Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95.607
Filter
Add more filters

Publication year range
1.
Cell ; 187(1): 204-215.e14, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38070508

ABSTRACT

Mounting evidence suggests metabolism instructs stem cell fate decisions. However, how fetal metabolism changes during development and how altered maternal metabolism shapes fetal metabolism remain unexplored. We present a descriptive atlas of in vivo fetal murine metabolism during mid-to-late gestation in normal and diabetic pregnancy. Using 13C-glucose and liquid chromatography-mass spectrometry (LC-MS), we profiled the metabolism of fetal brains, hearts, livers, and placentas harvested from pregnant dams between embryonic days (E)10.5 and 18.5. Our analysis revealed metabolic features specific to a hyperglycemic environment and signatures that may denote developmental transitions during euglycemic development. We observed sorbitol accumulation in fetal tissues and altered neurotransmitter levels in fetal brains isolated from hyperglycemic dams. Tracing 13C-glucose revealed disparate fetal nutrient sourcing depending on maternal glycemic states. Regardless of glycemic state, histidine-derived metabolites accumulated in late-stage fetal tissues. Our rich dataset presents a comprehensive overview of in vivo fetal tissue metabolism and alterations due to maternal hyperglycemia.


Subject(s)
Diabetes Mellitus , Diabetes, Gestational , Fetus , Animals , Female , Mice , Pregnancy , Diabetes Mellitus/metabolism , Fetus/metabolism , Glucose/metabolism , Placenta/metabolism , Diabetes, Gestational/metabolism
2.
Cell ; 186(2): 238-240, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36669471

ABSTRACT

Body temperature maintenance is an important regulator of glucose homeostasis. In this issue of Cell, Meng et al. discover a regulatory axis in which light activation of photoreceptive retinal ganglia stimulates the supraoptic nucleus (SON) to inhibit brown adipose tissue (BAT) thermogenesis and impair glucose homeostasis. This could explain the impact of constant light exposure on metabolism.


Subject(s)
Glucose , Supraoptic Nucleus , Glucose/metabolism , Supraoptic Nucleus/metabolism , Thermogenesis/physiology , Homeostasis , Adipose Tissue, Brown/metabolism , Energy Metabolism
3.
Cell ; 186(2): 398-412.e17, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36669474

ABSTRACT

Public health studies indicate that artificial light is a high-risk factor for metabolic disorders. However, the neural mechanism underlying metabolic modulation by light remains elusive. Here, we found that light can acutely decrease glucose tolerance (GT) in mice by activation of intrinsically photosensitive retinal ganglion cells (ipRGCs) innervating the hypothalamic supraoptic nucleus (SON). Vasopressin neurons in the SON project to the paraventricular nucleus, then to the GABAergic neurons in the solitary tract nucleus, and eventually to brown adipose tissue (BAT). Light activation of this neural circuit directly blocks adaptive thermogenesis in BAT, thereby decreasing GT. In humans, light also modulates GT at the temperature where BAT is active. Thus, our work unveils a retina-SON-BAT axis that mediates the effect of light on glucose metabolism, which may explain the connection between artificial light and metabolic dysregulation, suggesting a potential prevention and treatment strategy for managing glucose metabolic disorders.


Subject(s)
Adipose Tissue, Brown , Hypothalamus , Mice , Animals , Humans , Adipose Tissue, Brown/metabolism , Hypothalamus/metabolism , Thermogenesis/physiology , Retina , Retinal Ganglion Cells , Glucose/metabolism
4.
Cell ; 186(4): 748-763.e15, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36758548

ABSTRACT

Although many prokaryotes have glycolysis alternatives, it's considered as the only energy-generating glucose catabolic pathway in eukaryotes. Here, we managed to create a hybrid-glycolysis yeast. Subsequently, we identified an inositol pyrophosphatase encoded by OCA5 that could regulate glycolysis and respiration by adjusting 5-diphosphoinositol 1,2,3,4,6-pentakisphosphate (5-InsP7) levels. 5-InsP7 levels could regulate the expression of genes involved in glycolysis and respiration, representing a global mechanism that could sense ATP levels and regulate central carbon metabolism. The hybrid-glycolysis yeast did not produce ethanol during growth under excess glucose and could produce 2.68 g/L free fatty acids, which is the highest reported production in shake flask of Saccharomyces cerevisiae. This study demonstrated the significance of hybrid-glycolysis yeast and determined Oca5 as an inositol pyrophosphatase controlling the balance between glycolysis and respiration, which may shed light on the role of inositol pyrophosphates in regulating eukaryotic metabolism.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Diphosphates/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Inositol Phosphates/genetics , Inositol Phosphates/metabolism , Glycolysis/genetics , Respiration , Pyrophosphatases/metabolism , Glucose/metabolism
5.
Cell ; 186(1): 80-97.e26, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36608661

ABSTRACT

Glucose is a universal bioenergy source; however, its role in controlling protein interactions is unappreciated, as are its actions during differentiation-associated intracellular glucose elevation. Azido-glucose click chemistry identified glucose binding to a variety of RNA binding proteins (RBPs), including the DDX21 RNA helicase, which was found to be essential for epidermal differentiation. Glucose bound the ATP-binding domain of DDX21, altering protein conformation, inhibiting helicase activity, and dissociating DDX21 dimers. Glucose elevation during differentiation was associated with DDX21 re-localization from the nucleolus to the nucleoplasm where DDX21 assembled into larger protein complexes containing RNA splicing factors. DDX21 localized to specific SCUGSDGC motif in mRNA introns in a glucose-dependent manner and promoted the splicing of key pro-differentiation genes, including GRHL3, KLF4, OVOL1, and RBPJ. These findings uncover a biochemical mechanism of action for glucose in modulating the dimerization and function of an RNA helicase essential for tissue differentiation.


Subject(s)
DEAD-box RNA Helicases , Glucose , Keratinocytes , Cell Nucleolus/metabolism , Cell Nucleus/metabolism , DEAD-box RNA Helicases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Glucose/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Humans
6.
Annu Rev Biochem ; 90: 31-55, 2021 06 20.
Article in English | MEDLINE | ID: mdl-34153217

ABSTRACT

My graduate and postdoctoral training in metabolism and enzymology eventually led me to study the short- and long-term regulation of glucose and lipid metabolism. In the early phase of my career, my trainees and I identified, purified, and characterized a variety of phosphofructokinase enzymes from mammalian tissues. These studies led us to discover fructose 2,6-P2, the most potent activator of phosphofructokinase and glycolysis. The discovery of fructose 2,6-P2 led to the identification and characterization of the tissue-specific bifunctional enzyme 6-phosphofructo-2-kinase:fructose 2,6-bisphosphatase. We discovered a glucose signaling mechanism by which the liver maintains glucose homeostasis by regulating the activities of this bifunctional enzyme. With a rise in glucose, a signaling metabolite, xylulose 5-phosphate, triggers rapid activation of a specific protein phosphatase (PP2ABδC), which dephosphorylates the bifunctional enzyme, thereby increasing fructose 2,6-P2 levels and upregulating glycolysis. These endeavors paved the way for us to initiate the later phase of my career in which we discovered a new transcription factor termed the carbohydrate response element binding protein (ChREBP). Now ChREBP is recognized as the masterregulator controlling conversion of excess carbohydrates to storage of fat in the liver. ChREBP functions as a central metabolic coordinator that responds to nutrients independently of insulin. The ChREBP transcription factor facilitates metabolic adaptation to excess glucose, leading to obesity and its associated diseases.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Biochemistry/history , Fructosediphosphates/metabolism , Phosphofructokinase-2/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/chemistry , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gluconeogenesis/physiology , Glucose/metabolism , Glycolysis , History, 20th Century , History, 21st Century , Humans , Male , Mice , Phosphofructokinase-2/chemistry , Phosphofructokinases/chemistry , Phosphofructokinases/metabolism , Phosphorylation , United States
7.
Nat Rev Mol Cell Biol ; 24(4): 255-272, 2023 04.
Article in English | MEDLINE | ID: mdl-36316383

ABSTRACT

The classical role of AMP-activated protein kinase (AMPK) is as a cellular energy sensor activated by falling energy status, signalled by increases in AMP to ATP and ADP to ATP ratios. Once activated, AMPK acts to restore energy homeostasis by promoting ATP-producing catabolic pathways while inhibiting energy-consuming processes. In this Review, we provide an update on this canonical (AMP/ADP-dependent) activation mechanism, but focus mainly on recently described non-canonical pathways, including those by which AMPK senses the availability of glucose, glycogen or fatty acids and by which it senses damage to lysosomes and nuclear DNA. We also discuss new findings on the regulation of carbohydrate and lipid metabolism, mitochondrial and lysosomal homeostasis, and DNA repair. Finally, we discuss the role of AMPK in cancer, obesity, diabetes, nonalcoholic steatohepatitis (NASH) and other disorders where therapeutic targeting may exert beneficial effects.


Subject(s)
AMP-Activated Protein Kinases , Energy Metabolism , AMP-Activated Protein Kinases/metabolism , Lipid Metabolism , Glucose/metabolism , Adenosine Triphosphate/metabolism
8.
Cell ; 183(1): 258-268.e12, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32860739

ABSTRACT

Plasmodium species, the causative agent of malaria, rely on glucose for energy supply during blood stage. Inhibition of glucose uptake thus represents a potential strategy for the development of antimalarial drugs. Here, we present the crystal structures of PfHT1, the sole hexose transporter in the genome of Plasmodium species, at resolutions of 2.6 Å in complex with D-glucose and 3.7 Å with a moderately selective inhibitor, C3361. Although both structures exhibit occluded conformations, binding of C3361 induces marked rearrangements that result in an additional pocket. This inhibitor-binding-induced pocket presents an opportunity for the rational design of PfHT1-specific inhibitors. Among our designed C3361 derivatives, several exhibited improved inhibition of PfHT1 and cellular potency against P. falciparum, with excellent selectivity to human GLUT1. These findings serve as a proof of concept for the development of the next-generation antimalarial chemotherapeutics by simultaneously targeting the orthosteric and allosteric sites of PfHT1.


Subject(s)
Monosaccharide Transport Proteins/ultrastructure , Plasmodium falciparum/metabolism , Plasmodium falciparum/ultrastructure , Protozoan Proteins/ultrastructure , Amino Acid Sequence , Animals , Antimalarials , Biological Transport , Glucose/metabolism , Humans , Malaria , Malaria, Falciparum/parasitology , Monosaccharide Transport Proteins/chemistry , Monosaccharide Transport Proteins/metabolism , Parasites , Plasmodium falciparum/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Sugars/metabolism
9.
Cell ; 183(3): 684-701.e14, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33058756

ABSTRACT

Positive selection in Europeans at the 2q21.3 locus harboring the lactase gene has been attributed to selection for the ability of adults to digest milk to survive famine in ancient times. However, the 2q21.3 locus is also associated with obesity and type 2 diabetes in humans, raising the possibility that additional genetic elements in the locus may have contributed to evolutionary adaptation to famine by promoting energy storage, but which now confer susceptibility to metabolic diseases. We show here that the miR-128-1 microRNA, located at the center of the positively selected locus, represents a crucial metabolic regulator in mammals. Antisense targeting and genetic ablation of miR-128-1 in mouse metabolic disease models result in increased energy expenditure and amelioration of high-fat-diet-induced obesity and markedly improved glucose tolerance. A thrifty phenotype connected to miR-128-1-dependent energy storage may link ancient adaptation to famine and modern metabolic maladaptation associated with nutritional overabundance.


Subject(s)
Metabolic Diseases/genetics , MicroRNAs/genetics , Adipocytes, Brown/pathology , Adiposity , Alleles , Animals , Cell Differentiation , Cell Line , Cells, Cultured , Diet, High-Fat , Energy Metabolism , Epigenesis, Genetic , Genetic Loci , Glucose/metabolism , Homeostasis , Humans , Hypertrophy , Insulin Resistance , Leptin/deficiency , Leptin/metabolism , Male , Mammals/genetics , Mice, Inbred C57BL , Mice, Obese , MicroRNAs/metabolism , Obesity/genetics , Oligonucleotides/metabolism , Species Specificity
10.
Cell ; 180(6): 1178-1197.e20, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32200800

ABSTRACT

Social impairment is frequently associated with mitochondrial dysfunction and altered neurotransmission. Although mitochondrial function is crucial for brain homeostasis, it remains unknown whether mitochondrial disruption contributes to social behavioral deficits. Here, we show that Drosophila mutants in the homolog of the human CYFIP1, a gene linked to autism and schizophrenia, exhibit mitochondrial hyperactivity and altered group behavior. We identify the regulation of GABA availability by mitochondrial activity as a biologically relevant mechanism and demonstrate its contribution to social behavior. Specifically, increased mitochondrial activity causes gamma aminobutyric acid (GABA) sequestration in the mitochondria, reducing GABAergic signaling and resulting in social deficits. Pharmacological and genetic manipulation of mitochondrial activity or GABA signaling corrects the observed abnormalities. We identify Aralar as the mitochondrial transporter that sequesters GABA upon increased mitochondrial activity. This study increases our understanding of how mitochondria modulate neuronal homeostasis and social behavior under physiopathological conditions.


Subject(s)
Calcium-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Mitochondria/metabolism , gamma-Aminobutyric Acid/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Genetically Modified , Aspartic Acid/metabolism , Calcium/metabolism , Calcium-Binding Proteins/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Glucose/metabolism , Homeostasis , Humans , Male , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Proteins/metabolism , Neurons/metabolism , Social Behavior , Synaptic Transmission , gamma-Aminobutyric Acid/genetics
11.
Cell ; 180(6): 1198-1211.e19, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32200801

ABSTRACT

It has generally proven challenging to produce functional ß cells in vitro. Here, we describe a previously unidentified protein C receptor positive (Procr+) cell population in adult mouse pancreas through single-cell RNA sequencing (scRNA-seq). The cells reside in islets, do not express differentiation markers, and feature epithelial-to-mesenchymal transition characteristics. By genetic lineage tracing, Procr+ islet cells undergo clonal expansion and generate all four endocrine cell types during adult homeostasis. Sorted Procr+ cells, representing ∼1% of islet cells, can robustly form islet-like organoids when cultured at clonal density. Exponential expansion can be maintained over long periods by serial passaging, while differentiation can be induced at any time point in culture. ß cells dominate in differentiated islet organoids, while α, δ, and PP cells occur at lower frequencies. The organoids are glucose-responsive and insulin-secreting. Upon transplantation in diabetic mice, these organoids reverse disease. These findings demonstrate that the adult mouse pancreatic islet contains a population of Procr+ endocrine progenitors.


Subject(s)
Cell Culture Techniques/methods , Endothelial Protein C Receptor/metabolism , Islets of Langerhans/cytology , Animals , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Epithelial-Mesenchymal Transition/physiology , Female , Glucose/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Islets of Langerhans/growth & development , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Nude , Organoids/growth & development , Organoids/metabolism , Pancreas/cytology , Pancreas/metabolism , Protein C/metabolism , Stem Cells/cytology
12.
Cell ; 178(4): 807-819.e21, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398338

ABSTRACT

The NRF2 transcription factor controls a cell stress program that is implicated in cancer and there is great interest in targeting NRF2 for therapy. We show that NRF2 activity depends on Fructosamine-3-kinase (FN3K)-a kinase that triggers protein de-glycation. In its absence, NRF2 is extensively glycated, unstable, and defective at binding to small MAF proteins and transcriptional activation. Moreover, the development of hepatocellular carcinoma triggered by MYC and Keap1 inactivation depends on FN3K in vivo. N-acetyl cysteine treatment partially rescues the effects of FN3K loss on NRF2 driven tumor phenotypes indicating a key role for NRF2-mediated redox balance. Mass spectrometry reveals that other proteins undergo FN3K-sensitive glycation, including translation factors, heat shock proteins, and histones. How glycation affects their functions remains to be defined. In summary, our study reveals a surprising role for the glycation of cellular proteins and implicates FN3K as targetable modulator of NRF2 activity in cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , NF-E2-Related Factor 2/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Female , Gene Knockdown Techniques , Glucose/metabolism , Glycosylation , HEK293 Cells , Hep G2 Cells , Heterografts , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transduction, Genetic
13.
Nat Immunol ; 22(2): 179-192, 2021 02.
Article in English | MEDLINE | ID: mdl-33462452

ABSTRACT

Metabolic programming controls immune cell lineages and functions, but little is known about γδ T cell metabolism. Here, we found that γδ T cell subsets making either interferon-γ (IFN-γ) or interleukin (IL)-17 have intrinsically distinct metabolic requirements. Whereas IFN-γ+ γδ T cells were almost exclusively dependent on glycolysis, IL-17+ γδ T cells strongly engaged oxidative metabolism, with increased mitochondrial mass and activity. These distinct metabolic signatures were surprisingly imprinted early during thymic development and were stably maintained in the periphery and within tumors. Moreover, pro-tumoral IL-17+ γδ T cells selectively showed high lipid uptake and intracellular lipid storage and were expanded in obesity and in tumors of obese mice. Conversely, glucose supplementation enhanced the antitumor functions of IFN-γ+ γδ T cells and reduced tumor growth upon adoptive transfer. These findings have important implications for the differentiation of effector γδ T cells and their manipulation in cancer immunotherapy.


Subject(s)
Breast Neoplasms/metabolism , Colonic Neoplasms/metabolism , Energy Metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/metabolism , Thymus Gland/metabolism , Tumor Microenvironment , Animals , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Line, Tumor , Cell Lineage , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Female , Glucose/metabolism , Glycolysis , Humans , Immunotherapy, Adoptive , Interferon-gamma/metabolism , Interleukin-17/metabolism , Lipid Metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/transplantation , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Obesity/immunology , Obesity/metabolism , Organ Culture Techniques , Phenotype , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/transplantation , Thymus Gland/immunology , Tumor Burden
14.
Immunity ; 57(5): 1105-1123.e8, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38703775

ABSTRACT

Immunosuppressive macrophages restrict anti-cancer immunity in glioblastoma (GBM). Here, we studied the contribution of microglia (MGs) and monocyte-derived macrophages (MDMs) to immunosuppression and mechanisms underlying their regulatory function. MDMs outnumbered MGs at late tumor stages and suppressed T cell activity. Molecular and functional analysis identified a population of glycolytic MDM expressing GLUT1 with potent immunosuppressive activity. GBM-derived factors promoted high glycolysis, lactate, and interleukin-10 (IL-10) production in MDMs. Inhibition of glycolysis or lactate production in MDMs impaired IL-10 expression and T cell suppression. Mechanistically, intracellular lactate-driven histone lactylation promoted IL-10 expression, which was required to suppress T cell activity. GLUT1 expression on MDMs was induced downstream of tumor-derived factors that activated the PERK-ATF4 axis. PERK deletion in MDM abrogated histone lactylation, led to the accumulation of intratumoral T cells and tumor growth delay, and, in combination with immunotherapy, blocked GBM progression. Thus, PERK-driven glucose metabolism promotes MDM immunosuppressive activity via histone lactylation.


Subject(s)
Glioblastoma , Glucose , Histones , Macrophages , Glioblastoma/immunology , Glioblastoma/metabolism , Glioblastoma/pathology , Animals , Histones/metabolism , Mice , Macrophages/immunology , Macrophages/metabolism , Glucose/metabolism , Humans , Cell Line, Tumor , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 1/genetics , Interleukin-10/metabolism , Glycolysis , Microglia/metabolism , Microglia/immunology , Mice, Inbred C57BL , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Immune Tolerance
15.
Nat Rev Mol Cell Biol ; 22(2): 142-158, 2021 02.
Article in English | MEDLINE | ID: mdl-33398164

ABSTRACT

Metabolic homeostasis in mammals is tightly regulated by the complementary actions of insulin and glucagon. The secretion of these hormones from pancreatic ß-cells and α-cells, respectively, is controlled by metabolic, endocrine, and paracrine regulatory mechanisms and is essential for the control of blood levels of glucose. The deregulation of these mechanisms leads to various pathologies, most notably type 2 diabetes, which is driven by the combined lesions of impaired insulin action and a loss of the normal insulin secretion response to glucose. Glucose stimulates insulin secretion from ß-cells in a bi-modal fashion, and new insights about the underlying mechanisms, particularly relating to the second or amplifying phase of this secretory response, have been recently gained. Other recent work highlights the importance of α-cell-produced proglucagon-derived peptides, incretin hormones from the gastrointestinal tract and other dietary components, including certain amino acids and fatty acids, in priming and potentiation of the ß-cell glucose response. These advances provide a new perspective for the understanding of the ß-cell failure that triggers type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Glucose/metabolism , Homeostasis , Insulin Secretion , Insulin-Secreting Cells/physiology , Animals , Humans , Insulin-Secreting Cells/cytology
16.
Nat Rev Mol Cell Biol ; 22(6): 425-438, 2021 06.
Article in English | MEDLINE | ID: mdl-33772227

ABSTRACT

In animals, systemic control of metabolism is conducted by metabolic tissues and relies on the regulated circulation of a plethora of molecules, such as hormones and lipoprotein complexes. MicroRNAs (miRNAs) are a family of post-transcriptional gene repressors that are present throughout the animal kingdom and have been widely associated with the regulation of gene expression in various contexts, including virtually all aspects of systemic control of metabolism. Here we focus on glucose and lipid metabolism and review current knowledge of the role of miRNAs in their systemic regulation. We survey miRNA-mediated regulation of healthy metabolism as well as the contribution of miRNAs to metabolic dysfunction in disease, particularly diabetes, obesity and liver disease. Although most miRNAs act on the tissue they are produced in, it is now well established that miRNAs can also circulate in bodily fluids, including their intercellular transport by extracellular vesicles, and we discuss the role of such extracellular miRNAs in systemic metabolic control and as potential biomarkers of metabolic status and metabolic disease.


Subject(s)
Glucose/metabolism , MicroRNAs/metabolism , Animals , Humans , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Metabolic Diseases/metabolism
17.
Nat Rev Mol Cell Biol ; 22(11): 751-771, 2021 11.
Article in English | MEDLINE | ID: mdl-34285405

ABSTRACT

Insulin resistance, defined as a defect in insulin-mediated control of glucose metabolism in tissues - prominently in muscle, fat and liver - is one of the earliest manifestations of a constellation of human diseases that includes type 2 diabetes and cardiovascular disease. These diseases are typically associated with intertwined metabolic abnormalities, including obesity, hyperinsulinaemia, hyperglycaemia and hyperlipidaemia. Insulin resistance is caused by a combination of genetic and environmental factors. Recent genetic and biochemical studies suggest a key role for adipose tissue in the development of insulin resistance, potentially by releasing lipids and other circulating factors that promote insulin resistance in other organs. These extracellular factors perturb the intracellular concentration of a range of intermediates, including ceramide and other lipids, leading to defects in responsiveness of cells to insulin. Such intermediates may cause insulin resistance by inhibiting one or more of the proximal components in the signalling cascade downstream of insulin (insulin receptor, insulin receptor substrate (IRS) proteins or AKT). However, there is now evidence to support the view that insulin resistance is a heterogeneous disorder that may variably arise in a range of metabolic tissues and that the mechanism for this effect likely involves a unified insulin resistance pathway that affects a distal step in the insulin action pathway that is more closely linked to the terminal biological response. Identifying these targets is of major importance, as it will reveal potential new targets for treatments of diseases associated with insulin resistance.


Subject(s)
Antigens, CD/genetics , Diabetes Mellitus, Type 2/genetics , Insulin Resistance/genetics , Insulin/genetics , Receptor, Insulin/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Glucose/genetics , Glucose/metabolism , Humans , Insulin/metabolism , Liver/metabolism , Liver/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/genetics
18.
Cell ; 175(1): 117-132.e21, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30197082

ABSTRACT

The metabolic state of a cell is influenced by cell-extrinsic factors, including nutrient availability and growth factor signaling. Here, we present extracellular matrix (ECM) remodeling as another fundamental node of cell-extrinsic metabolic regulation. Unbiased analysis of glycolytic drivers identified the hyaluronan-mediated motility receptor as being among the most highly correlated with glycolysis in cancer. Confirming a mechanistic link between the ECM component hyaluronan and metabolism, treatment of cells and xenografts with hyaluronidase triggers a robust increase in glycolysis. This is largely achieved through rapid receptor tyrosine kinase-mediated induction of the mRNA decay factor ZFP36, which targets TXNIP transcripts for degradation. Because TXNIP promotes internalization of the glucose transporter GLUT1, its acute decline enriches GLUT1 at the plasma membrane. Functionally, induction of glycolysis by hyaluronidase is required for concomitant acceleration of cell migration. This interconnection between ECM remodeling and metabolism is exhibited in dynamic tissue states, including tumorigenesis and embryogenesis.


Subject(s)
Carrier Proteins/physiology , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Carbohydrate Metabolism/physiology , Carrier Proteins/metabolism , Cell Line, Tumor , Glucose/metabolism , Glucose Transporter Type 1 , Glycolysis/physiology , Humans , Hyaluronic Acid/physiology , Hyaluronoglucosaminidase/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Signal Transduction , Tristetraprolin/metabolism , Tristetraprolin/physiology
19.
Cell ; 174(6): 1549-1558.e14, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30100189

ABSTRACT

Engineering microorganisms for production of fuels and chemicals often requires major re-programming of metabolism to ensure high flux toward the product of interest. This is challenging, as millions of years of evolution have resulted in establishment of tight regulation of metabolism for optimal growth in the organism's natural habitat. Here, we show through metabolic engineering that it is possible to alter the metabolism of Saccharomyces cerevisiae from traditional ethanol fermentation to a pure lipogenesis metabolism, resulting in high-level production of free fatty acids. Through metabolic engineering and process design, we altered subcellular metabolic trafficking, fine-tuned NADPH and ATP supply, and decreased carbon flux to biomass, enabling production of 33.4 g/L extracellular free fatty acids. We further demonstrate that lipogenesis metabolism can replace ethanol fermentation by deletion of pyruvate decarboxylase enzymes followed by adaptive laboratory evolution. Genome sequencing of evolved strains showed that pyruvate kinase mutations were essential for this phenotype.


Subject(s)
Fatty Acids, Nonesterified/biosynthesis , Metabolic Engineering , Saccharomyces cerevisiae/metabolism , Acetyl Coenzyme A/metabolism , Glucose/metabolism , Glycolysis , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Lipogenesis , NADP/metabolism , Pentose Phosphate Pathway/genetics , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
20.
Cell ; 175(2): 502-513.e13, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30245009

ABSTRACT

Acetate is a major nutrient that supports acetyl-coenzyme A (Ac-CoA) metabolism and thus lipogenesis and protein acetylation. However, its source is unclear. Here, we report that pyruvate, the end product of glycolysis and key node in central carbon metabolism, quantitatively generates acetate in mammals. This phenomenon becomes more pronounced in the context of nutritional excess, such as during hyperactive glucose metabolism. Conversion of pyruvate to acetate occurs through two mechanisms: (1) coupling to reactive oxygen species (ROS) and (2) neomorphic enzyme activity from keto acid dehydrogenases that enable function as pyruvate decarboxylases. Further, we demonstrate that de novo acetate production sustains Ac-CoA pools and cell proliferation in limited metabolic environments, such as during mitochondrial dysfunction or ATP citrate lyase (ACLY) deficiency. By virtue of de novo acetate production being coupled to mitochondrial metabolism, there are numerous possible regulatory mechanisms and links to pathophysiology.


Subject(s)
Acetates/metabolism , Glucose/metabolism , Pyruvic Acid/metabolism , ATP Citrate (pro-S)-Lyase/physiology , Acetyl Coenzyme A/biosynthesis , Acetyl Coenzyme A/metabolism , Acetylation , Animals , Female , Glycolysis/physiology , Lipogenesis/physiology , Male , Mammals/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Oxidoreductases , Pyruvate Decarboxylase/physiology , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL