Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Mol Cell ; 75(4): 807-822.e8, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442424

ABSTRACT

mTORC2 controls glucose and lipid metabolism, but the mechanisms are unclear. Here, we show that conditionally deleting the essential mTORC2 subunit Rictor in murine brown adipocytes inhibits de novo lipid synthesis, promotes lipid catabolism and thermogenesis, and protects against diet-induced obesity and hepatic steatosis. AKT kinases are the canonical mTORC2 substrates; however, deleting Rictor in brown adipocytes appears to drive lipid catabolism by promoting FoxO1 deacetylation independently of AKT, and in a pathway distinct from its positive role in anabolic lipid synthesis. This facilitates FoxO1 nuclear retention, enhances lipid uptake and lipolysis, and potentiates UCP1 expression. We provide evidence that SIRT6 is the FoxO1 deacetylase suppressed by mTORC2 and show an endogenous interaction between SIRT6 and mTORC2 in both mouse and human cells. Our findings suggest a new paradigm of mTORC2 function filling an important gap in our understanding of this more mysterious mTOR complex.


Subject(s)
Adipocytes, Brown/metabolism , Forkhead Box Protein O1/metabolism , Lipolysis , Mechanistic Target of Rapamycin Complex 2/metabolism , Sirtuins/metabolism , Adipocytes, Brown/cytology , Animals , Forkhead Box Protein O1/genetics , HEK293 Cells , HeLa Cells , Humans , Mechanistic Target of Rapamycin Complex 2/genetics , Mice , Mice, Transgenic , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Sirtuins/genetics
2.
Nat Chem Biol ; 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38480981

ABSTRACT

A common approach for understanding how drugs induce their therapeutic effects is to identify the genetic determinants of drug sensitivity. Because 'chemo-genetic profiles' are performed in a pooled format, inference of gene function is subject to several confounding influences related to variation in growth rates between clones. In this study, we developed Method for Evaluating Death Using a Simulation-assisted Approach (MEDUSA), which uses time-resolved measurements, along with model-driven constraints, to reveal the combination of growth and death rates that generated the observed drug response. MEDUSA is uniquely effective at identifying death regulatory genes. We apply MEDUSA to characterize DNA damage-induced lethality in the presence and absence of p53. Loss of p53 switches the mechanism of DNA damage-induced death from apoptosis to a non-apoptotic death that requires high respiration. These findings demonstrate the utility of MEDUSA both for determining the genetic dependencies of lethality and for revealing opportunities to potentiate chemo-efficacy in a cancer-specific manner.

3.
Cell ; 146(3): 408-20, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21816276

ABSTRACT

The nutrient- and growth factor-responsive kinase mTOR complex 1 (mTORC1) regulates many processes that control growth, including protein synthesis, autophagy, and lipogenesis. Through unknown mechanisms, mTORC1 promotes the function of SREBP, a master regulator of lipo- and sterolgenic gene transcription. Here, we demonstrate that mTORC1 regulates SREBP by controlling the nuclear entry of lipin 1, a phosphatidic acid phosphatase. Dephosphorylated, nuclear, catalytically active lipin 1 promotes nuclear remodeling and mediates the effects of mTORC1 on SREBP target gene, SREBP promoter activity, and nuclear SREBP protein abundance. Inhibition of mTORC1 in the liver significantly impairs SREBP function and makes mice resistant, in a lipin 1-dependent fashion, to the hepatic steatosis and hypercholesterolemia induced by a high-fat and -cholesterol diet. These findings establish lipin 1 as a key component of the mTORC1-SREBP pathway.


Subject(s)
Nuclear Proteins/metabolism , Proteins/metabolism , Signal Transduction , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Animals , Humans , Lipid Metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes , Phosphatidate Phosphatase , TOR Serine-Threonine Kinases
4.
Mol Cell ; 72(6): 917-919, 2018 12 20.
Article in English | MEDLINE | ID: mdl-30576653

ABSTRACT

In this issue of Molecular Cell, Krzysiak et al. (2018) describe a mechanism by which insulin signaling represses the NAD+-dependent SIRT1 deacetylase by promoting PACS-2 binding and provide structural clues to understanding how SIRT1 activating compounds (STACs) work.


Subject(s)
Insulin , Sirtuin 1 , Signal Transduction
5.
J Biol Chem ; 298(10): 102379, 2022 10.
Article in English | MEDLINE | ID: mdl-35973513

ABSTRACT

Mechanistic target of rapamycin (mTOR) complex 2 (mTORC2) regulates metabolism, cell proliferation, and cell survival. mTORC2 activity is stimulated by growth factors, and it phosphorylates the hydrophobic motif site of the AGC kinases AKT, SGK, and PKC. However, the proteins that interact with mTORC2 to control its activity and localization remain poorly defined. To identify mTORC2-interacting proteins in living cells, we tagged endogenous RICTOR, an essential mTORC2 subunit, with the modified BirA biotin ligase BioID2 and performed live-cell proximity labeling. We identified 215 RICTOR-proximal proteins, including proteins with known mTORC2 pathway interactions, and 135 proteins (63%) not previously linked to mTORC2 signaling, including nuclear and cytoplasmic proteins. Our imaging and cell fractionation experiments suggest nearly 30% of RICTOR is in the nucleus, hinting at potential nuclear functions. We also identified 29 interactors containing RICTOR-dependent, insulin-stimulated phosphorylation sites, thus providing insight into mTORC2-dependent insulin signaling dynamics. Finally, we identify the endogenous ADP ribosylation factor 1 (ARF1) GTPase as an mTORC2-interacting protein. Through gain-of-function and loss-of-function studies, we provide functional evidence that ARF1 may negatively regulate mTORC2. In summary, we present a new method of studying endogenous mTORC2, a resource of RICTOR/mTORC2 protein interactions in living cells, and a potential mechanism of mTORC2 regulation by the ARF1 GTPase.


Subject(s)
ADP-Ribosylation Factor 1 , Protein Interaction Maps , Rapamycin-Insensitive Companion of mTOR Protein , TOR Serine-Threonine Kinases , Humans , ADP-Ribosylation Factor 1/metabolism , Insulin/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , TOR Serine-Threonine Kinases/metabolism , Protein Interaction Mapping/methods
6.
Immunity ; 39(6): 1043-56, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24315998

ABSTRACT

Naive T cells respond to antigen stimulation by exiting from quiescence and initiating clonal expansion and functional differentiation, but the control mechanism is elusive. Here we describe that Raptor-mTORC1-dependent metabolic reprogramming is a central determinant of this transitional process. Loss of Raptor abrogated T cell priming and T helper 2 (Th2) cell differentiation, although Raptor function is less important for continuous proliferation of actively cycling cells. mTORC1 coordinated multiple metabolic programs in T cells including glycolysis, lipid synthesis, and oxidative phosphorylation to mediate antigen-triggered exit from quiescence. mTORC1 further linked glucose metabolism to the initiation of Th2 cell differentiation by orchestrating cytokine receptor expression and cytokine responsiveness. Activation of Raptor-mTORC1 integrated T cell receptor and CD28 costimulatory signals in antigen-stimulated T cells. Our studies identify a Raptor-mTORC1-dependent pathway linking signal-dependent metabolic reprogramming to quiescence exit, and this in turn coordinates lymphocyte activation and fate decisions in adaptive immunity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation , Lymphocyte Activation/physiology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , TOR Serine-Threonine Kinases/metabolism , Th2 Cells/cytology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Gene Deletion , Glucose/metabolism , Mice , Mice, Inbred C57BL , Regulatory-Associated Protein of mTOR , Signal Transduction , TOR Serine-Threonine Kinases/genetics
7.
Mol Cell Proteomics ; 19(7): 1104-1119, 2020 07.
Article in English | MEDLINE | ID: mdl-32234964

ABSTRACT

Stimulating brown adipose tissue (BAT) activity represents a promising therapy for overcoming metabolic diseases. mTORC2 is important for regulating BAT metabolism, but its downstream targets have not been fully characterized. In this study, we apply proteomics and phosphoproteomics to investigate the downstream effectors of mTORC2 in brown adipocytes. We compare wild-type controls to isogenic cells with an induced knockout of the mTORC2 subunit RICTOR (Rictor-iKO) by stimulating each with insulin for a 30-min time course. In Rictor-iKO cells, we identify decreases to the abundance of glycolytic and de novo lipogenesis enzymes, and increases to mitochondrial proteins as well as a set of proteins known to increase upon interferon stimulation. We also observe significant differences to basal phosphorylation because of chronic RICTOR loss including decreased phosphorylation of the lipid droplet protein perilipin-1 in Rictor-iKO cells, suggesting that RICTOR could be involved with regulating basal lipolysis or droplet dynamics. Finally, we observe mild dampening of acute insulin signaling response in Rictor-iKO cells, and a subset of AKT substrates exhibiting statistically significant dependence on RICTOR.


Subject(s)
Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Insulin/pharmacokinetics , Mechanistic Target of Rapamycin Complex 2/metabolism , Proteome/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Animals , Chromatography, Liquid , Gene Knockout Techniques , Gene Ontology , Glycolysis/drug effects , Insulin/metabolism , Lipogenesis/drug effects , Mice , Mitochondria/drug effects , Phosphorylation , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Tandem Mass Spectrometry
8.
Nat Chem Biol ; 14(11): 1021-1031, 2018 11.
Article in English | MEDLINE | ID: mdl-30327559

ABSTRACT

Fatty acid synthase (FASN) predominantly generates straight-chain fatty acids using acetyl-CoA as the initiating substrate. However, monomethyl branched-chain fatty acids (mmBCFAs) are also present in mammals but are thought to be primarily diet derived. Here we demonstrate that mmBCFAs are de novo synthesized via mitochondrial BCAA catabolism, exported to the cytosol by adipose-specific expression of carnitine acetyltransferase (CrAT), and elongated by FASN. Brown fat exhibits the highest BCAA catabolic and mmBCFA synthesis fluxes, whereas these lipids are largely absent from liver and brain. mmBCFA synthesis is also sustained in the absence of microbiota. We identify hypoxia as a potent suppressor of BCAA catabolism that decreases mmBCFA synthesis in obese adipose tissue, such that mmBCFAs are significantly decreased in obese animals. These results identify adipose tissue mmBCFA synthesis as a novel link between BCAA metabolism and lipogenesis, highlighting roles for CrAT and FASN promiscuity influencing acyl-chain diversity in the lipidome.


Subject(s)
Adipose Tissue/enzymology , Amino Acids, Branched-Chain/metabolism , Fatty Acid Synthases/metabolism , Fatty Acids/biosynthesis , Obesity/enzymology , 3T3 Cells , Adipocytes/cytology , Animals , CRISPR-Cas Systems , Carnitine O-Acetyltransferase/metabolism , Cytosol/metabolism , Female , Hypoxia , Lentivirus/genetics , Lipogenesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , RNA, Small Interfering/metabolism
9.
Curr Diab Rep ; 19(11): 138, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31749022

ABSTRACT

PURPOSE OF REVIEW: Obesity is a major risk factor for type 2 diabetes. Although adipose tissue allows storage of excess calories in periods of overnutrition, in obesity, adipose tissue metabolism becomes dysregulated and can promote metabolic diseases. This review discusses recent advances in understandings how adipocyte metabolism impacts metabolic homeostasis. RECENT FINDINGS: The ability of adipocytes to synthesize lipids from glucose is a marker of metabolic fitness, e.g., low de novo lipogenesis (DNL) in adipocytes correlates with insulin resistance in obesity. Adipocyte DNL may promote synthesis of special "insulin sensitizing" signaling lipids that act hormonally. However, each metabolic intermediate in the DNL pathway (i.e., citrate, acetyl-CoA, malonyl-CoA, and palmitate) also has second messenger functions. Mounting evidence suggests these signaling functions may also be important for maintaining healthy adipocytes. While adipocyte DNL contributes to lipid storage, lipid precursors may have additional second messenger functions critical for maintaining adipocyte health, and thus systemic metabolic homeostasis.


Subject(s)
Adipocytes/physiology , Diabetes Mellitus, Type 2 , Insulin Resistance , Lipogenesis , Second Messenger Systems , Humans , Obesity/complications
10.
Nature ; 559(7712): 41-42, 2018 07.
Article in English | MEDLINE | ID: mdl-29959410
11.
Handb Exp Pharmacol ; 251: 3-36, 2019.
Article in English | MEDLINE | ID: mdl-30203328

ABSTRACT

Brown adipose tissue is well known to be a thermoregulatory organ particularly important in small rodents and human infants, but it was only recently that its existence and significance to metabolic fitness in adult humans have been widely realized. The ability of active brown fat to expend high amounts of energy has raised interest in stimulating thermogenesis therapeutically to treat metabolic diseases related to obesity and type 2 diabetes. In parallel, there has been a surge of research aimed at understanding the biology of rodent and human brown fat development, its remarkable metabolic properties, and the phenomenon of white fat browning, in which white adipocytes can be converted into brown like adipocytes with similar thermogenic properties. Here, we review the current understanding of the developmental and metabolic pathways involved in forming thermogenic adipocytes, and highlight some of the many unknown functions of brown fat that make its study a rich and exciting area for future research.


Subject(s)
Adipogenesis/physiology , Adipose Tissue, Brown , Adipose Tissue, White/metabolism , Diabetes Mellitus, Type 2 , Adult , Energy Metabolism , Humans , Thermogenesis/physiology
12.
Nucleic Acids Res ; 43(16): 7790-804, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26117538

ABSTRACT

Jumonji domain-containing protein 6 (JMJD6) is a nuclear protein involved in histone modification, transcription and RNA processing. Although JMJD6 is crucial for tissue development, the link between its molecular functions and its roles in any given differentiation process is unknown. We report that JMJD6 is required for adipogenic gene expression and differentiation in a manner independent of Jumonji C domain catalytic activity. JMJD6 knockdown led to a reduction of C/EBPß and C/EBPδ protein expression without affecting mRNA levels in the early phase of differentiation. However, ectopic expression of C/EBPß and C/EBPδ did not rescue differentiation. Further analysis demonstrated that JMJD6 was associated with the Pparγ2 and Cebpα loci and putative enhancers. JMJD6 was previously found associated with bromodomain and extra-terminal domain (BET) proteins, which can be targeted by the bromodomain inhibitor JQ1. JQ1 treatment prevented chromatin binding of JMJD6, Pparγ2 and Cebpα expression, and adipogenic differentiation, yet had no effect on C/EBPß and C/EBPδ expression or chromatin binding. These results indicate dual roles for JMJD6 in promoting adipogenic gene expression program by post-transcriptional regulation of C/EBPß and C/EBPδ and direct transcriptional activation of Pparγ2 and Cebpα during adipocyte differentiation.


Subject(s)
Adipocytes/metabolism , Adipogenesis/genetics , Gene Expression Regulation , Receptors, Cell Surface/metabolism , Adipose Tissue/metabolism , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line , Chromatin/metabolism , Female , Male , Mice , PPAR gamma/genetics , Protein Structure, Tertiary , RNA Polymerase II/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/physiology , Transcription, Genetic
13.
Biochim Biophys Acta ; 1842(3): 340-51, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23747579

ABSTRACT

The obesity epidemic has intensified efforts to understand the mechanisms controlling adipose tissue development. Adipose tissue is generally classified as white adipose tissue (WAT), the major energy storing tissue, or brown adipose tissue (BAT), which mediates non-shivering thermogenesis. It is hypothesized that brite adipocytes (brown in white) may represent a third adipocyte class. The recent realization that brown fat exist in adult humans suggests increasing brown fat energy expenditure could be a therapeutic strategy to combat obesity. To understand adipose tissue development, several groups are tracing the origins of mature adipocytes back to their adult precursor and embryonic ancestors. From these studies emerged a model that brown adipocytes originate from a precursor shared with skeletal muscle that expresses Myf5-Cre, while all white adipocytes originate from a Myf5-negative precursors. While this provided a rational explanation to why BAT is more metabolically favorable than WAT, recent work indicates the situation is more complex because subsets of white adipocytes also arise from Myf5-Cre expressing precursors. Lineage tracing studies further suggest that the vasculature may provide a niche supporting both brown and white adipocyte progenitors; however, the identity of the adipocyte progenitor cell is under debate. Differences in origin between adipocytes could explain metabolic heterogeneity between depots and/or influence body fat patterning particularly in lipodystrophy disorders. Here, we discuss recent insights into adipose tissue origins highlighting lineage-tracing studies in mice, how variations in metabolism or signaling between lineages could affect body fat distribution, and the questions that remain unresolved. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.


Subject(s)
Adipocytes/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Obesity/metabolism , Adipocytes/metabolism , Adipogenesis/genetics , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Animals , Body Fat Distribution , Cell Lineage , Humans , Mice , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Obesity/pathology , Thermogenesis/genetics
14.
Cancer Cell ; 12(1): 9-22, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17613433

ABSTRACT

The mammalian target of rapamycin (mTOR) has emerged as a critical effector in cell-signaling pathways commonly deregulated in human cancers. This has led to the prediction that mTOR inhibitors may be useful in oncology, and derivatives of one such molecule, rapamycin (from which mTOR derives its name), are currently in clinical development. In this review, we discuss recent progress in understanding mTOR signaling, paying particular attention to its relevance in cancer. We further discuss the use of rapamycin in oncology and conclude with a discussion on the future of mTOR-targeted therapy.


Subject(s)
Neoplasms/physiopathology , Protein Kinases/physiology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases
15.
Genome Res ; 21(3): 433-46, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21239477

ABSTRACT

The evolutionarily conserved target of rapamycin complex 1 (TORC1) controls cell growth in response to nutrient availability and growth factors. TORC1 signaling is hyperactive in cancer, and regulators of TORC1 signaling may represent therapeutic targets for human diseases. To identify novel regulators of TORC1 signaling, we performed a genome-scale RNA interference screen on microarrays of Drosophila melanogaster cells expressing human RPS6, a TORC1 effector whose phosphorylated form we detected by immunofluorescence. Our screen revealed that the TORC1-S6K-RPS6 signaling axis is regulated by many subcellular components, including the Class I vesicle coat (COPI), the spliceosome, the proteasome, the nuclear pore, and the translation initiation machinery. Using additional RNAi reagents, we confirmed 70 novel genes as significant on-target regulators of RPS6 phosphorylation, and we characterized them with extensive secondary assays probing various arms of the TORC1 pathways, identifying functional relationships among those genes. We conclude that cell-based microarrays are a useful platform for genome-scale and secondary screening in Drosophila, revealing regulators that may represent drug targets for cancers and other diseases of deregulated TORC1 signaling.


Subject(s)
Recombinant Proteins/metabolism , Ribosomal Protein S6/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western , Cells, Cultured , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Fluorescent Antibody Technique , Gene Regulatory Networks , Genome , Genomics , Humans , Microarray Analysis , Molecular Targeted Therapy , Phosphorylation , RNA Interference , Recombinant Proteins/genetics , Ribosomal Protein S6/genetics , Signal Transduction/genetics , Transcription Factors/genetics
16.
Proc Natl Acad Sci U S A ; 108(50): 20002-7, 2011 Dec 13.
Article in English | MEDLINE | ID: mdl-22135462

ABSTRACT

Soft-tissue sarcomas are heterogeneous cancers that can present with tissue-specific differentiation markers. To examine the cellular basis for this histopathological variation and to identify sarcoma-relevant molecular pathways, we generated a chimeric mouse model in which sarcoma-associated genetic lesions can be introduced into discrete, muscle-resident myogenic and mesenchymal cell lineages. Expression of Kirsten rat sarcoma viral oncogene [Kras(G12V)] and disruption of cyclin-dependent kinase inhibitor 2A (CDKN2A; p16p19) in prospectively isolated satellite cells gave rise to pleomorphic rhabdomyosarcomas (MyoD-, Myogenin- and Desmin-positive), whereas introduction of the same oncogenetic hits in nonmyogenic progenitors induced pleomorphic sarcomas lacking myogenic features. Transcriptional profiling demonstrated that myogenic and nonmyogenic Kras; p16p19(null) sarcomas recapitulate gene-expression signatures of human rhabdomyosarcomas and identified a cluster of genes that is concordantly up-regulated in both mouse and human sarcomas. This cluster includes genes associated with Ras and mechanistic target of rapamycin (mTOR) signaling, a finding consistent with activation of the Ras and mTOR pathways both in Kras; p16p19(null) sarcomas and in 26-50% of human rhabdomyosarcomas surveyed. Moreover, chemical inhibition of Ras or mTOR signaling arrested the growth of mouse Kras; p16p19(null) sarcomas and of human rhabdomyosarcoma cells in vitro and in vivo. Taken together, these data demonstrate the critical importance of lineage commitment within the tumor cell-of-origin in determining sarcoma histotype and introduce an experimental platform for rapid dissection of sarcoma-relevant cellular and molecular events.


Subject(s)
Cell Separation/methods , Muscle Cells/pathology , Muscle, Skeletal/pathology , Sarcoma/pathology , Animals , Biomarkers, Tumor/metabolism , Cell Lineage , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Muscle Development , Muscle Fibers, Skeletal/pathology , Muscle Neoplasms/pathology , Muscle, Skeletal/metabolism , Rats , Sarcoma/genetics , Signal Transduction/genetics , TOR Serine-Threonine Kinases/metabolism , Transcriptome , ras Proteins/metabolism
17.
Nat Rev Cancer ; 23(3): 156-172, 2023 03.
Article in English | MEDLINE | ID: mdl-36658431

ABSTRACT

Few metabolites can claim a more central and versatile role in cell metabolism than acetyl coenzyme A (acetyl-CoA). Acetyl-CoA is produced during nutrient catabolism to fuel the tricarboxylic acid cycle and is the essential building block for fatty acid and isoprenoid biosynthesis. It also functions as a signalling metabolite as the substrate for lysine acetylation reactions, enabling the modulation of protein functions in response to acetyl-CoA availability. Recent years have seen exciting advances in our understanding of acetyl-CoA metabolism in normal physiology and in cancer, buoyed by new mouse models, in vivo stable-isotope tracing approaches and improved methods for measuring acetyl-CoA, including in specific subcellular compartments. Efforts to target acetyl-CoA metabolic enzymes are also advancing, with one therapeutic agent targeting acetyl-CoA synthesis receiving approval from the US Food and Drug Administration. In this Review, we give an overview of the regulation and cancer relevance of major metabolic pathways in which acetyl-CoA participates. We further discuss recent advances in understanding acetyl-CoA metabolism in normal tissues and tumours and the potential for targeting these pathways therapeutically. We conclude with a commentary on emerging nodes of acetyl-CoA metabolism that may impact cancer biology.


Subject(s)
Acetyl Coenzyme A , Neoplasms , Animals , Humans , Mice , Acetyl Coenzyme A/metabolism , Metabolic Networks and Pathways , Neoplasms/drug therapy , Neoplasms/metabolism , Disease Models, Animal
18.
Curr Opin Genet Dev ; 83: 102112, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37703635

ABSTRACT

Nonshivering thermogenesis by brown adipose tissue (BAT) is an adaptive mechanism for maintaining body temperature in cold environments. BAT is critical in rodents and human infants and has substantial influence on adult human metabolism. Stimulating BAT therapeutically is also being investigated as a strategy against metabolic diseases because of its ability to function as a catabolic sink. Thus, understanding how brown adipocytes and the related brite/beige adipocytes use nutrients to fuel their demanding metabolism has both basic and translational implications. Recent advances in mass spectrometry and isotope tracing are improving the ability to study metabolic flux in vivo. Here, we review how such strategies are advancing our understanding of adipocyte thermogenesis and conclude with key future questions.


Subject(s)
Adipose Tissue, Brown , Obesity , Adult , Humans , Obesity/metabolism , Adipose Tissue, Brown/metabolism , Adipocytes, Brown/metabolism , Thermogenesis/genetics
19.
bioRxiv ; 2023 May 16.
Article in English | MEDLINE | ID: mdl-36712034

ABSTRACT

DNA damage can activate apoptotic and non-apoptotic forms of cell death; however, it remains unclear what features dictate which type of cell death is activated. We report that p53 controls the choice between apoptotic and non-apoptotic death following exposure to DNA damage. In contrast to the conventional model, which suggests that p53-deficient cells should be resistant to DNA damage-induced cell death, we find that p53-deficient cells die at high rates following DNA damage, but exclusively using non-apoptotic mechanisms. Our experimental data and computational modeling reveal that non-apoptotic death in p53-deficient cells has not been observed due to use of assays that are either insensitive to cell death, or that specifically score apoptotic cells. Using functional genetic screening - with an analysis that enables computational inference of the drug-induced death rate - we find in p53-deficient cells that DNA damage activates a mitochondrial respiration-dependent form of cell death, called MPT-driven necrosis. Cells deficient for p53 have high basal respiration, which primes MPT-driven necrosis. Finally, using metabolite profiling, we identified mitochondrial activity-dependent metabolic vulnerabilities that can be targeted to potentiate the lethality of DNA damage specifically in p53-deficient cells. Our findings reveal how the dual functions of p53 in regulating mitochondrial activity and the DNA damage response combine to facilitate the choice between apoptotic and non-apoptotic death.

20.
Nat Metab ; 5(7): 1204-1220, 2023 07.
Article in English | MEDLINE | ID: mdl-37337122

ABSTRACT

Adaptive thermogenesis by brown adipose tissue (BAT) dissipates calories as heat, making it an attractive anti-obesity target. Yet how BAT contributes to circulating metabolite exchange remains unclear. Here, we quantified metabolite exchange in BAT and skeletal muscle by arteriovenous metabolomics during cold exposure in fed male mice. This identified unexpected metabolites consumed, released and shared between organs. Quantitative analysis of tissue fluxes showed that glucose and lactate provide ~85% of carbon for adaptive thermogenesis and that cold and CL316,243 trigger markedly divergent fuel utilization profiles. In cold adaptation, BAT also dramatically increases nitrogen uptake by net consuming amino acids, except glutamine. Isotope tracing and functional studies suggest glutamine catabolism concurrent with synthesis via glutamine synthetase, which avoids ammonia buildup and boosts fuel oxidation. These data underscore the ability of BAT to function as a glucose and amino acid sink and provide a quantitative and comprehensive landscape of BAT fuel utilization to guide translational studies.


Subject(s)
Adipose Tissue, Brown , Glutamine , Male , Animals , Mice , Adipose Tissue, Brown/metabolism , Glutamine/metabolism , Glucose/metabolism , Thermogenesis/physiology , Muscle, Skeletal/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL