Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Invest Dermatol ; 141(4): 874-882.e6, 2021 04.
Article in English | MEDLINE | ID: mdl-32976879

ABSTRACT

Autosomal recessive congenital ichthyosis (ARCI) is a diverse group of cornification diseases associated with severe clinical complications and decreased quality of life. Germline mutations in the TGM1 gene, which encodes the enzyme TGM1, are the predominant cause of ARCI. These TGM1 mutations trigger the abnormal epidermal differentiation and impaired cutaneous barrier function observed in patients with ARCI. Unfortunately, current ARCI therapies focus solely on symptomatic relief. Thus, there is a significant unmet need for therapeutic strategies aimed at correcting the TGM1 deficiency underlying ARCI. In this study, we investigated the ability of KB105, a gene therapy vector encoding full-length human TGM1, to deliver functional human TGM1 to keratinocytes. In vitro, KB105 efficiently infected TGM1-deficient human keratinocytes, produced TGM1 protein, and rescued transglutaminase enzyme function. In vivo studies demonstrated that both single and repeated topical KB105 administration induced TGM1 protein expression in the target epidermal layer without triggering fibrosis, necrosis, or acute inflammation. Toxicity and biodistribution assessments on repeat dosing indicated that KB105 was well-tolerated and restricted to the dose site. Overall, our results demonstrate that rescuing TGM1 deficiency in patients with ARCI through topical KB105 application represents a promising strategy for safely and noninvasively treating this debilitating disease.


Subject(s)
Genetic Vectors/administration & dosage , Herpesvirus 1, Human/genetics , Ichthyosis, Lamellar/therapy , Transglutaminases/genetics , Animals , Biopsy , Cells, Cultured , Enzyme Assays , Female , Genetic Therapy/methods , Genetic Vectors/genetics , Germ-Line Mutation , Humans , Ichthyosis, Lamellar/genetics , Ichthyosis, Lamellar/pathology , Keratinocytes , Male , Mice , Models, Animal , Primary Cell Culture , Quality of Life , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Skin/enzymology , Skin/pathology , Transglutaminases/metabolism
2.
PLoS One ; 11(4): e0153933, 2016.
Article in English | MEDLINE | ID: mdl-27100181

ABSTRACT

Mutational processes and signatures that drive early tumorigenesis are centrally important for early cancer prevention. Yet, to date, biomarkers and risk factors for polyps (adenomas) that inordinately and rapidly develop into colon cancer remain poorly defined. Here, we describe surprisingly high mutational profiles through whole-genome sequence (WGS) analysis in 2 of 4 pairs of benign colorectal adenoma tissue samples. Unsupervised hierarchical clustered transcriptomic analysis of a further 7 pairs of adenomas reveals distinct mutational signatures regardless of adenoma size. Transitional single nucleotide substitutions of C:G>T:A predominate in the adenoma mutational spectrum. Strikingly, we observe mutations in the TGF-ß pathway and CEA-associated genes in 4 out of 11 adenomas, overlapping with the Wnt pathway. Immunohistochemical labeling reveals a nearly 5-fold increase in CEA levels in 23% of adenoma samples with a concomitant loss of TGF-ß signaling. We also define a functional role by which the CEA B3 domain interacts with TGFBR1, potentially inactivating the tumor suppressor function of TGF-ß signaling. Our study uncovers diverse mutational processes underlying the transition from early adenoma to cancer. This has broad implications for biomarker-driven targeting of CEA/TGF-ß in high-risk adenomas and may lead to early detection of aggressive adenoma to CRC progression.


Subject(s)
Adenoma/genetics , Carcinoembryonic Antigen/genetics , Colon/metabolism , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mutation/genetics , Transforming Growth Factor beta/genetics , Adenoma/metabolism , Adenoma/pathology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoembryonic Antigen/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Colon/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Progression , High-Throughput Nucleotide Sequencing/methods , Humans , Immunoenzyme Techniques , Immunoprecipitation , Signal Transduction , Transforming Growth Factor beta/metabolism
3.
Expert Opin Ther Targets ; 17(7): 743-60, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23651053

ABSTRACT

INTRODUCTION: The transforming growth factor-ß (TGF-ß) signaling pathway has a pivotal role in tumor suppression and yet, paradoxically, in tumor promotion. Functional context dependent insights into the TGF-ß pathway are crucial in developing TGF-ß-based therapeutics for cancer. AREAS COVERED: This review discusses the molecular mechanism of the TGF-ß pathway and describes the different ways of tumor suppression by TGF-ß. It is then explained how tumors can evade these effects and how TGF-ß contributes to further growing and spreading of some of the tumors. In the last part of the review, the data on targeting TGF-ß pathway for cancer treatment is assessed. This review focuses on anti-TGF-ß based treatment and other options targeting activated pathways in tumors where the TGF-ß tumor suppressor pathway is lost. Pre-clinical as well up to date results of the most recent clinical trials are given. EXPERT OPINION: Targeting the TGF-ß pathway can be a promising direction in cancer treatment. However, several challenges still exist, the most important are differentiating between the carcinogenic effects of TGF-ß and its other physiological roles, and delineating the tumor suppressive versus the tumor promoting roles of TGF-ß in each specific tumor. Future studies are needed in order to find safer and more effective TGF-ß-based drugs.


Subject(s)
Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , Animals , Humans , Signal Transduction
4.
Nat Rev Gastroenterol Hepatol ; 9(9): 530-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22710573

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common and lethal cancers worldwide. It arises from modulation of multiple genes by mutations, epigenetic regulation, noncoding RNAs and translational modifications of encoded proteins. Although >40% of HCCs are clonal and thought to arise from cancer stem cells (CSCs), the precise identification and mechanisms of CSC formation remain poorly understood. A functional role of transforming growth factor (TGF)-ß signalling in liver and intestinal stem cell niches has been demonstrated through mouse genetics. These studies demonstrate that loss of TGF-ß signalling yields a phenotype similar to a human CSC disorder, Beckwith-Wiedemann syndrome. Insights into this powerful pathway will be vital for developing new therapeutics in cancer. Current clinical approaches are aimed at establishing novel cancer drugs that target activated pathways when the TGF-ß tumour suppressor pathway is lost, and TGF-ß itself could potentially be targeted in metastases. Studies delineating key functional pathways in HCC and CSC formation could be important in preventing this disease and could lead to simple treatment strategies; for example, use of vitamin D might be effective when the TGF-ß pathway is lost or when wnt signalling is activated.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Liver Neoplasms/physiopathology , Neoplastic Stem Cells/pathology , Transforming Growth Factor beta/physiology , Animals , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Disease Progression , Humans , Liver/pathology , Liver Neoplasms/pathology , Mice , Signal Transduction/physiology
5.
J Biol Chem ; 286(49): 42749-42757, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-21998312

ABSTRACT

Cellular retinoic acid-binding protein II (CRABP-II) undergoes nuclear translocation upon binding of retinoic acid (RA). In the nucleus, CRABP-II directly binds to the nuclear receptor RAR to form a complex through which RA is "channeled" from the binding protein to the receptor. CRABP-II thus facilitates the ligation of RAR and markedly enhances its transcriptional activity. The primary sequence of CRABP-II contains three putative SUMOylation sites, centered at K45, K87, and K102. We show here that RA induces interactions of CRABP-II with the E2 SUMO ligase Ubc9 and triggers SUMOylation of the protein both in vitro and in cultured cells. Mutagenesis analyses demonstrate that K102 is the sole CRABP-II residue to be SUMOylated in response to RA. Mutation of this residue abolishes the ability of CRABP-II to undergo nuclear translocation in response RA and thus impairs CRABP-II-mediated activation of RAR. Additional observations demonstrate that apo-CRABP-II is associated with endoplasmic reticulum (ER), and that RA triggers the dissociation of CRABP-II from this location. Furthermore, we show that RA-induced dissociation of CRABP-II from the ER requires SUMOylation of K102. Hence, SUMOylation of K102 in response to RA binding is critical for dissociation of CRABP-II from ER and, consequently, for mobilization of the protein to nucleus and for its cooperation with RAR.


Subject(s)
Active Transport, Cell Nucleus , Receptors, Retinoic Acid/metabolism , Sumoylation , Tretinoin/metabolism , Animals , COS Cells , Cell Line, Tumor , Cell Nucleus/metabolism , Chlorocebus aethiops , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Transcription, Genetic , Ubiquitin-Conjugating Enzymes/metabolism
6.
Proc Natl Acad Sci U S A ; 108(11): 4340-5, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21368206

ABSTRACT

It currently is believed that vitamin A, retinol, functions through active metabolites: the visual chromophore 11-cis-retinal, and retinoic acids, which regulate gene transcription. Retinol circulates in blood bound to retinol-binding protein (RBP) and is transported into cells by a membrane protein termed "stimulated by retinoic acid 6" (STRA6). We show here that STRA6 not only is a vitamin A transporter but also is a cell-surface signaling receptor activated by the RBP-retinol complex. Association of RBP-retinol with STRA6 triggers tyrosine phosphorylation, resulting in recruitment and activation of JAK2 and the transcription factor STAT5. The RBP-retinol/STRA6/JAK2/STAT5 signaling cascade induces the expression of STAT target genes, including suppressor of cytokine signaling 3 (SOCS3), which inhibits insulin signaling, and peroxisome proliferator-activated receptor gamma (PPARγ), which enhances lipid accumulation. These observations establish that the parental vitamin A molecule is a transcriptional regulator in its own right, reveal that the scope of biological functions of the vitamin is broader than previously suspected, and provide a rationale for understanding how RBP and retinol regulate energy homeostasis and insulin responses.


Subject(s)
Gene Expression Regulation , Insulin/metabolism , Retinol-Binding Proteins/metabolism , Signal Transduction , Vitamin A/metabolism , Animals , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Janus Kinase 2/metabolism , Membrane Proteins/metabolism , Mice , Models, Biological , Phosphorylation/drug effects , Protein Binding/drug effects , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Triglycerides/metabolism
7.
Biochemistry ; 49(47): 10146-57, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21033716

ABSTRACT

In many tumor cells, the activation and activity of extracellular signal-regulated kinases (ERK1/2) are very high because of the constitutive activation of the Ras-mediated signaling pathway. Here, we ectopically expressed the human homologue of rat eukaryotic initiation factor 2-associated glycoprotein, p67/MetAP2, in EGF-treated mouse embryonic NIH3T3 fibroblasts and C2C12 myoblasts and NIH3T3 cell lines expressing the constitutively active form of MAP kinase kinase (MEK) to inhibit the activation and activity of ERK1/2 MAP kinases. In addition, we also ectopically expressed rat p67/MetAP2 in oncogenic Ras-induced transformed NIH3T3 fibroblasts and inhibited their transformed phenotype both in culture and in athymic nude mice possibly by inhibiting angiogenesis. This inhibition of ERK1/2 MAP kinases is due to the direct binding with rat p67/MetAP2, and this leads to the inhibition of activity of ERK1/2 MAP kinases both in vitro and in vivo. Furthermore, expression of p67/MetAP2 siRNA in both NIH3T3 fibroblasts and C2C12 myoblasts causes activation and activity of ERK1/2 MAP kinases. Our results thus suggest that ectopic expression of rat p67/MetAP2 in transformed cells can inhibit the tumorigenic phenotype by inhibiting the activation and activity of ERK1/2 MAP kinases and, thus, that p67/MetAP2 has tumor suppression activity.


Subject(s)
Aminopeptidases/physiology , Glycoproteins/physiology , Animals , Cell Line , Cyclohexanes/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Fatty Acids, Unsaturated/pharmacology , Genes, ras/genetics , Humans , Methionyl Aminopeptidases , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Peptide Fragments/metabolism , Rats , Sesquiterpenes/pharmacology , Signal Transduction/drug effects , Transfection
8.
Biochemistry ; 46(11): 3465-75, 2007 Mar 20.
Article in English | MEDLINE | ID: mdl-17311411

ABSTRACT

Eukaryotic initiation factor 2-associated glycoprotein, p67, plays important roles in the regulation of eIF2alpha phosphorylation and thus maintains cell growth and proliferation. The p67 sequence can be divided into two segments, the N-terminal segment of amino acids 1-107 (p26) and the downstream segment of amino acids 108-480 (p52). Comparison of the amino acid sequences of p67 from lower to higher organisms suggests that there is a progressive addition of several unique domains at the N-terminus of p67, and these unique domains, which are present in p26, play important roles in the modulation of eIF2alpha phosphorylation in mammalian cells. To test the hypothesis that the p26 segment is generated from p67 due to its autoproteolysis and whether p26 is required for the protection of eIF2alpha from phosphorylation, we have analyzed the time-dependent cleavage of functionally active rat recombinant p67 purified from either baculovirus-infected insect cells or transiently transfected mammalian cells. We noticed a regulated cleavage of p67 that generates several peptides along with the most stable p26 and p52 fragments. The p52 fragment has a low level of autoproteolysis activity that possibly increases the autoproteolysis of full-length p67. This activity could not be inhibited by a serine protease inhibitor, PMSF, but could be inhibited by a cocktail of protease inhibitors that includes bestatin, leupeptin, E64, AEBSF, and aprotinin. To provide evidence that the fragmentation of p67 is not due to the presence of any contaminant protease(s), we fractionated purified rat p67 with molecular sieve, anion exchange, and cation exchange chromatographic steps performed in the presence of different K+ ion concentrations. Our data show that the extent of cleavage of p67 into different fragments is higher in the presence of 0.75 M K+ ion and in samples stored at -80 degrees C. Under parallel conditions, p67's mutants, D251A and D262A, exhibited very little to no cleavage, whereas the H231E mutant exhibited extensive cleavage that generated a large amount of p26 fragment. The p26 fragment exhibited protection of eIF2alpha phosphorylation both in vivo and in vitro. Altogether, our data provide evidence that rat p67 has autoproteolytic activity that generates p26, which is required to block eIF2alpha from phosphorylation.


Subject(s)
Aminopeptidases/metabolism , Eukaryotic Initiation Factor-2/metabolism , Glycoproteins/metabolism , Peptide Fragments/metabolism , Amino Acid Sequence , Amino Acid Substitution , Aminopeptidases/genetics , Animals , Cells, Cultured , Glycoproteins/genetics , Methionyl Aminopeptidases , Mice , Peptide Hydrolases/metabolism , Phosphorylation/drug effects , Protease Inhibitors/pharmacology , Rats , Recombinant Proteins/metabolism , eIF-2 Kinase/metabolism
9.
Exp Cell Res ; 312(16): 3184-203, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16857189

ABSTRACT

Eukaryotic initiation factor 2 (eIF2)-associated glycoprotein, p67, plays an important role in protecting eIF2alpha from phosphorylation by eIF2alpha-specific kinases. To understand the molecular details of interaction between p67 and the subunits of eIF2, we applied several biochemical and mutational analyses to identify interacting domains within p67 and eIF2gamma. These studies were combined with functional in vivo and in vitro assays to address the importance of the interactions between p67 and eIF2gamma in eIF2alpha phosphorylation. Studies from yeast two-hybrid assays show that p67 interacts strongly with eIF2gamma, relatively weakly with eIF2alpha, and no interaction with eIF2beta. Further mutational analyses provided evidence that the N-terminal lysine-rich domain II and the 340-430 amino acid segment of p67 interact strongly with the C-terminal 409-472 amino acid segment of eIF2gamma. GST pull-down assays show that the interaction between p67 and eIF2gamma is direct. From co-immunoprecipitation studies, we find that the interaction between p67 and eIF2gamma could not only be detected in mammalian cells growing in growth medium, it could also be detected in transiently transfected cells with expression plasmids encoding p67 and eIF2gamma. However, this interaction could not be detected in p67 mutants lacking lysine-rich domain II and the 340-430 amino acid segment. We also find a very good correlation between p67 binding to eIF2gamma and the protection of eIF2alpha from phosphorylation. Altogether, our data provide genetic evidence for the interaction between p67 and eIF2gamma and that this interaction modulates the phosphorylation of eIF2alpha.


Subject(s)
Aminopeptidases/chemistry , Aminopeptidases/metabolism , Eukaryotic Initiation Factor-2/metabolism , Glycoproteins/chemistry , Glycoproteins/metabolism , Lysine/metabolism , Protein Subunits/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cells, Cultured , Eukaryotic Initiation Factor-2/chemistry , Immunoprecipitation , Mice , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Protein Subunits/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/cytology
10.
Arch Biochem Biophys ; 452(2): 138-48, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16843428

ABSTRACT

Phosphorylation of the alpha-subunit of eukaryotic initiation factor 2 is the major regulatory step in the initiation of protein synthesis in mammals. P67, a cellular glycoprotein, protects phosphorylation of eIF2alpha from kinases. Previously, we reported that the D6/2 mutant of p67 has higher levels of protection of eIF2alpha phosphorylation (POEP) activity. In this study, we report that the D6/2 mutant and its double mutants containing second-site alanine substitutions at the five conserved amino acid residues (D251, D262, H331, E364, and E459) show increased POEP activity in serum-starved rat tumor hepatoma cells. Serum-restoration to those cells did not abolish their increased POEP activity except the D6/2+H331A double mutant. The latter mutant shows slight inhibition of POEP activity during serum starvation and this inhibition increased significantly during serum restoration. KRC-7 cells constitutively expressing the D6/2 mutant showed slightly decreased levels of PKR phosphorylation and significantly low level of phosphorylation of ERKs 1 and 2. The D6/2 mutant also showed increased binding with eIF2alpha and eIF2gamma and almost similar binding with ERKs 1 and 2 as compared to wild type p67. Altogether, our data demonstrate that the increased binding of the D6/2 mutant with the subunits of eIF2 may be in part the cause for its high POEP activity.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Eukaryotic Initiation Factor-2/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Phosphoproteins/metabolism , Animals , Binding Sites , Cell Line, Tumor , Phosphorylation , Protein Binding , Protein Biosynthesis , Rats
11.
Exp Cell Res ; 303(1): 174-82, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15572037

ABSTRACT

Eukaryotic initiation factor 2-associated glycoprotein, p67, protects eIF2 from phosphorylation by its kinases. To understand the roles of p67 during skeletal muscle differentiation of mouse C2C12 myoblasts, we measured the level of p67 during myotube formation. We noticed that the level of p67 increases during myoblast differentiation and this increased level is controlled at the translational stage. The stability of p67 in the myotubes is due to its low turnover rate. The phosphorylation of the extracellular signal-regulated kinases (ERKs 1 and 2) is high in growth-factor-mediated cycling of C2C12 myoblasts and this phosphorylation decreases at 96 h when these myoblasts are grown in differentiation medium. At this time of differentiation, the level of p67 is higher compared to 0 h of differentiation. p67 binds to ERK2 and inhibits its activity in vitro. Taken together, these results suggest that the stability of p67 increases during myotube formation while inhibiting the phosphorylation of ERKs 1 and 2.


Subject(s)
Cell Differentiation/physiology , Eukaryotic Initiation Factor-2/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycoproteins/metabolism , Muscle, Skeletal/cytology , Animals , Mice , Muscle, Skeletal/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Phosphorylation , Recombinant Fusion Proteins/metabolism
12.
Biochemistry ; 43(46): 14821-31, 2004 Nov 23.
Article in English | MEDLINE | ID: mdl-15544353

ABSTRACT

Fumagillin, an angiogenic inhibitor, binds to methionine aminopeptidase 2, which is the same as eukaryotic initiation factor 2-associated glycoprotein, p67. p67 protects eIF2alpha from phosphorylation by its kinases. To understand the importance of fumagillin binding to p67, we measured the level of p67 in mouse C2C12 myoblasts treated with fumagillin. We show that fumagillin increases the stability of p67 by decreasing its turnover rate. The increased levels of p67 result in inhibition of phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERKs 1 and 2). p67 binds to these ERKs, and the 108-480 amino acid segment is sufficient for this binding. p67's affinity to ERKs 1 and 2 also increases in fumagillin-treated myoblasts while its affinity for eIF2alpha remains unchanged. A mutant at the conserved amino acid residue D251A increases the phosphorylation of ERKs 1 and 2 without affecting the binding to p67, thus indicating the importance of this residue in the regulation of the phosphorylation of these ERKs. These results suggest that fumagillin increases the stability of p67 and its affinity to ERKs 1 and 2 and causes the inhibition of the phosphorylation of ERKs 1 and 2.


Subject(s)
Aminopeptidases/metabolism , Angiogenesis Inhibitors/pharmacology , Eukaryotic Initiation Factor-2/metabolism , Fatty Acids, Unsaturated/pharmacology , Glycoproteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Alanine/genetics , Amino Acid Substitution/genetics , Aminopeptidases/biosynthesis , Aminopeptidases/genetics , Angiogenesis Inhibitors/metabolism , Animals , Aspartic Acid/genetics , Cell Line , Cyclohexanes , Enzyme Inhibitors/metabolism , Enzyme Stability/drug effects , Enzyme Stability/genetics , Fatty Acids, Unsaturated/metabolism , Female , Glycoproteins/biosynthesis , Glycoproteins/genetics , Inhibitory Concentration 50 , Methionyl Aminopeptidases , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/enzymology , Myoblasts, Skeletal/metabolism , Peptide Fragments/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Binding/genetics , Sesquiterpenes , Transfection
13.
Arch Biochem Biophys ; 427(1): 68-78, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15178489

ABSTRACT

Phosphorylation of the alpha-subunit of eukaryotic initiation factor 2 is the major regulatory step in the initiation of protein synthesis in mammals. P67, a cellular glycoprotein, protects phosphorylation of eIF2alpha from kinases. P67 has five conserved amino acid residues at the D251, D262, H331, E364, and E459 positions. To determine the roles of these conserved amino acid residues in eIF2alpha phosphorylation during serum-starved conditions, we constitutively expressed D251A, D262A, H331A, E364A, and E459A mutants in rat tumor hepatoma cells. We find that the point mutants D251A, H331A, and E364A lower the levels of eIF2alpha phosphorylation. These low levels of phosphorylation decrease when serum-starved cells are grown in medium containing serum. To understand the mechanism of action of the p67 mutants in eIF2alpha phosphorylation during serum-starvation, we performed detailed biochemical analyses with the D251A mutant. We find that neither the O-GlcNAc modification on the D251A mutant nor the binding of D251A mutant with eIF2gamma has significant effects on eIF2alpha phosphorylation during serum-starved conditions. However, the D251A mutant inhibits p67's activity to suppress the activity of ERK1/2. Our data suggest that both p67 and the D251A mutant bind to ERK1, thus strengthening the idea that p67 regulates the activity of ERK1. During serum-starvation conditions, both PKR and PERK are phosphorylated and the D251A mutant shows increased stability of PERK as well as a slight decrease in its activity. Altogether, our data provide evidence to suggest that p67 modulates the expression and activity of certain eIF2alpha-specific kinases.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Glycoproteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Blotting, Western , Cell Line , Culture Media, Serum-Free , Mitogen-Activated Protein Kinase 3 , Phosphorylation , Point Mutation , Precipitin Tests , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...