Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Biomolecules ; 10(10)2020 10 14.
Article in English | MEDLINE | ID: mdl-33066455

ABSTRACT

Besides ubiquitin (Ub), humans have a set of ubiquitin-like proteins (UBLs) that can also covalently modify target proteins. To date, less is known about UBLs than Ub and even less is known about the UBL called ubiquitin-fold modifier 1 (UFM1). Currently, our understanding of protein modification by UFM1 (UFMylation) is like a jigsaw puzzle with many missing pieces, and in some cases it is not even clear whether these pieces of data are in the right place. Here we review the current data on UFM1 from structural biology to biochemistry and cell biology. We believe that the physiological significance of protein modification by UFM1 is currently underestimated and there is more to it than meets the eye.


Subject(s)
Protein Processing, Post-Translational/physiology , Proteins/metabolism , Animals , Disease Progression , Humans , Models, Molecular , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Proteins/chemistry , Ubiquitin/chemistry , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes/physiology , Ubiquitin-Conjugating Enzymes/physiology , Ubiquitin-Protein Ligases/physiology , Ubiquitination/physiology
2.
Int J Biochem Cell Biol ; 123: 105746, 2020 06.
Article in English | MEDLINE | ID: mdl-32315770

ABSTRACT

Ubiquitin-activating enzyme E1, UBA1, functions at the apex of the enzymatic ubiquitylation cascade, catalysing ubiquitin activation. UBA1 is thus of fundamental importance to the modulation of ubiquitin homeostasis and to all downstream ubiquitylation-dependent cellular processes, including proteolysis through the ubiquitin-proteasome system and selective autophagy. The proteasome-dependent and -independent functions of UBA1 contribute significantly to a range of processes crucial to neuronal health. The significance of UBA1 activity to neuronal health is clear in light of accumulating evidence implicating impaired UBA1 activity in a range of neurodegenerative conditions, including Parkinson's disease, Alzheimer's disease, Huntington's disease and spinal muscular atrophy. Moreover, ubiquitylation-independent functions of UBA1 of importance to neuronal functioning have been proposed. Here, we summarise findings supporting the significant role of UBA1 in regulating neuronal functioning, and discuss the detrimental consequences of UBA1 impairment that contribute to neuronal dysfunction and degeneration.


Subject(s)
Neurodegenerative Diseases/enzymology , Neurons/enzymology , Ubiquitin-Activating Enzymes/metabolism , Ubiquitination , Animals , Autophagy/genetics , Humans , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/physiopathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/physiopathology , Neurodegenerative Diseases/therapy , Neurons/metabolism , Signal Transduction/genetics , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/physiology , Ubiquitination/genetics
3.
Mol Biol Cell ; 30(20): 2558-2570, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31411939

ABSTRACT

Protein quality mechanisms are fundamental for proteostasis of eukaryotic cells. Endoplasmic reticulum-associated degradation (ERAD) is a well-studied pathway that ensures quality control of secretory and endoplasmic reticulum (ER)-resident proteins. Different branches of ERAD are involved in degradation of malfolded secretory proteins, depending on the localization of the misfolded part, the ER lumen (ERAD-L), the ER membrane (ERAD-M), and the cytosol (ERAD-C). Here we report that modification of several ER transmembrane proteins with the photosensitive degron (psd) module resulted in light-dependent degradation of the membrane proteins via the ERAD-C pathway. We found dependency on the ubiquitylation machinery including the ubiquitin-activating enzyme Uba1, the ubiquitin--conjugating enzymes Ubc6 and Ubc7, and the ubiquitin-protein ligase Doa10. Moreover, we found involvement of the Cdc48 AAA-ATPase complex members Ufd1 and Npl4, as well as the proteasome, in degradation of Sec62-myc-psd. Thus, our work shows that ERAD-C substrates can be systematically generated via synthetic degron constructs, which facilitates future investigations of the ERAD-C pathway.


Subject(s)
Endoplasmic Reticulum-Associated Degradation/physiology , Membrane Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Membrane/metabolism , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/physiology , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Activating Enzymes/physiology , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
4.
J Cell Biochem ; 119(9): 7143-7150, 2018 09.
Article in English | MEDLINE | ID: mdl-29943840

ABSTRACT

This review evaluates the use of temperature-sensitive (ts) mutants to investigate functional molecules in mammalian cells. A series of studies were performed in which mammalian cells expressing functional molecules were isolated from ts mutants using complementation by the introduction and expression of the responsible protein tagged with the green fluorescent protein. The results showed that chromosome instability and cell-cycle arrest were caused by ts defects in the following three molecules: the largest subunit of RNA polymerase II, a protein involved in splicing, and ubiquitin-activating enzyme. The cells expressing functional protein were then isolated by introducing the responsible gene tagged with the green fluorescent protein to complement the ts phenotype. These cells proved to be useful in analyzing the dynamics of RNA polymerase II in living cells. Analyses of the functional interaction between proteins involved in splicing were also useful in the investigation of ts mutants and their derivatives. In addition, these cells demonstrated the functional localization of ubiquitin-activating enzyme in the nucleus. Mammalian ts mutants continue to show great potential to aid in understanding the functions of the essential molecules in cells. Therefore, it is highly important that studies on the identification and characterization of the genes responsible for the phenotype of a mutant are carried out.


Subject(s)
Chromosomal Instability , Chromosomal Proteins, Non-Histone/physiology , RNA Polymerase II/physiology , Reverse Genetics/methods , Ubiquitin-Activating Enzymes/physiology , Animals , CHO Cells , Cell Cycle Checkpoints , Cricetulus , Gene Editing/methods , Green Fluorescent Proteins , Mammals , Mutation , Temperature
5.
Asian Pac J Allergy Immunol ; 34(1): 77-85, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26994630

ABSTRACT

BACKGROUND: Autophagy-related genes ATG4B, ATG7, and ATG12 have been identified to play a critical role in viral replication. However, these genes have yet to be identified in hepatitis B virus (HBV). OBJECTIVE: To characterise the role of ATG4B, ATG7, and ATG12 genes in HBV infection. METHODS: The mRNA expression was examined by quantitative real-time RT-PCR and Western blotting. Short hairpin RNA (shRNA) of the target gene was used to examine the function of the gene in HBV replication. Evaluation of HBV DNA level was performed by real-time PCR. RESULTS: Our findings revealed that ATG12 gene expression was significantly up-regulated (p < 0.005), whereas ATG7 gene expression was down-regulated (p < 0.0001) in HepG2.2.15 cells when compared to HepG2 cells. However, no significant difference in mRNA level of ATG4B was observed. These results were consistent with protein level findings. Moreover, we analysed the function of ATG12 in HBV replication by using ATG12 shRNA and evaluated HBV DNA level. We found that the amount of HBV was decreased in ATG12-knockdown HepG2.2.15 cells when compared to control HepG2.2.15 cells (P < 0.05). The mRNA expression of interferon-alpha (IFN-α), interferon-beta (IFN-ß), and interferon-inducible genes (IFI) was also investigated. Our results showed that the expression of IFN-α, IFN-ß, and IFI27 genes were increased in ATG12-knockdown cells but not in Mx1 gene when compared to control cells (p < 0.005, p < 0.0001 and p < 0.005, respectively). CONCLUSION: These autophagy-related genes, ATG12 may play a role in HBV replication via impairing IFN pathway. However, the biological significance of other autophagic genes such as ATG7 warrants further study.


Subject(s)
Autophagy , Hepatitis B virus/physiology , Interferons/physiology , Signal Transduction/physiology , Virus Replication , Autophagy-Related Protein 12 , Autophagy-Related Protein 7 , DNA, Viral/analysis , Hep G2 Cells , Humans , Small Ubiquitin-Related Modifier Proteins/physiology , Ubiquitin-Activating Enzymes/physiology
6.
Trends Mol Med ; 21(10): 622-632, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26432019

ABSTRACT

Neurodegenerative diseases are a leading cause of disability and early death. A common feature of these conditions is disruption of protein homeostasis. Ubiquitin-like modifier activating enzyme 1 (UBA1), the E1 ubiquitin-activating enzyme, sits at the apex of the ubiquitin cascade and represents an important regulator of cellular protein homeostasis. Critical contributions of UBA1-dependent pathways to the regulation of homeostasis and degeneration in the nervous system are emerging, including specific disruption of UBA1 in spinal muscular atrophy (SMA) and Huntington's disease (HD). In this review we discuss recent findings that put UBA1 at the centre of cellular homeostasis and neurodegeneration, highlighting the potential for UBA1 to act as a promising therapeutic target for a range of neurodegenerative diseases.


Subject(s)
Homeostasis , Neurodegenerative Diseases , Ubiquitin-Activating Enzymes , Animals , Humans , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/therapy , Ubiquitin-Activating Enzymes/physiology
7.
Toxicol Lett ; 233(3): 239-45, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25639566

ABSTRACT

In the present study, the role of autophagy in sodium arsenite (arsenite)-induced neurotoxicity was investigated in rat primary cultured cortical neurons. Incubation with arsenite concentration-dependently increased LC3-II levels (a biomarker of autophagy), indicating that arsenite is capable of inducing autophagy. Co-localization of fluorescent puncta of monodansylcadaverine (a fluorescent dye of autophagic vacuoles) and LysoTracker Red (a fluorescent dye of lysosomes) as well as chloroquine-induced enhancement of arsenite-elevated LC3-II levels suggest that arsenite induced autolysosome formation in primary cultured cortical neurons. Incubation of 3-methyladenine (an autophagy inhibitor) prevented arsenite-induced LC3-II elevation, autolysosome formation, reduction in GAP 43 (a biomarker of neurite outgrowth), caspase 3 activation and neuronal cell loss. Furthermore, Atg7 siRNA transfection attenuated arsenite-induced autophagy and neurotoxicity. At the same time, Atg7siRNA transfection ameliorated arsenite-induced reduction in α-synuclein levels (a synaptic protein essential for neuroplasticity), suggesting that arsenite via autophagy may engulf α-synuclein. Cytotoxic activities as well as potencies in elevating LC3-II and reducing α-synuclein levels by arsenite, arsenate, monomethyl arsenite (MMA(III)), and dimethyl arsenate (DMA(V)) were compared as follows: MMA(III)>arsenite¼arsenate and DMA(V). Taken together, autophagy appears to play a pro-death role in arsenics-induced neurotoxicity. Moreover, autophagy and subsequent reduction in α-synuclein levels may be a vicious cycle in arsenics-induced neurotoxicity.


Subject(s)
Arsenites/toxicity , Autophagy/drug effects , Neurotoxicity Syndromes/etiology , alpha-Synuclein/physiology , Animals , Autophagy/physiology , Autophagy-Related Protein 7 , Female , Microtubule-Associated Proteins/analysis , Rats , Rats, Sprague-Dawley , Ubiquitin-Activating Enzymes/physiology , alpha-Synuclein/analysis
8.
Nat Neurosci ; 18(2): 239-51, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25581363

ABSTRACT

Neddylation is a ubiquitylation-like pathway that controls cell cycle and proliferation by covalently conjugating Nedd8 to specific targets. However, its role in neurons, nonreplicating postmitotic cells, remains unexplored. Here we report that Nedd8 conjugation increased during postnatal brain development and is active in mature synapses, where many proteins are neddylated. We show that neddylation controls spine development during neuronal maturation and spine stability in mature neurons. We found that neddylated PSD-95 was present in spines and that neddylation on Lys202 of PSD-95 is required for the proactive role of the scaffolding protein in spine maturation and synaptic transmission. Finally, we developed Nae1(CamKIIα-CreERT2) mice, in which neddylation is conditionally ablated in adult excitatory forebrain neurons. These mice showed synaptic loss, impaired neurotransmission and severe cognitive deficits. In summary, our results establish neddylation as an active post-translational modification in the synapse regulating the maturation, stability and function of dendritic spines.


Subject(s)
Brain/growth & development , Cognition Disorders/metabolism , Dendritic Spines/physiology , Guanylate Kinases/physiology , Membrane Proteins/physiology , Synapses/physiology , Synaptic Transmission/physiology , Ubiquitins/metabolism , Animals , Behavior, Animal/physiology , Brain/metabolism , Disks Large Homolog 4 Protein , Mice , Mice, Inbred C57BL , Mice, Knockout , NEDD8 Protein , Rats , Rats, Sprague-Dawley , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/physiology , Ubiquitins/antagonists & inhibitors
9.
Cancer Res ; 75(6): 1046-55, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25576084

ABSTRACT

Interferon regulatory factor-1 (IRF1) is a tumor suppressor that regulates cell fate in several cell types. Here, we report an inverse correlation in expression of nuclear IRF1 and the autophagy regulator ATG7 in human breast cancer cells that directly affects their cell fate. In mice harboring mutant Atg7, nuclear IRF1 was increased in mammary tumors, spleen, and kidney. Mechanistic investigations identified ATG7 and the cell death modulator beclin-1 (BECN1) as negative regulators of IRF1. Silencing ATG7 or BECN1 caused estrogen receptor-α to exit the nucleus at the time when IRF1 nuclear localization occurred. Conversely, silencing IRF1 promoted autophagy by increasing BECN1 and blunting IGF1 receptor and mTOR survival signaling. Loss of IRF1 promoted resistance to antiestrogens, whereas combined silencing of ATG7 and IRF1 restored sensitivity to these agents. Using a mathematical model to prompt signaling hypotheses, we developed evidence that ATG7 silencing could resensitize IRF1-attenuated cells to apoptosis through mechanisms that involve other estrogen-regulated genes. Overall, our work shows how inhibiting the autophagy proteins ATG7 and BECN1 can regulate IRF1-dependent and -independent signaling pathways in ways that engender a new therapeutic strategy to attack breast cancer.


Subject(s)
Apoptosis , Autophagy , Breast Neoplasms/pathology , Interferon Regulatory Factor-1/physiology , Signal Transduction/physiology , Animals , Apoptosis Regulatory Proteins/physiology , Autophagy-Related Protein 7 , Beclin-1 , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Lineage , Female , Humans , Membrane Proteins/physiology , Mice , Models, Theoretical , Ubiquitin-Activating Enzymes/physiology
10.
Biochim Biophys Acta ; 1855(1): 50-60, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25481052

ABSTRACT

Proteasome-mediated degradation is a common mechanism by which cells renew their intracellular proteins and maintain protein homeostasis. In this process, the E3 ubiquitin ligases are responsible for targeting specific substrates (proteins) for ubiquitin-mediated degradation. However, in cancer cells, the stability and the balance between oncoproteins and tumor suppressor proteins are disturbed in part due to deregulated proteasome-mediated degradation. This ultimately leads to either stabilization of oncoprotein(s) or increased degradation of tumor suppressor(s), contributing to tumorigenesis and cancer progression. Therefore, E3 ubiquitin ligases including the SCF types of ubiquitin ligases have recently evolved as promising therapeutic targets for the development of novel anti-cancer drugs. In this review, we highlighted the critical components along the ubiquitin pathway including E1, E2, various E3 enzymes and DUBs that could serve as potential drug targets and also described the available bioactive compounds that target the ubiquitin pathway to control various cancers.


Subject(s)
Molecular Targeted Therapy/methods , Neoplasms/therapy , Ubiquitin/metabolism , Animals , Humans , Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/therapeutic use , Signal Transduction , Ubiquitin/antagonists & inhibitors , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/physiology , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Ubiquitin-Conjugating Enzymes/physiology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/physiology , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/physiology , Ubiquitination/physiology
11.
Nat Commun ; 5: 4632, 2014 Aug 18.
Article in English | MEDLINE | ID: mdl-25130259

ABSTRACT

Gliomas are the most common primary tumours affecting the adult central nervous system and respond poorly to standard therapy. Myc is causally implicated in most human tumours and the majority of glioblastomas have elevated Myc levels. Using the Myc dominant negative Omomyc, we previously showed that Myc inhibition is a promising strategy for cancer therapy. Here, we preclinically validate Myc inhibition as a therapeutic strategy in mouse and human glioma, using a mouse model of spontaneous multifocal invasive astrocytoma and its derived neuroprogenitors, human glioblastoma cell lines, and patient-derived tumours both in vitro and in orthotopic xenografts. Across all these experimental models we find that Myc inhibition reduces proliferation, increases apoptosis and remarkably, elicits the formation of multinucleated cells that then arrest or die by mitotic catastrophe, revealing a new role for Myc in the proficient division of glioma cells.


Subject(s)
Astrocytoma/pathology , Brain Neoplasms/pathology , Glioblastoma/pathology , Glioma/pathology , Mitosis/physiology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Animals , Apoptosis/physiology , Astrocytoma/physiopathology , Astrocytoma/therapy , Brain Neoplasms/physiopathology , Brain Neoplasms/therapy , Cell Line, Tumor , Cell Proliferation/physiology , Disease Models, Animal , Glioblastoma/physiopathology , Glioblastoma/therapy , Glioma/physiopathology , Glioma/therapy , Heterografts , Humans , Mice , Mice, Transgenic , Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology , Proto-Oncogene Proteins c-myc/physiology , Ubiquitin-Activating Enzymes/physiology
12.
J Neurosci ; 34(25): 8411-22, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24948797

ABSTRACT

Huntington's disease (HD) belongs to a family of neurodegenerative diseases caused by misfolded proteins and shares the pathological hallmark of selective accumulation of misfolded proteins in neuronal cells. Polyglutamine expansion in the HD protein, huntingtin (Htt), causes selective neurodegeneration that is more severe in the striatum and cortex than in other brain regions, but the mechanism behind this selectivity is unknown. Here we report that in HD knock-in mice, the expression levels of mutant Htt (mHtt) are higher in brain tissues than in peripheral tissues. However, the expression of N-terminal mHtt via stereotaxic injection of viral vectors in mice also results in greater accumulation of mHtt in the striatum than in muscle. We developed an in vitro assay that revealed that extracts from the striatum and cortex promote the formation of high-molecular weight (HMW) mHtt compared with the relatively unaffected cerebellar and peripheral tissue extracts. Inhibition of ubiquitin-activating enzyme E1 (Ube1) increased the levels of HMW mHtt in the relatively unaffected tissues. Importantly, the expression levels of Ube1 are lower in brain tissues than peripheral tissues and decline in the nuclear fraction with age, which is correlated with the increased accumulation of mHtt in the brain and neuronal nuclei during aging. Our findings suggest that decreased targeting of misfolded Htt to the proteasome for degradation via Ube1 may underlie the preferential accumulation of toxic forms of mHtt in the brain and its selective neurodegeneration.


Subject(s)
Brain Chemistry/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ubiquitin-Activating Enzymes/physiology , Animals , Enzyme Activation/genetics , Female , Gene Knock-In Techniques , HEK293 Cells , Humans , Huntingtin Protein , Male , Mice , Mutation , Nerve Tissue Proteins/chemistry , Nuclear Proteins/chemistry , Tissue Distribution/genetics , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/genetics
13.
Autophagy ; 10(5): 878-88, 2014 May.
Article in English | MEDLINE | ID: mdl-24674921

ABSTRACT

In flowering plants, the tapetum, the innermost layer of the anther, provides both nutrient and lipid components to developing microspores, pollen grains, and the pollen coat. Though the programmed cell death of the tapetum is one of the most critical and sensitive steps for fertility and is affected by various environmental stresses, its regulatory mechanisms remain mostly unknown. Here we show that autophagy is required for the metabolic regulation and nutrient supply in anthers and that autophagic degradation within tapetum cells is essential for postmeiotic anther development in rice. Autophagosome-like structures and several vacuole-enclosed lipid bodies were observed in postmeiotic tapetum cells specifically at the uninucleate stage during pollen development, which were completely abolished in a retrotransposon-insertional OsATG7 (autophagy-related 7)-knockout mutant defective in autophagy, suggesting that autophagy is induced in tapetum cells. Surprisingly, the mutant showed complete sporophytic male sterility, failed to accumulate lipidic and starch components in pollen grains at the flowering stage, showed reduced pollen germination activity, and had limited anther dehiscence. Lipidomic analyses suggested impairment of editing of phosphatidylcholines and lipid desaturation in the mutant during pollen maturation. These results indicate a critical involvement of autophagy in a reproductive developmental process of rice, and shed light on the novel autophagy-mediated regulation of lipid metabolism in eukaryotic cells.


Subject(s)
Autophagy/genetics , Flowers/growth & development , Lipid Metabolism/genetics , Oryza , Plant Proteins/physiology , Ubiquitin-Activating Enzymes/physiology , Flowers/genetics , Flowers/metabolism , Meiosis/genetics , Oryza/genetics , Oryza/growth & development , Oryza/metabolism , Plants, Genetically Modified , Pollen/genetics , Pollen/metabolism
14.
PLoS Pathog ; 9(12): e1003809, 2013.
Article in English | MEDLINE | ID: mdl-24367261

ABSTRACT

Toxoplasma gondii resides in an intracellular compartment (parasitophorous vacuole) that excludes transmembrane molecules required for endosome-lysosome recruitment. Thus, the parasite survives by avoiding lysosomal degradation. However, autophagy can re-route the parasitophorous vacuole to the lysosomes and cause parasite killing. This raises the possibility that T. gondii may deploy a strategy to prevent autophagic targeting to maintain the non-fusogenic nature of the vacuole. We report that T. gondii activated EGFR in endothelial cells, retinal pigment epithelial cells and microglia. Blockade of EGFR or its downstream molecule, Akt, caused targeting of the parasite by LC3(+) structures, vacuole-lysosomal fusion, lysosomal degradation and killing of the parasite that were dependent on the autophagy proteins Atg7 and Beclin 1. Disassembly of GPCR or inhibition of metalloproteinases did not prevent EGFR-Akt activation. T. gondii micronemal proteins (MICs) containing EGF domains (EGF-MICs; MIC3 and MIC6) appeared to promote EGFR activation. Parasites defective in EGF-MICs (MIC1 ko, deficient in MIC1 and secretion of MIC6; MIC3 ko, deficient in MIC3; and MIC1-3 ko, deficient in MIC1, MIC3 and secretion of MIC6) caused impaired EGFR-Akt activation and recombinant EGF-MICs (MIC3 and MIC6) caused EGFR-Akt activation. In cells treated with autophagy stimulators (CD154, rapamycin) EGFR signaling inhibited LC3 accumulation around the parasite. Moreover, increased LC3 accumulation and parasite killing were noted in CD154-activated cells infected with MIC1-3 ko parasites. Finally, recombinant MIC3 and MIC6 inhibited parasite killing triggered by CD154 particularly against MIC1-3 ko parasites. Thus, our findings identified EGFR activation as a strategy used by T. gondii to maintain the non-fusogenic nature of the parasitophorous vacuole and suggest that EGF-MICs have a novel role in affecting signaling in host cells to promote parasite survival.


Subject(s)
Autophagy/physiology , ErbB Receptors/metabolism , Toxoplasma/physiology , Toxoplasmosis/enzymology , Animals , Apoptosis Regulatory Proteins/physiology , Autophagy-Related Protein 7 , Beclin-1 , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Enzyme Activation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Humans , Membrane Proteins/physiology , Mice , Oncogene Protein v-akt/metabolism , Toxoplasma/immunology , Toxoplasmosis/genetics , Toxoplasmosis/immunology , Ubiquitin-Activating Enzymes/physiology
15.
Mol Cell ; 50(2): 172-84, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23499007

ABSTRACT

The Uba6 (E1)-Use1 (E2) ubiquitin transfer cascade is a poorly understood alternative arm of the ubiquitin proteasome system (UPS) and is required for mouse embryonic development, independent of the canonical Uba1-E2-E3 pathway. Loss of neuronal Uba6 during embryonic development results in altered patterning of neurons in the hippocampus and the amygdala, decreased dendritic spine density, and numerous behavioral disorders. The levels of the E3 ubiquitin ligase Ube3a (E6-AP) and Shank3, both linked with dendritic spine function, are elevated in the amygdala of Uba6-deficient mice, while levels of the Ube3a substrate Arc are reduced. Uba6 and Use1 promote proteasomal turnover of Ube3a in mouse embryo fibroblasts (MEFs) and catalyze Ube3a ubiquitylation in vitro. These activities occur in parallel with an independent pathway involving Uba1-UbcH7, but in a spatially distinct manner in MEFs. These data reveal an unanticipated role for Uba6 in neuronal development, spine architecture, mouse behavior, and turnover of Ube3a.


Subject(s)
Amygdala/abnormalities , CA3 Region, Hippocampal/abnormalities , Qc-SNARE Proteins/deficiency , Ubiquitin-Activating Enzymes/deficiency , Ubiquitination , Amygdala/enzymology , Amygdala/pathology , Animals , Body Weight , CA3 Region, Hippocampal/enzymology , CA3 Region, Hippocampal/pathology , Cells, Cultured , Dendritic Spines/pathology , Embryonic Development , Energy Metabolism , Female , Genes, Lethal , Learning Disabilities/metabolism , Locomotion , Male , Memory Disorders/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins , Nerve Tissue Proteins/metabolism , Oxygen Consumption , Protein Stability , Protein Structure, Tertiary , Qc-SNARE Proteins/genetics , Qc-SNARE Proteins/physiology , SNARE Proteins , Social Behavior , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/physiology , Ubiquitin-Protein Ligases/metabolism , Vesicular Transport Proteins
16.
PLoS One ; 8(1): e32835, 2013.
Article in English | MEDLINE | ID: mdl-23382794

ABSTRACT

Neurodegenerative diseases cause tremendous suffering for those afflicted and their families. Many of these diseases involve accumulation of mis-folded or aggregated proteins thought to play a causal role in disease pathology. Ubiquitinated proteins are often found in these protein aggregates, and the aggregates themselves have been shown to inhibit the activity of the proteasome. These and other alterations in the Ubiquitin Pathway observed in neurodegenerative diseases have led to the question of whether impairment of the Ubiquitin Pathway on its own can increase mortality or if ongoing neurodegeneration alters Ubiquitin Pathway function as a side-effect. To address the role of the Ubiquitin Pathway in vivo, we studied loss-of-function mutations in the Drosophila Ubiquitin Activating Enzyme, Uba1 or E1, the most upstream enzyme in the Ubiquitin Pathway. Loss of only one functional copy of E1 caused a significant reduction in adult lifespan. Rare homozygous hypomorphic E1 mutants reached adulthood. These mutants exhibited further reduced lifespan and showed inappropriate Ras activation in the brain. Removing just one functional copy of Ras restored the lifespan of heterozygous E1 mutants to that of wild-type flies and increased the survival of homozygous E1 mutants. E1 homozygous mutants also showed severe motor impairment. Our findings suggest that processes that impair the Ubiquitin Pathway are sufficient to cause early mortality. Reduced lifespan and motor impairment are seen in the human disease X-linked Infantile Spinal Muscular Atrophy, which is associated with mutation in human E1 warranting further analysis of these mutants as a potential animal model for study of this disease.


Subject(s)
Drosophila Proteins , Drosophila melanogaster/enzymology , Longevity , Motor Activity , Spinal Muscular Atrophies of Childhood/genetics , Ubiquitin-Activating Enzymes , Animals , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/physiopathology , Humans , Longevity/genetics , Longevity/physiology , Motor Activity/genetics , Motor Activity/radiation effects , Mutation , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/physiology , Protein Folding , Spinal Muscular Atrophies of Childhood/physiopathology , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/physiology
17.
J Clin Oncol ; 31(9): 1231-8, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23358974

ABSTRACT

The ubiquitin proteasome system (UPS) regulates the ubiquitination, and thus degradation and turnover, of many proteins vital to cellular regulation and function. The UPS comprises a sequential series of enzymatic processes using four key enzyme families: E1 (ubiquitin-activating enzymes), E2 (ubiquitin-carrier proteins), E3 (ubiquitin-protein ligases), and E4 (ubiquitin chain assembly factors). Because the UPS is a crucial regulator of the cell cycle, and abnormal cell-cycle control can lead to oncogenesis, aberrancies within the UPS pathway can result in a malignant cellular phenotype and thus has become an attractive target for novel anticancer agents. This article will provide an overall review of the mechanics of the UPS, describe aberrancies leading to cancer, and give an overview of current drug therapies selectively targeting the UPS.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic , Proteasome Endopeptidase Complex/physiology , Ubiquitin-Activating Enzymes/physiology , Ubiquitin-Conjugating Enzymes/physiology , Ubiquitin-Protein Ligases/physiology , Antineoplastic Agents/therapeutic use , Cyclin-Dependent Kinase Inhibitor p27/physiology , DNA-Binding Proteins/physiology , Humans , Male , Mutation , Oncogene Proteins, Viral/physiology , Prostatic Neoplasms/genetics , Proteasome Endopeptidase Complex/drug effects , Proto-Oncogene Proteins c-mdm2/physiology , Tumor Suppressor Protein p53/physiology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics
18.
EMBO Rep ; 14(2): 143-51, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23337627

ABSTRACT

Autophagy and autophagy-related processes are fundamentally important in human health and disease. These processes are viewed primarily as cellular degradative pathways that recycle macromolecules and dysfunctional or redundant organelles into amino acids, sugars and lipids, especially during starvation. However, the ubiquitin-like autophagy proteins and other components of the autophagic machinery additionally participate in cellular reprogramming. We highlight these non-autophagic roles of autophagy proteins with the aim of drawing attention to this growing, but unexplored, research topic. We focus on the non-autophagic functions of autophagy proteins in cell survival and apoptosis, modulation of cellular traffic, protein secretion, cell signalling, transcription, translation and membrane reorganization.


Subject(s)
Autophagy , Intracellular Signaling Peptides and Proteins/physiology , Signal Transduction , Adaptor Proteins, Signal Transducing/physiology , Adipogenesis , Animals , Autophagy-Related Protein 12 , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Autophagy-Related Protein 8 Family , Exocytosis , Host-Pathogen Interactions , Humans , Microfilament Proteins/physiology , Microtubule-Associated Proteins/physiology , Protein Transport , Small Ubiquitin-Related Modifier Proteins/physiology , Ubiquitin-Activating Enzymes/physiology
19.
Cell Rep ; 1(4): 309-16, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-22832224

ABSTRACT

Inhibition of NEDD8-activating enzyme (NAE) has emerged as a highly promising approach to treat cancer through the adenosine sulfamate analog MLN4924. Here, we show that selective pressure results in HCT116 colorectal carcinoma cells with decreased MLN4924 sensitivity and identify a single-nucleotide transition that changes alanine 171 to threonine (A171T) of the NAE subunit UBA3. This reduces the enzyme's affinity for MLN4924 and ATP while increasing NEDD8 activation at physiological ATP concentrations. Expression of UBA3 A171T is sufficient to decrease MLN4924 sensitivity of naive HCT116 cells, indicating that it is a dominant suppressor of MLN4924-mediated cell death. Our data suggest that the on-target potency of MLN4924 selects for a point mutation in NAE that overcomes the molecule's inhibitory effects, allowing cancer cell survival.


Subject(s)
Cyclopentanes/pharmacology , Pyrimidines/pharmacology , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Amino Acid Sequence , Amino Acid Substitution , Cell Line, Tumor , Chromatography, Liquid , Cullin Proteins/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Drug Resistance, Neoplasm/genetics , HCT116 Cells , Humans , Molecular Sequence Data , Point Mutation , Sequence Alignment , Tandem Mass Spectrometry , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/physiology
20.
Development ; 139(15): 2751-62, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22745316

ABSTRACT

SUMOylation is a highly conserved post-translational modification shown to modulate target protein activity in a wide variety of cellular processes. Although the requirement for SUMO modification of specific substrates has received significant attention in vivo and in vitro, the developmental requirements for SUMOylation at the cell and tissue level remain poorly understood. Here, we show that in Drosophila melanogaster, both heterodimeric components of the SUMO E1-activating enzyme are zygotically required for mitotic progression but are dispensable for cell viability, homeostasis and DNA synthesis in non-dividing cells. Explaining the lack of more pleiotropic effects following a global block of SUMO conjugation, we further demonstrate that low levels of global substrate SUMOylation are detected in mutants lacking either or both E1 subunits. These results not only suggest that minimal SUMOylation persists in the absence of Aos1/Uba2, but also show that the process of cell division is selectively sensitive to reductions in global SUMOylation. Supporting this view, knockdown of SUMO or its E1 and E2 enzymes robustly disrupts proliferating cells in the developing eye, without any detectable effects on the development or differentiation of neighboring post-mitotic cells.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental , Nuclear Proteins/physiology , Photoreceptor Cells, Invertebrate/physiology , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Activating Enzymes/physiology , Animals , Cell Proliferation , Cell Survival , Crosses, Genetic , Drosophila Proteins/metabolism , Genomics , Homeostasis , Imaginal Discs/embryology , Ligases/metabolism , Mitosis , Models, Genetic , Mutation , Nuclear Proteins/metabolism , RNA Interference , Salivary Glands/embryology , Ubiquitin-Activating Enzymes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...