Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Hum Genet ; 140(8): 1229-1239, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34159400

ABSTRACT

The extensive clinical and genetic heterogeneity of congenital limb malformation calls for comprehensive genome-wide analysis of genetic variation. Genome sequencing (GS) has the potential to identify all genetic variants. Here we aim to determine the diagnostic potential of GS as a comprehensive one-test-for-all strategy in a cohort of undiagnosed patients with congenital limb malformations. We collected 69 cases (64 trios, 1 duo, 5 singletons) with congenital limb malformations with no molecular diagnosis after standard clinical genetic testing and performed genome sequencing. We also developed a framework to identify potential noncoding pathogenic variants. We identified likely pathogenic/disease-associated variants in 12 cases (17.4%) including four in known disease genes, and one repeat expansion in HOXD13. In three unrelated cases with ectrodactyly, we identified likely pathogenic variants in UBA2, establishing it as a novel disease gene. In addition, we found two complex structural variants (3%). We also identified likely causative variants in three novel high confidence candidate genes. We were not able to identify any noncoding variants. GS is a powerful strategy to identify all types of genomic variants associated with congenital limb malformation, including repeat expansions and complex structural variants missed by standard diagnostic approaches. In this cohort, no causative noncoding SNVs could be identified.


Subject(s)
Genetic Heterogeneity , Homeodomain Proteins/genetics , Limb Deformities, Congenital/genetics , Mutation , Transcription Factors/genetics , Ubiquitin-Activating Enzymes/genetics , Base Sequence , Cohort Studies , DNA Copy Number Variations , Gene Expression , Genetic Testing , Humans , Infant , Limb Deformities, Congenital/metabolism , Limb Deformities, Congenital/pathology , Male , Pedigree , Transcription Factors/deficiency , Ubiquitin-Activating Enzymes/deficiency , Whole Genome Sequencing
2.
J Clin Invest ; 131(8)2021 04 15.
Article in English | MEDLINE | ID: mdl-33651714

ABSTRACT

The excitability of interneurons requires Nav1.1, the α subunit of the voltage-gated sodium channel. Nav1.1 deficiency and mutations reduce interneuron excitability, a major pathological mechanism for epilepsy syndromes. However, the regulatory mechanisms of Nav1.1 expression remain unclear. Here, we provide evidence that neddylation is critical to Nav1.1 stability. Mutant mice lacking Nae1, an obligatory component of the E1 ligase for neddylation, in parvalbumin-positive interneurons (PVINs) exhibited spontaneous epileptic seizures and premature death. Electrophysiological studies indicate that Nae1 deletion reduced PVIN excitability and GABA release and consequently increased the network excitability of pyramidal neurons (PyNs). Further analysis revealed a reduction in sodium-current density, not a change in channel property, in mutant PVINs and decreased Nav1.1 protein levels. These results suggest that insufficient neddylation in PVINs reduces Nav1.1 stability and thus the excitability of PVINs; the ensuing increased PyN activity causes seizures in mice. Consistently, Nav1.1 was found reduced by proteomic analysis that revealed abnormality in synapses and metabolic pathways. Our findings describe a role of neddylation in maintaining Nav1.1 stability for PVIN excitability and reveal what we believe is a new mechanism in the pathogenesis of epilepsy.


Subject(s)
Action Potentials , Epilepsy/metabolism , Interneurons/metabolism , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Seizures/metabolism , Animals , Disease Models, Animal , Interneurons/pathology , Mice , Mice, Mutant Strains , NAV1.1 Voltage-Gated Sodium Channel/genetics , Protein Stability , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Seizures/genetics , Ubiquitin-Activating Enzymes/deficiency , Ubiquitin-Activating Enzymes/metabolism
3.
Cell Commun Signal ; 17(1): 82, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31345225

ABSTRACT

BACKGROUND: The SUMO-activating enzyme SAE1 is indispensable for protein SUMOylation. A dysregulation of SAE1 expression involves in progression of several human cancers. However, its biological roles of SAE1 in glioma are unclear by now. METHODS: The differential proteome between human glioma tissues and para-cancerous brain tissues were identified by LC-MS/MS. SAE1 expression was further assessed by immunohistochemistry. The patient overall survival versus SAE1 expression level was evaluated by Kaplan-Meier method. The glioma cell growth and migration were evaluated under SAE1 overexpression or inhibition by the CCK8, transwell assay and wound healing analysis. The SUMO1 modified target proteins were enriched from total cellular or tissue proteins by incubation with the anti-SUMO1 antibody on protein-A beads overnight, then the SUMOylated proteins were detected by Western blot. Cell apoptosis and cell cycle were analyzed by flow cytometry. The nude mouse xenograft was determined glioma growth and tumorigenicity in vivo. RESULTS: SAE1 is identified to increase in glioma tissues by a quantitative proteomic dissection, and SAE1 upregulation indicates a high level of tumor malignancy grade and a poor overall survival for glioma patients. SAE1 overexpression induces an increase of the SUMOylation and Ser473 phosphorylation of AKT, which promotes glioma cell growth in vitro and in nude mouse tumor model. On the contrary, SAE1 silence induces an obvious suppression of the SUMOylation and Ser473 phosphorylation of Akt, which inhibits glioma cell proliferation and the tumor xenograft growth through inducing cell cycle arrest at G2 phase and cell apoptosis driven by serial biochemical molecular events. CONCLUSION: SAE1 promotes glioma cancer progression via enhancing Akt SUMOylation-mediated signaling pathway, which indicates targeting SUMOylation is a promising therapeutic strategy for human glioma.


Subject(s)
Brain Neoplasms/pathology , Disease Progression , Glioma/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Sumoylation , Ubiquitin-Activating Enzymes/metabolism , Animals , Apoptosis , Carcinogenesis , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Transformation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Mice , Survival Analysis , Ubiquitin-Activating Enzymes/deficiency , Ubiquitin-Activating Enzymes/genetics , Up-Regulation
4.
Biochem Biophys Res Commun ; 514(2): 545-549, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31060776

ABSTRACT

UBA6 is an alternative enzyme for ubiquitin activation in vertebrates that plays a pivotal role in early mouse development. Previously, we reported that the Uba6 brain-specific knockout (NKO) mouse is a novel autism spectrum disorder (ASD) mouse model that displays decreased social behavior and communication. To determine the therapeutic impact of environmental stimulation in ASDs, we investigated the behavioral and molecular changes of the NKO and control mice after exposure to environmental enrichment and paired housing in different developmental phases. Our results demonstrated that early paired housing could diminish the ASD phenotypes of NKO mice such as impaired nest building and social interaction and anxiety. Additionally, increased histone acetylation in the amygdala was observed in NKO mice after paired housing without a change in Ube3a levels. Our data suggest that paired housing at an early time point can play a crucial role in ameliorating ASD behavior and can be applied in other ASD animal models or clinical settings.


Subject(s)
Amygdala/enzymology , Anxiety/genetics , Autism Spectrum Disorder/genetics , Housing, Animal , Ubiquitin-Activating Enzymes/genetics , Acetylation , Animals , Anxiety/enzymology , Anxiety/physiopathology , Anxiety/prevention & control , Autism Spectrum Disorder/enzymology , Autism Spectrum Disorder/physiopathology , Disease Models, Animal , Exploratory Behavior/physiology , Gene Expression , Histones/genetics , Histones/metabolism , Interpersonal Relations , Maze Learning/physiology , Mice , Mice, Knockout , Nesting Behavior/physiology , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes/deficiency , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
5.
Am J Hum Genet ; 99(3): 695-703, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27545681

ABSTRACT

Via whole-exome sequencing, we identified rare autosomal-recessive variants in UBA5 in five children from four unrelated families affected with a similar pattern of severe intellectual deficiency, microcephaly, movement disorders, and/or early-onset intractable epilepsy. UBA5 encodes the E1-activating enzyme of ubiquitin-fold modifier 1 (UFM1), a recently identified ubiquitin-like protein. Biochemical studies of mutant UBA5 proteins and studies in fibroblasts from affected individuals revealed that UBA5 mutations impair the process of ufmylation, resulting in an abnormal endoplasmic reticulum structure. In Caenorhabditis elegans, knockout of uba-5 and of human orthologous genes in the UFM1 cascade alter cholinergic, but not glutamatergic, neurotransmission. In addition, uba5 silencing in zebrafish decreased motility while inducing abnormal movements suggestive of seizures. These clinical, biochemical, and experimental findings support our finding of UBA5 mutations as a pathophysiological cause for early-onset encephalopathies due to abnormal protein ufmylation.


Subject(s)
Alleles , Brain Diseases/genetics , Mutation/genetics , Proteins/metabolism , Ubiquitin-Activating Enzymes/genetics , Age of Onset , Animals , Brain Mapping , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Child , Child, Preschool , Cholinergic Neurons/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Epilepsy/genetics , Exome/genetics , Female , Fibroblasts , Genes, Recessive/genetics , Humans , Intellectual Disability/genetics , Magnetic Resonance Imaging , Male , Microcephaly/genetics , Movement Disorders , Proteins/genetics , Synaptic Transmission/genetics , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes/deficiency , Ubiquitin-Activating Enzymes/metabolism , Ubiquitins/genetics , Ubiquitins/metabolism , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
6.
Nature ; 531(7595): 523-527, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-26982722

ABSTRACT

The integrated stress response (ISR) is a homeostatic mechanism by which eukaryotic cells sense and respond to stress-inducing signals, such as amino acid starvation. General controlled non-repressed (GCN2) kinase is a key orchestrator of the ISR, and modulates protein synthesis in response to amino acid starvation. Here we demonstrate in mice that GCN2 controls intestinal inflammation by suppressing inflammasome activation. Enhanced activation of ISR was observed in intestinal antigen presenting cells (APCs) and epithelial cells during amino acid starvation, or intestinal inflammation. Genetic deletion of Gcn2 (also known as Eif2ka4) in CD11c(+) APCs or intestinal epithelial cells resulted in enhanced intestinal inflammation and T helper 17 cell (TH17) responses, owing to enhanced inflammasome activation and interleukin (IL)-1ß production. This was caused by reduced autophagy in Gcn2(-/-) intestinal APCs and epithelial cells, leading to increased reactive oxygen species (ROS), a potent activator of inflammasomes. Thus, conditional ablation of Atg5 or Atg7 in intestinal APCs resulted in enhanced ROS and TH17 responses. Furthermore, in vivo blockade of ROS and IL-1ß resulted in inhibition of TH17 responses and reduced inflammation in Gcn2(-/-) mice. Importantly, acute amino acid starvation suppressed intestinal inflammation via a mechanism dependent on GCN2. These results reveal a mechanism that couples amino acid sensing with control of intestinal inflammation via GCN2.


Subject(s)
Amino Acids/metabolism , Colitis/metabolism , Inflammasomes/antagonists & inhibitors , Inflammation/metabolism , Intestinal Mucosa/metabolism , Intestines/pathology , Protein Serine-Threonine Kinases/metabolism , Amino Acids/administration & dosage , Amino Acids/deficiency , Amino Acids/pharmacology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Autophagy , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Colitis/etiology , Colitis/pathology , Colitis/prevention & control , Disease Models, Animal , Epithelial Cells/metabolism , Female , Humans , Inflammasomes/metabolism , Inflammation/etiology , Inflammation/pathology , Inflammation/prevention & control , Interleukin-1beta/immunology , Male , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Reactive Oxygen Species/metabolism , Stress, Physiological , Th17 Cells/immunology , Ubiquitin-Activating Enzymes/deficiency , Ubiquitin-Activating Enzymes/metabolism
7.
Behav Brain Res ; 281: 78-85, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25523030

ABSTRACT

The Uba6-Use1 ubiquitin enzyme cascade is a poorly understood arm of the ubiquitin-proteasome system required for mouse development. Recently, we reported that Uba6 brain-specific knockout (termed NKO) mice display abnormal social behavior and neuronal development due to a decreased spine density and accumulation of Ube3a and Shank3. To better characterize a potential role for NKO mice in autism spectrum disorders (ASDs), we performed a comprehensive behavioral characterization of the social behavior and communication of NKO mice. Our behavioral results confirmed that NKO mice display social impairments, as indicated by fewer vocalizations and decreased social interaction. We conclude that UBA6 NKO mice represent a novel ASD mouse model of anti-social and less verbal behavioral symptoms.


Subject(s)
Child Development Disorders, Pervasive/psychology , Social Behavior , Ubiquitin-Activating Enzymes/deficiency , Vocalization, Animal , Animals , Child Development Disorders, Pervasive/genetics , Disease Models, Animal , Female , Locomotion , Male , Mice , Mice, Knockout , Motor Activity , Nesting Behavior , Ubiquitin-Activating Enzymes/genetics
8.
Bull Cancer ; 100(7-8): 15-21, 2013.
Article in English | MEDLINE | ID: mdl-23823853

ABSTRACT

Cisplatin-(DDP)-based adjuvant chemotherapy is widely used for the treatment of esophageal cancer. However, DDP-based combinatorial treatments can eventually result in tumor resistance response. Therefore, new therapeutic strategies and/or new adjuvant drugs still need to be explored. In this study, we aimed to understand the role of autophagy in ESCC cells resistance to Cisplatin and discuss its potential therapeutic implication. We found that exposure to Cisplatin induced a significant increase in LC3 formation. While the proliferation of ESCC cells was inhibited upon Cisplatin exposure, inhibition of autophagy by ATG7 interference further increased the sensitivity to chemotherapy. Meanwhile, the Cisplatin-induced apoptotic cell death was significantly enhanced. These results suggest that autophagy may function importantly in ESCC cells resistance to Cisplatin. Intriguingly, the resistance could be recovered by autophagy inhibition. This also points to potential therapy for ESCC by perturbing autophagy.


Subject(s)
Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Carcinoma, Squamous Cell/drug therapy , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Esophageal Neoplasms/drug therapy , Ubiquitin-Activating Enzymes/deficiency , Apoptosis/drug effects , Apoptosis/physiology , Autophagy/physiology , Autophagy-Related Protein 7 , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Chemotherapy, Adjuvant/methods , Cytoplasmic Vesicles , Drug Resistance, Neoplasm/physiology , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Gene Knockdown Techniques , Humans , RNA Interference , Ubiquitin-Activating Enzymes/genetics
9.
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
10.
PLoS One ; 8(1): e55096, 2013.
Article in English | MEDLINE | ID: mdl-23383069

ABSTRACT

The phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway promotes melanoma tumor growth and survival while suppressing autophagy, a catabolic process through which cells collect and recycle cellular components to sustain energy homeostasis in starvation. Conversely, inhibitors of the PI3K/AKT/mTOR pathway, in particular the mTOR inhibitor temsirolimus (CCI-779), induce autophagy, which can promote tumor survival and thus, these agents potentially limit their own efficacy. We hypothesized that inhibition of autophagy in combination with mTOR inhibition would block this tumor survival mechanism and hence improve the cytotoxicity of mTOR inhibitors in melanoma. Here we found that melanoma cell lines of multiple genotypes exhibit high basal levels of autophagy. Knockdown of expression of the essential autophagy gene product ATG7 resulted in cell death, indicating that survival of melanoma cells is autophagy-dependent. We also found that the lysosomotropic agent and autophagy inhibitor hydroxychloroquine (HCQ) synergizes with CCI-779 and led to melanoma cell death via apoptosis. Combination treatment with CCI-779 and HCQ suppressed melanoma growth and induced cell death both in 3-dimensional (3D) spheroid cultures and in tumor xenografts. These data suggest that coordinate inhibition of the mTOR and autophagy pathways promotes apoptosis and could be a new therapeutic paradigm for the treatment of melanoma.


Subject(s)
Autophagy/drug effects , Melanoma/pathology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Allosteric Regulation/drug effects , Animals , Autophagy-Related Protein 7 , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic , Drug Synergism , Gene Knockdown Techniques , Humans , Hydroxychloroquine/pharmacology , Male , Mice , Mice, Nude , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Ubiquitin-Activating Enzymes/deficiency , Ubiquitin-Activating Enzymes/genetics
11.
Free Radic Biol Med ; 51(3): 664-70, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21684330

ABSTRACT

The ubiquitin-proteasome system governs the half-life of most cellular proteins. Calorie restriction (CR) extends the maximum life span of a variety of species and prevents oxidized protein accumulation. We studied the effects of CR on the ubiquitin-proteasome system and protein turnover in aging Saccharomyces cerevisiae. CR increased chronological life span as well as proteasome activity compared to control cells. The levels of protein carbonyls, a marker of protein oxidation, and those of polyubiquitinated proteins were modulated by CR. Controls, but not CR cells, exhibited a significant increase in oxidized proteins. In keeping with decreased proteasome activity, polyubiquitinated proteins were increased in young control cells compared to time-matched CR cells, but were profoundly decreased in aged control cells despite decreased proteasomal activity. This finding is related to a decreased polyubiquitination ability due to the impairment of the ubiquitin-activating enzyme in aged control cells, probably related to a more oxidative microenvironment. CR preserves the ubiquitin-proteasome system activity. Overall, we found that aging and CR modulate many aspects of protein modification and turnover.


Subject(s)
Aging/physiology , Caloric Restriction , Oxidative Stress , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae/physiology , Unfolded Protein Response , Cells, Cultured , Enzyme Activation , Oxidation-Reduction , Oxygen Consumption , Protein Carbonylation , Proteins/metabolism , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes/deficiency
12.
Blood Cells Mol Dis ; 45(2): 103-11, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20591702

ABSTRACT

Following bone marrow transplantation, donor stem cells are recruited from their quiescent status to promote the rapid reconstitution in recipients. This dynamic process is tightly regulated by a complex of internal and external signals. Protein modification by the ubiquitin like modifier ISG15 (ISGylation) is strongly induced by type I interferons (IFNs). There are higher levels of type I IFNs and protein ISGylation in the bone marrow of recipients shortly after transplantation. In order to clarify the physiological function of protein ISGylation, we generated a mouse model that lacks protein ISGylation due to deficiency of ISG15 conjugating enzyme Ube1L (Ube1L(-/-)). In this report, we focused on the analysis of the hematopoietic system in Ube1L(-)(/)(-) mice in steady-state hematopoiesis and its potential protective role during bone marrow reconstitution. Here we demonstrated that In Ube1L(-/-) mice, steady-state hematopoiesis was unperturbed. However, transplantation experiment revealed a 50% reduction in repopulation potential of Ube1L-deficient cells at 3weeks posttransplantation, but no differences at 6 and 12weeks. A competitive transplantation experiment magnified and extended this phenotype. Cell cycle analysis revealed that under the condition with high levels of IFNs and protein ISGylation, the Ube1L deficiency can cause G2/M phase block of cell cycle in hematopoietic multipotential progenitors. These observations indicate that although protein ISGylation is dispensable for steady-state hematopoiesis, it plays a significant role during interferon related stress response, such as bone marrow transplantation.


Subject(s)
Bone Marrow Transplantation , Cytokines/metabolism , Hematopoiesis/genetics , Ubiquitin-Activating Enzymes/metabolism , Animals , Cell Cycle , Cell Proliferation , Interferon Type I/metabolism , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Ubiquitin-Activating Enzymes/deficiency , Ubiquitin-Activating Enzymes/genetics , Ubiquitins/metabolism
13.
Autophagy ; 6(2): 248-55, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20083906

ABSTRACT

Photodynamic therapy (PDT) uses a photosensitizer, light and oxygen to produce extensive oxidative damage to organelles housing the photosensitizer. Although PDT is an efficient trigger of apoptosis, it also induces autophagy in many kinds of cells. Autophagy can serve as both a cell survival and a cell death mechanism. Our previous study indicates that autophagy contributes to cell death after PDT, especially in apoptosis-deficient cells. Here, we provide further evidence to support the role of autophagy in cell killing after PDT. Autophagy was blocked by knockdown of one essential factor, LC3 or Atg7, in MCF-7 cells. The cells were exposed to a range of doses of PDT sensitized by the phthalocyanine Pc 4; steps in autophagy were monitored by western blotting for LC3-II and by fluorescence microscopy for the uptake of monodansylcadaverine or for the distribution of transfected GFP-LC3; and overall cell death was monitored by MTT assay and by clonogenic assay. We find that blocking autophagy increased the survival of MCF-7 cells after PDT and increased the shoulder on the dose-response curve. In response to Pc 4-PDT, Atg7-deficient MCF-7 cells remained capable of robust accumulation of LC3-II, but were defective in comparison to Atg7(+) cells in the formation of autophagosomes. We conclude that apoptosis-deficient cells rely on autophagy for cell death after Pc 4-PDT and that the strong activation of LC3 maturation in response to PDT could occur even in cells with limited or no Atg7 expression.


Subject(s)
Breast Neoplasms , Cell Line, Tumor/radiation effects , Photochemotherapy , Ubiquitin-Activating Enzymes/deficiency , Apoptosis/physiology , Autophagy/physiology , Autophagy-Related Protein 7 , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Female , Humans , Indoles/therapeutic use , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Photosensitizing Agents/therapeutic use , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
15.
J Virol ; 83(2): 1147-51, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19004958

ABSTRACT

ISG15 functions as a critical antiviral molecule against influenza virus, with infection inducing both the conjugation of ISG15 to target proteins and production of free ISG15. Here, we report that mice lacking the ISG15 E1 enzyme UbE1L fail to form ISG15 conjugates. Both UbE1L(-/-) and ISG15(-/-) mice display increased susceptibility to influenza B virus infection, including non-mouse-adapted strains. Finally, we demonstrate that ISG15 controls influenza B virus infection through its action within radioresistant stromal cells and not bone marrow-derived cells. Thus, the conjugation of ISG15 to target proteins within stromal cells is critical to its activity against influenza virus.


Subject(s)
Cytokines/deficiency , Genetic Predisposition to Disease , Influenza B virus/immunology , Orthomyxoviridae Infections/immunology , Ubiquitin-Activating Enzymes/deficiency , Animals , Lung/virology , Mice , Survival Analysis , Ubiquitins/deficiency
16.
Biochem Biophys Res Commun ; 368(3): 643-9, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18249191

ABSTRACT

Autophagy is a bulk protein degradation system for the entire organelles and cytoplasmic proteins. Previously, we have shown the liver dysfunction by autophagy deficiency. To examine the pathological effect of autophagy deficiency, we examined protein composition and their levels in autophagy-deficient liver by the proteomic analysis. While impaired autophagy led to an increase in total protein mass, the protein composition was largely unchanged, consistent with non-selective proteins/organelles degradation of autophagy. However, a series of oxidative stress-inducible proteins, including glutathione S-transferase families, protein disulfide isomerase and glucose-regulated proteins were specifically increased in autophagy-deficient liver, probably due to enhanced gene expression, which is induced by accumulation of Nrf2 in the nuclei of mutant hepatocytes. Our results suggest that autophagy deficiency causes oxidative stress, and such stress might be the main cause of liver injury in autophagy-deficient liver.


Subject(s)
Autophagy , Heat-Shock Proteins/metabolism , Hepatocytes/metabolism , Liver/metabolism , Proteome/metabolism , Ubiquitin-Activating Enzymes/deficiency , Animals , Autophagy-Related Protein 7 , Cells, Cultured , Mice , Mice, Knockout , Mice, Transgenic , Proteomics/methods
17.
Mol Biol Cell ; 19(3): 797-806, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18094039

ABSTRACT

Autophagy has been proposed to promote cell death during lumen formation in three-dimensional mammary epithelial acini because numerous autophagic vacuoles are observed in the dying central cells during morphogenesis. Because these central cells die due to extracellular matrix (ECM) deprivation (anoikis), we have directly interrogated how matrix detachment regulates autophagy. Detachment induces autophagy in both nontumorigenic epithelial lines and in primary epithelial cells. RNA interference-mediated depletion of autophagy regulators (ATGs) inhibits detachment-induced autophagy, enhances apoptosis, and reduces clonogenic recovery after anoikis. Remarkably, matrix-detached cells still exhibit autophagy when apoptosis is blocked by Bcl-2 overexpression, and ATG depletion reduces the clonogenic survival of Bcl-2-expressing cells after detachment. Finally, stable reduction of ATG5 or ATG7 in MCF-10A acini enhances luminal apoptosis during morphogenesis and fails to elicit long-term luminal filling, even when combined with apoptotic inhibition mediated by Bcl-2 overexpression. Thus, autophagy promotes epithelial cell survival during anoikis, including detached cells harboring antiapoptotic lesions.


Subject(s)
Autophagy , Epithelial Cells/cytology , Extracellular Matrix/metabolism , Animals , Apoptosis/drug effects , Autophagy/drug effects , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Cell Adhesion/drug effects , Cell Line , Cell Survival/drug effects , Dogs , Epidermal Growth Factor/pharmacology , Epithelial Cells/drug effects , ErbB Receptors/metabolism , Extracellular Matrix/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Ubiquitin-Activating Enzymes/deficiency , Ubiquitin-Activating Enzymes/metabolism
18.
Mol Cell Biol ; 24(21): 9592-600, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15485925

ABSTRACT

Protein ISGylation is unique among ubiquitin-like conjugation systems in that the expression and conjugation processes are induced by specific stimuli, mainly via the alpha/beta interferon signaling pathway. It has been suggested that protein ISGylation plays a special role in the immune response, because of its interferon-signal dependency and its appearance only in higher eukaryotic organisms. Here, we report the identification of an ISG15-conjugating enzyme, Ubc8. Like other components of the protein ISGylation system (ISG15, UBE1L, and UBP43), Ubc8 is an interferon-inducible protein. Ubc8 clearly mediates protein ISGylation in transfection assays. The reduction of Ubc8 expression by small interfering RNA causes a decrease in protein ISGylation in HeLa cells upon interferon treatment. Neither UbcH7/UbcM4, the closest homologue of Ubc8 among known ubiquitin E2s, nor the small ubiquitin-like modifier E2 Ubc9 supports protein ISGylation. These findings strongly suggest that Ubc8 is a major ISG15-conjugating enzyme responsible for protein ISGylation upon interferon stimulation. Furthermore, we established an assay system to detect ISGylated target proteins by cotransfection of ISG15, UBE1L, and Ubc8 together with a target protein to be analyzed. This method provides an easy and effective way to identify new targets for the ISGylation system and will facilitate related studies.


Subject(s)
Cytokines/metabolism , Interferons/pharmacology , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitins/analogs & derivatives , Ubiquitins/metabolism , Animals , Base Sequence , Cells, Cultured , Cytokines/chemistry , Cytokines/genetics , DNA-Binding Proteins/metabolism , Humans , Mice , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Binding , Protein Processing, Post-Translational , RNA Interference , STAT1 Transcription Factor , Signal Transduction/drug effects , Trans-Activators/metabolism , Two-Hybrid System Techniques , Ubiquitin-Activating Enzymes/deficiency , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitins/chemistry , Ubiquitins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...