Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cancer Res Commun ; 3(5): 860-873, 2023 05.
Article in English | MEDLINE | ID: mdl-37377896

ABSTRACT

Immune checkpoint blockade therapy, one of the most promising cancer immunotherapies, has shown remarkable clinical impact in multiple cancer types. Despite the recent success of immune checkpoint blockade therapy, however, the response rates in patients with cancer are limited (∼20%-40%). To improve the success of immune checkpoint blockade therapy, relevant preclinical animal models are essential for the development and testing of multiple combination approaches and strategies. Companion dogs naturally develop several types of cancer that in many respects resemble clinical cancer in human patients. Therefore, the canine studies of immuno-oncology drugs can generate knowledge that informs and prioritizes new immuno-oncology therapy in humans. The challenge has been, however, that immunotherapeutic antibodies targeting canine immune checkpoint molecules such as canine PD-L1 (cPD-L1) have not been commercially available. Here, we developed a new cPD-L1 antibody as an immuno-oncology drug and characterized its functional and biological properties in multiple assays. We also evaluated the therapeutic efficacy of cPD-L1 antibodies in our unique caninized PD-L1 mice. Together, these in vitro and in vivo data, which include an initial safety profile in laboratory dogs, support development of this cPD-L1 antibody as an immune checkpoint inhibitor for studies in dogs with naturally occurring cancer for translational research. Our new therapeutic antibody and caninized PD-L1 mouse model will be essential translational research tools in raising the success rate of immunotherapy in both dogs and humans. Significance: Our cPD-L1 antibody and unique caninized mouse model will be critical research tools to improve the efficacy of immune checkpoint blockade therapy in both dogs and humans. Furthermore, these tools will open new perspectives for immunotherapy applications in cancer as well as other autoimmune diseases that could benefit a diverse and broader patient population.


Subject(s)
Neoplasms , Translational Research, Biomedical , Humans , Dogs , Animals , Mice , B7-H1 Antigen , Immune Checkpoint Inhibitors/pharmacology , Neoplasms/drug therapy , Immunotherapy , Antibodies
2.
Cells ; 11(18)2022 09 08.
Article in English | MEDLINE | ID: mdl-36139389

ABSTRACT

CtBP-interacting protein (CtIP) plays a critical role in controlling the homologous recombination-mediated DNA double-stranded break (DSB) repair pathway through DNA end resection, and recent studies suggest that it also plays a role in mitosis. However, the mechanism by which CtIP contributes to mitosis regulation remains elusive. Here, we show that depletion of CtIP leads to a delay in anaphase progression resulting in misaligned chromosomes, an aberrant number of centrosomes, and defects in chromosome segregation. Additionally, we demonstrate that CtIP binds and colocalizes with Targeting protein for Xklp2 (TPX2) during mitosis to regulate the recruitment of TPX2 to the spindle poles. Furthermore, depletion of CtIP resulted in both a lower concentration of Aurora A, its downstream target, and very low microtubule intensity at the spindle poles, suggesting an important role for the CtIP-TPX2-Auroa A complex in microtubule dynamics at the centrosomal spindles. Our findings reveal a novel function of CtIP in regulating spindle dynamics through interactions with TPX2 and indicate that CtIP is involved in the proper execution of the mitotic program, where deregulation may lead to chromosomal instability.


Subject(s)
Nuclear Proteins , Spindle Apparatus , DNA/metabolism , Microtubules/metabolism , Mitosis , Nuclear Proteins/metabolism , Spindle Apparatus/metabolism
4.
Cell Signal ; 80: 109926, 2021 04.
Article in English | MEDLINE | ID: mdl-33465404

ABSTRACT

The Neuroepithelial transforming gene 1 (Net1) is a RhoA subfamily guanine nucleotide exchange factor that is overexpressed in a number of cancers and contributes to cancer cell motility and proliferation. Net1 also plays a Rho GTPase independent role in mitotic progression, where it promotes centrosomal activation of Aurora A and Pak2, and aids in chromosome alignment during prometaphase. To understand regulatory mechanisms controlling the mitotic function of Net1, we examined whether it was phosphorylated by the mitotic kinase Cdk1. We observed that Cdk1 phosphorylated Net1 on multiple sites in its N-terminal regulatory domain and C-terminus in vitro. By raising phospho-specific antibodies to two of these sites, we also demonstrated that both endogenous and transfected Net1 were phosphorylated by Cdk1 in cells. Substitution of the major Cdk1 phosphorylation sites with aliphatic or acidic residues inhibited the interaction of Net1 with RhoA, and treatment of metaphase cells with a Cdk1 inhibitor increased Net1 activity. Cdk1 inhibition also increased Net1 localization to the plasma membrane and stimulated cortical F-actin accumulation. Moreover, Net1 overexpression caused spindle polarity defects that were reduced in frequency by acidic substitution of the major Cdk1 phosphorylation sites. These data indicate that Cdk1 phosphorylates Net1 during mitosis and suggest that this negatively regulates its ability to signal to RhoA and alter actin cytoskeletal organization.


Subject(s)
CDC2 Protein Kinase/metabolism , Mitosis , Oncogene Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Actin Cytoskeleton , Actins/metabolism , CDC2 Protein Kinase/antagonists & inhibitors , CDC2 Protein Kinase/genetics , Cell Membrane/metabolism , HeLa Cells , Humans , Mutagenesis, Site-Directed , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/genetics , Phosphorylation , Protein Stability , RNA Interference , RNA, Small Interfering/metabolism , Spindle Apparatus/physiology , rhoA GTP-Binding Protein/genetics
5.
Mol Carcinog ; 59(7): 691-700, 2020 07.
Article in English | MEDLINE | ID: mdl-32115801

ABSTRACT

Triple-negative breast cancer (TNBC) lacks a well-defined molecular target and is associated with poorer outcomes compared to other breast cancer subtypes. Programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) blockade therapy shows a 10% to 20% response rate in TNBC patients. Our previous studies show that PD-L1 proteins are heavily glycosylated in TNBC, and the glycosylation plays an important role in the PD-L1 protein's stability and immunosuppressive function. However, a strategy for PD-L1 deglycosylation in TNBC is poorly defined. Here we found that a saccharide analog, 2-deoxy- d-glucose (2-DG), inhibits glycosylation of PD-L1 and its immunosuppressive function by combining with EGFR inhibitor, gefitinib. Interestingly, 2-DG/gefitinib-induced deglycosylation of PD-L1 decreased the expression level of PD-L1 protein as well as its binding with PD-1. However, there was no significant decrease in 4-1BB expression and its binding with 4-1BBL by 2-DG/gefitinib. Furthermore, we demonstrated that the combination treatment of 2-DG/gefitinib and 4-1BB antibody enhances antitumor immunity in TNBC syngeneic murine models. Together, our results suggest a new immunotherapeutic strategy to enhance antitumor immunity by PD-L1 deglycosylation and 4-1BB stimulation in TNBC.


Subject(s)
Antineoplastic Agents/pharmacology , B7-H1 Antigen/metabolism , Deoxyglucose/pharmacology , Glucose/pharmacology , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/therapy , Animals , Antibodies/pharmacology , Cell Line , Cell Line, Tumor , Female , Gefitinib/pharmacology , HEK293 Cells , Humans , Immunotherapy/methods , Mice , Mice, Inbred BALB C
6.
J Cell Sci ; 131(3)2018 02 01.
Article in English | MEDLINE | ID: mdl-29361525

ABSTRACT

The neuroepithelial cell transforming gene 1A (Net1A, an isoform of Net1) is a RhoA subfamily guanine nucleotide exchange factor (GEF) that localizes to the nucleus in the absence of stimulation, preventing it from activating RhoA. Once relocalized in the cytosol, Net1A stimulates cell motility and extracellular matrix invasion. In the present work, we investigated mechanisms responsible for the cytosolic relocalization of Net1A. We demonstrate that inhibition of MAPK pathways blocks Net1A relocalization, with cells being most sensitive to JNK pathway inhibition. Moreover, activation of the JNK or p38 MAPK family pathway is sufficient to elicit Net1A cytosolic localization. Net1A relocalization stimulated by EGF or JNK activation requires nuclear export mediated by CRM1. JNK1 (also known as MAPK8) phosphorylates Net1A on serine 52, and alanine substitution at this site prevents Net1A relocalization caused by EGF or JNK activation. Glutamic acid substitution at this site is sufficient for Net1A relocalization and results in elevated RhoA signaling to stimulate myosin light chain 2 (MLC2, also known as MYL2) phosphorylation and F-actin accumulation. Net1A S52E expression stimulates cell motility, enables Matrigel invasion and promotes invadopodia formation. These data highlight a novel mechanism for controlling the subcellular localization of Net1A to regulate RhoA activation, cell motility, and invasion.


Subject(s)
Cell Movement , Karyopherins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Oncogene Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Stress, Physiological , Cell Movement/drug effects , Cytosol/drug effects , Cytosol/metabolism , Epidermal Growth Factor/pharmacology , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Glutamic Acid/metabolism , Humans , MCF-7 Cells , Mutation/genetics , Phosphorylation/drug effects , Phosphoserine/metabolism , Podosomes/drug effects , Podosomes/metabolism , Protein Transport/drug effects , Signal Transduction , Stress, Physiological/drug effects , Tumor Necrosis Factor-alpha/pharmacology , rhoA GTP-Binding Protein/metabolism , Exportin 1 Protein
7.
BMB Rep ; 50(7): 373-378, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28539160

ABSTRACT

The Jun activation-domain binding protein 1 (Jab1) induces p53 nuclear export and cytoplasmic degradation, but the underlying mechanism is poorly understood. Here, we show that phosphorylation at the threonine 155 residue is essential for Jab1-mediated p53 nuclear export. Jab1 stimulated phosphorylation of p53 at T155 was inhibited by curcumin, an inhibitor of COP9 signalosome (CSN)-associated kinases. The T155E mutant, which mimics phosphorylated p53, exhibited spontaneous cytoplasmic localization in the absence of Jab1. This process was prevented by leptinomycin B (LMB), but not by curcumin. The substitution of threonine 155 for valine (T155V) abrogated Jab1-mediated p53 nuclear export, indicating that phosphorylation at this site is essential for Jab1-mediated regulation of p53. Although T155E can be localized in the cytoplasm in the absence of Mdm2, the translocation of T155E was significantly enhanced by ectopic Hdm2 expression. Our data suggests that Jab1-mediated phosphorylation of p53 at Thr155 residue mediates nuclear export of p53. [BMB Reports 2017; 50(7): 373-378].


Subject(s)
COP9 Signalosome Complex/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Peptide Hydrolases/metabolism , Threonine/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Humans , Phosphorylation , Tumor Cells, Cultured
8.
Nucleic Acids Res ; 44(8): 3659-74, 2016 05 05.
Article in English | MEDLINE | ID: mdl-26762983

ABSTRACT

Trimethylated histone H3 lysine 27 (H3K27me3) is linked to gene silencing, whereas H3K4me3 is associated with gene activation. These two marks frequently co-occupy gene promoters, forming bivalent domains. Bivalency signifies repressed but activatable states of gene expression and can be resolved to active, H3K4me3-prevalent states during multiple cellular processes, including differentiation, development and epithelial mesenchymal transition. However, the molecular mechanism underlying bivalency resolution remains largely unknown. Here, we show that the H3K27 demethylase UTX (also called KDM6A) is required for the resolution and activation of numerous retinoic acid (RA)-inducible bivalent genes during the RA-driven differentiation of mouse embryonic stem cells (ESCs). Notably, UTX loss in mouse ESCs inhibited the RA-driven bivalency resolution and activation of most developmentally critical homeobox (Hox) a-d genes. The UTX-mediated resolution and activation of many bivalent Hox genes during mouse ESC differentiation were recapitulated during RA-driven differentiation of human NT2/D1 embryonal carcinoma cells. In support of the importance of UTX in bivalency resolution, Utx-null mouse ESCs and UTX-depleted NT2/D1 cells displayed defects in RA-driven cellular differentiation. Our results define UTX as a bivalency-resolving histone modifier necessary for stem cell differentiation.


Subject(s)
Cell Differentiation/genetics , Histone Demethylases/physiology , Nuclear Proteins/physiology , Promoter Regions, Genetic , Transcriptional Activation , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Genes, Homeobox , Histone Demethylases/metabolism , Humans , Mice , Nuclear Proteins/metabolism , Tretinoin/pharmacology
9.
Mol Endocrinol ; 30(3): 278-89, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26677753

ABSTRACT

Ras homolog (Rho) family small GTPases are critical regulators of actin cytoskeletal organization, cell motility, proliferation, and survival. Surprisingly, the large majority of the studies underlying our knowledge of Rho protein function have been carried out in cultured cells, and it is only recently that researchers have begun to assess Rho GTPase regulation and function in vivo. The purpose of this review is to evaluate our current knowledge of Rho GTPase function in mouse mammary gland development, tumorigenesis and metastasis. Although our knowledge is still incomplete, these studies are already uncovering important themes as to the physiological roles of Rho GTPase signaling in normal mammary gland development and function. Essential contributions of Rho proteins to breast cancer initiation, tumor progression, and metastatic dissemination have also been identified.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/pathology , rho GTP-Binding Proteins/metabolism , Animals , Disease Models, Animal , Female , Mice , Neoplasm Metastasis
10.
J Cell Sci ; 128(5): 913-22, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25588829

ABSTRACT

Net1 isoform A (Net1A) is a RhoA GEF that is required for cell motility and invasion in multiple cancers. Nuclear localization of Net1A negatively regulates its activity, and we have recently shown that Rac1 stimulates Net1A relocalization to the plasma membrane to promote RhoA activation and cytoskeletal reorganization. However, mechanisms controlling the subcellular localization of Net1A are not well understood. Here, we show that Net1A contains two nuclear localization signal (NLS) sequences within its N-terminus and that residues surrounding the second NLS sequence are acetylated. Treatment of cells with deacetylase inhibitors or expression of active Rac1 promotes Net1A acetylation. Deacetylase inhibition is sufficient for Net1A relocalization outside the nucleus, and replacement of the N-terminal acetylation sites with arginine residues prevents cytoplasmic accumulation of Net1A caused by deacetylase inhibition or EGF stimulation. By contrast, replacement of these sites with glutamine residues is sufficient for Net1A relocalization, RhoA activation and downstream signaling. Moreover, the N-terminal acetylation sites are required for rescue of F-actin accumulation and focal adhesion maturation in Net1 knockout MEFs. These data indicate that Net1A acetylation regulates its subcellular localization to impact on RhoA activity and actin cytoskeletal organization.


Subject(s)
Cell Membrane/metabolism , Cell Nucleus/metabolism , Cytoskeleton/metabolism , Oncogene Proteins/metabolism , Acetylation , Active Transport, Cell Nucleus/physiology , Animals , Cell Membrane/genetics , Cell Nucleus/genetics , Cytoskeleton/genetics , HeLa Cells , Humans , Mice , Mice, Knockout , Neuropeptides/genetics , Neuropeptides/metabolism , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Oncogene Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
11.
Cell Cycle ; 13(17): 2765-72, 2014.
Article in English | MEDLINE | ID: mdl-25486363

ABSTRACT

ATM activation following DNA damage is a critical event which is required for efficient DNA repair and cell survival, yet signaling mechanisms controlling its activation are incompletely understood. The RhoGEF Net1 has previously been reported to control Rho GTPase activation and downstream cell survival outcomes following double strand DNA damage. However the role of Net1 isoforms in controlling ATM-dependent cell signaling has not been assessed. In the present work we show that expression of the Net1A isoform is specifically required for efficient activation of ATM but not the related kinase DNA-PK after ionizing radiation. Surprisingly Net1A overexpression also potently suppresses ATM activation and phosphorylation of its substrate H2AX. This effect does not require catalytic activity towards RhoA or RhoB, and neither Rho GTPase affects ATM activation, on its own. Consistent with a role in controlling ATM activation, Net1A knockdown also impairs DNA repair and cell survival. Taken together these data indicate that Net1A plays a plays a previously unrecognized, Rho GTPase-independent role in controlling ATM activity and downstream signaling after DNA damage to impact cell survival.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Oncogene Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , rhoA GTP-Binding Protein/metabolism , rhoB GTP-Binding Protein/metabolism , Cell Survival/radiation effects , DNA Breaks, Double-Stranded , DNA Damage , Histones/metabolism , Humans , MCF-7 Cells , Protein Isoforms/metabolism , Radiation, Ionizing , Signal Transduction/radiation effects
12.
Cell Signal ; 26(12): 2998-3006, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25286227

ABSTRACT

Animal cell division is a fundamental process that requires complex changes in cytoskeletal organization and function. Aberrant cell division often has disastrous consequences for the cell and can lead to cell senescence, neoplastic transformation or death. As important regulators of the actin cytoskeleton, Rho GTPases play major roles in regulating many aspects of mitosis and cytokinesis. These include centrosome duplication and separation, generation of cortical rigidity, microtubule-kinetochore stabilization, cleavage furrow formation, contractile ring formation and constriction, and abscission. The ability of Rho proteins to function as regulators of cell division depends on their ability to cycle between their active, GTP-bound and inactive, GDP-bound states. However, Rho proteins are inherently inefficient at fulfilling this cycle and require the actions of regulatory proteins that enhance GTP binding (RhoGEFs), stimulate GTPase activity (RhoGAPs), and sequester inactive Rho proteins in the cytosol (RhoGDIs). The roles of these regulatory proteins in controlling cell division are an area of active investigation. In this review we will delineate the current state of knowledge of how specific RhoGEFs, RhoGAPs and RhoGDIs control mitosis and cytokinesis, and highlight the mechanisms by which their functions are controlled.


Subject(s)
Cells/cytology , Cells/enzymology , Mitosis , rho GTP-Binding Proteins/metabolism , Animals , Cytokinesis , Humans , Models, Biological
13.
Mol Biol Cell ; 24(17): 2655-67, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23864709

ABSTRACT

Neuroepithelial transforming gene 1 (Net1) is a RhoA-subfamily-specific guanine nucleotide exchange factor that is overexpressed in multiple human cancers and is required for proliferation. Molecular mechanisms underlying its role in cell proliferation are unknown. Here we show that overexpression or knockdown of Net1 causes mitotic defects. Net1 is required for chromosome congression during metaphase and generation of stable kinetochore microtubule attachments. Accordingly, inhibition of Net1 expression results in spindle assembly checkpoint activation. The ability of Net1 to control mitosis is independent of RhoA or RhoB activation, as knockdown of either GTPase does not phenocopy effects of Net1 knockdown on nuclear morphology, and effects of Net1 knockdown are effectively rescued by expression of catalytically inactive Net1. We also observe that Net1 expression is required for centrosomal activation of p21-activated kinase and its downstream kinase Aurora A, which are critical regulators of centrosome maturation and spindle assembly. These results identify Net1 as a novel regulator of mitosis and indicate that altered expression of Net1, as occurs in human cancers, may adversely affect genomic stability.


Subject(s)
Microtubules/chemistry , Mitosis/physiology , Oncogene Proteins/metabolism , Aurora Kinase A/metabolism , Cell Line, Tumor , Chromosome Segregation , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Genomic Instability , HeLa Cells , Humans , Microtubules/metabolism , Mitosis/genetics , Oncogene Proteins/genetics , p21-Activated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , rhoB GTP-Binding Protein/metabolism
14.
Mol Cell Biol ; 33(14): 2773-86, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23689132

ABSTRACT

Net1 is a RhoA guanine nucleotide exchange factor (GEF) that is overexpressed in a subset of human cancers and contributes to cancer cell motility and invasion in vitro. However, the molecular mechanism accounting for its role in cell motility and invasion has not been described. In the present work, we show that expression of both Net1 isoforms in breast cancer cells is required for efficient cell motility. Although loss of Net1 isoform expression only partially blocks RhoA activation, it inhibits lysophosphatidic acid (LPA)-stimulated migration as efficiently as knockdown of RhoA itself. However, we demonstrate that the Net1A isoform predominantly controls myosin light-chain phosphorylation and is required for trailing edge retraction during migration. Net1A interacts with focal adhesion kinase (FAK), localizes to focal adhesions, and is necessary for FAK activation and focal adhesion maturation during cell spreading. Net1A expression is also required for efficient invasion through a Matrigel matrix. Analysis of invading cells demonstrates that Net1A is required for amoeboid invasion, and loss of Net1A expression causes cells to shift to a mesenchymal phenotype characterized by high ß1-integrin activity and membrane type 1 matrix metalloproteinase (MT1-MMP) expression. These results demonstrate a previously unrecognized role for the Net1A isoform in controlling FAK activation during planar cell movement and amoeboid motility during extracellular matrix (ECM) invasion.


Subject(s)
Breast Neoplasms/enzymology , Focal Adhesion Kinase 1/metabolism , Oncogene Proteins/physiology , Breast Neoplasms/pathology , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Shape , Chemotaxis , Enzyme Activation , Extracellular Matrix/metabolism , Female , Focal Adhesions/metabolism , Guanine Nucleotide Exchange Factors/physiology , Humans , Lysophospholipids/physiology , Myosin Light Chains/metabolism , Neoplasm Invasiveness , Phosphorylation , Protein Binding , Protein Isoforms/physiology , Protein Processing, Post-Translational , Protein Transport , rhoA GTP-Binding Protein/metabolism
15.
J Virol ; 84(1): 426-36, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19846531

ABSTRACT

West Nile virus capsid protein (WNVCp) displays pathogenic toxicity via the apoptotic pathway. However, a cellular mechanism protective against this toxic effect has not been observed so far. Here, we identified Makorin ring finger protein 1 (MKRN1) as a novel E3 ubiquitin ligase for WNVCp. The cytotoxic effects of WNVCp as well as its expression levels were inhibited in U2OS cells that stably expressed MKRN1. Immunoprecipitation analyses revealed an interaction between MKRN1 and WNVCp. Domain analysis indicated that the C terminus of MKRN1 and the N terminus of WNVCp were required for the interaction. MKRN1 could induce WNVCp ubiquitination and degradation in a proteasome-dependent manner. Interestingly, the WNVCp mutant with amino acids 1 to 105 deleted WNVCp was degraded by MKRN1, whereas the mutant with amino acids 1 to 90 deleted was not. When three lysine sites at positions 101, 103, and 104 of WNVCp were replaced with alanine, MKRN1-mediated ubiquitination and degradation of the mutant were significantly inhibited, suggesting that these sites are required for the ubiquitination. Finally, U2OS cell lines stably expressing MKRN1 were resistant to cytotoxic effects of WNV. In contrast, cells depleted of MKRN1 were more susceptible to WNVCp cytotoxicity. Confirming this, overexpression of MKRN1 significantly reduced, but depletion of MKRN1 increased, WNV proliferation in 293T cells. Taken together, our results suggest that MKRN1 can protect cells from WNV by inducing WNVCp degradation.


Subject(s)
Capsid Proteins/metabolism , Nerve Tissue Proteins/metabolism , Ribonucleoproteins/metabolism , Ubiquitin-Protein Ligases/metabolism , West Nile virus/pathogenicity , Amino Acid Substitution , Binding Sites , Capsid Proteins/physiology , Cell Line, Tumor , Humans , Lysine , Nerve Tissue Proteins/genetics , Proteasome Endopeptidase Complex , Ribonucleoproteins/genetics , Ubiquitination , West Nile virus/chemistry
16.
J Cell Sci ; 122(Pt 15): 2613-22, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19567472

ABSTRACT

Maintaining proper telomere length requires the presence of the telomerase enzyme. Here we show that telomerase reverse transcriptase (TERT), a catalytic component of telomerase, is recruited to promyelocytic leukemia (PML) nuclear bodies through its interaction with PML-IV. Treatment of interferon-alpha (IFNalpha) in H1299 cells resulted in the increase of PML proteins with a concurrent decrease of telomerase activity, as previously reported. PML depletion, however, stimulated telomerase activity that had been inhibited by IFNalpha with no changes in TERT mRNA levels. Upon treatment with IFNalpha, exogenous TERT localized to PML nuclear bodies and binding between TERT and PML increased. Immunoprecipitation and immunofluorescence analyses showed that TERT specifically bound to PML-IV. Residues 553-633 of the C-terminal region of PML-IV were required for its interaction with the TERT region spanning residues 1-350 and 595-946. The expression of PML-IV and its deletion mutant, 553-633, suppressed intrinsic telomerase activity in H1299. TERT-mediated immunoprecipitation of PML or the 553-633 fragment demonstrated that these interactions inhibited telomerase activity. H1299 cell lines stably expressing PML-IV displayed decreased telomerase activity with no change of TERT mRNA levels. Accordingly, telomere length of PML-IV stable cell lines was shortened. These results indicate that PML-IV is a negative regulator of telomerase in the post-translational state.


Subject(s)
Liver Neoplasms/metabolism , Lung Neoplasms/metabolism , Nuclear Proteins/physiology , Telomerase/metabolism , Telomere/metabolism , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Down-Regulation , Fibroblasts/metabolism , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunoenzyme Techniques , Immunoprecipitation , Interferon-alpha/pharmacology , Liver Neoplasms/pathology , Lung Neoplasms/pathology , Promyelocytic Leukemia Protein , RNA, Messenger/metabolism , Telomerase/genetics , Tumor Cells, Cultured
17.
EMBO J ; 28(14): 2100-13, 2009 Jul 22.
Article in English | MEDLINE | ID: mdl-19536131

ABSTRACT

Makorin Ring Finger Protein 1 (MKRN1) is a transcriptional co-regulator and an E3 ligase. Here, we show that MKRN1 simultaneously functions as a differentially negative regulator of p53 and p21. In normal conditions, MKRN1 could destabilize both p53 and p21 through ubiquitination and proteasome-dependent degradation. As a result, depletion of MKRN1 induced growth arrest through activation of p53 and p21. Interestingly, MKRN1 used earlier unknown sites, K291 and K292, for p53 ubiquitination and subsequent degradation. Under severe stress conditions, however, MKRN1 primarily induced the efficient degradation of p21. This regulatory process contributed to the acceleration of DNA damage-induced apoptosis by eliminating p21. MKRN1 depletion diminished adriamycin or ultraviolet-induced cell death, whereas ectopic expression of MKRN1 facilitated apoptosis. Furthermore, MKRN1 stable cell lines that constantly produced low levels of p53 and p21 exhibited stabilization of p53, but not p21, with increased cell death on DNA damage. Our results indicate that MKRN1 exhibits dual functions of keeping cells alive by suppressing p53 under normal conditions and stimulating cell death by repressing p21 under stress conditions.


Subject(s)
Apoptosis , Cell Cycle , Nerve Tissue Proteins/metabolism , Ribonucleoproteins/metabolism , Tumor Suppressor Protein p53/metabolism , rho GTP-Binding Proteins/metabolism , Animals , DNA Damage , Gene Knockdown Techniques , Humans , Mice , Nerve Tissue Proteins/genetics , Proteasome Endopeptidase Complex , Ribonucleoproteins/genetics , Ubiquitination
18.
Cell Microbiol ; 10(1): 165-76, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17697133

ABSTRACT

The capsid protein of the West Nile virus (WNV) functions as an apoptotic agonist via the induction of mitochondrial dysfunction and the activation of caspases-9 and -3. Here, we have determined that the WNV capsid (WNVCp) is capable of binding to and sequestering HDM2 into the nucleolus. WNVCp was shown to interfere with the formation of the HDM2 and p53 complex, thereby causing the stabilization of p53 and the subsequent induction of its target apoptotic protein, Bax. Whereas WNVCp was capable of inducing the p53-dependent apoptotic process in wild-type mouse embryonic fibroblasts (MEF) or SH-SY5Y cells, it exerted no significant effects on p53-null MEF or on p53-knockdown SH-SY5Y cells. This suggests that WNVCp-mediated apoptosis requires p53. Furthermore, when WNV was transfected into cells, endogenous Hdm2 and WNVCp were able to interact physically. WNVCp expressed in wild-type MEF proved able to induce the translocation of the endogenous Hdm2 into the nucleolus. Consistently, WNV was highly pathogenic in the presence of p53, and was less so in the absence of p53. The results of these studies suggest that the apoptotic mechanism mediated by WNV might occur in accordance in a fashion similar to that of the tumour-suppressing mechanism mediated by ARF.


Subject(s)
Apoptosis , Capsid Proteins/metabolism , Cell Nucleolus/chemistry , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , West Nile virus/physiology , Animals , Cell Line , Cells, Cultured , Chlorocebus aethiops , Fibroblasts/virology , Humans , Mice , Protein Binding , bcl-2-Associated X Protein/biosynthesis
19.
Mol Cells ; 22(2): 133-40, 2006 Oct 31.
Article in English | MEDLINE | ID: mdl-17085963

ABSTRACT

Jun activation domain-binding protein 1 (Jab1) is involved in various cellular mechanisms including development in Drosophila and mouse, cell cycle control and signal transduction pathways. Recent studies also determined that Jab1 functions as a nuclear exporter and inducer of cytoplasmic degradation for several proteins including p53, p27, capsid of West Nile virus, and Smad4/7 proteins. In particular, p53 is shown to bind to and to be exported into the cytoplasm by Jab1, which helps to maintain low levels of p53 under normal conditions. This review was undertaken in an effort to understand the biological significance of the homeostasis of p53 as maintained in the presence of Jab1. Based on our observations, we have provided potential mechanistic hypotheses for the nuclear export of p53 in coordination with Jab1 and the role of other factors in these processes.


Subject(s)
Cell Nucleus/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Peptide Hydrolases/physiology , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus , Animals , COP9 Signalosome Complex , Cytoplasm/metabolism , Homeostasis , Humans , Karyopherins/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-mdm2/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Exportin 1 Protein
20.
J Biol Chem ; 281(40): 30166-74, 2006 Oct 06.
Article in English | MEDLINE | ID: mdl-16882664

ABSTRACT

The clinical manifestations of West Nile virus (WNV), a member of the Flavivirus family, include febrile illness, sporadic encephalitis, and paralysis. The capsid (Cp) of WNV is thought to participate in these processes by inducing apoptosis through mitochondrial dysfunction and activation of caspase-9 and caspase-3. To further identify the molecular mechanism of the WNV capsid protein (WNVCp), yeast two-hybrid assays were employed using WNV-Cp as bait. Jab1, the fifth subunit of the COP9 signalosome, was subsequently identified as a molecule that interacts with WNVCp. Immunoprecipitation and glutathione S-transferase pulldown assays confirmed that direct interaction could occur between WNVCp and Jab1. Immunofluorescence microscopy demonstrated that the overexpressed WNVCp, which localized to the nucleolus, was translocated to the cytoplasm upon its co-expression with Jab1. When treated with leptomycin B, Jab1-facilitated nuclear exclusion of WNVCp was prevented, which indicated that the CRM1 complex is required for Jab1-facilitated nuclear export of WNVCp. Moreover, Jab1 promoted the degradation of WNVCp in a proteasome-dependent way. Consistent with this, WNVCp-mediated cell cycle arrest at the G(2) phase in H1299 was prevented by exogenous Jab1. Finally, an analysis of WNVCp deletion mutants indicated that the first 15 amino acids were required for interaction with Jab1. Furthermore, the double-point mutant of the WNVCp, P5A/P8A, was incapable of binding to Jab1. These results indicate that Jab1 has a potential protective effect against pathogenic WNVCp and might provide a novel target site for the treatment of disease caused by WNV.


Subject(s)
Capsid Proteins/antagonists & inhibitors , Capsid Proteins/metabolism , Cytoplasm/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Peptide Hydrolases/physiology , West Nile virus/metabolism , Active Transport, Cell Nucleus/genetics , Amino Acid Sequence , COP9 Signalosome Complex , Capsid Proteins/biosynthesis , Capsid Proteins/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Nucleus/enzymology , Cell Nucleus/metabolism , Cytoplasm/enzymology , Cytoplasm/genetics , Gene Expression Regulation, Viral , Humans , Intracellular Signaling Peptides and Proteins/genetics , Molecular Sequence Data , Peptide Hydrolases/genetics , Signal Transduction/genetics , Two-Hybrid System Techniques , West Nile virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...