Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Cells ; 11(14)2022 07 11.
Article in English | MEDLINE | ID: mdl-35883611

ABSTRACT

Oxidative stress, caused by the accumulation of reactive oxygen species (ROS) during acute myocardial infarction (AMI), is one of the main factors leading to myocardial cell damage and programmed cell death. Phosphatidylinositol-3-kinase-AKT (PI3K-AKT) signaling is essential for regulating cell proliferation, differentiation, and apoptosis. Phosphoinositide-3-kinase (PI3K)-interacting protein 1 (PIK3IP1) is an intrinsic inhibitor of PI3K in various tissues, but its functional role during AMI remains unknown. In this study, the anti-ischemic role of PIK3IP1 in an in vitro AMI setting was evaluated using H9c2 cells. The MTT assay demonstrated that cell viability decreased significantly via treatment with H2O2 (200-500 µM). The TUNEL assay results revealed substantial cellular apoptosis following treatment with 200 µM H2O2. Under the same conditions, the expression levels of hypoxia-inducible factor (HIF-1α), endothelin-1 (ET-1), bcl-2-like protein 4 (BAX), and cleaved caspase-3 were elevated, whereas those of PIK3IP1, LC3II, p53, and Bcl-2 decreased significantly. PIK3IP1 overexpression inhibited H2O2-induced and PI3K-mediated apoptosis; however, PIK3IP1 knockdown reversed this effect, suggesting that PIK3IP1 functions as an anti-apoptotic molecule. To identify both the upstream and downstream molecules associated with PIK3IP1, ET-1 receptor type-specific antagonists (BQ-123 and BQ-788) and PI3K subtype-specific antagonists (LY294002 and IPI-549) were used to determine the participating isoforms. Co-immunoprecipitation was performed to identify the binding partners of PIK3IP1. Our results demonstrated that ROS-induced cardiac cell death may occur through the ETA-PI3Kγ-AKT axis, and that PIK3IP1 inhibits binding with both ETA and PI3Kγ. Taken together, these findings reveal that PIK3IP1 plays an anti-ischemic role by reducing the likelihood of programmed cell death via interaction with the ETA-PI3Kr-AKT axis.


Subject(s)
Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Hydrogen Peroxide/pharmacology , Phosphatidylinositol 3-Kinase , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism
2.
FASEB J ; 36(1): e22103, 2022 01.
Article in English | MEDLINE | ID: mdl-34921695

ABSTRACT

Ubiquitination has been shown to provide an essential regulatory role in modulating the duration and amplitude of the signaling activity in angiogenesis. While successive enzymatic reactions mediated by three distinct types of enzymes commonly known as E1, E2, and E3 are required for ubiquitination, the role of E3s which govern the final step of ubiquitination has been extensively analyzed in angiogenesis. In contrast, the role of E2s, which determine the context and functional consequences of ubiquitination, remains largely unknown with respect to angiogenesis. To better elucidate the role of E2s in modulating endothelial behaviors during angiogenesis, we first systematically analyze the expression pattern of E2s in endothelial cells (ECs) using previously published scRNA-seq data and identify ubiquitin-conjugating enzyme variant 1 (UBE2V1), an unconventional E2 without innate catalytic activity, as one of the most abundantly expressed E2s in ECs. While ubiquitously expressed in diverse cell types, abrogation of UBE2V1 significantly impairs proliferation and viability of human umbilical vein endothelial cells (HUVECs) without affecting other cell types, suggesting that UBE2V1 is likely to possess nonredundant functions in ECs. Consistent with this idea, UBE2V1 appears to be critical for morphogenesis and migration of ECs during angiogenesis. Interestingly, we find that UBE2V1 is essential for fibroblast growth factor 2 (FGF2)-induced angiogenesis, but appears to have minor effects on vascular endothelial growth factor-A-induced angiogenesis in vitro as well as in vivo. Therefore, it seems that UBE2V1 could enable ECs to distinguish two related yet distinct angiogenic cues. Mechanistically, we show that UBE2V1 promotes ubiquitination of MEK kinase 1, a key mediator of FGF2 signaling, to enhance phosphorylation of extracellular signal-regulated kinase 1/2 in HUVECs. Taken together, our results illustrate the unique role of UBE2V1 as a key modulator for angiogenic behaviors in ECs.


Subject(s)
Cell Proliferation , Endothelium, Vascular/metabolism , Fibroblast Growth Factor 2/metabolism , MAP Kinase Signaling System , Transcription Factors/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 2/genetics , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , PC-3 Cells , Transcription Factors/genetics , Ubiquitin-Conjugating Enzymes/genetics
3.
Circ Res ; 124(9): e63-e80, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30786847

ABSTRACT

RATIONALE: SERCA2a, sarco-endoplasmic reticulum Ca2+-ATPase, is a critical determinant of cardiac function. Reduced level and activity of SERCA2a are major features of heart failure. Accordingly, intensive efforts have been made to develop efficient modalities for SERCA2a activation. We showed that the activity of SERCA2a is enhanced by post-translational modification with SUMO1 (small ubiquitin-like modifier 1). However, the roles of other post-translational modifications on SERCA2a are still unknown. OBJECTIVE: In this study, we aim to assess the role of lysine acetylation on SERCA2a function and determine whether inhibition of lysine acetylation can improve cardiac function in the setting of heart failure. METHODS AND RESULTS: The acetylation of SERCA2a was significantly increased in failing hearts of humans, mice, and pigs, which is associated with the reduced level of SIRT1 (sirtuin 1), a class III histone deacetylase. Downregulation of SIRT1 increased the SERCA2a acetylation, which in turn led to SERCA2a dysfunction and cardiac defects at baseline. In contrast, pharmacological activation of SIRT1 reduced the SERCA2a acetylation, which was accompanied by recovery of SERCA2a function and cardiac defects in failing hearts. Lysine 492 (K492) was of critical importance for the regulation of SERCA2a activity via acetylation. Acetylation at K492 significantly reduced the SERCA2a activity, presumably through interfering with the binding of ATP to SERCA2a. In failing hearts, acetylation at K492 appeared to be mediated by p300 (histone acetyltransferase p300), a histone acetyltransferase. CONCLUSIONS: These results indicate that acetylation/deacetylation at K492, which is regulated by SIRT1 and p300, is critical for the regulation of SERCA2a activity in hearts. Pharmacological activation of SIRT1 can restore SERCA2a activity through deacetylation at K492. These findings might provide a novel strategy for the treatment of heart failure.


Subject(s)
Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sirtuin 1/metabolism , Acetylation , Adenosine Triphosphate/metabolism , Animals , Cell Line , Cells, Cultured , E1A-Associated p300 Protein/metabolism , Heart Failure/enzymology , Heart Failure/genetics , Humans , Lysine/genetics , Lysine/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocytes, Cardiac/pathology , Protein Processing, Post-Translational , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sirtuin 1/genetics , Swine
4.
Biochem Biophys Res Commun ; 506(1): 102-107, 2018 11 17.
Article in English | MEDLINE | ID: mdl-30336976

ABSTRACT

Ubiquitin-conjugating enzymes (E2) form thioester bonds with ubiquitin (Ub), which are subsequently transferred to target proteins for cellular progress. Ube2K/E2-25K (a class II E2 enzyme) contains a C-terminal ubiquitin-associated (UBA) domain that has been suggested to control ubiquitin recognition, dimerization, or poly-ubiquitin chain formation. Ube2K is a special E2 because it synthesizes K48-linked poly-ubiquitin chains without E3 ubiquitin ligase. We found that a novel interaction between the acceptor di-Ub (Ub2) and the auxiliary Ube2K promotes the discharging reaction and production of tri-Ub (Ub3), probably by guiding and positioning the K48 (in the distal Ub) of the acceptor Ub2 in the active site. We also determined the crystal structure of Ube2K-Ub2 at 2.47 Šresolution. Based on our structural and biochemical data, we proposed a structural model of Ub3 synthesis by Ube2K without E3.


Subject(s)
Lysine/chemistry , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitins/chemistry , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Lysine/metabolism , Molecular Docking Simulation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitins/genetics , Ubiquitins/metabolism
5.
J Immunol ; 199(9): 3051-3062, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28972088

ABSTRACT

The transcription factor NFAT1 plays a pivotal role in the homeostasis of T lymphocytes. However, its functional importance in non-CD4+ T cells, especially in systemic immune disorders, is largely unknown. In this study, we report that NFAT1 regulates dendritic cell (DC) tolerance and suppresses systemic autoimmunity using the experimental autoimmune myasthenia gravis (EAMG) as a model. Myasthenia gravis and EAMG are T cell-dependent, Ab-mediated autoimmune disorders in which the acetylcholine receptor is the major autoantigen. NFAT1-knockout mice showed higher susceptibility to EAMG development with enhanced Th1/Th17 cell responses. NFAT1 deficiency led to a phenotypic alteration of DCs that show hyperactivation of NF-κB-mediated signaling pathways and enhanced binding of NF-κB (p50) to the promoters of IL-6 and IL-12. As a result, NFAT1-knockout DCs produced much higher levels of proinflammatory cytokines such as IL-1ß, IL-6, IL-12, and TNF-α, which preferentially induce Th1/Th17 cell differentiation. Our data suggest that NFAT1 may limit the hyperactivation of the NF-κB-mediated proinflammatory response in DCs and suppress autoimmunity by serving as a key regulator of DC tolerance.


Subject(s)
Dendritic Cells/immunology , Lymphocyte Activation , Myasthenia Gravis, Autoimmune, Experimental/immunology , NFATC Transcription Factors/immunology , Signal Transduction/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/pathology , Immune Tolerance/genetics , Mice , Mice, Transgenic , Myasthenia Gravis, Autoimmune, Experimental/genetics , Myasthenia Gravis, Autoimmune, Experimental/pathology , NF-kappa B/genetics , NF-kappa B/immunology , NFATC Transcription Factors/genetics , Signal Transduction/genetics , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology
6.
J Cell Commun Signal ; 11(3): 265-273, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28321712

ABSTRACT

The human Ube2j1 and Ube2j2 are the only ubiquitin-conjugating enzymes (E2s) that are localized to endoplasmic reticulum (ER) through its C-terminal transmembrane domains. Ube2j1 is a known substrate of MAPK signalling pathway and it is phosphorylated at serine-184 during ER stress. Here, we demonstrate that Ube2j1, not Ube2j2 is essential for the recovery of cells from transient ER stress. The ectopic expression of wild-type Ube2j1 and phospho-mimic mutant, Ube2j1S184D but not phospho-mutant Ube2j1S184A can recover cells from ER stress. We also found that ubiquitin-ligase (E3), c-IAP1 preferentially interacts with phosphorylated Ube2j1. Moreover, we noticed that phosphorylated Ube2j1 is rapidly degraded by the proteasome during ER stress cell recovery. Taken together, these data suggest that Ube2j1 and its phosphorylation is important for transient ER stress cell recovery and the phosphorylated Ube2j1 is degraded by the proteasome.

7.
Mol Cell Biochem ; 425(1-2): 37-45, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27785701

ABSTRACT

The UBA-UBX domain-containing proteins can interact with ubiquitinated substrates and p97 during endoplasmic reticulum-associated degradation (ERAD). Here, we found that the expressions of all UBA-UBX genes p47, SAKS1, UBXD8, FAF1, and UBXD7 were elevated upon ER stress, albeit with different levels. Of which p47, SAKS1, and UBXD8 are 'immediate' respondents whereas FAF1 and UBXD7 were 'late' respondents to ER stress. Interestingly, the expression of specific UBA-UBX genes were altered in cells stably expressing three different ERAD substrates such as α-TCR, α1-antitrypsin, and δCD3. We first found that p47 and UBXD8 expression levels were increased in α-TCR and α1-antitrypsin stable cell lines, respectively, whereas SAKS1 expression level was reduced in all the three ERAD substrates tested. Of note, we also found p47 promotes, whereas SASK1 delays the degradation of a single ERAD substrate, α-TCR. Additionally, we found that SAKS1 selectively inhibits the degradation of ERAD substrates without affecting cytosolic proteasomal substrates. Taken together, our results identified that UBA-UBX proteins possess substrate selectivity and opposite role of two different UBA-UBX proteins in the degradation of a single ERAD substrate.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Endoplasmic Reticulum-Associated Degradation/physiology , Proteolysis , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Adaptor Proteins, Signal Transducing/genetics , Blood Proteins/genetics , Blood Proteins/metabolism , HeLa Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics
8.
Biochem Biophys Res Commun ; 469(4): 891-6, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26707646

ABSTRACT

Ubiquitination is a key regulatory mechanism in the immune deficiency (IMD) pathway in Drosophila. In this study, we first developed a simple immunoblot method to identify components involved in this pathway. Considering the emerging roles of ubiquitin-conjugating enzymes (E2s) in determining ubiquitin chain types and ubiquitination speed, we screened for E2s required for IMD activation. We found that UbcD4, in addition to the previously reported E2s Effete and Bendless, was required for activation of the IMD pathway. RNAi-mediated knockdown of the UbcD4 ortholog, E2-25K/Ube2K, inhibited TNFα- and LPS-mediated activation of the NF-κB pathway, implying that UbcD4 and E2-25K/Ube2K play a conserved role as positive regulators in both pathways.


Subject(s)
Drosophila Proteins/immunology , Drosophila/immunology , Signal Transduction/immunology , Ubiquitin-Conjugating Enzymes/immunology , Ubiquitination/immunology , Animals , Immunity, Innate/immunology
9.
Biol Reprod ; 93(3): 70, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26246218

ABSTRACT

The mammalian epididymis is a highly convoluted tubule that connects the testis to the vas deferens. Its proper functions in sperm transport, storage, and maturation are essential for male reproduction. One of the genes predominantly expressed in the epididymis is ADAM7 (a disintegrin and metalloprotease 7). Previous studies have shown that ADAM7 synthesized in the epididymis is secreted into the epididymal lumen and is then transferred to sperm membranes, where it forms a chaperone complex that is potentially involved in sperm fertility. In this study, we generated and analyzed mice with a targeted disruption in the Adam7 gene. We found that the fertility of male mice was modestly but significantly reduced by knockout of Adam7. Histological analyses revealed that the cell heights of the epithelium were dramatically decreased in the caput of the epididymis of Adam7-null mice, suggesting a requirement for ADAM7 in maintaining the integrity of the epididymal epithelium. We found that sperm from Adam7-null mice exhibit decreased motility, tail deformation, and altered tyrosine phosphorylation, indicating that the absence of ADAM7 leads to abnormal sperm functions and morphology. Western blot analyses revealed reduced levels of integral membrane protein 2B (ITM2B) and ADAM2 in sperm from Adam7-null mice, suggesting a requirement for ADAM7 in normal expression of sperm membrane proteins involved in sperm functions. Collectively, our study demonstrates for the first time that ADAM7 is required for normal fertility and is important for the maintenance of epididymal integrity and for sperm morphology, motility, and membrane proteins.


Subject(s)
ADAM Proteins/genetics , Epididymis/pathology , Infertility, Male/genetics , Infertility, Male/pathology , Membrane Proteins/genetics , Spermatozoa/pathology , Adaptor Proteins, Signal Transducing , Animals , Epithelial Cells/pathology , Epithelial Cells/ultrastructure , Female , Male , Mice , Mice, Knockout , Sperm Capacitation/genetics , Sperm Head/pathology , Sperm Motility/genetics , Sperm Tail/pathology
10.
Gene ; 565(1): 45-55, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25827285

ABSTRACT

The mammalian genome contains numerous genes encoding transcription factors that contain Krüppel-associated box (KRAB) and C2H2-type zinc finger (ZF) motifs (KRAB-ZF). In the present study, we identified KRAB-ZF genes expressed in the mouse testis or ovary, and selected three genes that exhibit gonad-specific or gonad-predominant expression. In vitro analyses showed that these gonadal KRAB-ZF proteins are localized in cell nuclei and are able to repress transcriptional activity. We further analyzed one of the gonad-specific reproductive genes, Zfp819, and found that it is expressed exclusively in spermatogenic cells. Overexpression of Zfp819 suppressed cell proliferation and induced apoptosis. Microarray analysis of Zfp819-overexpressing cells allowed us to identify numerous, potential target genes. A number of the down-regulated genes were found to show gene expression levels inversely correlated with Zfp819 during spermatogenesis. Some of the down-regulated genes were previously reported to play significant roles in spermatogenesis and apoptosis. Collectively, our study provides the first comprehensive information regarding the expression of reproductive KRAB-ZF genes in mice and reveals potential functions of Zfp819.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Gonads/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Animals , Cells, Cultured , DNA-Binding Proteins , Female , Gene Expression Profiling , Gene Expression Regulation , Gonads/cytology , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/genetics , Male , Mice , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Organ Specificity , Repressor Proteins/chemistry , Zinc Fingers
11.
Anal Biochem ; 443(2): 153-5, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24050967

ABSTRACT

Ubiquitin (Ub) is a posttranslational modifier, and total Ub (UbT) is always in dynamic equilibrium among free Ub (UbF), activated Ub (UbA), and conjugated Ub (UbC) in the forms of mono-Ub, thioester-bond-linked Ub, and peptide-bond-linked Ub, respectively. In this study, we developed a simple method to simultaneously determine the levels of UbT, UbF+UbA, and UbC in a single immunoblot and demonstrated its reliability and reproducibility by determining [UbT], [UbF+UbA], and [UbC] in various mouse tissues and cultured cells.


Subject(s)
Blotting, Western/methods , Ubiquitin/analysis , Animals , Cell Line , Humans , Mice , Peptides/analysis , Reproducibility of Results , Sulfides/analysis
12.
Sci Rep ; 3: 2623, 2013.
Article in English | MEDLINE | ID: mdl-24022007

ABSTRACT

Ubiquitin is involved in almost every cellular process, and it is also known to be a stress-inducible protein. Based on previous reports that many types of cancer display an elevated level of ubiquitin, we hypothesized that this increased amount of ubiquitin is essential for the growth of cancer cells and that, consequently, the downregulation of ubiquitin may be a potential anti-cancer treatment. We first found that the level of ubiquitin can be effectively downregulated via knockdown of a polyubiquitin gene, Ubb, with siRNA (Ubb-KD) and then demonstrated its anti-cancer effects in several cancer cell lines and xenograft mice. Ubb-KD resulted in the attenuation of TNFα-induced NF-κB activation, the stabilization of the tumor suppressor p53, and stress-sensitization. Taken together, downregulation of ubiquitin through Ubb-KD is a potential anti-cancer treatment by inhibiting ubiquitination at multiple sites related to oncogenic pathways and by weakening the ability of cancer cells to overcome increased stress.


Subject(s)
Gene Expression Regulation, Neoplastic , Polyubiquitin/genetics , Polyubiquitin/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Animals , Apoptosis/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Down-Regulation , Enzyme Activation/drug effects , Gene Knockdown Techniques , Humans , MCF-7 Cells , Male , Mice , NF-kappa B/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Proteolysis , RNA Interference , Tumor Burden/genetics , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitination , Xenograft Model Antitumor Assays
13.
Protein Expr Purif ; 86(1): 53-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22982229

ABSTRACT

Amyloid-ß peptide 1-42 (Aß(1-42)), the predominant form in senile plaques, plays important roles in the pathogenesis of Alzheimer's disease. Because Aß(1-42) has aggregation-prone nature, it has been difficult to produce in a soluble state in bacterial expression systems. In this study, we modified our expression system to increase the soluble fraction of Aß(1-42) in Escherichia coli (E. coli) cells. The expression level and solubility of recombinant Aß(1-42) induced at the low temperature (16°C) is highly increased compared to that induced at 37°C. To optimize expression temperature, the coding region of Aß(1-42) was constructed in a pCold vector, pCold-TF, which has a hexahistidine-tagged trigger factor (TF). Recombinant Aß(1-42) was expressed primarily as a soluble protein using pCold vector system and purified with a nickel-chelating resin. When the toxic effect of recombinant Aß(1-42) examined on human neuroblastoma SH-SY5Y cells, the purified Aß(1-42) induced cell toxicity on SH-SY5Y cells. In conclusion, the system developed in this study will provide a useful method for the production of aggregation prone-peptide such as Aß(1-42).


Subject(s)
Amyloid beta-Peptides/biosynthesis , Cold Temperature , Genetic Vectors/genetics , Peptide Fragments/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Amino Acid Sequence , Amyloid beta-Peptides/genetics , Cell Line, Tumor , Cell Survival , Chromatography, Affinity/methods , Escherichia coli/genetics , Humans , Neuroblastoma/pathology , Open Reading Frames , Peptide Fragments/genetics , Proteolysis , Recombinant Fusion Proteins/genetics , Solubility , Transformation, Genetic
14.
Bioorg Med Chem Lett ; 22(12): 4106-10, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22578456

ABSTRACT

Amyloid plaques, which are primarily composed of aggregated amyloid-beta (Aß) peptide, are the neuropathological hallmarks of Alzheimer's disease (AD). Fluorescent markers containing 2-styrylpyridazin-3(2H)-ones were developed to detect intracellular aggregated Aß peptides. Nine compounds exhibited a greater than 10-fold increase of in emission spectra before and after mixing with Aß aggregates compared with before mixing. Among these compounds, compound 9n exhibited the highest affinity for Aß aggregates (K(d)=1.84 µM) and selectively stained both aggregated intracellular Aß and Aß plaques in the transgenic AD model mice (APP/PS1). These preliminary results indicate that 2-styrylpyridazin-3(2H)-one derivatives are promising alternative fluorescence imaging agent for the study of AD.


Subject(s)
Alzheimer Disease/diagnosis , Amyloid beta-Peptides/analysis , Fluorescent Dyes , Plaque, Amyloid/chemistry , Pyridazines , Alzheimer Disease/pathology , Animals , Brain/pathology , Disease Models, Animal , Fluorescence , Fluorescent Dyes/chemistry , Humans , Mice , Mice, Transgenic , Pyridazines/chemistry , Sensitivity and Specificity , Spectrometry, Fluorescence , Staining and Labeling/methods
15.
J Mol Cell Cardiol ; 53(1): 53-63, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22449794

ABSTRACT

Protein kinase C (PKC)-interacting cousin of thioredoxin (PICOT) has distinct anti-hypertrophic and inotropic functions. We have previously shown that PICOT exerts its anti-hypertrophic effect by inhibiting calcineurin-NFAT signaling through its C-terminal glutaredoxin domain. However, the mechanism underlying the inotropic effect of PICOT is unknown. The results of protein pull-down experiments showed that PICOT directly binds to the catalytic domain of PKCζ through its N-terminal thioredoxin-like domain. Purified PICOT protein inhibited the kinase activity of PKCζ in vitro, which indicated that PICOT is an endogenous inhibitor of PKCζ. The inhibition of PKCζ activity with a PKCζ-specific pseudosubstrate peptide inhibitor was sufficient to increase the cardiac contractility in vitro and ex vivo. Overexpression of PICOT or inhibition of PKCζ activity down-regulated PKCα activity, which led to the elevation of sarcoplasmic reticulum Ca(2+)-ATPase (SERCA) 2a activity, concomitant with the increased phosphorylation of phospholamban (PLB). Overexpression of PICOT or inhibition of PKCζ activity also down-regulated protein phosphatase (PP) 2A activity, which subsequently resulted in the increased phosphorylation of troponin (Tn) I and T, key myofilament proteins associated with the regulation of contractility. PICOT appeared to inhibit PP2A activity through the disruption of the functional PKCζ/PP2A complex. In contrast to the overexpression of PICOT or inhibition of PKCζ, reduced PICOT expression resulted in up-regulation of PKCα and PP2A activities, followed by decreased phosphorylation of PLB, and TnI and T, respectively, supporting the physiological relevance of these events. Transgene- or adeno-associated virus (AAV)-mediated overexpression of PICOT restored the impaired contractility and prevented further morphological and functional deterioration of the failing hearts. Taken together, the results of the present study suggest that PICOT exerts its inotropic effect by negatively regulating PKCα and PP2A activities through the inhibition of PKCζ activity. This finding provides a novel insight into the regulation of cardiac contractility.


Subject(s)
Carrier Proteins/metabolism , Myocardial Contraction/physiology , Protein Kinase C/metabolism , Animals , Calcium-Binding Proteins/metabolism , Carrier Proteins/genetics , Cell Line , Enzyme Activation , Humans , Male , Mice , Models, Biological , Myocardial Contraction/drug effects , Myocardial Contraction/genetics , Peptide Fragments/pharmacology , Protein Binding , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-alpha/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Troponin I/metabolism , Troponin T/metabolism , tat Gene Products, Human Immunodeficiency Virus/pharmacology
16.
FEBS Lett ; 585(24): 3959-63, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22079664

ABSTRACT

In this study using non-reduced/reduced 2-dimensional electrophoresis (NR/R-2DE), we clearly demonstrated that E3-independent ubiquitination by Ube2K produced not only unanchored but also Ube2K-linked polyubiquitins through thioester and isopeptide bonds. E3-independent assembly of polyubiquitins on the catalytic cysteine of Ube2K strongly supports the possibility of 'en bloc transfer' for polyubiquitination. From the same analyses of E3-independent ubiquitination products by other E2s, we also found that different lengths of polyubiquitins were linked to different E2s through thioester bond; longer chains by Cdc34 like Ube2K, short chains by Ube2g2, and mono-ubiquitin by UbcH10. Our results suggest that E2s possess the different intrinsic catalytic activities for polyubiquitination.


Subject(s)
Biocatalysis , Electrophoresis, Gel, Two-Dimensional/methods , Polyubiquitin/isolation & purification , Polyubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Catalytic Domain , Cysteine , Esters
17.
PLoS One ; 5(12): e15645, 2010 Dec 20.
Article in English | MEDLINE | ID: mdl-21187975

ABSTRACT

BACKGROUND: The nuclear inclusion a (NIa) protease of turnip mosaic virus (TuMV) is responsible for the processing of the viral polyprotein into functional proteins. NIa was previously shown to possess a relatively strict substrate specificity with a preference for Val-Xaa-His-Gln↓, with the scissile bond located after Gln. The presence of the same consensus sequence, Val(12)-His-His-Gln(15), near the presumptive α-secretase cleavage site of the amyloid-ß (Aß) peptide led us to hypothesize that NIa could possess activity against Aß. METHODOLOGY/PRINCIPAL FINDINGS: Western blotting results showed that oligomeric as well as monomeric forms of Aß can be degraded by NIa in vitro. The specific cleavage of Aß was further confirmed by mass spectrometry analysis. NIa was shown to exist predominantly in the cytoplasm as observed by immunofluorescence microscopy. The overexpression of NIa in B103 neuroblastoma cells resulted in a significant reduction in cell death caused by both intracellularly generated and exogenously added Aß. Moreover, lentiviral-mediated expression of NIa in APP(sw)/PS1 transgenic mice significantly reduced the levels of Aß and plaques in the brain. CONCLUSIONS/SIGNIFICANCE: These results indicate that the degradation of Aß in the cytoplasm could be a novel strategy to control the levels of Aß, plaque formation, and the associated cell death.


Subject(s)
Amyloid beta-Peptides/metabolism , Brassica napus/virology , Endopeptidases/chemistry , Gene Expression Regulation, Enzymologic , Mosaic Viruses/enzymology , Viral Proteins/chemistry , Amino Acid Sequence , Amyloid beta-Peptides/chemistry , Animals , Cell Line, Tumor , Cell Survival , Cytoplasm/metabolism , Disease Progression , Mice , Mice, Transgenic , Molecular Sequence Data , Mosaic Viruses/genetics , Neurons/cytology , Rats , Subcellular Fractions/metabolism , Substrate Specificity
18.
J Biol Chem ; 285(46): 36070-80, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20826778

ABSTRACT

E2-25K/Hip2 is an unusual ubiquitin-conjugating enzyme that interacts with the frameshift mutant of ubiquitin B (UBB(+1)) and has been identified as a crucial factor regulating amyloid-ß neurotoxicity. To study the structural basis of the neurotoxicity mediated by the E2-25K-UBB(+1) interaction, we determined the three-dimensional structures of UBB(+1), E2-25K and the E2-25K/ubiquitin, and E2-25K/UBB(+1) complex. The structures revealed that ubiquitin or UBB(+1) is bound to E2-25K via the enzyme MGF motif and residues in α9 of the enzyme. Polyubiquitylation assays together with analyses of various E2-25K mutants showed that disrupting UBB(+1) binding markedly diminishes synthesis of neurotoxic UBB(+1)-anchored polyubiquitin. These results suggest that the interaction between E2-25K and UBB(+1) is critical for the synthesis and accumulation of UBB(+1)-anchored polyubiquitin, which results in proteasomal inhibition and neuronal cell death.


Subject(s)
Multiprotein Complexes/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Cell Line, Tumor , Cell Survival , Crystallography, X-Ray , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Hydrogen Bonding , Magnetic Resonance Spectroscopy , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes/chemistry , Mutation , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Ubiquitin/chemistry , Ubiquitin/genetics , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/genetics
19.
Biochem Biophys Res Commun ; 396(2): 425-8, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20417181

ABSTRACT

It is well known that S5a and hRpn13 are two major ubiquitin (Ub) receptors in the proteasome but little is known about their functional difference in recruiting ubiquitinated substrates. In this study using siRNA-mediated knockdown of S5a or hRpn13, we found that two Ub receptors had different substrate specificity although similar level of accumulation of high molecular weight Ub-conjugates was observed. Interesting enough, depletion of S5a, but not hRpn13, resulted in the Ub-containing aggregates and induced ER chaperones such as Grp78 and Grp94. ERAD substrates such as alpha-TCR and alpha1-antitrypsin were also stabilized by the depletion of S5a but not hRpn13. Our results suggest that there is different substrate specificity between S5a and hRpn13 at the level of delivery and S5a may be the major docking site for ERAD substrates.


Subject(s)
Membrane Glycoproteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Gene Knockdown Techniques , HSP70 Heat-Shock Proteins/metabolism , HeLa Cells , Heat-Shock Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Proteasome Endopeptidase Complex/genetics , Protein Stability , RNA, Small Interfering/genetics , RNA-Binding Proteins , Substrate Specificity
20.
Biochem Biophys Res Commun ; 388(4): 723-6, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19695230

ABSTRACT

It has been demonstrated that ubiquitin-conjugated proteins were accumulated by ectopically-expressed S5a as well as the ubiquitin-interacting motifs of S5a (S5a-UIMs). In this study, we further found that free S5a-UIMs stabilized only a subset of proteasomal substrates including p53, c-Fos, c-Jun, and p27 but not beta-catenin, p15, and ornithine decarboxylase. Both S5a-UIMs and epoxomicin inhibited the proliferation of A549 lung cancer cells but arrest at the different stages of cell cycle. Together, our results suggest a potential role of S5a-UIMs as an upstream proteasomal inhibitor by blocking the subset of substrates from delivery to the 26S proteasome.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Ubiquitin/metabolism , Amino Acid Motifs/genetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Humans , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RNA-Binding Proteins , Tumor Suppressor Protein p53/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...