Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Metab ; 5(11): 1969-1985, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37884694

ABSTRACT

T cell activation is associated with a profound and rapid metabolic response to meet increased energy demands for cell division, differentiation and development of effector function. Glucose uptake and engagement of the glycolytic pathway are major checkpoints for this event. Here we show that the low-affinity, concentration-dependent glucose transporter 2 (Glut2) regulates the development of CD8+ T cell effector responses in mice by promoting glucose uptake, glycolysis and glucose storage. Expression of Glut2 is modulated by environmental factors including glucose and oxygen availability and extracellular acidification. Glut2 is highly expressed by circulating, recently primed T cells, allowing efficient glucose uptake and storage. In glucose-deprived inflammatory environments, Glut2 becomes downregulated, thus preventing passive loss of intracellular glucose. Mechanistically, Glut2 expression is regulated by a combination of molecular interactions involving hypoxia-inducible factor-1 alpha, galectin-9 and stomatin. Finally, we show that human T cells also rely on this glucose transporter, thus providing a potential target for therapeutic immunomodulation.


Subject(s)
Glucose Transport Proteins, Facilitative , Glucose , Mice , Humans , Animals , Glucose/metabolism , Biological Transport/physiology , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/metabolism , Cell Differentiation , CD8-Positive T-Lymphocytes/metabolism
2.
Sci Transl Med ; 15(685): eabn5135, 2023 03.
Article in English | MEDLINE | ID: mdl-36857430

ABSTRACT

Patients with myelodysplastic syndrome and ring sideroblasts (MDS-RS) present with symptomatic anemia due to ineffective erythropoiesis that impedes their quality of life and increases morbidity. More than 80% of patients with MDS-RS harbor splicing factor 3B subunit 1 (SF3B1) mutations, the founder aberration driving MDS-RS disease. Here, we report how mis-splicing of coenzyme A synthase (COASY), induced by mutations in SF3B1, affects heme biosynthesis and erythropoiesis. Our data revealed that COASY was up-regulated during normal erythroid differentiation, and its silencing prevented the formation of erythroid colonies, impeded erythroid differentiation, and precluded heme accumulation. In patients with MDS-RS, loss of protein due to COASY mis-splicing led to depletion of both CoA and succinyl-CoA. Supplementation with COASY substrate (vitamin B5) rescued CoA and succinyl-CoA concentrations in SF3B1mut cells and mended erythropoiesis differentiation defects in MDS-RS primary patient cells. Our findings reveal a key role of the COASY pathway in erythroid maturation and identify upstream and downstream metabolites of COASY as a potential treatment for anemia in patients with MDS-RS.


Subject(s)
Anemia , Myelodysplastic Syndromes , Humans , Erythropoiesis , Pantothenic Acid , Quality of Life , Transcription Factors , Heme , RNA Splicing Factors , Phosphoproteins
3.
JCI Insight ; 7(10)2022 05 23.
Article in English | MEDLINE | ID: mdl-35472029

ABSTRACT

Voltage-gated hydrogen channel 1 (Hvcn1) is a voltage-gated proton channel, which reduces cytosol acidification and facilitates the production of ROS. The increased expression of this channel in some cancers has led to proposing Hvcn1 antagonists as potential therapeutics. While its role in most leukocytes has been studied in depth, the function of Hvcn1 in T cells remains poorly defined. We show that Hvcn1 plays a nonredundant role in protecting naive T cells from intracellular acidification during priming. Despite sharing overall functional impairment in vivo and in vitro, Hvcn1-deficient CD4+ and CD8+ T cells display profound differences during the transition from naive to primed T cells, including in the preservation of T cell receptor (TCR) signaling, cellular division, and death. These selective features result, at least in part, from a substantially different metabolic response to intracellular acidification associated with priming. While Hvcn1-deficient naive CD4+ T cells reprogram to rescue the glycolytic pathway, naive CD8+ T cells, which express high levels of this channel in the mitochondria, respond by metabolically compensating mitochondrial dysfunction, at least in part via AMPK activation. These observations imply heterogeneity between adaptation of naive CD4+ and CD8+ T cells to intracellular acidification during activation.


Subject(s)
Hydrogen , Protons , Hydrogen-Ion Concentration , Lymphocyte Count , Signal Transduction
4.
Nat Metab ; 2(11): 1223-1231, 2020 11.
Article in English | MEDLINE | ID: mdl-33106688

ABSTRACT

Cardiomyocytes rely on metabolic substrates, not only to fuel cardiac output, but also for growth and remodelling during stress. Here we show that mitochondrial pyruvate carrier (MPC) abundance mediates pathological cardiac hypertrophy. MPC abundance was reduced in failing hypertrophic human hearts, as well as in the myocardium of mice induced to fail by angiotensin II or through transverse aortic constriction. Constitutive knockout of cardiomyocyte MPC1/2 in mice resulted in cardiac hypertrophy and reduced survival, while tamoxifen-induced cardiomyocyte-specific reduction of MPC1/2 to the attenuated levels observed during pressure overload was sufficient to induce hypertrophy with impaired cardiac function. Failing hearts from cardiomyocyte-restricted knockout mice displayed increased abundance of anabolic metabolites, including amino acids and pentose phosphate pathway intermediates and reducing cofactors. These hearts showed a concomitant decrease in carbon flux into mitochondrial tricarboxylic acid cycle intermediates, as corroborated by complementary 1,2-[13C2]glucose tracer studies. In contrast, inducible cardiomyocyte overexpression of MPC1/2 resulted in increased tricarboxylic acid cycle intermediates, and sustained carrier expression during transverse aortic constriction protected against cardiac hypertrophy and failure. Collectively, our findings demonstrate that loss of the MPC1/2 causally mediates adverse cardiac remodelling.


Subject(s)
Anion Transport Proteins/metabolism , Cardiomegaly/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Monocarboxylic Acid Transporters/metabolism , Angiotensin II , Animals , Anion Transport Proteins/biosynthesis , Anion Transport Proteins/genetics , Cardiomegaly/pathology , Cell Proliferation , Citric Acid Cycle , Constriction, Pathologic , Female , Heart Failure/chemically induced , Heart Failure/metabolism , Heart Failure/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria, Heart/metabolism , Mitochondrial Membrane Transport Proteins/biosynthesis , Mitochondrial Membrane Transport Proteins/genetics , Monocarboxylic Acid Transporters/biosynthesis , Monocarboxylic Acid Transporters/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Pyruvic Acid/metabolism
5.
EMBO Rep ; 21(9): e48260, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32783398

ABSTRACT

IκB kinase ε (IKKε) is a key molecule at the crossroads of inflammation and cancer. Known to regulate cytokine secretion via NFκB and IRF3, the kinase is also a breast cancer oncogene, overexpressed in a variety of tumours. However, to what extent IKKε remodels cellular metabolism is currently unknown. Here, we used metabolic tracer analysis to show that IKKε orchestrates a complex metabolic reprogramming that affects mitochondrial metabolism and consequently serine biosynthesis independently of its canonical signalling role. We found that IKKε upregulates the serine biosynthesis pathway (SBP) indirectly, by limiting glucose-derived pyruvate utilisation in the TCA cycle, inhibiting oxidative phosphorylation. Inhibition of mitochondrial function induces activating transcription factor 4 (ATF4), which in turn drives upregulation of the expression of SBP genes. Importantly, pharmacological reversal of the IKKε-induced metabolic phenotype reduces proliferation of breast cancer cells. Finally, we show that in a highly proliferative set of ER negative, basal breast tumours, IKKε and PSAT1 are both overexpressed, corroborating the link between IKKε and the SBP in the clinical context.


Subject(s)
Breast Neoplasms , I-kappa B Kinase , Mitochondria , Serine/biosynthesis , Breast Neoplasms/genetics , Female , Humans , I-kappa B Kinase/genetics , Mitochondria/genetics , Mitochondria/metabolism , Oncogenes/genetics
6.
EMBO Mol Med ; 12(2): e10491, 2020 02 07.
Article in English | MEDLINE | ID: mdl-31930708

ABSTRACT

During obesity, macrophages infiltrate the breast tissue leading to low-grade chronic inflammation, a factor considered responsible for the higher risk of breast cancer associated with obesity. Here, we formally demonstrate that breast epithelial cells acquire malignant properties when exposed to medium conditioned by macrophages derived from human healthy donors. These effects were mediated by the breast cancer oncogene IKKε and its downstream target-the serine biosynthesis pathway as demonstrated by genetic or pharmacological tools. Furthermore, amlexanox, an FDA-approved drug targeting IKKε and its homologue TBK1, delayed in vivo tumour formation in a combined genetic mouse model of breast cancer and high-fat diet-induced obesity/inflammation. Finally, in human breast cancer tissues, we validated the link between inflammation-IKKε and alteration of cellular metabolism. Altogether, we identified a pathway connecting obesity-driven inflammation to breast cancer and a potential therapeutic strategy to reduce the risk of breast cancer associated with obesity.


Subject(s)
Breast Neoplasms/pathology , I-kappa B Kinase , Macrophages/cytology , Protein Serine-Threonine Kinases/metabolism , Serine , Aminopyridines/pharmacology , Animals , Culture Media, Conditioned , Epithelial Cells/pathology , Female , Humans , I-kappa B Kinase/metabolism , Inflammation , Mammary Glands, Human/pathology , Mice , Obesity , Serine/biosynthesis
7.
Mol Cell ; 73(3): 413-428.e7, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30598363

ABSTRACT

Receptor-interacting protein kinase (RIPK) 1 functions as a key mediator of tissue homeostasis via formation of Caspase-8 activating ripoptosome complexes, positively and negatively regulating apoptosis, necroptosis, and inflammation. Here, we report an unanticipated cell-death- and inflammation-independent function of RIPK1 and Caspase-8, promoting faithful chromosome alignment in mitosis and thereby ensuring genome stability. We find that ripoptosome complexes progressively form as cells enter mitosis, peaking at metaphase and disassembling as cells exit mitosis. Genetic deletion and mitosis-specific inhibition of Ripk1 or Caspase-8 results in chromosome alignment defects independently of MLKL. We found that Polo-like kinase 1 (PLK1) is recruited into mitotic ripoptosomes, where PLK1's activity is controlled via RIPK1-dependent recruitment and Caspase-8-mediated cleavage. A fine balance of ripoptosome assembly is required as deregulated ripoptosome activity modulates PLK1-dependent phosphorylation of downstream effectors, such as BUBR1. Our data suggest that ripoptosome-mediated regulation of PLK1 contributes to faithful chromosome segregation during mitosis.


Subject(s)
Caspase 8/metabolism , Chromosomal Instability , Colonic Neoplasms/enzymology , Fibroblasts/enzymology , Mitosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Aneuploidy , Animals , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosome Segregation , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Fibroblasts/pathology , HT29 Cells , Humans , Inflammation/enzymology , Inflammation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction , Polo-Like Kinase 1
8.
Mol Cell ; 69(4): 566-580.e5, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29452637

ABSTRACT

Tumor necrosis factor (TNF) can drive inflammation, cell survival, and death. While ubiquitylation-, phosphorylation-, and nuclear factor κB (NF-κB)-dependent checkpoints suppress the cytotoxic potential of TNF, it remains unclear whether ubiquitylation can directly repress TNF-induced death. Here, we show that ubiquitylation regulates RIPK1's cytotoxic potential not only via activation of downstream kinases and NF-kB transcriptional responses, but also by directly repressing RIPK1 kinase activity via ubiquitin-dependent inactivation. We find that the ubiquitin-associated (UBA) domain of cellular inhibitor of apoptosis (cIAP)1 is required for optimal ubiquitin-lysine occupancy and K48 ubiquitylation of RIPK1. Independently of IKK and MK2, cIAP1-mediated and UBA-assisted ubiquitylation suppresses RIPK1 kinase auto-activation and, in addition, marks it for proteasomal degradation. In the absence of a functional UBA domain of cIAP1, more active RIPK1 kinase accumulates in response to TNF, causing RIPK1 kinase-mediated cell death and systemic inflammatory response syndrome. These results reveal a direct role for cIAP-mediated ubiquitylation in controlling RIPK1 kinase activity and preventing TNF-mediated cytotoxicity.


Subject(s)
Baculoviral IAP Repeat-Containing 3 Protein/physiology , I-kappa B Kinase/metabolism , Inhibitor of Apoptosis Proteins/physiology , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Ubiquitin/metabolism , Animals , Apoptosis , HEK293 Cells , Humans , I-kappa B Kinase/genetics , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitination
9.
Cell Death Differ ; 25(3): 486-541, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29362479

ABSTRACT

Over the past decade, the Nomenclature Committee on Cell Death (NCCD) has formulated guidelines for the definition and interpretation of cell death from morphological, biochemical, and functional perspectives. Since the field continues to expand and novel mechanisms that orchestrate multiple cell death pathways are unveiled, we propose an updated classification of cell death subroutines focusing on mechanistic and essential (as opposed to correlative and dispensable) aspects of the process. As we provide molecularly oriented definitions of terms including intrinsic apoptosis, extrinsic apoptosis, mitochondrial permeability transition (MPT)-driven necrosis, necroptosis, ferroptosis, pyroptosis, parthanatos, entotic cell death, NETotic cell death, lysosome-dependent cell death, autophagy-dependent cell death, immunogenic cell death, cellular senescence, and mitotic catastrophe, we discuss the utility of neologisms that refer to highly specialized instances of these processes. The mission of the NCCD is to provide a widely accepted nomenclature on cell death in support of the continued development of the field.


Subject(s)
Cell Death , Animals , Humans , Lysosomes/metabolism , Lysosomes/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Necrosis/metabolism , Necrosis/pathology
10.
Nat Commun ; 7: 10972, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26960254

ABSTRACT

Caspases provide vital links in non-apoptotic regulatory networks controlling inflammation, compensatory proliferation, morphology and cell migration. How caspases are activated under non-apoptotic conditions and process a selective set of substrates without killing the cell remain enigmatic. Here we find that the Drosophila unconventional myosin CRINKLED (CK) selectively interacts with the initiator caspase DRONC and regulates some of its non-apoptotic functions. Loss of CK in the arista, border cells or proneural clusters of the wing imaginal discs affects DRONC-dependent patterning. Our data indicate that CK acts as substrate adaptor, recruiting SHAGGY46/GSK3-ß to DRONC, thereby facilitating caspase-mediated cleavage and localized modulation of kinase activity. Similarly, the mammalian CK counterpart, MYO7A, binds to and impinges on CASPASE-8, revealing a new regulatory axis affecting receptor interacting protein kinase-1 (RIPK1)>CASPASE-8 signalling. Together, our results expose a conserved role for unconventional myosins in transducing caspase-dependent regulation of kinases, allowing them to take part in specific signalling events.


Subject(s)
Caspase 8/metabolism , Caspases/metabolism , Drosophila Proteins/metabolism , Myosins/metabolism , Animals , Cell Line, Tumor , Drosophila melanogaster , Flow Cytometry , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Immunoprecipitation , Mice , Microscopy, Confocal , Myosin VIIa , NIH 3T3 Cells , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Wings, Animal
11.
Front Immunol ; 6: 227, 2015.
Article in English | MEDLINE | ID: mdl-26029212

ABSTRACT

Aerobic glycolysis has been generally associated with cancer cell proliferation, but fascinating and novel data show that it is also coupled to a series of further cellular functions. In this Mini Review, we will discuss some recent findings to illustrate newly defined roles for this process, in particular in non-malignant cells, supporting the idea that metabolism can be considered as an integral part of cellular signaling. Consequently, metabolism should be regarded as a plastic and highly dynamic determinant of a wide range of cellular specific functions.

13.
Mol Cell ; 43(3): 432-48, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21737329

ABSTRACT

A better understanding of the mechanisms through which anticancer drugs exert their effects is essential to improve combination therapies. While studying how genotoxic stress kills cancer cells, we discovered a large ∼2MDa cell death-inducing platform, referred to as "Ripoptosome." It contains the core components RIP1, FADD, and caspase-8, and assembles in response to genotoxic stress-induced depletion of XIAP, cIAP1 and cIAP2. Importantly, it forms independently of TNF, CD95L/FASL, TRAIL, death-receptors, and mitochondrial pathways. It also forms upon Smac-mimetic (SM) treatment without involvement of autocrine TNF. Ripoptosome assembly requires RIP1's kinase activity and can stimulate caspase-8-mediated apoptosis as well as caspase-independent necrosis. It is negatively regulated by FLIP, cIAP1, cIAP2, and XIAP. Mechanistically, IAPs target components of this complex for ubiquitylation and inactivation. Moreover, we find that etoposide-stimulated Ripoptosome formation converts proinflammatory cytokines into prodeath signals. Together, our observations shed new light on fundamental mechanisms by which chemotherapeutics may kill cancer cells.


Subject(s)
Apoptosis/physiology , Caspase 8/physiology , DNA Damage , Fas-Associated Death Domain Protein/physiology , Inhibitor of Apoptosis Proteins/genetics , Nuclear Pore Complex Proteins/physiology , RNA-Binding Proteins/physiology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Caspase 8/chemistry , Caspase 8/metabolism , Cell Line, Tumor , Enzyme Activation , Etoposide/pharmacology , Fas-Associated Death Domain Protein/chemistry , Fas-Associated Death Domain Protein/metabolism , Humans , Inhibitor of Apoptosis Proteins/physiology , Ligands , Mitochondria/metabolism , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Signal Transduction
14.
Exp Cell Res ; 316(13): 2071-86, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20420828

ABSTRACT

The mechanisms of secretory transport through the Golgi apparatus remain an issue of debate. The precise functional importance of calcium ions (Ca(2+)) for intra-Golgi transport has also been poorly studied. Here, using different approaches to measure free Ca(2+) concentrations in the cell cytosol ([Ca(2+)](cyt)) and inside the lumen of the Golgi apparatus ([Ca(2+)](GA)), we have revealed transient increases in [Ca(2+)](cyt) during the late phase of intra-Golgi transport that are concomitant with a decline in the maximal [Ca(2+)](GA) restoration ability. Thus, this redistribution of Ca(2+) from the Golgi apparatus into the cytosol during the movement of cargo through the Golgi apparatus appears to have a role in intra-Golgi transport, and mainly in the late Ca(2+)-dependent phase of SNARE-regulated fusion of Golgi compartments.


Subject(s)
Calcium/metabolism , Cytosol/metabolism , Golgi Apparatus/metabolism , Biological Transport , Calcium Signaling , Cells, Cultured , Fibroblasts/metabolism , HeLa Cells , Humans , Skin/cytology , Skin/metabolism , Subcellular Fractions
15.
Mol Cell ; 36(5): 736-42, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-20005838

ABSTRACT

A flurry of recent revelations is challenging the current dogma on how ubiquitin-dependent processes culminate in the activation of NF-kappaB by TNF. Here, we integrate these findings into a model for TNF-R1 signaling-and underscore the importance of individual components, including linear ubiquitin chains-which allows for the remarkable versatility of the ubiquitin system.


Subject(s)
Models, Biological , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Ubiquitin/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin/physiology
16.
Proc Natl Acad Sci U S A ; 105(4): 1226-31, 2008 Jan 29.
Article in English | MEDLINE | ID: mdl-18212116

ABSTRACT

Regeneration of mesenchymal tissues depends on a resident stem cell population, that in most cases remains elusive in terms of cellular identity and differentiation signals. We here show that primary cell cultures derived from adipose tissue or skeletal muscle differentiate into adipocytes when cultured in high glucose. High glucose induces ROS production and PKCbeta activation. These two events appear crucial steps in this differentiation process that can be directly induced by oxidizing agents and inhibited by PKCbeta siRNA silencing. The differentiated adipocytes, when implanted in vivo, form viable and vascularized adipose tissue. Overall, the data highlight a previously uncharacterized differentiation route triggered by high glucose that drives not only resident stem cells of the adipose tissue but also uncommitted precursors present in muscle cells to form adipose depots. This process may represent a feed-forward cycle between the regional increase in adiposity and insulin resistance that plays a key role in the pathogenesis of diabetes mellitus.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/drug effects , Cell Differentiation/drug effects , Glucose/pharmacology , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Adipocytes/transplantation , Adipocytes/ultrastructure , Adipogenesis/physiology , Adipose Tissue/blood supply , Adipose Tissue/cytology , Adipose Tissue/ultrastructure , Animals , Biomarkers/metabolism , Cell Differentiation/physiology , Cells, Cultured , Culture Media, Conditioned , Female , Glucose/metabolism , Humans , Muscle, Skeletal/ultrastructure , Rats , Rats, Nude , Stem Cells/ultrastructure
17.
J Clin Endocrinol Metab ; 92(12): 4810-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17785353

ABSTRACT

BACKGROUND: The endogenous cannabinoid system participates in the regulation of energy balance, and its dysregulation may be implicated in the pathogenesis of obesity. Adipose tissue endocannabinoids may produce metabolic and endocrine effects, but very few data are available in human adipose tissue and in primary human fat cells. EXPERIMENTAL DESIGN: We measured expression of type 1 and type 2 cannabinoid receptors (CNR), enzymes of cannabinoids synthesis and degradation in human omental, sc abdominal, and gluteal adipose tissue from lean and obese subjects. Furthermore, we assessed the effect of CNR1 stimulation on glucose uptake and intracellular transduction mechanisms in primary human adipocytes. Then we assessed the reciprocal regulation between CNR1 and peroxisome proliferator-activated receptor-gamma (PPARgamma). Finally, we tested whether leptin and adiponectin are regulated by CNR1 in human adipocytes. RESULTS: We found that most genes of the endocannabinoid system are down-regulated in gluteal fat and up-regulated in visceral and sc abdominal adipose tissue of obese patients. Treatment of adipocytes with rosiglitazone markedly down-regulated CNR1 expression, whereas Win 55,212 up-regulated PPARgamma. Win 55,212 increased (+50%) glucose uptake, the translocation of glucose transporter 4, and intracellular calcium in fat cells. All these effects were inhibited by SR141716 and wortmannin and by removing extracellular calcium. Win 55,212 and SR141716 had no effect on expression of adiponectin and leptin. CONCLUSIONS: These results indicate a role for the local endocannabinoids in the regulation of glucose metabolism in human adipocytes and suggest a role in channeling excess energy fuels to adipose tissue in obese humans.


Subject(s)
Adipocytes/metabolism , Calcium/physiology , Cannabinoid Receptor Modulators/physiology , Glucose/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Adipose Tissue/chemistry , Adipose Tissue/metabolism , Adult , Body Mass Index , Calcium/metabolism , Cell Differentiation/physiology , Female , Humans , Hypoglycemic Agents/pharmacology , Male , Microscopy, Confocal , Obesity/metabolism , PPAR gamma/metabolism , Protein Transport , RNA/biosynthesis , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone , Thiazolidinediones/pharmacology
18.
Mol Cell ; 25(2): 193-205, 2007 Jan 26.
Article in English | MEDLINE | ID: mdl-17244528

ABSTRACT

Macroautophagy is an evolutionary conserved lysosomal pathway involved in the turnover of cellular macromolecules and organelles. In spite of its essential role in tissue homeostasis, the molecular mechanisms regulating mammalian macroautophagy are poorly understood. Here, we demonstrate that a rise in the free cytosolic calcium ([Ca(2+)](c)) is a potent inducer of macroautophagy. Various Ca(2+) mobilizing agents (vitamin D(3) compounds, ionomycin, ATP, and thapsigargin) inhibit the activity of mammalian target of rapamycin, a negative regulator of macroautophagy, and induce massive accumulation of autophagosomes in a Beclin 1- and Atg7-dependent manner. This process is mediated by Ca(2+)/calmodulin-dependent kinase kinase-beta and AMP-activated protein kinase and inhibited by ectopic Bcl-2 located in the endoplasmatic reticulum (ER), where it lowers the [Ca(2+)](ER) and attenuates agonist-induced Ca(2+) fluxes. Thus, an increase in the [Ca(2+)](c) serves as a potent inducer of macroautophagy and as a target for the antiautophagy action of ER-located Bcl-2.


Subject(s)
Autophagy/drug effects , Autophagy/physiology , Calcium/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , AMP-Activated Protein Kinases , Adenosine Triphosphate/pharmacology , Autophagy-Related Protein 7 , Base Sequence , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Kinase , Cell Line , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Ionomycin/pharmacology , Microscopy, Electron , Models, Biological , Multienzyme Complexes/metabolism , Protein Kinases/metabolism , RNA, Small Interfering/genetics , Signal Transduction , TOR Serine-Threonine Kinases , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism
19.
J Cell Biol ; 175(6): 901-11, 2006 Dec 18.
Article in English | MEDLINE | ID: mdl-17178908

ABSTRACT

The voltage-dependent anion channel (VDAC) of the outer mitochondrial membrane mediates metabolic flow, Ca(2+), and cell death signaling between the endoplasmic reticulum (ER) and mitochondrial networks. We demonstrate that VDAC1 is physically linked to the endoplasmic reticulum Ca(2+)-release channel inositol 1,4,5-trisphosphate receptor (IP(3)R) through the molecular chaperone glucose-regulated protein 75 (grp75). Functional interaction between the channels was shown by the recombinant expression of the ligand-binding domain of the IP(3)R on the ER or mitochondrial surface, which directly enhanced Ca(2+) accumulation in mitochondria. Knockdown of grp75 abolished the stimulatory effect, highlighting chaperone-mediated conformational coupling between the IP(3)R and the mitochondrial Ca(2+) uptake machinery. Because organelle Ca(2+) homeostasis influences fundamentally cellular functions and death signaling, the central location of grp75 may represent an important control point of cell fate and pathogenesis.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Molecular Chaperones/metabolism , Voltage-Dependent Anion Channel 1/metabolism , Aequorin , Animals , Calcium Signaling , HSP70 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Liver/metabolism , Membrane Proteins/metabolism , Mice , Protein Conformation , Rats , Two-Hybrid System Techniques
20.
J Cell Biol ; 174(7): 985-96, 2006 Sep 25.
Article in English | MEDLINE | ID: mdl-16982800

ABSTRACT

Replication of human cytomegalovirus (CMV) requires the expression of the viral mitochondria-localized inhibitor of apoptosis (vMIA). vMIA inhibits apoptosis by recruiting Bax to mitochondria, resulting in its neutralization. We show that vMIA decreases cell size, reduces actin polymerization, and induces cell rounding. As compared with vMIA-expressing CMV, vMIA-deficient CMV, which replicates in fibroblasts expressing the adenoviral apoptosis suppressor E1B19K, induces less cytopathic effects. These vMIA effects can be separated from its cell death-inhibitory function because vMIA modulates cellular morphology in Bax-deficient cells. Expression of vMIA coincided with a reduction in the cellular adenosine triphosphate (ATP) level. vMIA selectively inhibited one component of the ATP synthasome, namely, the mitochondrial phosphate carrier. Exposure of cells to inhibitors of oxidative phosphorylation produced similar effects, such as an ATP level reduced by 30%, smaller cell size, and deficient actin polymerization. Similarly, knockdown of the phosphate carrier reduced cell size. Our data suggest that the cytopathic effect of CMV can be explained by vMIA effects on mitochondrial bioenergetics.


Subject(s)
Apoptosis , Cytomegalovirus Infections/metabolism , Cytomegalovirus/physiology , Immediate-Early Proteins/physiology , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Viral Proteins/physiology , Actins/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Cytomegalovirus/genetics , Cytopathogenic Effect, Viral , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/pathology , Fibroblasts/virology , HeLa Cells , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/toxicity , Mice , Mitochondrial Proteins/genetics , NIH 3T3 Cells , Oxidative Phosphorylation/drug effects , Polymers/metabolism , Viral Proteins/genetics , Viral Proteins/toxicity , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-2-Associated X Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...