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










Publication year range
1.
bioRxiv ; 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38659932

ABSTRACT

E3-ubiquitin ligases (E3s) are main components of the ubiquitin-proteasome system (UPS), as they determine substrate specificity in response to internal and external cues to regulate protein homeostasis. However, the regulation of membrane protein ubiquitination by E3s within distinct cell membrane compartments or organelles is not well understood. We show that FBXO10, the interchangeable component of the SKP1/CUL1/F-box ubiquitin ligase complex (SCF-E3), undergoes lipid-modification with geranylgeranyl isoprenoid at Cysteine953 (C953), facilitating its dynamic trafficking to the outer mitochondrial membrane (OMM). FBXO10 polypeptide does not contain a canonical mitochondrial targeting sequence (MTS); instead, its geranylgeranylation at C953 and the interaction with two cytosolic factors, PDE6δ (a prenyl group-binding protein), and HSP90 (a mitochondrial chaperone) orchestrate specific OMM targeting of prenyl-FBXO10 across diverse membrane compartments. The geranylgeranylation-deficient FBXO10(C953S) mutant redistributes away from the OMM, leading to impaired mitochondrial ATP production, decreased mitochondrial membrane potential, and increased mitochondrial fragmentation. Phosphoglycerate mutase 5 (PGAM5) was identified as a potential substrate of FBXO10 at the OMM using comparative quantitative mass spectrometry analyses of enriched mitochondria (LFQ-MS/MS), leveraging the redistribution of FBXO10(C953S). FBXO10, but not FBXO10(C953S), promoted polyubiquitylation and degradation of PGAM5. Examination of the role of this pathway in a physiological context revealed that the loss of FBXO10 or expression of prenylation-deficient-FBXO10(C953S) inhibited PGAM5 degradation, disrupted mitochondrial homeostasis, and impaired myogenic differentiation of human iPSCs and murine myoblasts. Our studies identify a mechanism for selective E3-ligase mediated regulation of mitochondrial membrane proteostasis and metabolic health, potentially amenable to therapeutic intervention.

2.
Cell Metab ; 35(11): 2060-2076.e9, 2023 11 07.
Article in English | MEDLINE | ID: mdl-37852255

ABSTRACT

A high-fat diet (HFD) promotes metastasis through increased uptake of saturated fatty acids (SFAs). The fatty acid transporter CD36 has been implicated in this process, but a detailed understanding of CD36 function is lacking. During matrix detachment, endoplasmic reticulum (ER) stress reduces SCD1 protein, resulting in increased lipid saturation. Subsequently, CD36 is induced in a p38- and AMPK-dependent manner to promote preferential uptake of monounsaturated fatty acids (MUFAs), thereby maintaining a balance between SFAs and MUFAs. In attached cells, CD36 palmitoylation is required for MUFA uptake and protection from palmitate-induced lipotoxicity. In breast cancer mouse models, CD36-deficiency induced ER stress while diminishing the pro-metastatic effect of HFD, and only a palmitoylation-proficient CD36 rescued this effect. Finally, AMPK-deficient tumors have reduced CD36 expression and are metastatically impaired, but ectopic CD36 expression restores their metastatic potential. Our results suggest that, rather than facilitating HFD-driven tumorigenesis, CD36 plays a supportive role by preventing SFA-induced lipotoxicity.


Subject(s)
AMP-Activated Protein Kinases , Fatty Acids, Monounsaturated , Animals , Mice , Fatty Acids, Monounsaturated/metabolism , AMP-Activated Protein Kinases/metabolism , Fatty Acids/metabolism , Biological Transport , Homeostasis
3.
STAR Protoc ; 4(3): 102416, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37405928

ABSTRACT

Prenylation and palmitoylation are two major lipid modifications of cellular proteins that anchor proteins to cell membranes. Here, we present a protocol for detecting these modifications in cellular proteins by radioactive metabolic labeling. We describe steps for metabolic labeling of cells, cell harvesting for carrying out immunoprecipitations, subjecting immunocomplexes to SDS-PAGE, and transferring them to polyvinylidine flouride (PVDF) membranes. We then detail detection of labeled target proteins by exposing PVDF membranes to phosphor screens and using a phosphor imager machine. For complete details of this protocol, please refer to Liang et al.1.


Subject(s)
Fluorocarbon Polymers , Membrane Lipids , Proteins , Proteins/metabolism , Polyvinyls/metabolism , Lipid Metabolism
4.
Curr Opin Pharmacol ; 69: 102360, 2023 04.
Article in English | MEDLINE | ID: mdl-36870180
5.
Cell Rep ; 42(1): 111999, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36662618

ABSTRACT

Substrate degradation by the ubiquitin proteasome system (UPS) in specific membrane compartments remains elusive. Here, we show that the interplay of two lipid modifications and PDE6δ regulates compartmental substrate targeting via the SCFFBXL2. FBXL2 is palmitoylated in a prenylation-dependent manner on cysteines 417 and 419 juxtaposed to the CaaX motif. Palmitoylation/depalmitoylation regulates its subcellular trafficking for substrate engagement and degradation. To control its subcellular distribution, lipid-modified FBXL2 interacts with PDE6δ. Perturbing the equilibrium between FBXL2 and PDE6δ disrupts the delivery of FBXL2 to all membrane compartments, whereas depalmitoylated FBXL2 is enriched on the endoplasmic reticulum (ER). Depalmitoylated FBXL2(C417S/C419S) promotes the degradation of IP3R3 at the ER, inhibits IP3R3-dependent mitochondrial calcium overload, and counteracts calcium-dependent cell death upon oxidative stress. In contrast, disrupting the PDE6δ-FBXL2 equilibrium has the opposite effect. These findings describe a mechanism underlying spatially-restricted substrate degradation and suggest that inhibition of FBXL2 palmitoylation and/or binding to PDE6δ may offer therapeutic benefits.


Subject(s)
F-Box Proteins , F-Box Proteins/metabolism , Calcium/metabolism , Lipoylation , Ubiquitination , Lipids
6.
Curr Opin Pharmacol ; 67: 102310, 2022 12.
Article in English | MEDLINE | ID: mdl-36288660

ABSTRACT

The ubiquitin proteasome system (UPS) is a proteolytic machinery for the degradation of protein substrates that are post-translationally conjugated with ubiquitin polymers through the enzymatic action of ubiquitin ligases, in a process termed ubiquitylation. Ubiquitylation of substrates precedes their proteolysis via proteasomes, a hierarchical feature of UPS. E3-ubiquitin ligases recruit protein substrates providing specificity for ubiquitylation. Innate and adaptive immune system networks are regulated by ubiquitylation and substrate degradation via E3-ligases/UPS. Deregulation of E3-ligases/UPS components in immune cells is involved in the development of lymphomas, neurodevelopmental abnormalities, and cancers. Targeting E3-ligases for therapeutic intervention provides opportunities to mitigate the unintended broad effects of 26S proteasome inhibition. Recently, bifunctional moieties such as PROTACs and molecular glues have been developed to re-purpose E3-ligases for targeted degradation of unwanted aberrant proteins, with a potential for clinical use. Here, we summarize the involvement of E3-ligases/UPS components in immune-related diseases with perspectives.


Subject(s)
Proteasome Endopeptidase Complex , Ubiquitin , Humans , Ubiquitin/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Protein Ligases/metabolism , Proteolysis , Proteins/metabolism
7.
Nat Struct Mol Biol ; 26(7): 628-636, 2019 07.
Article in English | MEDLINE | ID: mdl-31209342

ABSTRACT

Protein prenylation is believed to be catalyzed by three heterodimeric enzymes: FTase, GGTase1 and GGTase2. Here we report the identification of a previously unknown human prenyltransferase complex consisting of an orphan prenyltransferase α-subunit, PTAR1, and the catalytic ß-subunit of GGTase2, RabGGTB. This enzyme, which we named GGTase3, geranylgeranylates FBXL2 to allow its localization at cell membranes, where this ubiquitin ligase mediates the polyubiquitylation of membrane-anchored proteins. In cells, FBXL2 is specifically recognized by GGTase3 despite having a typical carboxy-terminal CaaX prenylation motif that is predicted to be recognized by GGTase1. Our crystal structure analysis of the full-length GGTase3-FBXL2-SKP1 complex reveals an extensive multivalent interface specifically formed between the leucine-rich repeat domain of FBXL2 and PTAR1, which unmasks the structural basis of the substrate-enzyme specificity. By uncovering a missing prenyltransferase and its unique mode of substrate recognition, our findings call for a revision of the 'prenylation code'.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Dimethylallyltranstransferase/metabolism , F-Box Proteins/metabolism , Alkyl and Aryl Transferases/chemistry , Cell Line , Crystallography, X-Ray , Dimethylallyltranstransferase/chemistry , F-Box Proteins/chemistry , HeLa Cells , Humans , Models, Molecular , Polyubiquitin/metabolism , Protein Conformation , Protein Prenylation , Protein Subunits/chemistry , Protein Subunits/metabolism
8.
Cell Rep ; 25(4): 833-840.e3, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30355490

ABSTRACT

FBXL2 targets IP3R3 for ubiquitin-mediated degradation to limit Ca2+ flux to mitochondria and, consequently, apoptosis. Efficient replication of hepatitis C virus (HCV) requires geranylgeranylation of FBXL2. Here, we show that the viral protein NS5A forms a trimeric complex with IP3R3 and FBXL2, unmasking IP3R3's degron in the absence of inositol 1,4,5-trisphosphate (IP3) stimulation. FBXL2 knockdown or expression of a stable IP3R3 mutant causes persistent Ca2+ flux and sensitizes cells to apoptosis, resulting in the inhibition of viral replication. Importantly, the effect of FBXL2 silencing is rescued by depleting IP3R3, but not p85ß, another established FBXL2 substrate, indicating that the anti-HCV effect of FBXL2 knockdown is largely due to IP3R3 stabilization. Finally, disruption of the FBXL2-NS5A-IP3R3 complex using somatic cell genetics or pharmacologic inhibition results in IP3R3 stabilization and suppression of HCV replication. This study reveals an IP3-independent molecular mechanism through which HCV promotes IP3R3 degradation, thereby inhibiting virus-induced apoptosis and establishing chronic infection.


Subject(s)
Apoptosis , Hepacivirus/physiology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Proteolysis , Viral Nonstructural Proteins/metabolism , Animals , Apoptosis/drug effects , Carbamates , Cell Line , F-Box Proteins/metabolism , Hepacivirus/drug effects , Hepatitis C/metabolism , Hepatitis C/pathology , Hepatitis C/virology , Humans , Imidazoles/pharmacology , Leucine/analogs & derivatives , Leucine/pharmacology , Protein Multimerization/drug effects , Proteolysis/drug effects , Pyrrolidines , Valine/analogs & derivatives , Virus Replication/drug effects
9.
Cell Death Differ ; 24(10): 1694-1704, 2017 10.
Article in English | MEDLINE | ID: mdl-28665402

ABSTRACT

Carriers of heterozygous germline BAP1 mutations (BAP1+/-) develop cancer. We studied plasma from 16 BAP1+/- individuals from 2 families carrying different germline BAP1 mutations and 30 BAP1 wild-type (BAP1WT) controls from these same families. Plasma samples were analyzed by liquid chromatography time-of-flight mass spectrometry (LC-TOF-MS), ultra-performance liquid chromatography triple quadrupole mass spectrometry (UPLC-TQ-MS), and gas chromatography time-of-flight mass spectrometry (GC-TOF-MS). We found a clear separation in the metabolic profile between BAP1WT and BAP1+/- individuals. We confirmed the specificity of the data in vitro using 12 cell cultures of primary fibroblasts we derived from skin punch biopsies from 12/46 of these same individuals, 6 BAP1+/- carriers and 6 controls from both families. BAP1+/- fibroblasts displayed increased aerobic glycolysis and lactate secretion, and reduced mitochondrial respiration and ATP production compared with BAP1WT. siRNA-mediated downregulation of BAP1 in primary BAP1WT fibroblasts and in primary human mesothelial cells, led to the same reduced mitochondrial respiration and increased aerobic glycolysis as we detected in primary fibroblasts from carriers of BAP1+/- mutations. The plasma and cell culture results were highly reproducible and were specifically and only linked to BAP1 status and not to gender, age or family, or cell type, and required an intact BAP1 catalytic activity. Accordingly, we were able to build a metabolomic model capable of predicting BAP1 status with 100% accuracy using data from human plasma. Our data provide the first experimental evidence supporting the hypothesis that aerobic glycolysis, also known as the 'Warburg effect', does not necessarily occur as an adaptive process that is consequence of carcinogenesis, but rather that it may also predate malignancy by many years and facilitate carcinogenesis.


Subject(s)
Mitochondria/genetics , Mutation/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Germ Cells/metabolism , Heterozygote , Humans , Mitochondria/metabolism , Skin/pathology
10.
J Cell Biol ; 216(8): 2329-2338, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28619714

ABSTRACT

The product of the KRAS oncogene, KRAS4B, promotes tumor growth when associated with the plasma membrane (PM). PM association is mediated, in part, by farnesylation of KRAS4B, but trafficking of nascent KRAS4B to the PM is incompletely understood. We performed a genome-wide screen to identify genes required for KRAS4B membrane association and identified a G protein-coupled receptor, GPR31. GPR31 associated with KRAS4B on cellular membranes in a farnesylation-dependent fashion, and retention of GPR31 on the endoplasmic reticulum inhibited delivery of KRAS4B to the PM. Silencing of GPR31 expression partially mislocalized KRAS4B, slowed the growth of KRAS-dependent tumor cells, and blocked KRAS-stimulated macropinocytosis. Our data suggest that GPR31 acts as a secretory pathway chaperone for KRAS4B.


Subject(s)
Cell Membrane/enzymology , Molecular Chaperones/metabolism , Neoplasms/enzymology , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, G-Protein-Coupled/metabolism , A549 Cells , Cell Proliferation , Endoplasmic Reticulum/enzymology , HCT116 Cells , HeLa Cells , Humans , Molecular Chaperones/genetics , Mutation , Neoplasms/genetics , Pinocytosis , Prenylation , Protein Binding , Protein Isoforms , Protein Transport , Proto-Oncogene Proteins p21(ras)/genetics , RNA Interference , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Transfection , Tumor Burden
11.
Nature ; 546(7659): 554-558, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28614300

ABSTRACT

In response to environmental cues that promote IP3 (inositol 1,4,5-trisphosphate) generation, IP3 receptors (IP3Rs) located on the endoplasmic reticulum allow the 'quasisynaptical' feeding of calcium to the mitochondria to promote oxidative phosphorylation. However, persistent Ca2+ release results in mitochondrial Ca2+ overload and consequent apoptosis. Among the three mammalian IP3Rs, IP3R3 appears to be the major player in Ca2+-dependent apoptosis. Here we show that the F-box protein FBXL2 (the receptor subunit of one of 69 human SCF (SKP1, CUL1, F-box protein) ubiquitin ligase complexes) binds IP3R3 and targets it for ubiquitin-, p97- and proteasome-mediated degradation to limit Ca2+ influx into mitochondria. FBXL2-knockdown cells and FBXL2-insensitive IP3R3 mutant knock-in clones display increased cytosolic Ca2+ release from the endoplasmic reticulum and sensitization to Ca2+-dependent apoptotic stimuli. The phosphatase and tensin homologue (PTEN) gene is frequently mutated or lost in human tumours and syndromes that predispose individuals to cancer. We found that PTEN competes with FBXL2 for IP3R3 binding, and the FBXL2-dependent degradation of IP3R3 is accelerated in Pten-/- mouse embryonic fibroblasts and PTEN-null cancer cells. Reconstitution of PTEN-null cells with either wild-type PTEN or a catalytically dead mutant stabilizes IP3R3 and induces persistent Ca2+ mobilization and apoptosis. IP3R3 and PTEN protein levels directly correlate in human prostate cancer. Both in cell culture and xenograft models, a non-degradable IP3R3 mutant sensitizes tumour cells with low or no PTEN expression to photodynamic therapy, which is based on the ability of photosensitizer drugs to cause Ca2+-dependent cytotoxicity after irradiation with visible light. Similarly, disruption of FBXL2 localization with GGTi-2418, a geranylgeranyl transferase inhibitor, sensitizes xenotransplanted tumours to photodynamic therapy. In summary, we identify a novel molecular mechanism that limits mitochondrial Ca2+ overload to prevent cell death. Notably, we provide proof-of-principle that inhibiting IP3R3 degradation in PTEN-deregulated cancers represents a valid therapeutic strategy.


Subject(s)
Apoptosis , Calcium/metabolism , F-Box Proteins/antagonists & inhibitors , Inositol 1,4,5-Trisphosphate Receptors/metabolism , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Binding, Competitive , Calcium Signaling , Endoplasmic Reticulum/metabolism , F-Box Proteins/genetics , F-Box Proteins/metabolism , Fibroblasts , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/deficiency , Inositol 1,4,5-Trisphosphate Receptors/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/metabolism , Mutation , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Photochemotherapy , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , Ubiquitin/metabolism , Xenograft Model Antitumor Assays
12.
Nature ; 546(7659): 549-553, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28614305

ABSTRACT

BRCA1-associated protein 1 (BAP1) is a potent tumour suppressor gene that modulates environmental carcinogenesis. All carriers of inherited heterozygous germline BAP1-inactivating mutations (BAP1+/-) developed one and often several BAP1-/- malignancies in their lifetime, mostly malignant mesothelioma, uveal melanoma, and so on. Moreover, BAP1-acquired biallelic mutations are frequent in human cancers. BAP1 tumour suppressor activity has been attributed to its nuclear localization, where it helps to maintain genome integrity. The possible activity of BAP1 in the cytoplasm is unknown. Cells with reduced levels of BAP1 exhibit chromosomal abnormalities and decreased DNA repair by homologous recombination, indicating that BAP1 dosage is critical. Cells with extensive DNA damage should die and not grow into malignancies. Here we discover that BAP1 localizes at the endoplasmic reticulum. Here, it binds, deubiquitylates, and stabilizes type 3 inositol-1,4,5-trisphosphate receptor (IP3R3), modulating calcium (Ca2+) release from the endoplasmic reticulum into the cytosol and mitochondria, promoting apoptosis. Reduced levels of BAP1 in BAP1+/- carriers cause reduction both of IP3R3 levels and of Ca2+ flux, preventing BAP1+/- cells that accumulate DNA damage from executing apoptosis. A higher fraction of cells exposed to either ionizing or ultraviolet radiation, or to asbestos, survive genotoxic stress, resulting in a higher rate of cellular transformation. We propose that the high incidence of cancers in BAP1+/- carriers results from the combined reduced nuclear and cytoplasmic activities of BAP1. Our data provide a mechanistic rationale for the powerful ability of BAP1 to regulate gene-environment interaction in human carcinogenesis.


Subject(s)
Calcium/metabolism , Cell Transformation, Neoplastic , Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mitochondria/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Apoptosis/genetics , Asbestos/toxicity , Calcium Signaling , Cell Nucleus/metabolism , Cell Survival , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/radiation effects , Cells, Cultured , DNA Damage , Epithelium , Fibroblasts , Gene-Environment Interaction , Humans , Protein Binding , Protein Stability , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Ubiquitin/metabolism , Ubiquitin Thiolesterase/deficiency , Ubiquitin Thiolesterase/genetics
13.
Nat Cell Biol ; 15(5): 472-80, 2013 May.
Article in English | MEDLINE | ID: mdl-23604317

ABSTRACT

F-box proteins are the substrate-recognition subunits of SCF (Skp1/Cul1/F-box protein) ubiquitin ligase complexes. Purification of the F-box protein FBXL2 identified the PI(3)K regulatory subunit p85ß and tyrosine phosphatase PTPL1 as interacting proteins. FBXL2 interacts with the pool of p85ß that is free of p110 PI(3)K catalytic subunits and targets this pool for ubiquitylation and subsequent proteasomal degradation. FBXL2-mediated degradation of p85ß is dependent on the integrity of its CaaX motif. Whereas most SCF substrates require phosphorylation to interact with their F-box proteins, phosphorylation of p85ß on Tyr 655, which is adjacent to the degron, inhibits p85ß binding to FBXL2. Dephosphorylation of phospho-Tyr-655 by PTPL1 stimulates p85ß binding to and degradation through FBXL2. Finally, defects in the FBXL2-mediated degradation of p85ß inhibit the binding of p110 subunits to IRS1, attenuate the PI(3)K signalling cascade and promote autophagy. We propose that FBXL2 and PTPL1 suppress p85ß levels, preventing the inhibition of PI(3)K by an excess of free p85 that could compete with p85-p110 heterodimers for IRS1.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , F-Box Proteins/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 13/metabolism , Signal Transduction , Autophagy , Class Ia Phosphatidylinositol 3-Kinase/genetics , Enzyme Activation , Enzyme Stability , F-Box Proteins/genetics , Gene Silencing , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Insulin Receptor Substrate Proteins/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Interaction Mapping , Protein Tyrosine Phosphatase, Non-Receptor Type 13/genetics , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transfection , Tyrosine/metabolism , Ubiquitination
14.
Clin Cases Miner Bone Metab ; 9(2): 118-20, 2012 May.
Article in English | MEDLINE | ID: mdl-23087723

ABSTRACT

Vitamin D is increasingly recognized to have several beneficial effects. Its toxicity, causing hypercalcemia, is considered as extremely rare. We report case series of 15 patients (most of them being elderly subjects) with iatrogenic symptomatic hypercalcemia in whom toxicity occurred due to empirical excessive administration of vitamin D by oral and parenteral route.

15.
J Biol Chem ; 287(16): 13182-93, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22367208

ABSTRACT

Calpains are calcium-regulated cysteine proteases that have been implicated in the regulation of cell death pathways. Here, we used our calpain-1 null mouse model to evaluate the function of calpain-1 in neural degeneration following a rodent model of traumatic brain injury. In vivo, calpain-1 null mice show significantly less neural degeneration and apoptosis and a smaller contusion 3 days post-injury than wild type littermates. Protection from traumatic brain injury corroborated with the resistance of calpain-1 neurons to apoptosis induced by oxidative stress. Biochemical analysis revealed that caspase-3 activation, extracellular calcium entry, mitochondrial membrane permeability, and release of apoptosis-inducing factor from mitochondria are partially blocked in the calpain-1 null neurons. These findings suggest that the calpain-1 knock-out mice may serve as a useful model system for neuronal protection and apoptosis in traumatic brain injury and other neurodegenerative disorders in which oxidative stress plays a role.


Subject(s)
Apoptosis/physiology , Brain Injuries/metabolism , Brain Injuries/pathology , Calpain/genetics , Calpain/metabolism , Oxidative Stress/physiology , Animals , Apoptosis Inducing Factor/metabolism , Calcium/metabolism , Caspase 3/metabolism , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Membranes/metabolism , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/pathology , RNA, Small Interfering/genetics
16.
Mol Cell ; 44(2): 317-24, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-22017877

ABSTRACT

DEPTOR is a recently identified inhibitor of the mTOR kinase that is highly regulated at the posttranslational level. In response to mitogens, we found that DEPTOR was rapidly phosphorylated on three serines in a conserved degron, facilitating binding and ubiquitylation by the F box protein ßTrCP, with consequent proteasomal degradation of DEPTOR. Phosphorylation of the ßTrCP degron in DEPTOR is executed by CK1α after a priming phosphorylation event mediated by either the mTORC1 or mTORC2 complexes. Blocking the ßTrCP-dependent degradation of DEPTOR via ßTrCP knockdown or expression of a stable DEPTOR mutant that is unable to bind ßTrCP results in mTOR inhibition. Our findings reveal that mTOR cooperates with CK1α and ßTrCP to generate an auto-amplification loop to promote its own full activation. Moreover, our results suggest that pharmacologic inhibition of CK1 may be a viable therapeutic option for the treatment of cancers characterized by activation of mTOR-signaling pathways.


Subject(s)
Casein Kinase Ialpha/metabolism , TOR Serine-Threonine Kinases/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , Cell Line , Humans , Intracellular Signaling Peptides and Proteins , Models, Biological , Phosphorylation , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Transfection , beta-Transducin Repeat-Containing Proteins/genetics
17.
Proc Natl Acad Sci U S A ; 106(47): 19842-7, 2009 Nov 24.
Article in English | MEDLINE | ID: mdl-19897731

ABSTRACT

As mediators of innate immunity, neutrophils respond to chemoattractants by adopting a highly polarized morphology. Efficient chemotaxis requires the formation of one prominent pseudopod at the cell front characterized by actin polymerization, while local inhibition suppresses the formation of rear and lateral protrusions. This asymmetric control of signaling pathways is required for directional migration along a chemotactic gradient. Here, we identify the MAGUK protein p55/MPP1 as a mediator of the frontness signal required for neutrophil polarization. We developed a p55 knockout (p55(-/-)) mouse model, and demonstrate that p55(-/-) neutrophils form multiple transient pseudopods upon chemotactic stimulation, and do not migrate efficiently in vitro. Upon agonist stimulation, p55 is rapidly recruited to the leading edge of neutrophils in mice and humans. Total F-actin polymerization, along with Rac1 and RhoA activation, appear to be normal in p55(-/-) neutrophils. Importantly, phosphorylation of Akt is significantly decreased in p55(-/-) neutrophils upon chemotactic stimulation. The activity of immunoprecipitated phosphatidylinositol 3-kinase gamma (PI3Kgamma), responsible for chemoattractant-induced synthesis of PIP(3) and Akt phosphorylation, is unperturbed in p55(-/-) neutrophils. Although the total amount of PIP(3) is normal in p55(-/-) neutrophils, PIP(3) is diffusely localized and forms punctate aggregates in activated p55(-/-) neutrophils, as compared to its accumulation at the leading edge membrane in the wild type neutrophils. Together, these results show that p55 is required for neutrophil polarization by regulating Akt phosphorylation through a mechanism that is independent of PI3Kgamma activity.


Subject(s)
Cell Polarity , Guanylate Kinases/metabolism , Neutrophils , Actins/metabolism , Animals , Chemotaxis, Leukocyte , Class Ib Phosphatidylinositol 3-Kinase , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Enzyme Activation , Female , GTP Phosphohydrolases/metabolism , Guanylate Kinases/genetics , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Isoenzymes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Stem Cell Transplantation , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein , rhoA GTP-Binding Protein/metabolism
18.
Cell Cycle ; 7(22): 3539-47, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-19001877

ABSTRACT

Heterochromatin plays an essential role in the preservation of epigenetic information, the transcriptional repression of repetitive DNA elements and inactive genes, and the proper segregation of chromosomes during mitosis. Here we identify KDM2A, a JmjC-domain containing histone demethylase, as a heterochromatin-associated and HP1-interacting protein that promotes HP1 localization to chromatin. We show that KDM2A is required to maintain the heterochromatic state, as determined using a candidate-based approach coupled to an in vivo epigenetic reporter system. Remarkably, a parallel and independent siRNA screen also detected a role for KDM2A in epigenetic silencing. Moreover, we demonstrate that KDM2A associates with centromeres and represses transcription of small non-coding RNAs that are encoded by the clusters of satellite repeats at the centromere. Dissecting the relationship between heterochromatin and centromeric RNA transcription is the basis of ongoing studies. We demonstrate that forced expression of these satellite RNA transcripts compromise the heterochromatic state and HP1 localization to chromatin. Finally, we show that KDM2A is required to sustain centromeric integrity and genomic stability, particularly during mitosis. Since the disruption of epigenetic control mechanisms contributes to cellular transformation, these results, together with the low levels of KDM2A found in prostate carcinomas, suggest a role for KDM2A in cancer development.


Subject(s)
DNA, Satellite/genetics , Heterochromatin/genetics , Oxidoreductases, N-Demethylating/physiology , Transcription, Genetic , 3T3 Cells , Animals , Cell Transformation, Neoplastic , Centromere/genetics , Chromatin Assembly and Disassembly/genetics , F-Box Proteins , Genomic Instability , HeLa Cells , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases , Mice , Transfection
19.
Brain Res ; 1207: 9-18, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18377878

ABSTRACT

Glutamic acid decarboxylase (GAD) is the rate-limiting enzyme for gamma-aminobutyric acid (GABA) biosynthesis. Previously, we reported the presence of truncated forms of GAD in vivo and in vitro. In addition, an unidentified endogenous protease responsible for proteolytic cleavage of full-length GAD (fGAD) to its truncated form (tGAD) was also observed. In this communication, we report that mu-calpain is a good candidate for conversion of fGAD(67) to tGAD(67). This conclusion is based on the following observations: 1. purified recombinant GAD(67) is cleaved by mu-calpain at specific sites; 2. in brain synaptosomal preparation, GAD(67) is cleaved to its truncated form by an endogenous protease which is inhibited by specific calpain inhibitors; 3. in mu-calpain knockout mice, the level of tGAD in the brain is greatly reduced compared with the wild type; 4. when mu-calpain gene is silenced by siRNA, the level of tGAD is also markedly reduced compared to the control group; and 5. mu-calpain is activated by neuronal stimulation and Ca(2+)-influx. The physiological significance of calpain in regulation of GABA synthesis and GABAergic neurotransmission is also discussed.


Subject(s)
Brain/metabolism , Calpain/physiology , Glutamate Decarboxylase/metabolism , Animals , Brain/drug effects , Brain/ultrastructure , Calcium/metabolism , Calpain/pharmacology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Gene Deletion , Glutamate Decarboxylase/drug effects , Mice , Mice, Knockout/genetics , Neurons/drug effects , Neurons/physiology , Protein Processing, Post-Translational , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Subcellular Fractions , Synaptosomes/drug effects , Synaptosomes/metabolism , Transfection/methods
20.
Mol Cell Biol ; 27(17): 6038-52, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17576811

ABSTRACT

Calpains are ubiquitous calcium-regulated cysteine proteases that have been implicated in cytoskeletal organization, cell proliferation, apoptosis, cell motility, and hemostasis. Gene targeting was used to evaluate the physiological function of mouse calpain-1 and establish that its inactivation results in reduced platelet aggregation and clot retraction potentially by causing dephosphorylation of platelet proteins. Here, we report that calpain-1 null (Capn1-/-) platelets accumulate protein tyrosine phosphatase 1B (PTP1B), which correlates with enhanced tyrosine phosphatase activity and dephosphorylation of multiple substrates. Treatment of Capn1-/- platelets with bis(N,N-dimethylhydroxamido)hydroxooxovanadate, an inhibitor of tyrosine phosphatases, corrected the aggregation defect and recovered impaired clot retraction. More importantly, platelet aggregation, clot retraction, and tyrosine dephosphorylation defects were rescued in the double knockout mice lacking both calpain-1 and PTP1B. Further evaluation of mutant mice by the ferric chloride-induced arterial injury model suggests that the Capn1-/- mice are relatively resistant to thrombosis in vivo. Together, our results demonstrate that PTP1B is a physiological target of calpain-1 and suggest that a similar mechanism may regulate calpain-1-mediated tyrosine dephosphorylation in other cells.


Subject(s)
Blood Platelets/metabolism , Calpain/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Animals , Blood Coagulation , Calpain/genetics , Mice , Mice, Knockout , Platelet Aggregation/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Signal Transduction/physiology , Thrombosis/metabolism , Vanadates/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...