Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Blood ; 133(11): 1171-1185, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30587525

ABSTRACT

Aberrant activation of mTOR signaling in acute myeloid leukemia (AML) results in a survival advantage that promotes the malignant phenotype. To improve our understanding of factors that contribute to mammalian target of rapamycin (mTOR) signaling activation and identify novel therapeutic targets, we searched for unique interactors of mTOR complexes through proteomics analyses. We identify cyclin dependent kinase 9 (CDK9) as a novel binding partner of the mTOR complex scaffold protein, mLST8. Our studies demonstrate that CDK9 is present in distinct mTOR-like (CTOR) complexes in the cytoplasm and nucleus. In the nucleus, CDK9 binds to RAPTOR and mLST8, forming CTORC1, to promote transcription of genes important for leukemogenesis. In the cytoplasm, CDK9 binds to RICTOR, SIN1, and mLST8, forming CTORC2, and controls messenger RNA (mRNA) translation through phosphorylation of LARP1 and rpS6. Pharmacological targeting of CTORC complexes results in suppression of growth of primitive human AML progenitors in vitro and elicits strong antileukemic responses in AML xenografts in vivo.


Subject(s)
Carcinogenesis/drug effects , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , RNA, Messenger/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Biomarkers, Tumor/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Cytarabine/pharmacology , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Nude , Phosphorylation , Protein Biosynthesis , Proteome/analysis , RNA, Messenger/drug effects , RNA, Messenger/genetics , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
J Clin Invest ; 127(4): 1316-1320, 2017 Apr 03.
Article in English | MEDLINE | ID: mdl-28240607

ABSTRACT

Primary myelofibrosis (PMF) is a clonal hematologic malignancy characterized by BM fibrosis, extramedullary hematopoiesis, circulating CD34+ cells, splenomegaly, and a propensity to evolve to acute myeloid leukemia. Moreover, the spleen and BM of patients harbor atypical, clustered megakaryocytes, which contribute to the disease by secreting profibrotic cytokines. Here, we have revealed that megakaryocytes in PMF show impaired maturation that is associated with reduced GATA1 protein. In investigating the cause of GATA1 downregulation, our gene-expression study revealed the presence of the RPS14-deficient gene signature, which is associated with defective ribosomal protein function and linked to the erythroid lineage in 5q deletion myelodysplastic syndrome. Surprisingly, reduced GATA1 expression and impaired differentiation were limited to megakaryocytes, consistent with a proproliferative effect of a GATA1 deficiency on this lineage. Importantly, expression of GATA1 effectively rescued maturation of PMF megakaryocytes. Together, these results suggest that ribosomal deficiency contributes to impaired megakaryopoiesis in myeloproliferative neoplasms.


Subject(s)
Down-Regulation , GATA1 Transcription Factor/biosynthesis , Megakaryocytes/metabolism , Primary Myelofibrosis/metabolism , Thrombopoiesis , Animals , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Chromosomes, Human, Pair 5/metabolism , GATA1 Transcription Factor/genetics , Humans , Megakaryocytes/pathology , Mice , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Ribosomal Proteins/biosynthesis , Ribosomal Proteins/genetics
3.
Mol Cancer Res ; 14(10): 984-993, 2016 10.
Article in English | MEDLINE | ID: mdl-27364770

ABSTRACT

Glioblastoma multiforme remains the deadliest malignant brain tumor, with glioma stem cells (GSC) contributing to treatment resistance and tumor recurrence. We have identified MAPK-interacting kinases (MNK) as potential targets for the GSC population in glioblastoma multiforme. Isoform-level subtyping using The Cancer Genome Atlas revealed that both MNK genes (MKNK1 and MKNK2) are upregulated in mesenchymal glioblastoma multiforme as compared with other subtypes. Expression of MKNK1 is associated with increased glioma grade and correlated with the mesenchymal GSC marker, CD44, and coexpression of MKNK1 and CD44 predicts poor survival in glioblastoma multiforme. In established and patient-derived cell lines, pharmacologic MNK inhibition using LY2801653 (merestinib) inhibited phosphorylation of the eukaryotic translation initiation factor 4E, a crucial effector for MNK-induced mRNA translation in cancer cells and a marker of transformation. Importantly, merestinib inhibited growth of GSCs grown as neurospheres as determined by extreme limiting dilution analysis. When the effects of merestinib were assessed in vivo using an intracranial xenograft mouse model, improved overall survival was observed in merestinib-treated mice. Taken together, these data provide strong preclinical evidence that pharmacologic MNK inhibition targets mesenchymal glioblastoma multiforme and its GSC population. IMPLICATIONS: These findings raise the possibility of MNK inhibition as a viable therapeutic approach to target the mesenchymal subtype of glioblastoma multiforme. Mol Cancer Res; 14(10); 984-93. ©2016 AACR.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Indazoles/administration & dosage , Intracellular Signaling Peptides and Proteins/genetics , Neoplastic Stem Cells/enzymology , Niacinamide/analogs & derivatives , Protein Serine-Threonine Kinases/genetics , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Hyaluronan Receptors , Indazoles/pharmacology , Mice , Neoplasm Grading , Niacinamide/administration & dosage , Niacinamide/pharmacology , Phosphorylation/drug effects , Survival Analysis , Xenograft Model Antitumor Assays
4.
J Biol Chem ; 291(5): 2389-96, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26645692

ABSTRACT

We provide evidence for a unique pathway engaged by the type II IFN receptor, involving mTORC2/AKT-mediated downstream regulation of mTORC1 and effectors. These events are required for formation of the eukaryotic translation initiation factor 4F complex (eIF4F) and initiation of mRNA translation of type II interferon-stimulated genes. Our studies establish that Rictor is essential for the generation of type II IFN-dependent antiviral and antiproliferative responses and that it controls the generation of type II IFN-suppressive effects on normal and malignant hematopoiesis. Together, our findings establish a central role for mTORC2 in IFNγ signaling and type II IFN responses.


Subject(s)
Carrier Proteins/metabolism , Eukaryotic Initiation Factor-4F/metabolism , Interferon-gamma/metabolism , Multiprotein Complexes/metabolism , Receptors, Interferon/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Chemokine CXCL10/metabolism , Fibroblasts/metabolism , Gene Expression Regulation , Hematopoiesis , Hematopoietic Stem Cells/cytology , Humans , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Knockout , Phosphorylation , Polyribosomes/metabolism , Protein Biosynthesis , Rapamycin-Insensitive Companion of mTOR Protein , U937 Cells
5.
Oncotarget ; 6(32): 33206-16, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26375673

ABSTRACT

We investigated the efficacy of targeting the PIM kinase pathway in Philadelphia chromosome-positive (Ph+) leukemias. We provide evidence that inhibition of PIM, with the pan-PIM inhibitor SGI-1776, results in suppression of classic PIM effectors and also elements of the mTOR pathway, suggesting interplay between PIM and mTOR signals. Our data demonstrate that PIM inhibition enhances the effects of imatinib mesylate on Ph+ leukemia cells. We also found that PIM inhibition results in suppression of leukemic cell proliferation and induction of apoptosis of Ph+ leukemia cells, including those resistant to imatinib mesylate. Importantly, inhibition of PIM results in enhanced suppression of primary leukemic progenitors from patients with CML. Altogether these findings suggest that pharmacological PIM targeting may provide a unique therapeutic approach for the treatment of Ph+ leukemias.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Genes, abl/genetics , Imidazoles/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcr/genetics , Pyridazines/pharmacology , Dose-Response Relationship, Drug , Humans , Imatinib Mesylate/pharmacology , K562 Cells , Mutation , Philadelphia Chromosome , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Tumor Cells, Cultured
6.
Mol Cell Biol ; 35(15): 2684-98, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26012550

ABSTRACT

We provide evidence that human SLFN5, an interferon (IFN)-inducible member of the Schlafen (SLFN) family of proteins, exhibits key roles in controlling motility and invasiveness of renal cell carcinoma (RCC) cells. Our studies define the mechanism by which this occurs, demonstrating that SLFN5 negatively controls expression of the matrix metalloproteinase 1 gene (MMP-1), MMP-13, and several other genes involved in the control of malignant cell motility. Importantly, our data establish that SLFN5 expression correlates with a better overall survival in a large cohort of patients with RCC. The inverse relationship between SLFN5 expression and RCC aggressiveness raises the possibility of developing unique therapeutic approaches in the treatment of RCC, by modulating SLFN5 expression.


Subject(s)
Carcinoma, Renal Cell/pathology , Cell Cycle Proteins/genetics , Kidney Neoplasms/pathology , Matrix Metalloproteinase 13/biosynthesis , Matrix Metalloproteinase 1/biosynthesis , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/mortality , Cell Cycle Proteins/biosynthesis , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Interferon-alpha/pharmacology , Kidney Neoplasms/genetics , Kidney Neoplasms/mortality , Neoplasm Invasiveness/genetics , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering
7.
Proc Natl Acad Sci U S A ; 111(31): 11377-82, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25049393

ABSTRACT

We provide evidence that S6 kinase 1 (S6K1) Aly/REF-like target (SKAR) is engaged in IFN-α signaling and plays a key role in the generation of IFN responses. Our data demonstrate that IFN-α induces phosphorylation of SKAR, which is mediated by either the p90 ribosomal protein S6 kinase (RSK) or p70 S6 kinase (S6K1), in a cell type-specific manner. This type I IFN-inducible phosphorylation of SKAR results in enhanced interaction with the eukaryotic initiation factor (eIF)4G and recruitment of activated RSK1 to 5' cap mRNA. Our studies also establish that SKAR is present in cap-binding CBP80 immune complexes and that this interaction is mediated by eIF4G. We demonstrate that inducible protein expression of key IFN-α-regulated protein products such as ISG15 and p21(WAF1/CIP1) requires SKAR activity. Importantly, our studies define a requirement for SKAR in the generation of IFN-α-dependent inhibitory effects on malignant hematopoietic progenitors from patients with chronic myeloid leukemia or myeloproliferative neoplasms. Taken altogether, these findings establish critical and essential roles for SKAR in the regulation of mRNA translation of IFN-sensitive genes and induction of IFN-α biological responses.


Subject(s)
Interferon-alpha/metabolism , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytokines/metabolism , Guanosine/analogs & derivatives , Guanosine/metabolism , Humans , Mice , Nuclear Cap-Binding Protein Complex/metabolism , Phosphorylation/drug effects , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/drug effects , Ubiquitins/metabolism
8.
J Interferon Cytokine Res ; 34(4): 289-96, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24559173

ABSTRACT

Interferons (IFNs) are released by cells on exposure to various stimuli, including viruses, double-stranded RNA, and other cytokines and various polypeptides. These IFNs play important physiological and pathophysiological roles in humans. Many clinical studies have established activity for these cytokines in the treatment of several malignancies, viral syndromes, and autoimmune disorders. In this review, the regulatory effects of type I and II IFN receptors on the translation-initiation process mediated by mechanistic target of rapamycin (mTOR) and mitogen-activated protein kinase (MAPK) pathways and the known mechanisms of control of mRNA translation of IFN-stimulated genes are summarized and discussed.


Subject(s)
Autoimmune Diseases/immunology , Interferon Type I/metabolism , Interferon-gamma/metabolism , Neoplasms/immunology , RNA, Messenger/metabolism , Receptor, Interferon alpha-beta/metabolism , Virus Diseases/immunology , Animals , Gene Expression Regulation , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , Receptors, Interferon/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
9.
J Biol Chem ; 289(10): 6581-6591, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24469448

ABSTRACT

We provide evidence that type I IFN-induced STAT activation is diminished in cells with targeted disruption of the Rictor gene, whose protein product is a key element of mTOR complex 2. Our studies show that transient or stable knockdown of Rictor or Sin1 results in defects in activation of elements of the STAT pathway and reduced STAT-DNA binding complexes. This leads to decreased expression of several IFN-inducible genes that mediate important biological functions. Our studies also demonstrate that Rictor and Sin1 play essential roles in the generation of the suppressive effects of IFNα on malignant erythroid precursors from patients with myeloproliferative neoplasms. Altogether, these findings provide evidence for critical functions for Rictor/Sin1 complexes in type I IFN signaling and the generation of type I IFN antineoplastic responses.


Subject(s)
Antineoplastic Agents/pharmacology , Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Interferon Type I/pharmacology , Transcription, Genetic/drug effects , Animals , Carrier Proteins/genetics , Cells, Cultured , Fibroblasts/drug effects , Gene Knockdown Techniques , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Humans , Mice , Phosphorylation , Polycythemia Vera/metabolism , Polycythemia Vera/pathology , Rapamycin-Insensitive Companion of mTOR Protein , Signal Transduction
10.
PLoS One ; 8(11): e78780, 2013.
Article in English | MEDLINE | ID: mdl-24260131

ABSTRACT

Chronic myeloid leukemia (CML) and Ph+ acute lymphoblastic leukemia (ALL) are characterized by the presence of the BCR-ABL oncoprotein, which leads to activation of a plethora of pro-mitogenic and pro-survival pathways, including the mTOR signaling cascade. We provide evidence that in BCR-ABL expressing cells, treatment with tyrosine kinase inhibitors (TKIs) results in upregulation of mRNA levels and protein expression of sestrin3 (SESN3), a unique cellular inhibitor of mTOR complex 1 (mTORC1). Such upregulation appears to be mediated by regulatory effects on mTOR, as catalytic inhibition of the mTOR kinase also induces SESN3. Catalytic mTOR inhibition also results in upregulation of SESN3 expression in cells harboring the TKI-insensitive T315I-BCR-ABL mutant, which is resistant to imatinib mesylate. Overexpression of SESN3 results in inhibitory effects on different Ph+ leukemic cell lines including KT-1-derived leukemic precursors, indicating that SESN3 mediates anti-leukemic responses in Ph+ cells. Altogether, our findings suggest the existence of a novel mechanism for the generation of antileukemic responses in CML cells, involving upregulation of SESN3 expression.


Subject(s)
Fusion Proteins, bcr-abl/biosynthesis , Gene Expression Regulation, Leukemic , Heat-Shock Proteins/biosynthesis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Animals , Cell Line, Tumor , Fusion Proteins, bcr-abl/genetics , Heat-Shock Proteins/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Up-Regulation/genetics
11.
J Biol Chem ; 288(46): 33006-15, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24089532

ABSTRACT

There is emerging evidence that the IFN-inducible family of Slfn genes and proteins play important roles in cell cycle progression and control of cellular proliferation, but the precise functional roles of different Slfn members in the regulation of tumorigenesis remain unclear. In the present study, we undertook a systematic analysis on the expression and functional relevance of different mouse Slfn genes in malignant melanoma and renal cell carcinoma cells. Our studies demonstrate that several mouse Slfn genes are up-regulated in response to IFN treatment of mouse melanoma and renal cell carcinoma cells, including Slfn1, Slfn2, Slfn4, Slfn5, and Slfn8. Our data show that Slfn2 and Slfn3 play essential roles in the control of mouse malignant melanoma cell proliferation and/or anchorage-independent growth, suggesting key and non-overlapping roles for these genes in the control of malignant melanoma tumorigenesis. In renal cell carcinoma cells, in addition to Slfn2 and Slfn3, Slfn5 also exhibits important antineoplastic effects. Altogether, our findings indicate important functions for distinct mouse Slfn genes in the control of tumorigenesis and provide evidence for differential involvement of distinct members of this gene family in controlling tumorigenesis. They also raise the potential of future therapeutic approaches involving modulation of expression of members of this family of genes in malignant melanoma and renal cell carcinoma.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cell Cycle Proteins/metabolism , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/metabolism , Melanoma/metabolism , Neoplasm Proteins/metabolism , Animals , Antiviral Agents/pharmacology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Interferon-gamma/pharmacology , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Melanoma/genetics , Melanoma/pathology , Mice , Neoplasm Proteins/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
12.
J Biomol Screen ; 18(2): 180-90, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22972848

ABSTRACT

Alternative splicing (AS) is an efficient mechanism that involves the generation of transcriptome and protein diversity from a single gene. Defects in pre-messenger RNA (mRNA) splicing are an important cause of numerous diseases, including cancer. AS of pre-mRNA as a target for cancer therapy has not been well studied. We have reported previously that a splicing factor, polypyrimidine tract-binding protein (PTB), is overexpressed in ovarian tumors compared with matched normal controls, and knockdown of PTB expression by short-hairpin RNA impairs ovarian tumor cell growth, colony formation, and invasiveness. Given the complexity of PTB's molecular functions, a chemical method for controlling PTB activity might provide a therapeutic and experimental tool. However, no commercially available PTB inhibitors have yet been described. To expand our ability to find novel inhibitors, we developed a robust, fluorometric, cell-based high-throughput screening assay in 96-well plates that reports on the splicing activity of PTB. In an attempt to use the cells for large-scale chemical screens to identify PTB modulators, we established cell lines stably expressing the reporter gene. Our results suggest that this high-throughput assay could be used to identify small-molecule modulators of PTB activity. Based on these findings and the role that upregulated PTB has on cell proliferation and malignant properties of tumors, targeting PTB for inhibition with small molecules offers a promising strategy for cancer therapy.


Subject(s)
Alternative Splicing , High-Throughput Screening Assays/methods , Small Molecule Libraries , Alternative Splicing/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Gene Expression Regulation/drug effects , Genes, Reporter , Humans , Nerve Tissue Proteins/genetics , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , RNA Precursors/genetics , Reproducibility of Results
13.
Mol Pharmacol ; 79(4): 735-41, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21252292

ABSTRACT

Nuclear factor (NF)-YB, a subunit of the transcription factor nuclear factor Y (NF-Y) complex, binds and activates CCAAT-containing promoters. Our previous work suggested that NF-YB may be a mediator of topoisomerase IIα (Top2α), working through the Top2α promoter. DNA topoisomerase II (Top2) is an essential nuclear enzyme and the primary target for several clinically important anticancer drugs. Our teniposide-resistant human lymphoblastic leukemia CEM cells (CEM/VM-1-5) express reduced Top2α protein compared with parental CEM cells. To study the regulation of Top2α during the development of drug resistance, we found that NF-YB protein expression is increased in CEM/VM-1-5 cells compared with parental CEM cells. This further suggests that increased NF-YB may be a negative regulator of Top2α in CEM/VM-1-5 cells. We asked what causes the up-regulation of NF-YB in CEM/VM-1-5 cells. We found by microRNA profiling that hsa-miR-485-3p is lower in CEM/VM-1-5 cells compared with CEM cells. MicroRNA target prediction programs revealed that the 3'-untranslated region (3'-UTR) of NF-YB harbors a putative hsa-miR-485-3p binding site. We thus hypothesized that hsa-miR-485-3p mediates drug responsiveness by decreasing NF-YB expression, which in turn negatively regulates Top2α expression. To test this, we overexpressed miR-485-3p in CEM/VM-1-5 cells and found that this led to reduced expression of NF-YB, a corresponding up-regulation of Top2α, and increased sensitivity to the Top2 inhibitors. Results in CEM cells were replicated in drug-sensitive and -resistant human rhabdomyosarcoma Rh30 cells, suggesting that our findings represent a general phenomenon. Ours is the first study to show that miR-485-3p mediates Top2α down-regulation in part by altered regulation of NF-YB.


Subject(s)
Antigens, Neoplasm/biosynthesis , CCAAT-Binding Factor/metabolism , DNA Topoisomerases, Type II/biosynthesis , DNA-Binding Proteins/biosynthesis , Etoposide/toxicity , Gene Expression Regulation, Enzymologic/drug effects , MicroRNAs/physiology , Antigens, Neoplasm/genetics , Antineoplastic Agents/toxicity , Cell Line, Tumor , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Down-Regulation/drug effects , Drug Resistance, Neoplasm , Gene Expression Regulation, Enzymologic/physiology , Humans , Poly-ADP-Ribose Binding Proteins , Teniposide/toxicity , Up-Regulation/drug effects
14.
J Pineal Res ; 46(1): 87-94, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18798788

ABSTRACT

Through inhibitory G protein-coupled melatonin receptors, melatonin regulates intracellular signaling systems and also the transcriptional activity of certain genes. Clock genes are proposed as regulatory factors in forming dopamine-related behaviors and mood and melatonin has the ability to regulate these processes. Melatonin-mediated changes in clock gene expression have been reported in brain regions, including the striatum, that are crucial for the development of dopaminergic behaviors and mood. However, it is not known whether melatonin receptors present in striatum mediate these effects. Therefore, we investigated the role of the melatonin/melatonin receptor system on clock gene expression using a model of primary neuronal cultures prepared from striatum. We found that melatonin at the receptor affinity range (i.e., nm) affects the expression of the clock genes mPer1, mClock, mBmal1 and mNPAS2 (neuronal PAS domain protein 2) differentially in a pertussis toxin-sensitive manner: a decrease in Per1 and Clock, an increase in NPAS2 and no change in Bmal1 expression. Furthermore, mutating MT1 melatonin receptor (i.e., MT1 knockouts, MT1(-/-)) reversed melatonin-induced changes, indicating the involvement of MT1 receptor in the regulatory action of melatonin on neuronal clock gene expression. Therefore, by controlling clock gene expression we propose melatonin receptors (i.e., MT1) as novel therapeutic targets for the pathobiologies of dopamine-related behaviors and mood.


Subject(s)
CLOCK Proteins/genetics , Corpus Striatum/physiology , Melatonin/physiology , Neurons/physiology , Receptor, Melatonin, MT1/genetics , Analysis of Variance , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , CLOCK Proteins/biosynthesis , CLOCK Proteins/metabolism , Cells, Cultured , Corpus Striatum/cytology , Corpus Striatum/metabolism , Cyclic AMP/metabolism , Female , Male , Melatonin/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred ICR , Mice, Knockout , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Period Circadian Proteins/biosynthesis , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Pertussis Toxin/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Melatonin, MT1/biosynthesis , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/biosynthesis , Receptor, Melatonin, MT2/genetics , Receptor, Melatonin, MT2/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL