Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Neurol Res ; 46(1): 89-97, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37703903

ABSTRACT

OBJECTIVE: One of the important causes of death in cancer patients is malignant metastasis, invasion, and metastasis of tumor cells. Metastasis is also the most basic physiological characteristics and pathogenesis of various tumors. Previously published studies have suggested that autocrine motor factor receptor (AMFR) is the key regulator of tumor cell migration and invasion. Meanwhile, AMFR is highly expressed in esophageal tumors, gastrointestinal tumors, and bladder cancer, and it is also involved in its pathogenesis. However, the role of AMFR in glioblastoma has not been reported. METHODS: In order to study the role of AMFR in the cell migration and invasion of glioblastoma, AMFR was silenced using siRNA and overexpressed using cDNA. Immunoblotting analysis and real-time quantitative polymerase chain reaction (PCR) were employed to assess the expression of AMFR. We conducted wound healing assay, cell migration assay, and tumorsphere formation assay to detect the invasion and metastatic ability of glioblastoma. RESULTS: This study found that the level of AMFR expression was significantly correlated with the malignant degree of glioma tissue in clinic samples. AMFR silencing decreased cell migration and invasion of LN229. Overexpression of AMFR significantly increased cell migration and invasion of U251. CONCLUSION: This study suggests that AMFR could be used as a therapeutic strategy for the clinical treatment of glioblastoma.


Subject(s)
Glioblastoma , Humans , Receptors, Autocrine Motility Factor/genetics , Receptors, Autocrine Motility Factor/metabolism , Glioblastoma/genetics , Glioblastoma/pathology , RNA, Small Interfering/genetics , Cell Movement , Cell Proliferation , Cell Line, Tumor , Neoplasm Invasiveness
2.
Cell Death Dis ; 14(12): 810, 2023 12 08.
Article in English | MEDLINE | ID: mdl-38065978

ABSTRACT

Ferroptosis, which is driven by iron-dependent lipid peroxidation, plays an essential role in liver ischemia-reperfusion injury (IRI) during liver transplantation (LT). Gp78, an E3 ligase, has been implicated in lipid metabolism and inflammation. However, its role in liver IRI and ferroptosis remains unknown. Here, hepatocyte-specific gp78 knockout (HKO) or overexpressed (OE) mice were generated to examine the effect of gp78 on liver IRI, and a multi-omics approach (transcriptomics, proteomics, and metabolomics) was performed to explore the potential mechanism. Gp78 expression decreased after reperfusion in LT patients and mice with IRI, and gp78 expression was positively correlated with liver damage. Gp78 absence from hepatocytes alleviated liver damage in mice with IRI, ameliorating inflammation. However, mice with hepatic gp78 overexpression showed the opposite phenotype. Mechanistically, gp78 overexpression disturbed lipid homeostasis, remodeling polyunsaturated fatty acid (PUFA) metabolism, causing oxidized lipids accumulation and ferroptosis, partly by promoting ACSL4 expression. Chemical inhibition of ferroptosis or ACSL4 abrogated the effects of gp78 on ferroptosis and liver IRI. Our findings reveal a role of gp78 in liver IRI pathogenesis and uncover a mechanism by which gp78 promotes hepatocyte ferroptosis by ACSL4, suggesting the gp78-ACSL4 axis as a feasible target for the treatment of IRI-associated liver damage.


Subject(s)
Ferroptosis , Hepatocytes , Liver Diseases , Receptors, Autocrine Motility Factor , Reperfusion Injury , Animals , Humans , Mice , Hepatocytes/enzymology , Inflammation/metabolism , Liver Diseases/metabolism , Reperfusion Injury/metabolism , Liver Transplantation , Receptors, Autocrine Motility Factor/genetics , Receptors, Autocrine Motility Factor/metabolism , Coenzyme A Ligases
3.
Acta Neuropathol ; 146(2): 353-368, 2023 08.
Article in English | MEDLINE | ID: mdl-37119330

ABSTRACT

Hereditary spastic paraplegias (HSP) are rare, inherited neurodegenerative or neurodevelopmental disorders that mainly present with lower limb spasticity and muscle weakness due to motor neuron dysfunction. Whole genome sequencing identified bi-allelic truncating variants in AMFR, encoding a RING-H2 finger E3 ubiquitin ligase anchored at the membrane of the endoplasmic reticulum (ER), in two previously genetically unexplained HSP-affected siblings. Subsequently, international collaboration recognized additional HSP-affected individuals with similar bi-allelic truncating AMFR variants, resulting in a cohort of 20 individuals from 8 unrelated, consanguineous families. Variants segregated with a phenotype of mainly pure but also complex HSP consisting of global developmental delay, mild intellectual disability, motor dysfunction, and progressive spasticity. Patient-derived fibroblasts, neural stem cells (NSCs), and in vivo zebrafish modeling were used to investigate pathomechanisms, including initial preclinical therapy assessment. The absence of AMFR disturbs lipid homeostasis, causing lipid droplet accumulation in NSCs and patient-derived fibroblasts which is rescued upon AMFR re-expression. Electron microscopy indicates ER morphology alterations in the absence of AMFR. Similar findings are seen in amfra-/- zebrafish larvae, in addition to altered touch-evoked escape response and defects in motor neuron branching, phenocopying the HSP observed in patients. Interestingly, administration of FDA-approved statins improves touch-evoked escape response and motor neuron branching defects in amfra-/- zebrafish larvae, suggesting potential therapeutic implications. Our genetic and functional studies identify bi-allelic truncating variants in AMFR as a cause of a novel autosomal recessive HSP by altering lipid metabolism, which may potentially be therapeutically modulated using precision medicine with statins.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Spastic Paraplegia, Hereditary , Animals , Humans , Spastic Paraplegia, Hereditary/drug therapy , Spastic Paraplegia, Hereditary/genetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Zebrafish , Mutation , Motor Neurons , Receptors, Autocrine Motility Factor/genetics
4.
J Biomol NMR ; 74(4-5): 223-228, 2020 May.
Article in English | MEDLINE | ID: mdl-32333192

ABSTRACT

Recent methyl adiabatic relaxation dispersion experiments provide examination of conformational dynamics across a very wide timescale (102-105 s-1) and, particularly, provide insight into the hydrophobic core of proteins and allosteric effects associated with modulators. The experiments require efficient decoupling of 1H and 13C spin interactions, and some artifacts have been discovered, which are associated with the design of the proton decoupling scheme. The experimental data suggest that the original design is valid; however, pulse sequences with either no proton decoupling or proton decoupling with imperfect pulses can potentially exhibit complications in the experiments. Here, we demonstrate that pulse imperfections in the proton decoupling scheme can be dramatically alleviated by using a single composite π pulse and provide pure single-exponential relaxation data. It allows the opportunity to access high-quality methyl adiabatic relaxation dispersion data by removing the cross-correlation between dipole-dipole interaction and chemical shift anisotropy. The resulting high-quality data is illustrated with the binding of an allosteric modulator (G2BR) to the ubiquitin conjugating enzyme Ube2g2.


Subject(s)
Artifacts , Nuclear Magnetic Resonance, Biomolecular/methods , Peptide Fragments/chemistry , Protein Conformation , Receptors, Autocrine Motility Factor/chemistry , Ubiquitin-Conjugating Enzymes/chemistry , Allosteric Regulation , Binding Sites , Models, Molecular , Peptide Fragments/genetics , Peptide Fragments/metabolism , Point Mutation , Protein Folding , Protons , Receptors, Autocrine Motility Factor/genetics , Recombinant Fusion Proteins/metabolism , Thermodynamics , Ubiquitin-Conjugating Enzymes/metabolism
5.
Nat Commun ; 11(1): 379, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31953408

ABSTRACT

Insig-2 is an ER membrane protein negatively controlling lipid biosynthesis. Here, we find that Insig-2 is increased in the tissues, including liver, but unaltered in the muscle of gp78-deficient mice. In hepatocytes and undifferentiated C2C12 myoblasts, Insig-2 is ubiquitylated on Cys215 by gp78 and degraded. However, the C215 residue is oxidized by elevated reactive oxygen species (ROS) during C2C12 myoblasts differentiating into myotubes, preventing Insig-2 from ubiquitylation and degradation. The stabilized Insig-2 downregulates lipogenesis through inhibiting the SREBP pathway, helping to channel the carbon flux to ATP generation and protecting myotubes from lipid over-accumulation. Evolutionary analysis shows that the YECK (in which C represents Cys215 in human Insig-2) tetrapeptide sequence in Insig-2 is highly conserved in amniotes but not in aquatic amphibians and fishes, suggesting it may have been shaped by differential selection. Together, this study suggests that competitive oxidation-ubiquitylation on Cys215 of Insig-2 senses ROS and prevents muscle cells from lipid accumulation.


Subject(s)
Cysteine/metabolism , Membrane Proteins/metabolism , Receptors, Autocrine Motility Factor/metabolism , Ubiquitination , Amphibians , Animals , CHO Cells , Cell Line , Cricetulus , Down-Regulation , Evolution, Molecular , Fishes , Hepatocytes/metabolism , Humans , Lipid Metabolism , Lipogenesis , Liver/metabolism , Male , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Cells/metabolism , Muscle Fibers, Skeletal/metabolism , Myoblasts/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Receptors, Autocrine Motility Factor/genetics , Sequence Analysis, Protein , Sterol Regulatory Element Binding Proteins/metabolism , Transcriptome
6.
J Biol Chem ; 294(52): 20084-20096, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31748412

ABSTRACT

The endoplasmic reticulum-associated degradation (ERAD) pathway mediates the endoplasmic reticulum-to-cytosol retrotranslocation of defective proteins through protein complexes called retrotranslocons. Defective proteins usually have complex conformations and topologies, and it is unclear how ERAD can thread these conformationally diverse protein substrates through the retrotranslocons. Here, we investigated the substrate conformation flexibility necessary for transport via retrotranslocons on the ERAD-L, ERAD-M, and HIV-encoded protein Vpu-hijacked ERAD branches. To this end, we appended various ERAD substrates with specific domains whose conformations were tunable in flexibility or tightness by binding to appropriate ligands. With this technique, we could define the capacity of specific retrotranslocons in disentangling very tight, less tight but well-folded, and unstructured conformations. The Hrd1 complex, the retrotranslocon on the ERAD-L branch, permitted the passage of substrates with a proteinase K-resistant tight conformation, whereas the E3 ligase gp78-mediated ERAD-M allowed passage only of nearly completely disordered but not well-folded substrates and thus may have the least unfoldase activity. Vpu-mediated ERAD, containing a potential retrotranslocon, could unfold well-folded substrates for successful retrotranslocation. However, substrate retrotranslocation in Vpu-mediated ERAD was blocked by enhanced conformational tightness of the substrate. On the basis of these findings, we propose a mechanism underlying polypeptide movement through the endoplasmic reticulum membrane. We anticipate that our biochemical system paves the way for identifying the factors necessary for the retrotranslocation of membrane proteins.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum-Associated Degradation/drug effects , HEK293 Cells , Human Immunodeficiency Virus Proteins/genetics , Human Immunodeficiency Virus Proteins/metabolism , Humans , Leupeptins/pharmacology , Proteasome Endopeptidase Complex/metabolism , Protein Unfolding , Receptors, Autocrine Motility Factor/genetics , Receptors, Autocrine Motility Factor/metabolism , Substrate Specificity , Trimetrexate/pharmacology , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism
7.
Mol Pharmacol ; 96(5): 641-654, 2019 11.
Article in English | MEDLINE | ID: mdl-31492698

ABSTRACT

The hepatic endoplasmic reticulum (ER)-anchored monotopic proteins, cytochromes P450 (P450s), are enzymes that metabolize endobiotics (physiologically active steroids and fatty acids), as well as xenobiotics including therapeutic/chemotherapeutic drugs, nutrients, carcinogens, and toxins. Alterations of hepatic P450 content through synthesis, inactivation, or proteolytic turnover influence their metabolic function. P450 proteolytic turnover occurs via ER-associated degradation (ERAD) involving ubiquitin (Ub)-dependent proteasomal degradation (UPD) as a major pathway. UPD critically involves P450 protein ubiquitination by E2/E3 Ub-ligase complexes. We have previously identified the ER-polytopic gp78/AMFR (autocrine motility factor receptor) as a relevant E3 in CYP3A4, CYP3A23, and CYP2E1 UPD. We now document that liver-conditional genetic ablation of gp78/AMFR in male mice disrupts P450 ERAD, resulting in statistically significant stabilization of Cyp2a5 and Cyp2c, in addition to that of Cyp3a and Cyp2e1. More importantly, we establish that such stabilization is of the functionally active P450 proteins, leading to corresponding statistically significant enhancement of their drug-metabolizing capacities. Our findings, with clinically relevant therapeutic drugs (nicotine, coumarin, chlorzoxazone, and acetaminophen) and the prodrug (tamoxifen) as P450 substrates, reveal that P450 ERAD disruption could influence therapeutic drug response and/or toxicity, warranting serious consideration as a potential source of clinically relevant drug-drug interactions (DDIs). Because gp78/AMFR is not only an E3 Ub-ligase, but also a cell-surface prometastatic oncogene that is upregulated in various malignant cancers, our finding that hepatic gp78/AMFR knockout can enhance P450-dependent bioactivation of relevant cancer chemotherapeutic prodrugs is of therapeutic relevance and noteworthy in prospective drug design and development. SIGNIFICANCE STATEMENT: The cell-surface and ER transmembrane protein gp78/AMFR, a receptor for the prometastatic autocrine motility factor (AMF), as well as an E3 ubiquitin-ligase involved in the ER-associated degradation (ERAD) of not only the tumor metastatic suppressor KAI1 but also of hepatic cytochromes P450, is upregulated in various human cancers, enhancing their invasiveness, metastatic potential, and poor prognosis. Liver-specific gp78/AMFR genetic ablation results in functional protein stabilization of several hepatic P450s and consequently enhanced drug and prodrug metabolism, a feature that could be therapeutically exploited in the bioactivation of chemotherapeutic prodrugs through design and development of novel short-term gp78/AMFR chemical inhibitors.


Subject(s)
Cytochrome P-450 Enzyme Inducers/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Gene Deletion , Hepatocytes/metabolism , Receptors, Autocrine Motility Factor/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Aspirin/pharmacology , Cells, Cultured , Cytochrome P-450 Enzyme System/genetics , Enzyme Induction/drug effects , Enzyme Induction/physiology , Hepatocytes/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Stability/drug effects , Receptors, Autocrine Motility Factor/genetics , Tamoxifen/pharmacology , Ubiquitin-Protein Ligases/genetics
8.
Nat Metab ; 1(5): 570-583, 2019 05.
Article in English | MEDLINE | ID: mdl-32694855

ABSTRACT

Metabolism in mammals is regulated by complex interplay among different organs. Fatty acid synthesis is increased in white adipose tissue (WAT) when it is inhibited in the liver. Here we identify glycoprotein non-metastatic melanoma protein B (Gpnmb) as one liver-WAT cross-talk factor involved in lipogenesis. Inhibition of the hepatic sterol regulatory element-binding protein pathway leads to increased transcription of Gpnmb and promotes processing of the membrane protein to a secreted form. Gpnmb stimulates lipogenesis in WAT and exacerbates diet-induced obesity and insulin resistance. In humans, Gpnmb is tightly associated with body mass index and is a strong risk factor for obesity. Gpnmb inhibition by a neutralizing antibody or liver-specific knockdown improves metabolic parameters, including weight gain reduction and increased insulin sensitivity, probably by promoting the beiging of WAT. These results suggest that Gpnmb is a liver-secreted factor regulating lipogenesis in WAT, and that Gpnmb inhibition may provide a therapeutic strategy in obesity and diabetes.


Subject(s)
Adipose Tissue, White/metabolism , Eye Proteins/metabolism , Insulin Resistance , Liver/metabolism , Membrane Glycoproteins/metabolism , Obesity/metabolism , Animals , Eye Proteins/genetics , Eye Proteins/physiology , Homeostasis , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipid Metabolism , Lipogenesis/genetics , Lipogenesis/physiology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Receptors, Autocrine Motility Factor/genetics , Receptors, Autocrine Motility Factor/metabolism , Up-Regulation
9.
FASEB J ; 33(2): 1927-1945, 2019 02.
Article in English | MEDLINE | ID: mdl-30230921

ABSTRACT

The mechanism by which the endoplasmic reticulum (ER) ubiquitin ligases sense stress to potentiate their activity is poorly understood. GP78, an ER E3 ligase, is best known for its role in ER-associated protein degradation, although its activity is also linked to mitophagy, ER-mitochondria junctions, and MAPK signaling, thus highlighting the importance of understanding its regulation. In healthy cells, Mahogunin really interesting new gene (RING) finger 1 (MGRN1) interacts with GP78 and proteasomally degrades it to alleviate mitophagy. Here, we identify calmodulin (CaM) as the adapter protein that senses fluctuating cytosolic Ca2+ levels and modulates the Ca2+-dependent MGRN1-GP78 interactions. When stress elevates cytosolic Ca2+ levels in cultured and primary neuronal cells, CaM binds to both E3 ligases and inhibits their interaction. Molecular docking, simulation, and biophysical studies show that CaM interacts with both proteins with different affinities and binding modes. The physiological impact of this interaction switch manifests in the regulation of ER-associated protein degradation, ER-mitochondria junctions, and relative distribution of smooth ER and rough ER.-Mukherjee, R., Bhattacharya, A., Sau, A., Basu, S., Chakrabarti, S., Chakrabarti, O. Calmodulin regulates MGRN1-GP78 interaction mediated ubiquitin proteasomal degradation system.


Subject(s)
Calmodulin/metabolism , Neurons/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Receptors, Autocrine Motility Factor/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Calcium Signaling , Calmodulin/chemistry , Calmodulin/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Mice , Molecular Docking Simulation , Neurons/cytology , Proteasome Endopeptidase Complex/genetics , Receptors, Autocrine Motility Factor/chemistry , Receptors, Autocrine Motility Factor/genetics , Ubiquitin/genetics , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics
10.
Biochem Biophys Res Commun ; 509(2): 348-353, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30585151

ABSTRACT

The E3 ubiquitin (Ub) ligase gp78 plays an important role in endoplasmic reticulum (ER)-associated degradation (ERAD) and regulation of lipid biogenesis. Although a variety of substrates of gp78 have been described, the regulation of the degradation of gp78 itself remains poorly understood. To address this problem, we used co-immunoprecipitation-coupled liquid chromatography-tandem mass spectrometry (Co-IP/LC-MS/MS) to identify novel proteins interacting with gp78. One of the proteins identified in this study is the deubiquitylating (DUB) enzyme USP34 (Ub-specific protease 34). We demonstrate that knockdown of USP34 facilitates proteasomal degradation of gp78 and consequently impairs the function of gp78 in regulating lipid droplet formation. This study unveils a previously unknown function of USP34 in regulating the metabolic stability of gp78 and adds to our understanding of the relevance of partnering of DUBs and E3s in regulation of protein ubiquitylation.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Hepatocytes/metabolism , Receptors, Autocrine Motility Factor/genetics , Ubiquitin-Specific Proteases/genetics , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , HEK293 Cells , Hepatocytes/cytology , Humans , Lipid Droplets/metabolism , Lipid Metabolism/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Autocrine Motility Factor/metabolism , Signal Transduction , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/metabolism , Ubiquitination
11.
Elife ; 72018 12 13.
Article in English | MEDLINE | ID: mdl-30543180

ABSTRACT

Mammalian HMG-CoA reductase (HMGCR), the rate-limiting enzyme of the cholesterol biosynthetic pathway and the therapeutic target of statins, is post-transcriptionally regulated by sterol-accelerated degradation. Under cholesterol-replete conditions, HMGCR is ubiquitinated and degraded, but the identity of the E3 ubiquitin ligase(s) responsible for mammalian HMGCR turnover remains controversial. Using systematic, unbiased CRISPR/Cas9 genome-wide screens with a sterol-sensitive endogenous HMGCR reporter, we comprehensively map the E3 ligase landscape required for sterol-accelerated HMGCR degradation. We find that RNF145 and gp78 independently co-ordinate HMGCR ubiquitination and degradation. RNF145, a sterol-responsive ER-resident E3 ligase, is unstable but accumulates following sterol depletion. Sterol addition triggers RNF145 recruitment to HMGCR via Insigs, promoting HMGCR ubiquitination and proteasome-mediated degradation. In the absence of both RNF145 and gp78, Hrd1, a third UBE2G2-dependent E3 ligase, partially regulates HMGCR activity. Our findings reveal a critical role for the sterol-responsive RNF145 in HMGCR regulation and elucidate the complexity of sterol-accelerated HMGCR degradation. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).


Subject(s)
Hydroxymethylglutaryl CoA Reductases/genetics , Membrane Proteins/genetics , Receptors, Autocrine Motility Factor/genetics , Ubiquitin-Protein Ligases/genetics , Animals , CRISPR-Cas Systems , Cholesterol/metabolism , Humans , Membrane Proteins/metabolism , Mice , Proteolysis , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitination
12.
Neuro Oncol ; 20(12): 1594-1605, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30053149

ABSTRACT

Background: Aerobic glycolysis confers several advantages to tumor cells, including shunting of metabolites into anabolic pathways. In glioblastoma cells, hypoxia induces a flux shift from the pentose phosphate pathway toward glycolysis and a switch from proliferation to migration. The mechanistic link between glycolysis and migration is poorly understood. Since glucose-6-phosphate isomerase (GPI) is identical to the secreted cytokine autocrine motility factor (AMF), we investigated whether GPI/AMF regulates glioblastoma cell invasion. Methods: The expression and hypoxic regulation of GPI/AMF and its receptor AMFR were analyzed in glioblastoma tissue and cell lines. Functional effects were studied in vitro and in xenograft models. Results: High GPI/AMF expression in glioblastomas was found to be associated with a worse patient prognosis, and levels were highest in hypoxic pseudopalisades. Hypoxia upregulated both GPI/AMF and AMFR expression as well as GPI/AMF secretion in vitro. GPI/AMF stimulated cell migration in an autocrine fashion, and GPI/AMF expression was upregulated in migratory cells but reduced in rapidly proliferating cells. Knockdown or inhibition of GPI/AMF reduced glioblastoma cell migration but in part stimulated proliferation. In a highly invasive orthotopic glioblastoma model, GPI/AMF knockdown reduced tumor cell invasion but did not prolong survival. In a highly proliferative model, knockdown tumors were even larger and more proliferative than controls; however, perivascular invasion, provoked by simultaneous bevacizumab treatment, was reduced. Conclusions: GPI/AMF is a potent motogen for glioblastoma cells, explaining in part the association between glycolysis and migration. Targeting GPI/AMF is, however, problematic, since beneficial anti-invasive effects may be outweighed by unintended mitogenic effects. Key Points: 1.Increased glycolysis is linked with increased cell migration and invasion in glioblastoma cells. 2.The glycolysis enzyme GPI/AMF may serve as a target for antimetabolic and anti-invasive therapy. 3.Despite reducing tumor invasion, GPI/AMF targeting may have unwanted growth stimulatory effects.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Movement , Cell Proliferation , Glioblastoma/pathology , Glucose-6-Phosphate Isomerase/metabolism , Receptors, Autocrine Motility Factor/metabolism , Animals , Apoptosis , Autocrine Communication , Biomarkers, Tumor/genetics , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/metabolism , Glucose-6-Phosphate Isomerase/genetics , Humans , Mice , Neoplasm Invasiveness , Prognosis , Receptors, Autocrine Motility Factor/genetics , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
J Cell Physiol ; 233(10): 6352-6368, 2018 10.
Article in English | MEDLINE | ID: mdl-29741771

ABSTRACT

In cells, protein synthesis and degradation are normal processes, which are tightly regulated by various cellular metabolic pathways. Cellular protein quality control (PQC) mechanisms always present a continuous and rigorous check over all intracellular proteins before they can participate in various cellular physiological processes with the help of PQC pathways like autophagy and ubiquitin proteasome system (UPS). The UPS employs few selective E3 ubiquitin ligases for the intracellular degradation of cyclin-dependent kinase inhibitor 1B (p27Kip1 ) that tightly controls cell cycle progression. But, the complex mechanistic interactions and the interplay between E3 ubiquitin ligases involved in the functional regulation as well as expression of p27 are not well known. Here, we demonstrate that cell surface glycoprotein Gp78, a putative E3 ubiquitin ligase, is involved in the stabilization of intracellular steady-state levels of p27. Transient overexpression of Gp78 increases the accumulation of p27 in cells in the form of massive inclusions like structures, which could be due to its cumulative increased stability in cells. We have also monitored how under stress condition, E3 ubiquitin ligase Gp78 regulates endogenous levels of p27 in cells. ER stress treatment generates a marginal increase in Gp78 endogenous levels, and this elevation effect was prominent for intracellular accumulation of p27 in cells. Taken together, our current findings suggest a valuable multifactorial regulatory mechanism and linkage of p27 with UPS pathway.


Subject(s)
Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Proteasome Endopeptidase Complex/genetics , Receptors, Autocrine Motility Factor/genetics , A549 Cells , Animals , Autophagy/genetics , COS Cells , Cell Cycle Proteins/genetics , Chlorocebus aethiops , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation/genetics , Gene Knockdown Techniques , Humans , Metabolic Networks and Pathways , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin-Protein Ligases/genetics
14.
J Biol Chem ; 293(11): 4047-4055, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29374057

ABSTRACT

Cholesterol biosynthesis is tightly regulated in the cell. For example, high sterol concentrations can stimulate degradation of the rate-limiting cholesterol biosynthetic enzyme 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMG-CoA reductase, HMGCR). HMGCR is broken down by the endoplasmic reticulum membrane-associated protein complexes consisting of insulin-induced genes (Insigs) and the E3 ubiquitin ligase gp78. Here we found that HMGCR degradation is partially blunted in Chinese hamster ovary (CHO) cells lacking gp78 (gp78-KO). To identify other ubiquitin ligase(s) that may function together with gp78 in triggering HMGCR degradation, we performed a small-scale short hairpin RNA-based screening targeting endoplasmic reticulum-localized E3s. We found that knockdown of both ring finger protein 145 (Rnf145) and gp78 genes abrogates sterol-induced degradation of HMGCR in CHO cells. We also observed that RNF145 interacts with Insig-1 and -2 proteins and ubiquitinates HMGCR. Moreover, the tetrapeptide sequence YLYF in the sterol-sensing domain and the Cys-537 residue in the RING finger domain were essential for RNF145 binding to Insigs and RNF145 E3 activity, respectively. Of note, amino acid substitutions in the YLYF or of Cys-537 completely abolished RNF145-mediated HMGCR degradation. In summary, our study reveals that RNF145, along with gp78, promotes HMGCR degradation in response to elevated sterol levels and identifies residues essential for RNF145 function.


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Hydroxymethylglutaryl CoA Reductases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Proteolysis , Receptors, Autocrine Motility Factor/metabolism , Sterols/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Endoplasmic Reticulum/drug effects , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Receptors, Autocrine Motility Factor/genetics , Ubiquitin/metabolism , Ubiquitination
15.
Hellenic J Cardiol ; 59(1): 8-13, 2018.
Article in English | MEDLINE | ID: mdl-28212872

ABSTRACT

OBJECTIVE: To study the correlation between the GP78 gene polymorphism and blood fat, blood glucose, blood pressure and coronary atherosclerotic heart disease. METHODS: A total of 72 patients with coronary atherosclerotic heart disease were selected as the observation group, and 68 healthy participants were selected as the control group. The gp78 gene polymorphism of both groups was studied via polymerase chain reaction-restriction fragment length polymorphism (RFLP). At the same time, the multiple expression quantities of the GP78 gene in the tissues of both groups were tested via fluorogenic quantitative PCR, enzyme-linked immunosorbent assay (ELISA) and Western-blotting assay. Furthermore, the blood fat, blood glucose and blood pressure of subjects in both groups were tested. RESULTS: The percentages of the gp78 gene polymorphisms of Arg/Arg, Arg/Gly and Gly/Gly at the 145 locus of the study subjects in the observation group were 12.3%, 43.2% and 44.5%, respectively, while those in the control group were 74.3%, 11.2% and 14.5%, respectively, and there were significant differences between both groups. Based on the test results of the blood fat, blood glucose and blood pressure of the objects in the observation group and control group, significant differences were found between the two groups (P<0.05). CONCLUSION: There was a significant correlation between the 145 locus of the gp89 gene and coronary atherosclerotic heart disease, indexes of blood fat, blood glucose and blood pressure.


Subject(s)
Atherosclerosis/genetics , Coronary Artery Disease/genetics , DNA/genetics , Polymorphism, Genetic , Receptors, Autocrine Motility Factor/genetics , Adult , Atherosclerosis/metabolism , Blotting, Western , Coronary Artery Disease/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Follow-Up Studies , Genotype , Humans , Male , Middle Aged , Polymerase Chain Reaction , Receptors, Autocrine Motility Factor/metabolism , Retrospective Studies
16.
Oncogene ; 36(47): 6555-6567, 2017 11 23.
Article in English | MEDLINE | ID: mdl-28759037

ABSTRACT

The cellular prion protein (PrPC) is associated with metastasis, tumor progression and recurrence; however, the precise mechanisms underlying its action is not well understood. Our study found that PrPC degradation decreased tumor progression in colorectal cancer (CRC). In a CRC cell line and human CRC tissue exposed to hypoxia, induced heat-shock 70-kDa protein-1-like (HSPA1L) expression stabilized hypoxia-inducible factor-1α (HIF-1α) protein and promoted PrPC accumulation and tumorigenicity in vivo. PrPC was degraded via the proteasome pathway mediated by the ubiquitin-protein E3 ligase glycoprotein 78 (GP78), which interacts directly with PrPC. However, hypoxia-induced HSPA1L interacted with GP78 and inhibited its functions. HSPA1L knockdown facilitated the interaction of GP78 and PrPC, thereby increasing PrPC ubiquitination. Thus, GP78 was identified as the ubiquitinase for PrPC, thereby revealing an essential mechanism that controls PrPC levels in CRC. Our results suggest that the HSPA1L/HIF-1α/GP78 axis has a crucial role in PrPC accumulation during tumor progression.


Subject(s)
Carcinogenesis/metabolism , Colorectal Neoplasms/pathology , HSP70 Heat-Shock Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Prion Proteins/metabolism , Receptors, Autocrine Motility Factor/metabolism , Cell Culture Techniques , Colorectal Neoplasms/drug therapy , Disease Progression , Flow Cytometry , Gene Knockdown Techniques , HSP70 Heat-Shock Proteins/genetics , HT29 Cells , Humans , Molecular Targeted Therapy/methods , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RNA Interference , RNA, Small Interfering , Receptors, Autocrine Motility Factor/genetics , Signal Transduction , Ubiquitination
17.
Nat Cell Biol ; 19(7): 808-819, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28604676

ABSTRACT

Ubiquitin linkage to cysteine is an unconventional modification targeting protein for degradation. However, the physiological regulation of cysteine ubiquitylation is still mysterious. Here we found that ACAT2, a cellular enzyme converting cholesterol and fatty acid to cholesteryl esters, was ubiquitylated on Cys277 for degradation when the lipid level was low. gp78-Insigs catalysed Lys48-linked polyubiquitylation on this Cys277. A high concentration of cholesterol and fatty acid, however, induced cellular reactive oxygen species (ROS) that oxidized Cys277, resulting in ACAT2 stabilization and subsequently elevated cholesteryl esters. Furthermore, ACAT2 knockout mice were more susceptible to high-fat diet-associated insulin resistance. By contrast, expression of a constitutively stable form of ACAT2 (C277A) resulted in higher insulin sensitivity. Together, these data indicate that lipid-induced stabilization of ACAT2 ameliorates lipotoxicity from excessive cholesterol and fatty acid. This unconventional cysteine ubiquitylation of ACAT2 constitutes an important mechanism for sensing lipid-overload-induced ROS and fine-tuning lipid homeostasis.


Subject(s)
Cholesterol/metabolism , Fatty Acids/metabolism , Liver/enzymology , Sterol O-Acyltransferase/metabolism , Animals , CHO Cells , Cholesterol Esters/metabolism , Cricetulus , Cysteine , Diet, High-Fat , Disease Models, Animal , Genotype , Hep G2 Cells , Homeostasis , Humans , Insulin Resistance , Male , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Phenotype , Proteolysis , RNA Interference , Reactive Oxygen Species/metabolism , Receptors, Autocrine Motility Factor/genetics , Receptors, Autocrine Motility Factor/metabolism , Sterol O-Acyltransferase/deficiency , Sterol O-Acyltransferase/genetics , Time Factors , Transfection , Ubiquitination , Sterol O-Acyltransferase 2
18.
J Biol Chem ; 291(35): 18252-62, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27387505

ABSTRACT

The endoplasmic reticulum (ER) network comprises sheets and tubules that are connected by dynamic three-way junctions. Lunapark (Lnp) localizes to and stabilizes ER three-way junctions by antagonizing the small GTPase Atlastin, but how Lnp shapes the ER network is unclear. Here, we used an affinity purification approach and mass spectrometry to identify Lnp as an interacting partner of the ER protein quality control ubiquitin ligase gp78. Accordingly, Lnp purified from mammalian cells has a ubiquitin ligase activity in vitro Intriguingly, biochemical analyses show that this activity can be attributed not only to associated ubiquitin ligase, but also to an intrinsic ubiquitin ligase activity borne by Lnp itself. This activity is contained in the N-terminal 45 amino acids of Lnp although this segment does not share homology to any known ubiquitin ligase motifs. Despite its interaction with gp78, Lnp does not seem to have a broad function in degradation of misfolded ER proteins. On the other hand, the N-terminal ubiquitin ligase-bearing motif is required for the ER three-way junction localization of Lnp. Our study identifies a new type of ubiquitin ligase and reveals a potential link between ubiquitin and ER morphology regulation.


Subject(s)
Endoplasmic Reticulum/metabolism , Homeodomain Proteins/metabolism , Receptors, Autocrine Motility Factor/metabolism , Amino Acid Motifs , Animals , COS Cells , Chlorocebus aethiops , Endoplasmic Reticulum/genetics , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , HeLa Cells , Homeodomain Proteins/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Transport , Receptors, Autocrine Motility Factor/genetics
19.
Genet Mol Res ; 15(2)2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27173213

ABSTRACT

Our study clarifies the role of the autocrine motility factor receptor (AMFR) gene in porcine preadipocyte differentiation. AMFR-siRNA was transfected into porcine preadipocytes and the preadipocytes were induced to differentiation. Subsequently, qRT-PCR was conducted to examine changes in mRNA expression of a series of genes in porcine preadipocytes, including AMFR, sterol-regulatory element-binding protein-1a (SREBP1a), SREBP2, insulin-induced gene 1 (Insig1), and Insig2. Expression changes in the mRNA of genes regulating adipocyte differentiation were also analyzed using qRT-PCR, including peroxisome proliferator-activated receptor gamma (PPARγ), CCAAT/enhancer-binding protein alpha (C/EBPα), and Kruppel-like factor 2 (KLF2). Western blot analysis was conducted to examine the changes in AMFR protein expression in porcine preadipocytes. Additionally, morphological changes in differentiated porcine preadipocytes were examined by oil red O staining, and changes in optical density (OD) values were measured using an ultraviolet spectrophotometer. At 24 h after transfection with AMFR-siRNA, AMFR mRNA expression significantly reduced (P < 0.01), and AMFR protein expression markedly decreased (P < 0.05). The mRNA expression of SREBP1a, SREBP2, Insig1, and C/EBPα was significantly reduced (P < 0.01), whereas the expression of KLF2 mRNA was significantly elevated (P < 0.01). After induction of preadipocyte differentiation, the number of lipid droplets decreased in the AMFR-silenced group, and the OD value markedly reduced (P < 0.05). In addition, the expression of C/EBPα mRNA significantly decreased (P < 0.05), whereas the expression of KLF2 mRNA considerably increased (P < 0.05). Taken together, silencing of the AMFR gene inhibits the differentiation of porcine preadipocytes.


Subject(s)
Adipocytes/metabolism , Cell Differentiation , Receptors, Autocrine Motility Factor/metabolism , Adipocytes/cytology , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cells, Cultured , Gene Silencing , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Receptors, Autocrine Motility Factor/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Swine
20.
Proteomics Clin Appl ; 10(6): 681-90, 2016 06.
Article in English | MEDLINE | ID: mdl-25689831

ABSTRACT

PURPOSE: Affinity proteomic approaches by antibody bead arrays enable multiplexed analysis of proteins in body fluids. In the presented study, we investigated blood plasma within osteoporosis to discovery differential protein profiles and to propose novel biomarkers candidates for subsequent studies. EXPERIMENTAL DESIGN: Starting with 4608 antibodies and plasma samples from 22 women for an untargeted screening, a set of 72 proteins were suggested for further analysis. Complementing these with targets from literature and other studies, a targeted bead array of 180 antibodies was built to profile for 92 proteins in plasma samples of 180 women from two independent population-based studies. RESULTS: Differential profiles between osteoporosis patients and matched controls were discovered for 12 proteins in at least one of the two study sets. Among these targets, the levels of autocrine motility factor receptor (AMFR) were concordantly lower in plasma of female osteoporosis patients. Subsequently, verification of anti-AMFR antibody selectivity was conducted using high-density peptide and protein arrays, and Western blotting. CONCLUSIONS AND CLINICAL RELEVANCE: Further validation in additional study sets will be needed to determine the clinical value of the observed decrease in AMFR plasma levels in osteoporosis patients, but AMFR may aid our understanding of disease mechanisms and could support existing tools for diagnosis and monitoring of patient mobility within osteoporosis.


Subject(s)
Osteoporosis/diagnosis , Proteomics/methods , Receptors, Autocrine Motility Factor/genetics , Aged , Amino Acid Sequence , Antibodies/chemistry , Antibody Specificity , Biomarkers/blood , Case-Control Studies , Female , Gene Expression , Humans , Middle Aged , Osteoporosis/blood , Osteoporosis/genetics , Protein Array Analysis , Receptors, Autocrine Motility Factor/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...