Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Nat Immunol ; 20(12): 1668-1680, 2019 12.
Article in English | MEDLINE | ID: mdl-31636464

ABSTRACT

Lymph node fibroblastic reticular cells (FRCs) respond to signals from activated T cells by releasing nitric oxide, which inhibits T cell proliferation and restricts the size of the expanding T cell pool. Whether interactions with FRCs also support the function or differentiation of activated CD8+ T cells is not known. Here we report that encounters with FRCs enhanced cytokine production and remodeled chromatin accessibility in newly activated CD8+ T cells via interleukin-6. These epigenetic changes facilitated metabolic reprogramming and amplified the activity of pro-survival pathways through differential transcription factor activity. Accordingly, FRC conditioning significantly enhanced the persistence of virus-specific CD8+ T cells in vivo and augmented their differentiation into tissue-resident memory T cells. Our study demonstrates that FRCs play a role beyond restricting T cell expansion-they can also shape the fate and function of CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Fibroblasts/physiology , Lymph Nodes/immunology , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Cellular Reprogramming , Chromatin Assembly and Disassembly , Cytotoxicity, Immunologic , Epigenesis, Genetic , Gene Expression Regulation , Immunologic Memory , Interleukin-6/genetics , Interleukin-6/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism
2.
Cell ; 158(1): 41-53, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24995977

ABSTRACT

A hallmark of type 2 diabetes mellitus (T2DM) is the development of pancreatic ß cell failure, which results in insulinopenia and hyperglycemia. We show that the adipokine adipsin has a beneficial role in maintaining ß cell function. Animals genetically lacking adipsin have glucose intolerance due to insulinopenia; isolated islets from these mice have reduced glucose-stimulated insulin secretion. Replenishment of adipsin to diabetic mice treated hyperglycemia by boosting insulin secretion. We identify C3a, a peptide generated by adipsin, as a potent insulin secretagogue and show that the C3a receptor is required for these beneficial effects of adipsin. C3a acts on islets by augmenting ATP levels, respiration, and cytosolic free Ca(2+). Finally, we demonstrate that T2DM patients with ß cell failure are deficient in adipsin. These findings indicate that the adipsin/C3a pathway connects adipocyte function to ß cell physiology, and manipulation of this molecular switch may serve as a therapy in T2DM.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Adipose Tissue/metabolism , Animals , Complement C3a/metabolism , Complement Factor D/genetics , Complement Factor D/metabolism , Diabetes Mellitus, Type 2/physiopathology , Diet, High-Fat , Glucose/metabolism , Humans , Inflammation/metabolism , Insulin/metabolism , Insulin Secretion , Mice
3.
Cell ; 152(1-2): 304-15, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23332762

ABSTRACT

The IκB kinase complex (IKK) is a key regulator of immune responses, inflammation, cell survival, and tumorigenesis. The prosurvival function of IKK centers on activation of the transcription factor NF-κB, whose target gene products inhibit caspases and prevent prolonged JNK activation. Here, we report that inactivation of the BH3-only protein BAD by IKK independently of NF-κB activation suppresses TNFα-induced apoptosis. TNFα-treated Ikkß(-/-) mouse embryonic fibroblasts (MEFs) undergo apoptosis significantly faster than MEFs deficient in both RelA and cRel due to lack of inhibition of BAD by IKK. IKK phosphorylates BAD at serine-26 (Ser26) and primes it for inactivation. Elimination of Ser26 phosphorylation promotes BAD proapoptotic activity, thereby accelerating TNFα-induced apoptosis in cultured cells and increasing mortality in animals. Our results reveal that IKK inhibits TNFα-induced apoptosis through two distinct but cooperative mechanisms: activation of the survival factor NF-κB and inactivation of the proapoptotic BH3-only BAD protein.


Subject(s)
Apoptosis , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , bcl-Associated Death Protein/metabolism , Animals , Fibroblasts/cytology , I-kappa B Kinase/genetics , Mice , Mice, Knockout , Phosphorylation , Serine/metabolism , bcl-Associated Death Protein/chemistry , bcl-Associated Death Protein/genetics , bcl-X Protein/metabolism
4.
Mol Cell ; 77(6): 1251-1264.e9, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32023484

ABSTRACT

Lipid droplets (LDs) store lipids for energy and are central to cellular lipid homeostasis. The mechanisms coordinating lipid storage in LDs with cellular metabolism are unclear but relevant to obesity-related diseases. Here we utilized genome-wide screening to identify genes that modulate lipid storage in macrophages, a cell type involved in metabolic diseases. Among ∼550 identified screen hits is MLX, a basic helix-loop-helix leucine-zipper transcription factor that regulates metabolic processes. We show that MLX and glucose-sensing family members MLXIP/MondoA and MLXIPL/ChREBP bind LDs via C-terminal amphipathic helices. When LDs accumulate in cells, these transcription factors bind to LDs, reducing their availability for transcriptional activity and attenuating the response to glucose. Conversely, the absence of LDs results in hyperactivation of MLX target genes. Our findings uncover a paradigm for a lipid storage response in which binding of MLX transcription factors to LD surfaces adjusts the expression of metabolic genes to lipid storage levels.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation , Glucose/metabolism , Lipid Droplets/metabolism , Proteome/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cells, Cultured , Genetic Testing , Humans , Macrophages/cytology , Macrophages/metabolism , Protein Binding , Proteome/analysis , RNA, Small Interfering , Transcription, Genetic
5.
EMBO J ; 42(11): e111901, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36917141

ABSTRACT

Changes in mitochondrial morphology are associated with nutrient utilization, but the precise causalities and the underlying mechanisms remain unknown. Here, using cellular models representing a wide variety of mitochondrial shapes, we show a strong linear correlation between mitochondrial fragmentation and increased fatty acid oxidation (FAO) rates. Forced mitochondrial elongation following MFN2 over-expression or DRP1 depletion diminishes FAO, while forced fragmentation upon knockdown or knockout of MFN2 augments FAO as evident from respirometry and metabolic tracing. Remarkably, the genetic induction of fragmentation phenocopies distinct cell type-specific biological functions of enhanced FAO. These include stimulation of gluconeogenesis in hepatocytes, induction of insulin secretion in islet ß-cells exposed to fatty acids, and survival of FAO-dependent lymphoma subtypes. We find that fragmentation increases long-chain but not short-chain FAO, identifying carnitine O-palmitoyltransferase 1 (CPT1) as the downstream effector of mitochondrial morphology in regulation of FAO. Mechanistically, we determined that fragmentation reduces malonyl-CoA inhibition of CPT1, while elongation increases CPT1 sensitivity to malonyl-CoA inhibition. Overall, these findings underscore a physiologic role for fragmentation as a mechanism whereby cellular fuel preference and FAO capacity are determined.


Subject(s)
Fatty Acids , Malonyl Coenzyme A , Fatty Acids/metabolism , Malonyl Coenzyme A/metabolism , Malonyl Coenzyme A/pharmacology , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Oxidation-Reduction , Mitochondria/metabolism
6.
Mol Cell ; 75(2): 357-371.e7, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31227231

ABSTRACT

Carbohydrate response element binding protein (ChREBP) is a key transcriptional regulator of de novo lipogenesis (DNL) in response to carbohydrates and in hepatic steatosis. Mechanisms underlying nutrient modulation of ChREBP are under active investigation. Here we identify host cell factor 1 (HCF-1) as a previously unknown ChREBP-interacting protein that is enriched in liver biopsies of nonalcoholic steatohepatitis (NASH) patients. Biochemical and genetic studies show that HCF-1 is O-GlcNAcylated in response to glucose as a prerequisite for its binding to ChREBP and subsequent recruitment of OGT, ChREBP O-GlcNAcylation, and activation. The HCF-1:ChREBP complex resides at lipogenic gene promoters, where HCF-1 regulates H3K4 trimethylation to prime recruitment of the Jumonji C domain-containing histone demethylase PHF2 for epigenetic activation of these promoters. Overall, these findings define HCF-1's interaction with ChREBP as a previously unappreciated mechanism whereby glucose signals are both relayed to ChREBP and transmitted for epigenetic regulation of lipogenic genes.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Homeodomain Proteins/genetics , Host Cell Factor C1/genetics , Lipogenesis/genetics , Non-alcoholic Fatty Liver Disease/genetics , Animals , Carbohydrates/genetics , Epigenesis, Genetic , Gene Expression Regulation , Glucose/metabolism , Hexosamines/genetics , Hexosamines/metabolism , Humans , Liver/metabolism , Mice , Non-alcoholic Fatty Liver Disease/pathology , Promoter Regions, Genetic/genetics , Protein Interaction Maps/genetics
7.
Genes Dev ; 28(5): 479-90, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24589777

ABSTRACT

Mutations in isocitrate dehydrogenase 1 and 2 (IDH1/2) have been discovered in several cancer types and cause the neurometabolic syndrome D2-hydroxyglutaric aciduria (D2HGA). The mutant enzymes exhibit neomorphic activity resulting in production of D2-hydroxyglutaric acid (D-2HG). To study the pathophysiological consequences of the accumulation of D-2HG, we generated transgenic mice with conditionally activated IDH2(R140Q) and IDH2(R172K) alleles. Global induction of mutant IDH2 expression in adults resulted in dilated cardiomyopathy, white matter abnormalities throughout the central nervous system (CNS), and muscular dystrophy. Embryonic activation of mutant IDH2 resulted in more pronounced phenotypes, including runting, hydrocephalus, and shortened life span, recapitulating the abnormalities observed in D2HGA patients. The diseased hearts exhibited mitochondrial damage and glycogen accumulation with a concordant up-regulation of genes involved in glycogen biosynthesis. Notably, mild cardiac hypertrophy was also observed in nude mice implanted with IDH2(R140Q)-expressing xenografts, suggesting that 2HG may potentially act in a paracrine fashion. Finally, we show that silencing of IDH2(R140Q) in mice with an inducible transgene restores heart function by lowering 2HG levels. Together, these findings indicate that inhibitors of mutant IDH2 may be beneficial in the treatment of D2HGA and suggest that 2HG produced by IDH mutant tumors has the potential to provoke a paraneoplastic condition.


Subject(s)
Cardiomyopathies/genetics , Glutarates/metabolism , Isocitrate Dehydrogenase/genetics , Mutation , Neurodegenerative Diseases/genetics , Animals , Cardiomyopathies/enzymology , Cardiomyopathies/pathology , Cell Line , Gene Expression Profiling , Gene Expression Regulation, Developmental , Heart/physiopathology , Humans , Isocitrate Dehydrogenase/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/pathology
8.
Epilepsia ; 59(1): e1-e4, 2018 01.
Article in English | MEDLINE | ID: mdl-29171006

ABSTRACT

Metabolic alteration, either through the ketogenic diet (KD) or by genetic alteration of the BAD protein, can produce seizure protection in acute chemoconvulsant models of epilepsy. To assess the seizure-protective role of knocking out (KO) the Bad gene in a chronic epilepsy model, we used the Kcna1-/- model of epilepsy, which displays progressively increased seizure severity and recapitulates the early death seen in sudden unexplained death in epilepsy (SUDEP). Beginning on postnatal day 24 (P24), we continuously video monitored Kcna1-/- and Kcna1-/- Bad-/- double knockout mice to assess survival and seizure severity. We found that Kcna1-/- Bad-/- mice outlived Kcna1-/- mice by approximately 2 weeks. Kcna1-/- Bad-/- mice also spent significantly less time in seizure than Kcna1-/- mice on P24 and the day of death, showing that BadKO provides seizure resistance in a genetic model of chronic epilepsy.


Subject(s)
Brugada Syndrome/etiology , Brugada Syndrome/genetics , Epilepsy , Kv1.1 Potassium Channel/genetics , bcl-Associated Death Protein/deficiency , Age Factors , Animals , Brugada Syndrome/metabolism , Disease Models, Animal , Electroencephalography , Epilepsy/complications , Epilepsy/genetics , Epilepsy/prevention & control , Female , Kv1.1 Potassium Channel/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , bcl-Associated Death Protein/genetics
9.
Nature ; 491(7425): 608-12, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-23135403

ABSTRACT

Defects in the availability of haem substrates or the catalytic activity of the terminal enzyme in haem biosynthesis, ferrochelatase (Fech), impair haem synthesis and thus cause human congenital anaemias. The interdependent functions of regulators of mitochondrial homeostasis and enzymes responsible for haem synthesis are largely unknown. To investigate this we used zebrafish genetic screens and cloned mitochondrial ATPase inhibitory factor 1 (atpif1) from a zebrafish mutant with profound anaemia, pinotage (pnt (tq209)). Here we describe a direct mechanism establishing that Atpif1 regulates the catalytic efficiency of vertebrate Fech to synthesize haem. The loss of Atpif1 impairs haemoglobin synthesis in zebrafish, mouse and human haematopoietic models as a consequence of diminished Fech activity and elevated mitochondrial pH. To understand the relationship between mitochondrial pH, redox potential, [2Fe-2S] clusters and Fech activity, we used genetic complementation studies of Fech constructs with or without [2Fe-2S] clusters in pnt, as well as pharmacological agents modulating mitochondrial pH and redox potential. The presence of [2Fe-2S] cluster renders vertebrate Fech vulnerable to perturbations in Atpif1-regulated mitochondrial pH and redox potential. Therefore, Atpif1 deficiency reduces the efficiency of vertebrate Fech to synthesize haem, resulting in anaemia. The identification of mitochondrial Atpif1 as a regulator of haem synthesis advances our understanding of the mechanisms regulating mitochondrial haem homeostasis and red blood cell development. An ATPIF1 deficiency may contribute to important human diseases, such as congenital sideroblastic anaemias and mitochondriopathies.


Subject(s)
Erythroblasts/metabolism , Erythropoiesis , Heme/biosynthesis , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Proteins/metabolism , Anemia, Sideroblastic/genetics , Anemia, Sideroblastic/metabolism , Anemia, Sideroblastic/pathology , Animals , Disease Models, Animal , Erythroblasts/cytology , Ferrochelatase/metabolism , Genetic Complementation Test , Humans , Hydrogen-Ion Concentration , Mice , Mitochondria/pathology , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Oxidation-Reduction , Proteins/genetics , Zebrafish/metabolism , ATPase Inhibitory Protein
10.
Mol Cell ; 40(5): 687-8, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21145478

ABSTRACT

The BCL-2 family member Noxa induces apoptosis by antagonizing the prosurvival protein MCL-1. In this issue of Molecular Cell, Lowman et al. (2010) uncover a glucose-dependent phosphoregulatory mechanism that inactivates Noxa's apoptotic function and triggers its capacity to modulate glucose metabolism.

11.
PLoS Genet ; 8(8): e1002902, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22927828

ABSTRACT

Obesity-associated metabolic complications are generally considered to emerge from abnormalities in carbohydrate and lipid metabolism, whereas the status of protein metabolism is not well studied. Here, we performed comparative polysome and associated transcriptional profiling analyses to study the dynamics and functional implications of endoplasmic reticulum (ER)-associated protein synthesis in the mouse liver under conditions of obesity and nutrient deprivation. We discovered that ER from livers of obese mice exhibits a general reduction in protein synthesis, and comprehensive analysis of polysome-bound transcripts revealed extensive down-regulation of protein synthesis machinery, mitochondrial components, and bile acid metabolism in the obese translatome. Nutrient availability also plays an important but distinct role in remodeling the hepatic ER translatome in lean and obese mice. Fasting in obese mice partially reversed the overall translatomic differences between lean and obese nonfasted controls, whereas fasting of the lean mice mimicked many of the translatomic changes induced by the development of obesity. The strongest examples of such regulations were the reduction in Cyp7b1 and Slco1a1, molecules involved in bile acid metabolism. Exogenous expression of either gene significantly lowered plasma glucose levels, improved hepatic steatosis, but also caused cholestasis, indicating the fine balance bile acids play in regulating metabolism and health. Together, our work defines dynamic regulation of the liver translatome by obesity and nutrient availability, and it identifies a novel role for bile acid metabolism in the pathogenesis of metabolic abnormalities associated with obesity.


Subject(s)
Endoplasmic Reticulum/metabolism , Liver/metabolism , Obesity/metabolism , Polyribosomes/metabolism , Protein Biosynthesis , Animals , Bile Acids and Salts/metabolism , Cholestasis/metabolism , Cytochrome P450 Family 7 , Fasting/metabolism , Fatty Liver/metabolism , Glucose/metabolism , Liver/pathology , Male , Mice , Mice, Obese , Obesity/genetics , Specific Pathogen-Free Organisms , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism
12.
Sci Adv ; 8(39): eabq0117, 2022 Sep 30.
Article in English | MEDLINE | ID: mdl-36179030

ABSTRACT

The fate of pyruvate is a defining feature in many cell types. One major fate is mitochondrial entry via the mitochondrial pyruvate carrier (MPC). We found that diffuse large B cell lymphomas (DLBCLs) consume mitochondrial pyruvate via glutamate-pyruvate transaminase 2 to enable α-ketoglutarate production as part of glutaminolysis. This led us to discover that glutamine exceeds pyruvate as a carbon source for the tricarboxylic acid cycle in DLBCLs. As a result, MPC inhibition led to decreased glutaminolysis in DLBCLs, opposite to previous observations in other cell types. We also found that MPC inhibition or genetic depletion decreased DLBCL proliferation in an extracellular matrix (ECM)-like environment and xenografts, but not in a suspension environment. Moreover, the metabolic profile of DLBCL cells in ECM is markedly different from cells in a suspension environment. Thus, we conclude that the synergistic consumption and assimilation of glutamine and pyruvate enables DLBCL proliferation in an extracellular environment-dependent manner.

13.
Cell Rep ; 41(1): 111445, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36198266

ABSTRACT

MCL-1 is an anti-apoptotic BCL-2 family protein essential for survival of diverse cell types and is a major driver of cancer and chemoresistance. The mechanistic basis for the oncogenic supremacy of MCL-1 among its anti-apoptotic homologs is unclear and implicates physiologic roles of MCL-1 beyond apoptotic suppression. Here we find that MCL-1-dependent hematologic cancer cells specifically rely on fatty acid oxidation (FAO) as a fuel source because of metabolic wiring enforced by MCL-1 itself. We demonstrate that FAO regulation by MCL-1 is independent of its anti-apoptotic activity, based on metabolomic, proteomic, and genomic profiling of MCL-1-dependent leukemia cells lacking an intact apoptotic pathway. Genetic deletion of Mcl-1 results in transcriptional downregulation of FAO pathway proteins such that glucose withdrawal triggers cell death despite apoptotic blockade. Our data reveal that MCL-1 is a master regulator of FAO, rendering MCL-1-driven cancer cells uniquely susceptible to treatment with FAO inhibitors.


Subject(s)
Neoplasms , Proteomics , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Fatty Acids , Glucose , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasms/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
14.
Biochemistry ; 50(12): 2213-22, 2011 Mar 29.
Article in English | MEDLINE | ID: mdl-21299233

ABSTRACT

Proteolysis plays a key role in regulating the levels and activity of peptide hormones. Characterization of the proteolytic pathways that cleave peptide hormones is of basic interest and can, in some cases, spur the development of novel therapeutics. The lack, however, of an efficient approach to identify endogenous fragments of peptide hormones has hindered the elucidation of these proteolytic pathways. Here, we apply a mass spectrometry (MS) based peptidomics approach to characterize the intestinal fragments of peptide histidine isoleucine (PHI), a hormone that promotes glucose-stimulated insulin secretion (GSIS). Our approach reveals a proteolytic pathway in the intestine that truncates PHI at its C-terminus to produce a PHI fragment that is inactive in a GSIS assay, a result that provides a potential mechanism of PHI regulation in vivo. Differences between these in vivo peptidomics studies and in vitro lysate experiments, which showed N- and C-terminal processing of PHI, underscore the effectiveness of this approach to discover physiologically relevant proteolytic pathways. Moreover, integrating this peptidomics approach with bioassays (i.e., GSIS) provides a general strategy to reveal proteolytic pathways that may regulate the activity of peptide hormones.


Subject(s)
Peptide Hormones/metabolism , Proteomics/methods , Amino Acid Sequence , Animals , Binding, Competitive , Dipeptidyl Peptidase 4/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Hormones/chemistry , Peptide Hydrolases/metabolism , Peptide PHI/chemistry , Peptide PHI/metabolism , Tissue Extracts/metabolism
15.
Nat Commun ; 12(1): 2939, 2021 05 19.
Article in English | MEDLINE | ID: mdl-34011960

ABSTRACT

Elucidation of non-canonical protein functions can identify novel tissue homeostasis pathways. Herein, we describe a role for the Bcl-2 family member BAD in postnatal mammary gland morphogenesis. In Bad3SA knock-in mice, where BAD cannot undergo phosphorylation at 3 key serine residues, pubertal gland development is delayed due to aberrant tubulogenesis of the ductal epithelium. Proteomic and RPPA analyses identify that BAD regulates focal adhesions and the mRNA translation repressor, 4E-BP1. These results suggest that BAD modulates localized translation that drives focal adhesion maturation and cell motility. Consistent with this, cells within Bad3SA organoids contain unstable protrusions with decreased compartmentalized mRNA translation and focal adhesions, and exhibit reduced cell migration and tubulogenesis. Critically, protrusion stability is rescued by 4E-BP1 depletion. Together our results confirm an unexpected role of BAD in controlling localized translation and cell migration during mammary gland development.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mammary Glands, Human/growth & development , Mammary Glands, Human/metabolism , bcl-Associated Death Protein/metabolism , Amino Acid Substitution , Animals , Cell Line , Cell Movement/genetics , Female , Gene Knock-In Techniques , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Morphogenesis , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Organoids/growth & development , Organoids/metabolism , Phosphorylation , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serine/chemistry , bcl-Associated Death Protein/deficiency , bcl-Associated Death Protein/genetics
16.
Cell Rep ; 37(8): 110037, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34818536

ABSTRACT

Glucose metabolism modulates the islet ß cell responses to diabetogenic stress, including inflammation. Here, we probed the metabolic mechanisms that underlie the protective effect of glucose in inflammation by interrogating the metabolite profiles of primary islets from human donors and identified de novo glutathione synthesis as a prominent glucose-driven pro-survival pathway. We find that pyruvate carboxylase is required for glutathione synthesis in islets and promotes their antioxidant capacity to counter inflammation and nitrosative stress. Loss- and gain-of-function studies indicate that pyruvate carboxylase is necessary and sufficient to mediate the metabolic input from glucose into glutathione synthesis and the oxidative stress response. Altered redox metabolism and cellular capacity to replenish glutathione pools are relevant in multiple pathologies beyond obesity and diabetes. Our findings reveal a direct interplay between glucose metabolism and glutathione biosynthesis via pyruvate carboxylase. This metabolic axis may also have implications in other settings where sustaining glutathione is essential.


Subject(s)
Glucose/metabolism , Glutathione/biosynthesis , Pyruvate Carboxylase/metabolism , Adult , Animals , Antioxidants/physiology , Female , Glutathione/metabolism , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Oxidation-Reduction , Oxidative Stress/physiology , Primary Cell Culture
17.
Nat Metab ; 3(5): 604-617, 2021 05.
Article in English | MEDLINE | ID: mdl-34002097

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD), the most prevalent liver pathology worldwide, is intimately linked with obesity and type 2 diabetes. Liver inflammation is a hallmark of NAFLD and is thought to contribute to tissue fibrosis and disease pathogenesis. Uncoupling protein 1 (UCP1) is exclusively expressed in brown and beige adipocytes, and has been extensively studied for its capacity to elevate thermogenesis and reverse obesity. Here we identify an endocrine pathway regulated by UCP1 that antagonizes liver inflammation and pathology, independent of effects on obesity. We show that, without UCP1, brown and beige fat exhibit a diminished capacity to clear succinate from the circulation. Moreover, UCP1KO mice exhibit elevated extracellular succinate in liver tissue that drives inflammation through ligation of its cognate receptor succinate receptor 1 (SUCNR1) in liver-resident stellate cell and macrophage populations. Conversely, increasing brown and beige adipocyte content in mice antagonizes SUCNR1-dependent inflammatory signalling in the liver. We show that this UCP1-succinate-SUCNR1 axis is necessary to regulate liver immune cell infiltration and pathology, and systemic glucose intolerance in an obesogenic environment. As such, the therapeutic use of brown and beige adipocytes and UCP1 extends beyond thermogenesis and may be leveraged to antagonize NAFLD and SUCNR1-dependent liver inflammation.


Subject(s)
Disease Susceptibility , Hepatitis/etiology , Hepatitis/metabolism , Succinic Acid/metabolism , Uncoupling Protein 1/genetics , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/metabolism , Animals , Extracellular Space/metabolism , Glucose/metabolism , Glucose Intolerance/metabolism , Hepatitis/pathology , Humans , Metabolic Networks and Pathways , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Uncoupling Protein 1/metabolism
18.
Biosens Bioelectron ; 178: 113031, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33571808

ABSTRACT

Aberrant production of reactive oxygen species (ROS) leads to tissue damage accumulation, which is associated with a myriad of human pathologies. Although several sensors have been developed for ROS quantification, their applications for ROS-related human physiologies and pathologies still remain problematic due to the unstable nature of ROS. Herein, we developed Trx1-cpYFP-fRMsr (TYfR), a genetically-encoded fluorescent biosensor with the remarkable specificity and sensitivity toward fMetRO (free Methionine-R-sulfoxide), allowing for dynamic quantification of physiological levels of fMetRO, a novel indicator of ROS and methionine redox status in vitro and in vivo. Moreover, using the sensor, we observed a significant fMetRO enrichment in serum from patients with acute coronary syndrome, one of the most severe cardiovascular diseases, which becomes more evident following percutaneous coronary intervention. Collectively, this study proposes that fMetRO is a novel biomarker of tissue damage accumulation in ROS-associated human pathologies, and that TYfR is a promising tool for quantifying fMetRO with potentials in versatile applications.


Subject(s)
Biosensing Techniques , Methionine Sulfoxide Reductases , Humans , Methionine , Methionine Sulfoxide Reductases/metabolism , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species
19.
Mol Cell Biol ; 27(4): 1222-35, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17145774

ABSTRACT

How cells die in the absence of oxygen (anoxia) is not understood. Here we report that cells deficient in Bax and Bak or caspase-9 do not undergo anoxia-induced cell death. However, the caspase-9 null cells do not survive reoxygenation due to the generation of mitochondrial reactive oxygen species. The individual loss of Bim, Bid, Puma, Noxa, Bad, caspase-2, or hypoxia-inducible factor 1beta, which are potential upstream regulators of Bax or Bak, did not prevent anoxia-induced cell death. Anoxia triggered the loss of the Mcl-1 protein upstream of Bax/Bak activation. Cells containing a mitochondrial DNA cytochrome b 4-base-pair deletion ([rho(-)] cells) and cells depleted of their entire mitochondrial DNA ([rho(0)] cells) are oxidative phosphorylation incompetent and displayed loss of the Mcl-1 protein under anoxia. [rho(0)] cells, in contrast to [rho(-)] cells, did not die under anoxia. However, [rho(0)] cells did undergo cell death in the presence of the Bad BH3 peptide, an inhibitor of Bcl-X(L)/Bcl-2 proteins. These results indicate that [rho(0)] cells survive under anoxia despite the loss of Mcl-1 protein due to residual prosurvival activity of the Bcl-X(L)/Bcl-2 proteins. Collectively, these results demonstrate that anoxia-induced cell death requires the loss of Mcl-1 protein and inhibition of the electron transport chain to negate Bcl-X(L)/Bcl-2 proteins.


Subject(s)
Fibroblasts/cytology , Neoplasm Proteins/deficiency , Proto-Oncogene Proteins c-bcl-2/deficiency , Adaptation, Physiological/drug effects , Animals , Caspase 9/deficiency , Caspase 9/metabolism , Cell Death/drug effects , Cell Hypoxia/drug effects , Cytochromes b/genetics , DNA, Mitochondrial/metabolism , Electron Transport/drug effects , Fibroblasts/drug effects , Fibroblasts/enzymology , Glycolysis/drug effects , Humans , Hypoxia-Inducible Factor 1/metabolism , Mice , Models, Biological , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/metabolism , Oxygen/pharmacology , Protein Structure, Tertiary/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Sequence Deletion , Tumor Cells, Cultured
20.
Nature ; 424(6951): 952-6, 2003 Aug 21.
Article in English | MEDLINE | ID: mdl-12931191

ABSTRACT

Glycolysis and apoptosis are considered major but independent pathways that are critical for cell survival. The activity of BAD, a pro-apoptotic BCL-2 family member, is regulated by phosphorylation in response to growth/survival factors. Here we undertook a proteomic analysis to assess whether BAD might also participate in mitochondrial physiology. In liver mitochondria, BAD resides in a functional holoenzyme complex together with protein kinase A and protein phosphatase 1 (PP1) catalytic units, Wiskott-Aldrich family member WAVE-1 as an A kinase anchoring protein, and glucokinase (hexokinase IV). BAD is required to assemble the complex in that Bad-deficient hepatocytes lack this complex, resulting in diminished mitochondria-based glucokinase activity and blunted mitochondrial respiration in response to glucose. Glucose deprivation results in dephosphorylation of BAD, and BAD-dependent cell death. Moreover, the phosphorylation status of BAD helps regulate glucokinase activity. Mice deficient for BAD or bearing a non-phosphorylatable BAD(3SA) mutant display abnormal glucose homeostasis including profound defects in glucose tolerance. This combination of proteomics, genetics and physiology indicates an unanticipated role for BAD in integrating pathways of glucose metabolism and apoptosis.


Subject(s)
Apoptosis , Carrier Proteins/metabolism , Glucokinase/metabolism , Glycolysis , Mitochondria, Liver/chemistry , Mitochondria, Liver/metabolism , Animals , Carrier Proteins/genetics , Gene Deletion , Glucose/metabolism , Holoenzymes/metabolism , Homeostasis , Macromolecular Substances , Mice , Mice, Knockout , Mitochondria, Liver/enzymology , Phosphorylation , Proteomics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-Associated Death Protein
SELECTION OF CITATIONS
SEARCH DETAIL