Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 11(482)2019 03 06.
Article in English | MEDLINE | ID: mdl-30842315

ABSTRACT

Epstein-Barr virus (EBV) is a DNA tumor virus responsible for 1 to 2% of human cancers including subtypes of Burkitt's lymphoma, Hodgkin's lymphoma, gastric carcinoma, and nasopharyngeal carcinoma (NPC). Persistent latent infection drives EBV-associated tumorigenesis. Epstein-Barr nuclear antigen 1 (EBNA1) is the only viral protein consistently expressed in all EBV-associated tumors and is therefore an attractive target for therapeutic intervention. It is a multifunctional DNA binding protein critical for viral replication, genome maintenance, viral gene expression, and host cell survival. Using a fragment-based approach and x-ray crystallography, we identify a 2,3-disubstituted benzoic acid series that selectively inhibits the DNA binding activity of EBNA1. We characterize these inhibitors biochemically and in cell-based assays, including chromatin immunoprecipitation and DNA replication assays. In addition, we demonstrate the potency of EBNA1 inhibitors to suppress tumor growth in several EBV-dependent xenograft models, including patient-derived xenografts for NPC. These inhibitors selectively block EBV gene transcription and alter the cellular transforming growth factor-ß (TGF-ß) signaling pathway in NPC tumor xenografts. These EBNA1-specific inhibitors show favorable pharmacological properties and have the potential to be further developed for the treatment of EBV-associated malignancies.


Subject(s)
DNA, Viral/metabolism , Drug Design , Epstein-Barr Virus Nuclear Antigens/metabolism , Herpesvirus 4, Human/physiology , Nasopharyngeal Neoplasms/virology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Virus Latency/drug effects , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Viral/drug effects , Humans , Mice, Nude , Nasopharyngeal Neoplasms/pathology , Structure-Activity Relationship
2.
Cell Cycle ; 17(4): 468-478, 2018.
Article in English | MEDLINE | ID: mdl-29157092

ABSTRACT

ONC201, founding member of the imipridone class of small molecules, is currently being evaluated in advancer cancer clinical trials. We explored single agent and combinatorial efficacy of ONC201 in preclinical models of hematological malignancies. ONC201 demonstrated (GI50 1-8 µM) dose- and time-dependent efficacy in acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), Burkitt's lymphoma, anaplastic large cell lymphoma (ALCL), cutaneous T-cell lymphoma (CTCL), Hodgkin's lymphoma (nodular sclerosis) and multiple myeloma (MM) cell lines including cells resistant to standard of care (dexamethasone in MM) and primary samples. ONC201 induced caspase-dependent apoptosis that involved activation of the integrated stress response (ATF4/CHOP) pathway, inhibition of Akt phosphorylation, Foxo3a activation, downregulation of cyclin D1, IAP and Bcl-2 family members. ONC201 synergistically reduced cell viability in combination with cytarabine and 5-azacytidine in AML cells. ONC201 combined with cytarabine in a Burkitt's lymphoma xenograft model induced tumor growth inhibition that was superior to either agent alone. ONC201 synergistically combined with bortezomib in MM, MCL and ALCL cells and with ixazomib or dexamethasone in MM cells. ONC201 combined with bortezomib in a Burkitt's lymphoma xenograft model reduced tumor cell density and improved CHOP induction compared to either agent alone. These results serve as a rationale for ONC201 single-agent trials in relapsed/refractory acute leukemia, non-Hodgkin's lymphoma, MM and combination trial with dexamethasone in MM, provide pharmacodynamic biomarkers and identify further synergistic combinatorial regimens that can be explored in the clinic.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Activating Transcription Factor 4/metabolism , Animals , Antineoplastic Agents/therapeutic use , Azacitidine/pharmacology , Boron Compounds/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , G1 Phase Cell Cycle Checkpoints/drug effects , Glycine/analogs & derivatives , Glycine/pharmacology , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Humans , Imidazoles , Mice , Mice, SCID , Pyridines , Pyrimidines , Transcription Factor CHOP/metabolism , Transplantation, Heterologous
3.
PLoS One ; 12(8): e0180541, 2017.
Article in English | MEDLINE | ID: mdl-28767654

ABSTRACT

Cancer stem cells (CSCs) correlate with recurrence, metastasis and poor survival in clinical studies. Encouraging results from clinical trials of CSC inhibitors have further validated CSCs as therapeutic targets. ONC201 is a first-in-class small molecule imipridone in Phase I/II clinical trials for advanced cancer. We have previously shown that ONC201 targets self-renewing, chemotherapy-resistant colorectal CSCs via Akt/ERK inhibition and DR5/TRAIL induction. In this study, we demonstrate that the anti-CSC effects of ONC201 involve early changes in stem cell-related gene expression prior to tumor cell death induction. A targeted network analysis of gene expression profiles in colorectal cancer cells revealed that ONC201 downregulates stem cell pathways such as Wnt signaling and modulates genes (ID1, ID2, ID3 and ALDH7A1) known to regulate self-renewal in colorectal, prostate cancer and glioblastoma. ONC201-mediated changes in CSC-related gene expression were validated at the RNA and protein level for each tumor type. Accordingly, we observed inhibition of self-renewal and CSC markers in prostate cancer cell lines and patient-derived glioblastoma cells upon ONC201 treatment. Interestingly, ONC201-mediated CSC depletion does not occur in colorectal cancer cells with acquired resistance to ONC201. Finally, we observed that basal expression of CSC-related genes (ID1, CD44, HES7 and TCF3) significantly correlate with ONC201 efficacy in >1000 cancer cell lines and combining the expression of multiple genes leads to a stronger overall prediction. These proof-of-concept studies provide a rationale for testing CSC expression at the RNA and protein level as a predictive and pharmacodynamic biomarker of ONC201 response in ongoing clinical studies.


Subject(s)
Biomarkers, Tumor/genetics , Central Nervous System Neoplasms/physiopathology , Colorectal Neoplasms/physiopathology , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/physiopathology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Neoplastic Stem Cells/drug effects , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Central Nervous System Neoplasms/genetics , Colorectal Neoplasms/genetics , Glioblastoma/genetics , HCT116 Cells , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Imidazoles , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Neoplastic Stem Cells/metabolism , Pyridines , Pyrimidines , Transcriptome , Wnt Signaling Pathway/drug effects
4.
Sci Signal ; 9(415): ra18, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26884600

ABSTRACT

ONC201 (also called TIC10) is a small molecule that inactivates the cell proliferation- and cell survival-promoting kinases Akt and ERK and induces cell death through the proapoptotic protein TRAIL. ONC201 is currently in early-phase clinical testing for various malignancies. We found through gene expression and protein analyses that ONC201 triggered an increase in TRAIL abundance and cell death through an integrated stress response (ISR) involving the transcription factor ATF4, the transactivator CHOP, and the TRAIL receptor DR5. ATF4 was not activated in ONC201-resistant cancer cells, and in ONC201-sensitive cells, knockdown of ATF4 or CHOP partially abrogated ONC201-induced cytotoxicity and diminished the ONC201-stimulated increase in DR5 abundance. The activation of ATF4 in response to ONC201 required the kinases HRI and PKR, which phosphorylate and activate the translation initiation factor eIF2α. ONC201 rapidly triggered cell cycle arrest, which was associated with decreased abundance of cyclin D1, decreased activity of the kinase complex mTORC1, and dephosphorylation of the retinoblastoma (Rb) protein. The abundance of X-linked inhibitor of apoptosis protein (XIAP) negatively correlated with the extent of apoptosis in response to ONC201. These effects of ONC201 were independent of whether cancer cells had normal or mutant p53. Thus, ONC201 induces cell death through the coordinated induction of TRAIL by an ISR pathway.


Subject(s)
Activating Transcription Factor 4/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Neoplasms, Experimental/drug therapy , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , eIF-2 Kinase/metabolism , Activating Transcription Factor 4/genetics , Animals , Cell Line, Tumor , Humans , Imidazoles , Mice , Mice, Nude , Mutation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Pyridines , Pyrimidines , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays , eIF-2 Kinase/genetics
5.
Cancer Res ; 75(18): 3842-52, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26294215

ABSTRACT

The tumor-suppressor p53 prevents cancer development via initiating cell-cycle arrest, cell death, repair, or antiangiogenesis processes. Over 50% of human cancers harbor cancer-causing mutant p53. p53 mutations not only abrogate its tumor-suppressor function, but also endow mutant p53 with a gain of function (GOF), creating a proto-oncogene that contributes to tumorigenesis, tumor progression, and chemo- or radiotherapy resistance. Thus, targeting mutant p53 to restore a wild-type p53 signaling pathway provides an attractive strategy for cancer therapy. We demonstrate that small-molecule NSC59984 not only restores wild-type p53 signaling, but also depletes mutant p53 GOF. NSC59984 induces mutant p53 protein degradation via MDM2 and the ubiquitin-proteasome pathway. NSC59984 restores wild-type p53 signaling via p73 activation, specifically in mutant p53-expressing colorectal cancer cells. At therapeutic doses, NSC59984 induces p73-dependent cell death in cancer cells with minimal genotoxicity and without evident toxicity toward normal cells. NSC59984 synergizes with CPT11 to induce cell death in mutant p53-expressing colorectal cancer cells and inhibits mutant p53-associated colon tumor xenograft growth in a p73-dependent manner in vivo. We hypothesize that specific targeting of mutant p53 may be essential for anticancer strategies that involve the stimulation of p73 in order to efficiently restore tumor suppression. Taken together, our data identify NSC59984 as a promising lead compound for anticancer therapy that acts by targeting GOF-mutant p53 and stimulates p73 to restore the p53 pathway signaling.


Subject(s)
Antineoplastic Agents/pharmacology , DNA-Binding Proteins/physiology , Neoplasm Proteins/physiology , Nitrofurans/pharmacology , Nuclear Proteins/physiology , Piperazines/pharmacology , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/physiology , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/pathology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Drug Screening Assays, Antitumor , Drug Synergism , Gene Knockdown Techniques , Genes, p53 , Humans , Irinotecan , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Nitrofurans/chemistry , Nitrofurans/toxicity , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Piperazines/chemistry , Piperazines/toxicity , Proteolysis , Proto-Oncogene Mas , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/metabolism , Tumor Protein p73 , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
6.
Cancer Res ; 75(8): 1668-74, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25681273

ABSTRACT

ONC201/TIC10 is a small-molecule inducer of the TRAIL gene under current investigation as a novel anticancer agent. In this study, we identify critical molecular determinants of ONC201 sensitivity offering potential utility as pharmacodynamic or predictive response markers. By screening a library of kinase siRNAs in combination with a subcytotoxic dose of ONC201, we identified several kinases that ablated tumor cell sensitivity, including the MAPK pathway-inducer KSR1. Unexpectedly, KSR1 silencing did not affect MAPK signaling in the presence or absence of ONC201, but instead reduced expression of the antiapoptotic proteins FLIP, Mcl-1, Bcl-2, cIAP1, cIAP2, and survivin. In parallel to this work, we also conducted a synergy screen in which ONC201 was combined with approved small-molecule anticancer drugs. In multiple cancer cell populations, ONC201 synergized with diverse drug classes, including the multikinase inhibitor sorafenib. Notably, combining ONC201 and sorafenib led to synergistic induction of TRAIL and its receptor DR5 along with a potent induction of cell death. In a mouse xenograft model of hepatocellular carcinoma, we demonstrated that ONC201 and sorafenib cooperatively and safely triggered tumor regressions. Overall, our results established a set of determinants for ONC201 sensitivity that may predict therapeutic response, particularly in settings of sorafenib cotreatment to enhance anticancer responses.


Subject(s)
Antineoplastic Agents/therapeutic use , CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Drug Resistance, Neoplasm/genetics , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Inhibitor of Apoptosis Proteins/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , Baculoviral IAP Repeat-Containing 3 Protein , Cells, Cultured , Drug Screening Assays, Antitumor , Genetic Association Studies , HCT116 Cells , Hep G2 Cells , Humans , Imidazoles , Mice , Mice, Nude , Pyridines , Pyrimidines , Small Molecule Libraries , Ubiquitin-Protein Ligases/physiology
7.
Curr Drug Targets ; 15(1): 80-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24387333

ABSTRACT

p53 is one of the most important tumor suppressor genes that is frequently mutated in human cancers. Generally, p53 functions as a transcription factor that is stabilized and activated by various genotoxic and cellular stress signals, such as DNA damage, hypoxia, oncogene activation and nutrient deprivation, consequently leading to cell cycle arrest, apoptosis, senescence and metabolic adaptation. p53 not only becomes functionally deficient in most cancers, but not infrequently mutant p53 also acquires dominant negative activity and oncogenic properties. p53 has remained an attractive target for cancer therapy. Strategies targeting p53 have been developed including gene therapy to restore p53 function, inhibition of p53-MDM2 interaction, restoration of mutant p53 to wild-type p53, targeting p53 family proteins, eliminating mutant p53, as well as p53-based vaccines. Some of these p53-targeted therapies have entered clinical trials. We discuss the therapeutic potential of p53, with particular focus on the therapeutic strategies to rescue p53 inactivation in human cancers. In addition, we discuss the challenges of p53-targeted therapy and new opportunities for the future.


Subject(s)
Genes, Tumor Suppressor , Genes, p53 , Neoplasms/therapy , Humans , Neoplasms/genetics , Signal Transduction
8.
Cancer Res ; 74(4): 1153-65, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24247721

ABSTRACT

p53 reactivation offers a broad-based strategy for cancer therapy. In this study, we report the identification of prodigiosin that can reactivate p53 family-dependent transcriptional activity in p53-deficient human colon cancer cells. Prodigiosin and its structural analogue (compound R) induced the expression of p53 target genes accompanied by cell-cycle arrest and apoptosis in p53-deficient cancer cells. Prodigiosin restored p53 signaling in cancer cells harboring hotspot TP53 mutations, with little to no detectable cytotoxicity in normal human fibroblasts and with no genotoxicity. Prodigiosin induced the expression of p73 and disrupted its interaction with mutant p53, thereby rescuing p53 pathway deficiency and promoting antitumor effects. The disruption of mutant p53/p73 interaction was specific to prodigiosin and not related to mTOR inhibition. Our findings suggest that mutant p53 needs to be targeted in the context of p73 stimulation to allow efficient restoration of the p53 pathway. In exhibiting this capability, prodigiosin and its analogue provide lead compounds to rescue deficiencies in the p53 pathway in cancer cells by upregulating p73 and targeting mutant p53/p73 interaction there.


Subject(s)
DNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Genes, p53/drug effects , Nuclear Proteins/genetics , Prodigiosin/pharmacology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Animals , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Genes, Tumor Suppressor/drug effects , HCT116 Cells , Humans , Mice , Mice, Nude , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nuclear Proteins/metabolism , Protein Binding/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics , Xenograft Model Antitumor Assays
9.
Cancer Biol Ther ; 13(12): 1185-94, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22892846

ABSTRACT

One of the hallmarks of cancer is metabolic deregulation. Many tumors display increased glucose uptake and breakdown through the process of aerobic glycolysis, also known as the Warburg effect. Less studied in cancer development and progression is the importance of the glutamine (Gln) pathway, which provides cells with a variety of essential products to sustain cell proliferation, such as ATP and macromolecules for biosynthesis. To this end Gln dependency was assessed in a panel of non-small cell lung cancer lines (NSCLC). Gln was found to be essential for the growth of cells with high rates of glutaminolysis, and after exploring multiple genes in the Gln pathway, GLS1 was found to be the key enzyme associated with this dependence. This dependence was confirmed by observing the rescue of decreased growth by exogenous addition of downstream metabolites of glutaminolysis. Expression of the GLS1 splice variant KGA was found to be decreased in tumors compared with normal lung tissue. Transient knock down of GLS1 splice variants indicated that loss of GAC had the most detrimental effect on cancer cell growth. In conclusion, NSCLC cell lines depend on Gln for glutaminolysis to a varying degree, in which the GLS1 splice variant GAC plays an essential role and is a potential target for cancer metabolism-directed therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Glutaminase , Glutamine/metabolism , Protein Isoforms , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation , Glutaminase/genetics , Glutaminase/metabolism , Glycolysis , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Molecular Targeted Therapy , Protein Isoforms/genetics , Protein Isoforms/metabolism
10.
PLoS One ; 6(11): e27425, 2011.
Article in English | MEDLINE | ID: mdl-22087314

ABSTRACT

Non-acute transforming retroviruses like mouse mammary tumor virus (MMTV) cause cancer, at least in part, through integration near cellular genes involved in growth control, thereby de-regulating their expression. It is well-established that MMTV commonly integrates near and activates expression of members of the Wnt and Fgf pathways in mammary tumors. However, there are a significant number of tumors for which the proviral integration sites have not been identified. Here, we used high through-put screening to identify common integration sites (CISs) in MMTV-induced tumors from C3H/HeN and BALB/c mice. As expected, members of both the Wnt and Fgf families were identified in this screen. In addition, a number of novel CISs were found, including Tcf7l2, Antxr1/Tem8, and Arhgap18. We show here that expression of these three putative oncogenes in normal murine mammary gland cells altered their growth kinetics and caused their morphological transformation when grown in three dimensional cultures. Additionally, expression of Tcf7l2 and Antxr1/Tem8 sensitized cells to exogenous WNT ligand. As Tcf7l2, Antxr1/Tem8, and Arhgap18 have been associated with human breast and other cancers, these data demonstrate that MMTV-induced insertional mutation remains an important means for identifying genes involved in breast cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, Neoplasm , Mammary Neoplasms, Animal/virology , Mammary Tumor Virus, Mouse/physiology , Virus Integration , Animals , Biomarkers, Tumor/genetics , Cell Proliferation , Cell Shape , Female , Hepatocyte Nuclear Factor 1-alpha , Mammary Neoplasms, Animal/genetics , Mice , Microfilament Proteins , Mutagenesis, Insertional , Receptors, Cell Surface , Receptors, Peptide/genetics , T Cell Transcription Factor 1/genetics , Tumor Cells, Cultured
11.
Biochem J ; 385(Pt 3): 795-802, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15458387

ABSTRACT

Protein kinase B can phoshorylate and thereby inactivate the FOXO (forkhead box O) family of transcription factors. When active, FOXO factors can bind to DNA in promoter sequences and subsequently regulate gene expression. We have used DNA microarray analysis to identify potential gene targets of FOXO. In the present study we demonstrate that caveolin-1 is directly controlled by FOXO. Firstly, caveolin-1 expression was increased upon induction or over-expression of FOXO factors at both mRNA and protein levels. Second, we show that endogenous regulation of FOXO activity regulates caveolin-1 levels and that this can be inhibited by dominant-negative FOXO. Third, FOXO activates transcription from the caveolin-1 promoter, and using chromatin immunoprecipitations we demonstrated that this activation occurs via direct interaction of FOXO with the promoter. Finally, we demonstrate FOXO-mediated attenuation of EGF (epidermal growth factor)-induced signalling, which in part is mediated by caveolin-1 expression, as suggested by previous studies [Park, Park, Cho, Kim, Ko, Seo and Park (2000) J. Biol. Chem. 275, 20847-20852]. These findings suggest a novel mechanism by which FOXO factors can exert their cellular effects via transcriptional activation of caveolin-1.


Subject(s)
Caveolins/genetics , Caveolins/metabolism , Gene Expression Regulation , Transcription Factors/metabolism , Animals , Caveolin 1 , Cell Cycle , Cell Line , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Epidermal Growth Factor/pharmacology , Fibroblasts , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Transcription Factors/genetics
12.
J Cell Sci ; 115(Pt 20): 3957-66, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12244133

ABSTRACT

The serine/threonine kinase protein kinase B (PKB/c-Akt) acts downstream of the lipid kinase phosphoinositide 3-kinase (PI3K) and functions as an essential mediator in many growth-factor-induced cellular responses such as cell cycle regulation, cell survival and transcriptional regulation. PI3K activation generates 3'-phosphorylated phosphatidylinositol lipids (PtdIns3P) and PKB activation requires PtdIns3P-dependent membrane translocation and phosphorylation by upstream kinases. However PKB activation and function is also regulated by interaction with other proteins. Here we show binding of PKB to periplakin, a member of the plakin family of cytolinker proteins. Interaction between PKB and periplakin was mapped to part of the pleckstrin homology (PH) domain of PKB, which is probably not involved in lipid binding, and indeed binding to periplakin did not affect PKB activation. We therefore investigated the possibility that periplakin may act as a scaffold or localization signal for PKB. In cells endogenous periplakin localizes to different cellular compartments, including plasma membrane, intermediate filament structures, the nucleus and mitochondria. Overexpression of the C-terminal part of periplakin, encompassing the PKB binding region, results in predominant intermediate filament localization and little nuclear staining. This also resulted in inhibition of nuclear PKB signalling as indicated by inhibition of PKB-dependent Forkhead transcription factor regulation. These results suggest a possible role for periplakin as a localization signal in PKB-mediated signalling.


Subject(s)
Cytoskeletal Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Actins/metabolism , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Cell Line , Chlorocebus aethiops , Conserved Sequence , DNA-Binding Proteins/metabolism , Humans , Mitochondria/metabolism , Mutation , Plakins , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Sequence Deletion , Signal Transduction , Tumor Cells, Cultured , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...