Prox1 in the control of energy homeostasis.


Subject(s)
Energy Metabolism , Homeodomain Proteins/metabolism , Receptors, Estrogen/metabolism , Trans-Activators/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Gene Expression Regulation , Genome-Wide Association Study , Hep G2 Cells , Homeodomain Proteins/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Receptors, Estrogen/genetics , Regulon/genetics , Trans-Activators/genetics , Transcription Factors , Tumor Suppressor Proteins/genetics , ERRalpha Estrogen-Related Receptor
6.
Proc Natl Acad Sci U S A ; 110(44): 17975-80, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24127579

ABSTRACT

Estrogen-related receptor α (ERRα) is a key regulator of mitochondrial function and metabolism essential for energy-driven cellular processes in both normal and cancer cells. ERRα has also been shown to mediate bone-derived macrophage activation by proinflammatory cytokines. However, the role of ERRα in cancer in which inflammation acts as a tumor promoter has yet to be investigated. Herein we show that global loss of ERRα accelerates the development of diethylnitrosamine (DEN)-induced hepatocellular carcinoma. Biochemical and metabolomics studies revealed that loss of ERRα promotes hepatocyte necrosis over apoptosis in response to DEN due to a deficiency in energy production. We further show that increased hepatocyte death and associated compensatory proliferation observed in DEN-injured ERRα-null livers is concomitant with increased nuclear factor κB (NF-κB)-dependent transcriptional control of cytokine expression in Kupffer cells. In particular, we demonstrate that loss of ERRα-dependent regulation of the NF-κB inhibitor IκBα leads to enhanced NF-κB activity and cytokine gene activation. Our work thus shows that global loss of ERRα activity promotes hepatocellular carcinoma by independent but synergistic mechanisms in hepatocytes and Kupffer cells, implying that pharmacological manipulation of ERRα activity may have a significant clinical impact on carcinogen-induced cancers.


Subject(s)
Carcinogenesis/metabolism , Hepatocytes/pathology , Liver Neoplasms/metabolism , Receptors, Estrogen/deficiency , Animals , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Chromatography, Liquid , DNA Primers/genetics , Diethylnitrosamine/administration & dosage , Diethylnitrosamine/toxicity , I-kappa B Proteins/metabolism , Immunohistochemistry , Kupffer Cells , Liver Neoplasms/chemically induced , Metabolomics , Mice , Mice, Knockout , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Necrosis , Real-Time Polymerase Chain Reaction , Receptors, Estrogen/genetics , Tandem Mass Spectrometry , ERRalpha Estrogen-Related Receptor
7.
PLoS Genet ; 7(6): e1002143, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21731503

ABSTRACT

Metabolic homeostasis and circadian rhythms are closely intertwined biological processes. Nuclear receptors, as sensors of hormonal and nutrient status, are actively implicated in maintaining this physiological relationship. Although the orphan nuclear receptor estrogen-related receptor α (ERRα, NR3B1) plays a central role in the control of energy metabolism and its expression is known to be cyclic in the liver, its role in temporal control of metabolic networks is unknown. Here we report that ERRα directly regulates all major components of the molecular clock. ERRα-null mice also display deregulated locomotor activity rhythms and circadian period lengths under free-running conditions, as well as altered circulating diurnal bile acid and lipid profiles. In addition, the ERRα-null mice exhibit time-dependent hypoglycemia and hypoinsulinemia, suggesting a role for ERRα in modulating insulin sensitivity and glucose handling during the 24-hour light/dark cycle. We also provide evidence that the newly identified ERRα corepressor PROX1 is implicated in rhythmic control of metabolic outputs. To help uncover the molecular basis of these phenotypes, we performed genome-wide location analyses of binding events by ERRα, PROX1, and BMAL1, an integral component of the molecular clock. These studies revealed the existence of transcriptional regulatory loops among ERRα, PROX1, and BMAL1, as well as extensive overlaps in their target genes, implicating these three factors in the control of clock and metabolic gene networks in the liver. Genomic convergence of ERRα, PROX1, and BMAL1 transcriptional activity thus identified a novel node in the molecular circuitry controlling the daily timing of metabolic processes.


Subject(s)
Homeodomain Proteins/metabolism , Liver/metabolism , Receptors, Estrogen/metabolism , Tumor Suppressor Proteins/metabolism , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , Bile Acids and Salts/blood , Blood Glucose/analysis , Blotting, Western , CLOCK Proteins/metabolism , COS Cells , Chlorocebus aethiops , Cholesterol/blood , Circadian Rhythm , Gene Expression Profiling , Gene Expression Regulation , Gluconeogenesis , Glycolysis , Hep G2 Cells , Homeodomain Proteins/genetics , Homeostasis , Humans , Insulin/blood , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Photoperiod , Promoter Regions, Genetic , Protein Binding , RNA Interference , Receptors, Estrogen/genetics , Triglycerides/blood , Tumor Suppressor Proteins/genetics , ERRalpha Estrogen-Related Receptor
8.
Cell Rep ; 43(1): 113615, 2024 01 23.
Article in English | MEDLINE | ID: mdl-38159280

ABSTRACT

The integrated stress response (ISR) is critical for cell survival under stress. In response to diverse environmental cues, eIF2α becomes phosphorylated, engendering a dramatic change in mRNA translation. The activation of ISR plays a pivotal role in the early embryogenesis, but the eIF2-dependent translational landscape in pluripotent embryonic stem cells (ESCs) is largely unexplored. We employ a multi-omics approach consisting of ribosome profiling, proteomics, and metabolomics in wild-type (eIF2α+/+) and phosphorylation-deficient mutant eIF2α (eIF2αA/A) mouse ESCs (mESCs) to investigate phosphorylated (p)-eIF2α-dependent translational control of naive pluripotency. We show a transient increase in p-eIF2α in the naive epiblast layer of E4.5 embryos. Absence of eIF2α phosphorylation engenders an exit from naive pluripotency following 2i (two chemical inhibitors of MEK1/2 and GSK3α/ß) withdrawal. p-eIF2α controls translation of mRNAs encoding proteins that govern pluripotency, chromatin organization, and glutathione synthesis. Thus, p-eIF2α acts as a key regulator of the naive pluripotency gene regulatory network.


Subject(s)
Mouse Embryonic Stem Cells , Pluripotent Stem Cells , Animals , Mice , Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/metabolism , Phosphorylation , Pluripotent Stem Cells/metabolism , RNA, Messenger/metabolism , Eukaryotic Initiation Factor-2/metabolism
9.
Mol Metab ; 78: 101814, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37802398

ABSTRACT

OBJECTIVE: Estrogen related receptor α (ERRα) occupies a central node in the transcriptional control of energy metabolism, including in skeletal muscle, but whether modulation of its activity can directly contribute to extend endurance to exercise remains to be investigated. The goal of this study was to characterize the benefit of mice engineered to express a physiologically relevant activated form of ERRα on skeletal muscle exercise metabolism and performance. METHODS: We recently shown that mutational inactivation of three regulated phosphosites in the amino terminal domain of the nuclear receptor ERRα impedes its degradation, leading to an accumulation of ERRα proteins and perturbation of metabolic homeostasis in ERRα3SA mutant mice. Herein, we used a multi-omics approach in combination with physical endurance tests to ascertain the consequences of expressing the constitutively active phospho-deficient ERRα3SA form on muscle exercise performance and energy metabolism. RESULTS: Genetic heightening of ERRα activity enhanced exercise capacity, fatigue-resistance, and endurance. This phenotype resulted from extensive reprogramming of ERRα global DNA occupancy and transcriptome in muscle leading to an increase in oxidative fibers, mitochondrial biogenesis, fatty acid oxidation, and lactate homeostasis. CONCLUSION: Our findings support the potential to enhance physical performance and exercise-induced health benefits by targeting molecular pathways regulating ERRα transcriptional activity.


Subject(s)
Muscle, Skeletal , Running , Mice , Animals , Muscle, Skeletal/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Gene Expression Regulation , ERRalpha Estrogen-Related Receptor
10.
Mol Cancer Res ; 21(10): 1050-1063, 2023 10 02.
Article in English | MEDLINE | ID: mdl-37409967

ABSTRACT

Dysregulation of mTOR signaling plays a critical role in promoting prostate cancer growth. HOXB13, a homeodomain transcription factor, is known to influence the androgen response and prostate cancer development. Recently, HOXB13 was found to complex with mTOR on chromatin. However, the functional crosstalk between HOXB13 and mTOR remains elusive. We now report that mTOR directly interacts with and hierarchically phosphorylates HOXB13 at threonine 8 and 41 then serine 31 to promote its interaction with the E3 ligase SKP2 while enhancing its oncogenic properties. Expression of HOXB13 harboring phosphomimetic mutations at the mTOR-targeted sites stimulates prostate cancer cellular growth both in vitro and in murine xenografts. Transcriptional profiling studies revealed a phospho-HOXB13-dependent gene signature capable of robustly discriminating between normal prostate tissues, primary and metastatic prostate cancer samples. This work uncovers a previously unanticipated molecular cascade by which mTOR directly phosphorylates HOXB13 to dictate a specific gene program with oncogenic implications in prostate cancer. IMPLICATIONS: Control of HOXB13 transcriptional activity via its direct phosphorylation by the mTOR kinase is a potential therapeutic avenue for the management of advanced prostate cancer.


Subject(s)
Homeodomain Proteins , Prostatic Neoplasms , Male , Humans , Animals , Mice , Phosphorylation , Cell Line, Tumor , Cell Proliferation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Prostatic Neoplasms/pathology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
11.
Nat Commun ; 14(1): 6982, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37914694

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is epidemiologically associated with obesity and diabetes and can lead to liver cirrhosis and hepatocellular carcinoma if left untreated. The intricate signaling pathways that orchestrate hepatocyte energy metabolism and cellular stress, intrahepatic cell crosstalk, as well as interplay between peripheral tissues remain elusive and are crucial for the development of anti-NASH therapies. Herein, we reveal E3 ligase FBXW7 as a key factor regulating hepatic catabolism, stress responses, systemic energy homeostasis, and NASH pathogenesis with attenuated FBXW7 expression as a feature of advanced NASH. Multiomics and pharmacological intervention showed that FBXW7 loss-of-function in hepatocytes disrupts a metabolic transcriptional axis conjointly controlled by the nutrient-sensing nuclear receptors ERRα and PPARα, resulting in suppression of fatty acid oxidation, elevated ER stress, apoptosis, immune infiltration, fibrogenesis, and ultimately NASH progression in male mice. These results provide the foundation for developing alternative strategies co-targeting ERRα and PPARα for the treatment of NASH.


Subject(s)
Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Animals , Male , Mice , F-Box-WD Repeat-Containing Protein 7/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , Hepatocytes/metabolism , Homeostasis , Liver/metabolism , Liver Neoplasms/metabolism , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Nutrients , PPAR alpha/genetics , PPAR alpha/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism
12.
Cell Metab ; 6(1): 25-37, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17618854

ABSTRACT

Downregulation and functional deactivation of the transcriptional coactivator PGC-1alpha has been implicated in heart failure pathogenesis. We hypothesized that the estrogen-related receptor alpha (ERRalpha), which recruits PGC-1alpha to metabolic target genes in heart, exerts protective effects in the context of stressors known to cause heart failure. ERRalpha(-/-) mice subjected to left ventricular (LV) pressure overload developed signatures of heart failure including chamber dilatation and reduced LV fractional shortening. (31)P-NMR studies revealed abnormal phosphocreatine depletion in ERRalpha(-/-) hearts subjected to hemodynamic stress, indicative of a defect in ATP reserve. Mitochondrial respiration studies demonstrated reduced maximal ATP synthesis rates in ERRalpha(-/-) hearts. Cardiac ERRalpha target genes involved in energy substrate oxidation, ATP synthesis, and phosphate transfer were downregulated in ERRalpha(-/-) mice at baseline or with pressure overload. These results demonstrate that the nuclear receptor ERRalpha is required for the adaptive bioenergetic response to hemodynamic stressors known to cause heart failure.


Subject(s)
Heart/physiopathology , Receptors, Estrogen/physiology , Ventricular Pressure/physiology , Ventricular Remodeling/physiology , Adaptation, Physiological , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Biomarkers/metabolism , Blood Pressure , Cardiac Output, Low , Cardiomegaly/physiopathology , Energy Metabolism , Female , Gene Expression Profiling , Heart/embryology , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , ERRalpha Estrogen-Related Receptor
13.
Cell Metab ; 6(1): 13-24, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17618853

ABSTRACT

At birth, the heart undergoes a critical metabolic switch from a predominant dependence on carbohydrates during fetal life to a greater dependence on postnatal oxidative metabolism. This remains the principle metabolic state throughout life, although pathologic conditions such as heart failure and cardiac hypertrophy reactivate components of the fetal genetic program to increase carbohydrate utilization. Disruption of the ERRgamma gene (Esrrg), which is expressed at high levels in the fetal and postnatal mouse heart, blocks this switch, resulting in lactatemia, electrocardiographic abnormalities, and death during the first week of life. Genomic ChIP-on-chip and expression analysis identifies ERRgamma as both a direct and an indirect regulator of a nuclear-encoded mitochondrial genetic network that coordinates the postnatal metabolic transition. These findings reveal an unexpected and essential molecular genetic component of the oxidative metabolic gene program in the heart and highlight ERRgamma in the study of cardiac hypertrophy and failure.


Subject(s)
Cardiomegaly/metabolism , Electron Transport/physiology , Energy Metabolism/physiology , Gene Expression Regulation, Developmental/physiology , Genes, Mitochondrial/physiology , Heart/embryology , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Estrogen/physiology , Animals , Cardiomegaly/pathology , Chromatin Immunoprecipitation , Electrocardiography , Female , Gene Expression Profiling , Heart/physiopathology , Lactates/blood , Male , Mice , Mice, Knockout , Myocytes, Cardiac , Oligonucleotide Array Sequence Analysis , Patch-Clamp Techniques , Sodium/metabolism , Ventricular Function
14.
Cell Metab ; 5(5): 345-56, 2007 May.
Article in English | MEDLINE | ID: mdl-17488637

ABSTRACT

Orphan nuclear receptor ERRalpha (NR3B1) is recognized as a key regulator of mitochondrial biogenesis, but it is not known whether ERRalpha and other ERR isoforms play a broader role in cardiac energetics and function. We used genome-wide location analysis and expression profiling to appraise the role of ERRalpha and gamma (NR3B3) in the adult heart. Our data indicate that the two receptors, acting as nonobligatory heterodimers, target a common set of promoters involved in the uptake of energy substrates, production and transport of ATP across the mitochondrial membranes, and intracellular fuel sensing, as well as Ca(2+) handling and contractile work. Motif-finding algorithms assisted by functional studies indicated that ERR target promoters are enriched for NRF-1, CREB, and STAT3 binding sites. Our study thus reveals that the ERRs orchestrate a comprehensive cardiac transcriptional program and further suggests that modulation of ERR activities could be used to manage cardiomyopathies.


Subject(s)
Gene Expression Regulation , Genome/genetics , Heart/physiology , Promoter Regions, Genetic/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/metabolism , Animals , Chromatin Immunoprecipitation , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Profiling , Male , Mice , Mice, Knockout , NF-E2-Related Factor 1/metabolism , Promoter Regions, Genetic/genetics , STAT3 Transcription Factor/metabolism , ERRalpha Estrogen-Related Receptor
15.
STAR Protoc ; 3(2): 101434, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35693211

ABSTRACT

Rapid immunoprecipitation mass spectrometry of endogenous protein (RIME) is a technique to study protein complexes on chromatin. The protocol below describes specific steps for RIME analysis of the male human-derived prostate cancer cell line LNCaP. This approach can also be applied to other prostate cancer cell lines such as 22Rv1, DU145, and PC3. For other cell types, we recommend optimizing the number of cell culture plates to ensure adequate sample for mass spectrometry protein detection. For complete details on the use and execution of this protocol, please refer to Mohammed et al. (2016) and Dufour et al. (2022).


Subject(s)
Chromatin , Prostatic Neoplasms , Humans , Immunoprecipitation , Male , Mass Spectrometry/methods , Prostate/metabolism , Prostatic Neoplasms/metabolism
16.
Nat Commun ; 13(1): 2105, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440636

ABSTRACT

Insulin resistance, a harbinger of the metabolic syndrome, is a state of compromised hormonal response resulting from the dysregulation of a wide range of insulin-controlled cellular processes. However, how insulin affects cellular energy metabolism via long-term transcriptional regulation and whether boosting mitochondrial function alleviates insulin resistance remains to be elucidated. Herein we reveal that insulin directly enhances the activity of the nuclear receptor ERRα via a GSK3ß/FBXW7 signaling axis. Liver-specific deletion of GSK3ß or FBXW7 and mice harboring mutations of ERRα phosphosites (ERRα3SA) co-targeted by GSK3ß/FBXW7 result in accumulated ERRα proteins that no longer respond to fluctuating insulin levels. ERRα3SA mice display reprogrammed liver and muscle transcriptomes, resulting in compromised energy homeostasis and reduced insulin sensitivity despite improved mitochondrial function. This crossroad of insulin signaling and transcriptional control by a nuclear receptor offers a framework to better understand the complex cellular processes contributing to the development of insulin resistance.


Subject(s)
Insulin Resistance , Animals , F-Box-WD Repeat-Containing Protein 7/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Insulin/metabolism , Insulin Resistance/genetics , Mice , Receptors, Estrogen/metabolism , ERRalpha Estrogen-Related Receptor
17.
Cell Rep ; 38(12): 110534, 2022 03 22.
Article in English | MEDLINE | ID: mdl-35320709

ABSTRACT

A growing number of studies support a direct role for nuclear mTOR in gene regulation and chromatin structure. Still, the scarcity of known chromatin-bound mTOR partners limits our understanding of how nuclear mTOR controls transcription. Herein, comprehensive mapping of the mTOR chromatin-bound interactome in both androgen-dependent and -independent cellular models of prostate cancer (PCa) identifies a conserved 67-protein interaction network enriched for chromatin modifiers, transcription factors, and SUMOylation machinery. SUMO2/3 and nuclear pore protein NUP210 are among the strongest interactors, while the androgen receptor (AR) is the dominant androgen-inducible mTOR partner. Further investigation reveals that NUP210 facilitates mTOR nuclear trafficking, that mTOR and AR form a functional transcriptional module with the nucleosome remodeling and deacetylase (NuRD) complex, and that androgens specify mTOR-SUMO2/3 promoter-enhancer association. This work identifies a vast network of mTOR-associated nuclear complexes advocating innovative molecular strategies to modulate mTOR-dependent gene regulation with conceivable implications for PCa and other diseases.


Subject(s)
Chromatin , Prostatic Neoplasms , Androgens/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , TOR Serine-Threonine Kinases/metabolism
18.
Commun Biol ; 5(1): 955, 2022 09 12.
Article in English | MEDLINE | ID: mdl-36097051

ABSTRACT

Functional oncogenic links between ErbB2 and ERRα in HER2+ breast cancer patients support a therapeutic benefit of co-targeted therapies. However, ErbB2 and ERRα also play key roles in heart physiology, and this approach could pose a potential liability to cardiovascular health. Herein, using integrated phosphoproteomic, transcriptomic and metabolic profiling, we uncovered molecular mechanisms associated with the adverse remodeling of cardiac functions in mice with combined attenuation of ErbB2 and ERRα activity. Genetic disruption of both effectors results in profound effects on cardiomyocyte architecture, inflammatory response and metabolism, the latter leading to a decrease in fatty acyl-carnitine species further increasing the reliance on glucose as a metabolic fuel, a hallmark of failing hearts. Furthermore, integrated omics signatures of ERRα loss-of-function and doxorubicin treatment exhibit common features of chemotherapeutic cardiotoxicity. These findings thus reveal potential cardiovascular risks in discrete combination therapies in the treatment of breast and other cancers.


Subject(s)
Receptors, Estrogen , Ventricular Remodeling , Animals , Doxorubicin/pharmacology , Mice , Myocytes, Cardiac/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , ERRalpha Estrogen-Related Receptor
19.
Sci Rep ; 11(1): 21268, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34711912

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is the most frequent liver disease worldwide and can progress to non-alcoholic steatohepatitis (NASH), which is characterized by triglyceride accumulation, inflammation, and fibrosis. No pharmacological agents are currently approved to treat these conditions, but it is clear now that modulation of lipid synthesis and autophagy are key biological mechanisms that could help reduce or prevent these liver diseases. The folliculin (FLCN) protein has been recently identified as a central regulatory node governing whole body energy homeostasis, and we hypothesized that FLCN regulates highly metabolic tissues like the liver. We thus generated a liver specific Flcn knockout mouse model to study its role in liver disease progression. Using the methionine- and choline-deficient diet to mimic liver fibrosis, we demonstrate that loss of Flcn reduced triglyceride accumulation, fibrosis, and inflammation in mice. In this aggressive liver disease setting, loss of Flcn led to activation of transcription factors TFEB and TFE3 to promote autophagy, promoting the degradation of intracellular lipid stores, ultimately resulting in reduced hepatocellular damage and inflammation. Hence, the activity of FLCN could be a promising target for small molecule drugs to treat liver fibrosis by specifically activating autophagy. Collectively, these results show an unexpected role for Flcn in fatty liver disease progression and highlight new potential treatment strategies.


Subject(s)
Autophagy/genetics , Hepatitis/etiology , Hepatitis/metabolism , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Proto-Oncogene Proteins/deficiency , Signal Transduction , Tumor Suppressor Proteins/deficiency , Animals , Biomarkers , Biopsy , Computational Biology , Diet, High-Fat , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Genetic Predisposition to Disease , Hepatitis/pathology , Immunohistochemistry , Liver Cirrhosis/pathology , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Transcriptome
20.
Oncogene ; 39(41): 6406-6420, 2020 10.
Article in English | MEDLINE | ID: mdl-32855526

ABSTRACT

DNA methylation is implicated in the acquisition of malignant phenotypes, and the use of epigenetic modulating drugs is a promising anti-cancer therapeutic strategy. 5-aza-2'deoxycytidine (decitabine, 5-azadC) is an FDA-approved DNA methyltransferase (DNMT) inhibitor with proven effectiveness against hematological malignancies and more recently triple-negative breast cancer (BC). Herein, genetic or pharmacological studies uncovered a hitherto unknown feedforward molecular link between DNMT1 and the estrogen related receptor α (ERRα), a key transcriptional regulator of cellular metabolism. Mechanistically, DNMT1 promotes ERRα stability which in turn couples DNMT1 transcription with that of the methionine cycle and S-adenosylmethionine synthesis to drive DNA methylation. In vitro and in vivo investigation using a pre-clinical mouse model of BC demonstrated a clear therapeutic advantage for combined administration of the ERRα inhibitor C29 with 5-azadC. A large-scale bisulfite genomic sequencing analysis revealed specific methylation perturbations fostering the discovery that reversal of promoter hypermethylation and consequently derepression of the tumor suppressor gene, IRF4, is a factor underlying the observed BC suppressive effects. This work thus uncovers a critical role of ERRα in the crosstalk between transcriptional control of metabolism and epigenetics and illustrates the potential for targeting ERRα in combination with DNMT inhibitors for BC treatment and other epigenetics-driven malignancies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Interferon Regulatory Factors/genetics , Receptors, Estrogen/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , CpG Islands/genetics , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation/drug effects , Decitabine/pharmacology , Decitabine/therapeutic use , Epigenesis, Genetic/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Promoter Regions, Genetic/genetics , Protein Stability , Receptors, Estrogen/antagonists & inhibitors , S-Adenosylmethionine/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays , ERRalpha Estrogen-Related Receptor
SELECTION OF CITATIONS
SEARCH DETAIL