32 results on '"Arslan, Ad"'
Search Results
2. Downregulation of GATA1 drives impaired hematopoiesis in primary myelofibrosis
- Author
-
Sandeep Gurbuxani, John D. Crispino, William Vainchenker, Laure Gilles, Leonidas C. Platanias, Qiong Yang, Brady L. Stein, Katerina Konstantinoff, Qiang Jeremy Wen, Ahmet Dirim Arslan, Christian Marinaccio, Sriram Sundaravel, Maureen McNulty, Isabelle Plo, Priyanka Arya, Ayalew Tefferi, Amittha Wickrema, Anna Rita Migliaccio, Jonathan C. Zhao, Terra L. Lasho, Animesh Pardanani, Gilles, L, Arslan, Ad, Marinaccio, C, Wen, Qj, Arya, P, Mcnulty, M, Yang, Q, Zhao, Jc, Konstantinoff, K, Lasho, T, Pardanani, A, Stein, B, Plo, I, Sundaravel, S, Wickrema, A, FRANCO MIGLIACCIO, ANNA RITA, Gurbuxani, S, Vainchenker, W, Platanias, Lc, Tefferi, A, and Crispino, J. D.
- Subjects
Ribosomal Proteins ,0301 basic medicine ,CD34 ,Down-Regulation ,Biology ,Thrombopoiesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,medicine ,Animals ,Humans ,Downregulation GATA1 hematopoiesis myelofibrosis ,GATA1 Transcription Factor ,Myelofibrosis ,Megakaryopoiesis ,Brief Report ,Myeloid leukemia ,GATA1 ,General Medicine ,medicine.disease ,Extramedullary hematopoiesis ,Haematopoiesis ,030104 developmental biology ,Primary Myelofibrosis ,030220 oncology & carcinogenesis ,Cancer research ,Chromosomes, Human, Pair 5 ,Chromosome Deletion ,Megakaryocytes - 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.
- Published
- 2017
3. Schlafen 5 as a novel therapeutic target in pancreatic ductal adenocarcinoma.
- Author
-
Fischietti M, Eckerdt F, Blyth GT, Arslan AD, Mati WM, Oku CV, Perez RE, Lee-Chang C, Kosciuczuk EM, Saleiro D, Beauchamp EM, Lesniak MS, Verzella D, Sun L, Fish EN, Yang GY, Qiang W, and Platanias LC
- Subjects
- Animals, Humans, Mice, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Line, Tumor, Cell Proliferation, Gene Expression Regulation, Neoplastic, Molecular Targeted Therapy methods, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal pathology, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal drug therapy, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Pancreatic Neoplasms metabolism
- Abstract
We provide evidence that a member of the human Schlafen (SLFN) family of proteins, SLFN5, is overexpressed in human pancreatic ductal adenocarcinoma (PDAC). Targeted deletion of SLFN5 results in decreased PDAC cell proliferation and suppresses PDAC tumorigenesis in in vivo PDAC models. Importantly, high expression levels of SLFN5 correlate with worse outcomes in PDAC patients, implicating SLFN5 in the pathophysiology of PDAC that leads to poor outcomes. Our studies establish novel regulatory effects of SLFN5 on cell cycle progression through binding/blocking of the transcriptional repressor E2F7, promoting transcription of key genes that stimulate S phase progression. Together, our studies suggest an essential role for SLFN5 in PDAC and support the potential for developing new therapeutic approaches for the treatment of pancreatic cancer through SLFN5 targeting.
- Published
- 2021
- Full Text
- View/download PDF
4. Identification and targeting of novel CDK9 complexes in acute myeloid leukemia.
- Author
-
Beauchamp EM, Abedin SM, Radecki SG, Fischietti M, Arslan AD, Blyth GT, Yang A, Lantz C, Nelson A, Goo YA, Akpan I, Eklund EA, Frankfurt O, Fish EN, Thomas PM, Altman JK, and Platanias LC
- Subjects
- 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, 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
- 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.
- Published
- 2019
- Full Text
- View/download PDF
5. IFN-γ-inducible antiviral responses require ULK1-mediated activation of MLK3 and ERK5.
- Author
-
Saleiro D, Blyth GT, Kosciuczuk EM, Ozark PA, Majchrzak-Kita B, Arslan AD, Fischietti M, Reddy NK, Horvath CM, Davis RJ, Fish EN, and Platanias LC
- Subjects
- Animals, Autophagy, Beclin-1 metabolism, Cell Line, Tumor, Class III Phosphatidylinositol 3-Kinases metabolism, Cytokines metabolism, Gene Expression Profiling, Humans, Immunity, Innate, Mice, Multigene Family, Phosphorylation, Protein Binding, Receptors, Interferon metabolism, Recombinant Proteins metabolism, Signal Transduction, Transcription, Genetic, U937 Cells, Virus Diseases metabolism, Interferon gamma Receptor, Mitogen-Activated Protein Kinase Kinase Kinase 11, Autophagy-Related Protein-1 Homolog metabolism, Interferon-gamma metabolism, Intracellular Signaling Peptides and Proteins metabolism, MAP Kinase Kinase Kinases metabolism, Mitogen-Activated Protein Kinase 7 metabolism
- Abstract
It is well established that activation of the transcription factor signal transducer and activator of transcription 1 (STAT1) is required for the interferon-γ (IFN-γ)-mediated antiviral response. Here, we found that IFN-γ receptor stimulation also activated Unc-51-like kinase 1 (ULK1), an initiator of Beclin-1-mediated autophagy. Furthermore, the interaction between ULK1 and the mitogen-activated protein kinase kinase kinase MLK3 (mixed lineage kinase 3) was necessary for MLK3 phosphorylation and downstream activation of the kinase ERK5. This autophagy-independent activity of ULK1 promoted the transcription of key antiviral IFN-stimulated genes (ISGs) and was essential for IFN-γ-dependent antiviral effects. These findings define a previously unknown IFN-γ pathway that appears to be a key element of the antiviral response., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2018
- Full Text
- View/download PDF
6. Slfn2 Regulates Type I Interferon Responses by Modulating the NF-κB Pathway.
- Author
-
Fischietti M, Arslan AD, Sassano A, Saleiro D, Majchrzak-Kita B, Ebine K, Munshi HG, Fish EN, and Platanias LC
- Subjects
- Animals, Binding Sites genetics, Cell Cycle Proteins deficiency, Cell Cycle Proteins genetics, Cells, Cultured, Gene Expression, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Models, Biological, NIH 3T3 Cells, Phosphoprotein Phosphatases metabolism, Promoter Regions, Genetic, Signal Transduction, Cell Cycle Proteins metabolism, Interferon Type I metabolism, NF-kappa B metabolism
- Abstract
Although members of the Slfn family have been implicated in the regulation of type I interferon (IFN) responses, the mechanisms by which they mediate their effects remain unknown. In the present study, we provide evidence that targeted disruption of the Slfn2 gene leads to increased transcription of IFN-stimulated genes (ISGs) and enhanced type I IFN-mediated antiviral responses. We demonstrate that Slfn2 interacts with protein phosphatase 6 regulatory subunit 1 (PPP6R1), leading to reduced type I IFN-induced activation of nuclear factor kappa B (NF-κB) signaling, resulting in reduced expression of ISGs. Altogether, these data suggest a novel mechanism by which Slfn2 controls ISG expression and provide evidence for a critical role for Slfn2 in the regulation of IFN-mediated biological responses., (Copyright © 2018 American Society for Microbiology.)
- Published
- 2018
- Full Text
- View/download PDF
7. Human SLFN5 is a transcriptional co-repressor of STAT1-mediated interferon responses and promotes the malignant phenotype in glioblastoma.
- Author
-
Arslan AD, Sassano A, Saleiro D, Lisowski P, Kosciuczuk EM, Fischietti M, Eckerdt F, Fish EN, and Platanias LC
- Subjects
- Carcinogenesis genetics, Cell Cycle Proteins metabolism, Female, Gene Expression Regulation, Neoplastic, Glioblastoma drug therapy, Glioblastoma pathology, Humans, Interferons therapeutic use, Male, Multiprotein Complexes genetics, STAT1 Transcription Factor metabolism, Transcription, Genetic, Tumor Cells, Cultured, Cell Cycle Proteins genetics, Glioblastoma genetics, Interferons genetics, STAT1 Transcription Factor genetics
- Abstract
We provide evidence that the IFN-regulated member of the Schlafen (SLFN) family of proteins, SLFN5, promotes the malignant phenotype in glioblastoma multiforme (GBM). Our studies indicate that SLFN5 expression promotes motility and invasiveness of GBM cells, and that high levels of SLFN5 expression correlate with high-grade gliomas and shorter overall survival in patients suffering from GBM. In efforts to uncover the mechanism by which SLFN5 promotes GBM tumorigenesis, we found that this protein is a transcriptional co-repressor of STAT1. Type-I IFN treatment triggers the interaction of STAT1 with SLFN5, and the resulting complex negatively controls STAT1-mediated gene transcription via interferon stimulated response elements. Thus, SLFN5 is both an IFN-stimulated response gene and a repressor of IFN-gene transcription, suggesting the existence of a negative-feedback regulatory loop that may account for suppression of antitumor immune responses in glioblastoma.
- Published
- 2017
- Full Text
- View/download PDF
8. Downregulation of GATA1 drives impaired hematopoiesis in primary myelofibrosis.
- Author
-
Gilles L, Arslan AD, Marinaccio C, Wen QJ, Arya P, McNulty M, Yang Q, Zhao JC, Konstantinoff K, Lasho T, Pardanani A, Stein B, Plo I, Sundaravel S, Wickrema A, Migliaccio A, Gurbuxani S, Vainchenker W, Platanias LC, Tefferi A, and Crispino JD
- Subjects
- 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, Down-Regulation, GATA1 Transcription Factor biosynthesis, Megakaryocytes metabolism, Primary Myelofibrosis metabolism, Thrombopoiesis
- 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.
- Published
- 2017
- Full Text
- View/download PDF
9. MNK Inhibition Disrupts Mesenchymal Glioma Stem Cells and Prolongs Survival in a Mouse Model of Glioblastoma.
- Author
-
Bell JB, Eckerdt FD, Alley K, Magnusson LP, Hussain H, Bi Y, Arslan AD, Clymer J, Alvarez AA, Goldman S, Cheng SY, Nakano I, Horbinski C, Davuluri RV, James CD, and Platanias LC
- Subjects
- 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, 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
- 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., Competing Interests: None, (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
10. Differential Regulation of ZEB1 and EMT by MAPK-Interacting Protein Kinases (MNK) and eIF4E in Pancreatic Cancer.
- Author
-
Kumar K, Chow CR, Ebine K, Arslan AD, Kwok B, Bentrem DJ, Eckerdt FD, Platanias LC, and Munshi HG
- Subjects
- Carcinoma, Pancreatic Ductal metabolism, Cell Culture Techniques, Cell Line, Tumor, Cell Movement drug effects, Gene Expression Regulation, Neoplastic drug effects, Humans, MicroRNAs genetics, Pancreatic Neoplasms metabolism, Phosphorylation, Protein Kinase Inhibitors pharmacology, Zinc Finger E-box-Binding Homeobox 1, Carcinoma, Pancreatic Ductal genetics, Epithelial-Mesenchymal Transition, Eukaryotic Initiation Factor-4E metabolism, Homeodomain Proteins genetics, Pancreatic Neoplasms genetics, Protein Serine-Threonine Kinases metabolism, Transcription Factors genetics
- Abstract
Unlabelled: Human pancreatic ductal adenocarcinoma (PDAC) tumors are associated with dysregulation of mRNA translation. In this report, it is demonstrated that PDAC cells grown in collagen exhibit increased activation of the MAPK-interacting protein kinases (MNK) that mediate eIF4E phosphorylation. Pharmacologic and genetic targeting of MNKs reverse epithelial-mesenchymal transition (EMT), decrease cell migration, and reduce protein expression of the EMT-regulator ZEB1 without affecting ZEB1 mRNA levels. Paradoxically, targeting eIF4E, the best-characterized effector of MNKs, increases ZEB1 mRNA expression through repression of ZEB1-targeting miRNAs, miR-200c and miR-141. In contrast, targeting the MNK effector hnRNPA1, which can function as a translational repressor, increases ZEB1 protein without increasing ZEB1 mRNA levels. Importantly, treatment with MNK inhibitors blocks growth of chemoresistant PDAC cells in collagen and decreases the number of aldehyde dehydrogenase activity-positive (Aldefluor+) cells. Significantly, MNK inhibitors increase E-cadherin mRNA levels and decrease vimentin mRNA levels in human PDAC organoids without affecting ZEB1 mRNA levels. Importantly, MNK inhibitors also decrease growth of human PDAC organoids., Implications: These results demonstrate differential regulation of ZEB1 and EMT by MNKs and eIF4E, and identify MNKs as potential targets in pancreatic cancer., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
11. Interferon γ (IFNγ) Signaling via Mechanistic Target of Rapamycin Complex 2 (mTORC2) and Regulatory Effects in the Generation of Type II Interferon Biological Responses.
- Author
-
Kroczynska B, Rafidi RL, Majchrzak-Kita B, Kosciuczuk EM, Blyth GT, Jemielity J, Warminska Z, Saleiro D, Mehrotra S, Arslan AD, Fish EN, and Platanias LC
- Subjects
- 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, Carrier Proteins metabolism, Eukaryotic Initiation Factor-4F metabolism, Interferon-gamma metabolism, Multiprotein Complexes metabolism, Receptors, Interferon metabolism, Signal Transduction, TOR Serine-Threonine Kinases metabolism
- 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., (© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
12. A simple, low-cost staining method for rapid-throughput analysis of tumor spheroids.
- Author
-
Eckerdt F, Alvarez A, Bell J, Arvanitis C, Iqbal A, Arslan AD, Hu B, Cheng SY, Goldman S, and Platanias LC
- Subjects
- Acridine Orange chemistry, Biological Assay methods, Brain Neoplasms diagnosis, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Glioblastoma diagnosis, Humans, Neoplastic Stem Cells pathology, Spheroids, Cellular pathology, Brain Neoplasms pathology, Glioblastoma pathology, Staining and Labeling methods
- Abstract
Tumor spheroids are becoming an important tool for the investigation of cancer stem cell (CSC) function in tumors; thus, low-cost and high-throughput methods for drug screening of tumor spheroids are needed. Using neurospheres as non-adherent three-dimensional (3-D) cultures, we developed a simple, low-cost acridine orange (AO)-based method that allows for rapid analysis of live neurospheres by fluorescence microscopy in a 96-well format. This assay measures the cross-section area of a spheroid, which corresponds to cell viability. Our novel method allows rapid screening of a panel of anti-proliferative drugs to assess inhibitory effects on the growth of cancer stem cells in 3-D cultures.
- Published
- 2016
- Full Text
- View/download PDF
13. Pre-clinical evidence of PIM kinase inhibitor activity in BCR-ABL1 unmutated and mutated Philadelphia chromosome-positive (Ph+) leukemias.
- Author
-
Curi DA, Beauchamp EM, Blyth GT, Arslan AD, Donato NJ, Giles FJ, Altman JK, and Platanias LC
- Subjects
- Dose-Response Relationship, Drug, Humans, Imatinib Mesylate pharmacology, K562 Cells, Mutation, Philadelphia Chromosome, Proto-Oncogene Proteins c-pim-1 antagonists & inhibitors, Tumor Cells, Cultured, 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
- 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.
- Published
- 2015
- Full Text
- View/download PDF
14. Human Schlafen 5 (SLFN5) Is a Regulator of Motility and Invasiveness of Renal Cell Carcinoma Cells.
- Author
-
Sassano A, Mavrommatis E, Arslan AD, Kroczynska B, Beauchamp EM, Khuon S, Chew TL, Green KJ, Munshi HG, Verma AK, and Platanias LC
- Subjects
- 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, Carcinoma, Renal Cell pathology, Cell Cycle Proteins genetics, Kidney Neoplasms pathology, Matrix Metalloproteinase 1 biosynthesis, Matrix Metalloproteinase 13 biosynthesis
- 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., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
15. Regulatory effects of SKAR in interferon α signaling and its role in the generation of type I IFN responses.
- Author
-
Kroczynska B, Mehrotra S, Majchrzak-Kita B, Arslan AD, Altman JK, Stein BL, McMahon B, Kozlowski P, Kahle PJ, Eklund EA, Fish EN, and Platanias LC
- Subjects
- 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, Ubiquitins metabolism, Interferon-alpha metabolism, Nuclear Proteins metabolism, RNA-Binding Proteins metabolism, Signal Transduction drug effects
- 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.
- Published
- 2014
- Full Text
- View/download PDF
16. Regulation of interferon-dependent mRNA translation of target genes.
- Author
-
Kroczynska B, Mehrotra S, Arslan AD, Kaur S, and Platanias LC
- Subjects
- 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, Autoimmune Diseases immunology, Interferon Type I metabolism, Interferon-gamma metabolism, Neoplasms immunology, RNA, Messenger metabolism, Receptor, Interferon alpha-beta metabolism, Virus Diseases immunology
- 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.
- Published
- 2014
- Full Text
- View/download PDF
17. Critical roles for Rictor/Sin1 complexes in interferon-dependent gene transcription and generation of antiproliferative responses.
- Author
-
Kaur S, Kroczynska B, Sharma B, Sassano A, Arslan AD, Majchrzak-Kita B, Stein BL, McMahon B, Altman JK, Su B, Calogero RA, Fish EN, and Platanias LC
- Subjects
- 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, Antineoplastic Agents pharmacology, Carrier Proteins metabolism, Gene Expression Regulation, Neoplastic drug effects, Interferon Type I pharmacology, Transcription, Genetic drug effects
- 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.
- Published
- 2014
- Full Text
- View/download PDF
18. Involvement of polypyrimidine tract-binding protein (PTBP1) in maintaining breast cancer cell growth and malignant properties.
- Author
-
He X, Arslan AD, Ho TT, Yuan C, Stampfer MR, and Beck WT
- Abstract
We have investigated some roles of splicing factor polypyrimidine tract-binding protein (PTBP1) in human breast cancer. We found that PTBP1 was upregulated in progressively transformed human mammary epithelial cells (HMECs), as well as in breast tumor cell lines compared with HMECs with finite growth potential and found that the level of PTBP1 correlated with the transformation state of HMECs. Knockdown of PTBP1 expression substantially inhibited tumor cell growth, colony formation in soft agar and in vitro invasiveness of breast cancer cell lines, a result similar to what we have reported in ovarian cancer. However, ectopic expression of PTBP1 (as a PTBP1-EGFP fusion protein) did not enhance the proliferation of immortalized HMEC. Rather, PTBP1 expression promoted anchorage-independent growth of an immortalized HMEC as assessed by increased colony formation in soft agar. In addition, we found that knockdown of PTBP1 expression led to upregulation of the expression of the M1 isoform of pyruvate kinase (PKM1) and increase of the ratio of PKM1 vs PKM2. PKM1 has been reported to promote oxidative phosphorylation and reduce tumorigenesis. Correspondingly, we observed increased oxygen consumption in PTBP1-knockdown breast cancer cells. Together, these results suggest that PTBP1 is associated with breast tumorigenesis and appears to be required for tumor cell growth and maintenance of transformed properties. PTBP1 exerts these effects, in part, by regulating the splicing of pyruvate kinase, and consequently alters glucose metabolism and contributes to the Warburg effect.
- Published
- 2014
- Full Text
- View/download PDF
19. Concordance of deregulated mechanisms unveiled in underpowered experiments: PTBP1 knockdown case study.
- Author
-
Gardeux V, Arslan AD, Achour I, Ho TT, Beck WT, and Lussier YA
- Subjects
- Cell Line, Tumor, Humans, Molecular Sequence Annotation, Neurons cytology, Neurons metabolism, RNA, Small Interfering genetics, Sequence Analysis, RNA, Computational Biology methods, Gene Expression Profiling, Gene Knockdown Techniques, Heterogeneous-Nuclear Ribonucleoproteins deficiency, Heterogeneous-Nuclear Ribonucleoproteins genetics, Polypyrimidine Tract-Binding Protein deficiency, Polypyrimidine Tract-Binding Protein genetics
- Abstract
Background: Genome-wide transcriptome profiling generated by microarray and RNA-Seq often provides deregulated genes or pathways applicable only to larger cohort. On the other hand, individualized interpretation of transcriptomes is increasely pursued to improve diagnosis, prognosis, and patient treatment processes. Yet, robust and accurate methods based on a single paired-sample remain an unmet challenge., Methods: "N-of-1-pathways" translates gene expression data profiles into mechanism-level profiles on single pairs of samples (one p-value per geneset). It relies on three principles: i) statistical universe is a single paired sample, which serves as its own control; ii) statistics can be derived from multiple gene expression measures that share common biological mechanisms assimilated to genesets; iii) semantic similarity metric takes into account inter-mechanisms' relationships to better assess commonality and differences, within and cross study-samples (e.g. patients, cell-lines, tissues, etc.), which helps the interpretation of the underpinning biology., Results: In the context of underpowered experiments, N-of-1-pathways predictions perform better or comparable to those of GSEA and Differentially Expressed Genes enrichment (DEG enrichment), within-and cross-datasets. N-of-1-pathways uncovered concordant PTBP1-dependent mechanisms across datasets (Odds-Ratios≥13, p-values≤1 × 10-5), such as RNA splicing and cell cycle. In addition, it unveils tissue-specific mechanisms of alternatively transcribed PTBP1-dependent genesets. Furthermore, we demonstrate that GSEA and DEG Enrichment preclude accurate analysis on single paired samples., Conclusions: N-of-1-pathways enables robust and biologically relevant mechanism-level classifiers with small cohorts and one single paired samples that surpasses conventional methods. Further, it identifies unique sample/ patient mechanisms, a requirement for precision medicine.
- Published
- 2014
- Full Text
- View/download PDF
20. Regulatory effects of sestrin 3 (SESN3) in BCR-ABL expressing cells.
- Author
-
Vakana E, Arslan AD, Szilard A, Altman JK, and Platanias LC
- Subjects
- 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, Fusion Proteins, bcr-abl biosynthesis, Gene Expression Regulation, Leukemic, Heat-Shock Proteins biosynthesis, Leukemia, Myelogenous, Chronic, BCR-ABL Positive metabolism
- 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.
- Published
- 2013
- Full Text
- View/download PDF
21. Expression and regulatory effects of murine Schlafen (Slfn) genes in malignant melanoma and renal cell carcinoma.
- Author
-
Mavrommatis E, Arslan AD, Sassano A, Hua Y, Kroczynska B, and Platanias LC
- Subjects
- 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, Carcinoma, Renal Cell metabolism, Cell Cycle Proteins metabolism, Gene Expression Regulation, Neoplastic, Kidney Neoplasms metabolism, Melanoma metabolism, Neoplasm Proteins metabolism
- 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.
- Published
- 2013
- Full Text
- View/download PDF
22. A high-throughput assay to identify small-molecule modulators of alternative pre-mRNA splicing.
- Author
-
Arslan AD, He X, Wang M, Rumschlag-Booms E, Rong L, and Beck WT
- Subjects
- 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, Alternative Splicing drug effects, High-Throughput Screening Assays methods, Small Molecule Libraries
- 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.
- Published
- 2013
- Full Text
- View/download PDF
23. Novel regulation of nuclear factor-YB by miR-485-3p affects the expression of DNA topoisomerase IIα and drug responsiveness.
- Author
-
Chen CF, He X, Arslan AD, Mo YY, Reinhold WC, Pommier Y, and Beck WT
- Subjects
- 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, 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
- 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.
- Published
- 2011
- Full Text
- View/download PDF
24. Knockdown of splicing factor SRp20 causes apoptosis in ovarian cancer cells and its expression is associated with malignancy of epithelial ovarian cancer.
- Author
-
He X, Arslan AD, Pool MD, Ho TT, Darcy KM, Coon JS, and Beck WT
- Subjects
- Base Sequence, Cell Division genetics, DNA Primers, Female, Humans, Immunohistochemistry, Neoplasms, Glandular and Epithelial genetics, Ovarian Neoplasms genetics, RNA Interference, RNA-Binding Proteins genetics, Serine-Arginine Splicing Factors, Gene Knockdown Techniques, Neoplasms, Glandular and Epithelial pathology, Ovarian Neoplasms pathology, RNA-Binding Proteins physiology
- Abstract
Our previous study revealed that two splicing factors, polypyrimidine tract-binding protein (PTB) and SRp20, were upregulated in epithelial ovarian cancer (EOC) and knockdown of PTB expression inhibited ovarian tumor cell growth and transformation properties. In this report, we show that knockdown of SRp20 expression in ovarian cancer cells also causes substantial inhibition of tumor cell growth and colony formation in soft agar and the extent of such inhibition appeared to correlate with the extent of suppression of SRp20. Massive knockdown of SRp20 expression triggered remarkable apoptosis in these cells. These results suggest that overexpression of SRp20 is required for ovarian tumor cell growth and survival. Immunohistochemical staining for PTB and SRp20 of two specialized tissue microarrays, one containing benign ovarian tumors, borderline/low malignant potential (LMP) ovarian tumors as well as invasive EOC and the other containing invasive EOC ranging from stage I to stage IV disease, reveals that PTB and SRp20 are both expressed differentially between benign tumors and invasive EOC, and between borderline/LMP tumors and invasive EOC. There were more all-negative or mixed staining cases (at least two evaluable section cores per case) in benign tumors than in invasive EOC, whereas there were more all-positive staining cases in invasive EOC than in the other two disease classifications. Among invasive EOC, the majority of cases were stained all positive for both PTB and SRp20, and there were no significant differences in average staining or frequency of positive cancer cells between any of the tumor stages. Therefore, the expression of PTB and SRp20 is associated with malignancy of ovarian tumors but not with stage of invasive EOC.
- Published
- 2011
- Full Text
- View/download PDF
25. The melatonin receptor MT1 is required for the differential regulatory actions of melatonin on neuronal 'clock' gene expression in striatal neurons in vitro.
- Author
-
Imbesi M, Arslan AD, Yildiz S, Sharma R, Gavin D, Tun N, Manev H, and Uz T
- Subjects
- 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, CLOCK Proteins genetics, Corpus Striatum physiology, Melatonin physiology, Neurons physiology, Receptor, Melatonin, MT1 genetics
- 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.
- Published
- 2009
- Full Text
- View/download PDF
26. A new approach in the treatment of skeletal open bite: vertically activated bite block.
- Author
-
Arat ZM, Sezer FE, and Arslan AD
- Subjects
- Adolescent, Adult, Cephalometry, Child, Female, Humans, Male, Molar physiopathology, Orthodontic Appliance Design, Vertical Dimension, Open Bite therapy, Orthodontic Appliances, Removable, Tooth Movement Techniques instrumentation
- Abstract
This study examined the effects of the vertically activated bite block, a new appliance designed for maxillary molar intrusion in open bite malocclusion, on the dentofacial structures in 1 adolescent patient and 1 adult patient. The vertically activated bite block consists of 2 parts, the main block and an auxiliary segment. The auxilliary segment is separated bilaterally from the main block in the posterior region and connected bilaterally to the main block in the palatal region by open jackscrews. Closing the jackscrews moves the auxiliary segment toward the palate, thereby inducing molar intrusion. The present study achieved a reduction in open bite, using the VABB through: (1) active intrusion of the maxillary molars; (2) control of posterior vertical alveolar development and/or compensatory eruption of the mandibular molars; (3) stimulation of anterior vertical alveolar development by restriction of the tongue; and (4) anterior repositioning of mandible. Achieving these dentoalveolar and skeletal changes are required treatment objectives in cases of skeletal open bite. As a result of attaining these objectives, the open bite was eliminated in 1 early growth and 1 late growth patient by 14 mm and 6 mm, respectively.
- Published
- 2006
27. Skeletal and dental effects of a mini maxillary protraction appliance.
- Author
-
Altug Z and Arslan AD
- Subjects
- Cephalometry, Child, Female, Follow-Up Studies, Humans, Incisor pathology, Male, Malocclusion, Angle Class I pathology, Malocclusion, Angle Class III pathology, Malocclusion, Angle Class III therapy, Mandible pathology, Maxilla pathology, Maxillofacial Development, Palatal Expansion Technique instrumentation, Rotation, Vertical Dimension, Extraoral Traction Appliances, Facial Bones pathology, Orthodontic Appliance Design, Tooth pathology
- Abstract
The aim of this study was to evaluate the skeletal and dentoalveolar changes achieved by a modified protractor in growing skeletal and dental Class III patients and to compare these changes with normal growth in a Class I untreated control group. The study group consisted of 25 patients (11 girls, 14 boys; mean age 11.74 +/- 1.81 years). The control group was composed of 20 patients (15 girls, five boys; mean age 11.89 +/- 1.08 years). The Class III patients were treated with a bonded acrylic cap splint type expander and a modified maxillary protractor until a positive overjet was achieved. The mean observation period was 0.65 years. Changes in study and control groups and differences between the groups were analyzed statistically. The results showed that protraction appliance produced a significant positive improvement in maxillo-mandibular relations. The forward movement of the maxilla was significant in treated Class III patients, but a slight difference was present between the two groups regarding maxillary rotation. The effective length of the maxilla was significantly increased in the Class III patients. The mandible was positioned backward, and posterior rotation of the mandible was significant in the treatment group. There was a significant increase in lower anterior facial height of treated Class III patients. The dentoalveolar measurements showed that the maxillary incisors proclined and the mandibular incisors significantly retroclined in the Class III group. A modified maxillary protractor treatment is effective for correcting skeletal Class III malocclusion.
- Published
- 2006
- Full Text
- View/download PDF
28. Impaired hippocampal long-term potentiation in melatonin MT2 receptor-deficient mice.
- Author
-
Larson J, Jessen RE, Uz T, Arslan AD, Kurtuncu M, Imbesi M, and Manev H
- Subjects
- Animals, Behavior, Animal, Dose-Response Relationship, Radiation, Electric Stimulation methods, Excitatory Postsynaptic Potentials genetics, Excitatory Postsynaptic Potentials radiation effects, In Vitro Techniques, Long-Term Potentiation radiation effects, Male, Maze Learning physiology, Mice, Mice, Knockout, Reaction Time genetics, Hippocampus physiology, Long-Term Potentiation physiology, Receptor, Melatonin, MT2 deficiency, Receptor, Melatonin, MT2 physiology
- Abstract
The pineal product melatonin that acts on specific melatonin receptors has been implicated in pathobiological mechanisms of neuropsychiatric disorders including Alzheimer's disease. We used mice lacking melatonin MT(2) receptors (MT(2) knockouts) to investigate the role of these receptors in synaptic plasticity and learning-dependent behavior. In field CA1 of hippocampal slices from wild-type mice, theta burst stimulation induced robust and stable long-term potentiation that was smaller and decremental in slices from MT(2) knockouts. Tested in an elevated plus-maze on two consecutive days, wild-type mice showed shorter transfer latencies to enter a closed arm on the second day; this experience-dependent behavior did not occur in MT(2) knockouts. These results suggest that MT(2) receptors participate in hippocampal synaptic plasticity and in memory processes.
- Published
- 2006
- Full Text
- View/download PDF
29. Drug- and region-specific effects of protracted antidepressant and cocaine treatment on the content of melatonin MT(1) and MT(2) receptor mRNA in the mouse brain.
- Author
-
Imbesi M, Uz T, Yildiz S, Arslan AD, and Manev H
- Abstract
OBJECTIVES: In the mammalian brain, G protein-coupled MT(1) and MT(2) melatonin receptors may be involved in Alzheimer's pathology, long-term potentiation, depression, and in the behavioral effects of psychoactive drugs. These drugs; e.g. antidepressants and drugs of abuse, are typically used over long periods of time and may alter neuroplasticity and gene expression. We hypothesized that such antidepressant- and cocaine-altered expression of melatonin receptor mRNA may occur in the hippocampus and striatum. METHODOLOGY: Male C3H/HeJ mice were treated with the antidepressants fluoxetine, desipramine, and clomipramine, with the psychostimulant cocaine, and with a vehicle either a single time or once a day for 14 days. Brain samples were collected 24 h after the last injection and the content of MT(1) and MT(2) mRNA was assayed. RESULTS: A single drug injection did not alter the MT(1) and MT(2) mRNA content. In the hippocampus, protracted treatment with antidepressants increased the amount of MT(1) mRNA (with the exception of fluoxetine) but decreased MT(2) mRNA content; cocaine did not produce any alterations. In the striatum, antidepressants produced the opposite effect on MT(1) mRNA content; they decreased it. They did not significantly alter striatal MT(2) mRNA (we observed a nonsignificant trend to a decrease). Cocaine also decreased striatal MT(1) mRNA content without affecting MT(2) mRNA. CONCLUSION: These results suggest that drug- and region-specific alterations of MT(1)/MT(2) mRNA produced by protracted antidepressants and cocaine treatment may alter MT1/MT2 expression and contribute to long-term neuroplastic effects of these drugs.
- Published
- 2006
30. A mini-maxillary protractor for Class III correction.
- Author
-
Altug Z and Arslan AD
- Subjects
- Cephalometry, Child, Dental Stress Analysis, Humans, Male, Extraoral Traction Appliances, Malocclusion, Angle Class III therapy, Orthodontics, Corrective instrumentation, Palatal Expansion Technique instrumentation
- Published
- 2005
31. The regional and cellular expression profile of the melatonin receptor MT1 in the central dopaminergic system.
- Author
-
Uz T, Arslan AD, Kurtuncu M, Imbesi M, Akhisaroglu M, Dwivedi Y, Pandey GN, and Manev H
- Subjects
- Animals, Blotting, Northern methods, Blotting, Western methods, Circadian Rhythm physiology, Humans, Immunohistochemistry methods, Male, Mice, Microdissection methods, RNA, Messenger biosynthesis, Rats, Receptor, Melatonin, MT1 genetics, Receptors, Dopamine D2 genetics, Receptors, Dopamine D2 metabolism, Reverse Transcriptase Polymerase Chain Reaction methods, Tyrosine 3-Monooxygenase metabolism, Brain cytology, Dopamine metabolism, Gene Expression Regulation physiology, Neurons metabolism, Receptor, Melatonin, MT1 metabolism
- Abstract
The physiological effects of pineal melatonin are primarily mediated by melatonin receptors located in the brain and periphery. Even though there are a number of studies demonstrating the regulatory role of melatonin in the development of dopaminergic behaviors, such as psychostimulant-induced diurnal locomotor sensitization or drug seeking, little is known about the contribution of melatonin receptors (i.e., MT1) to this role. Therefore, as a first step in understanding the functional role of melatonin receptors in dopaminergic behaviors, we focused on determining the expression pattern of MT1 receptors in the dopaminergic system of the human and rodent brain. Regional (e.g., nucleus accumbens shell) and cellular (e.g., tyrosine hydroxylase immunopositive cells) expression of MT1 mRNA was characterized by applying the immuno-laser capture microdissection (immuno-LCM) technique coupled with nested RT-PCR. Moreover, employing quantitative Western immunoblotting and RT-PCR, we found that the mouse MT1 receptor expression presents diurnal variations (i.e., low mRNA and high protein levels at night, ZT21). The dopaminergic system-based presence of MT1 receptor proteins was not limited to rodents; we found these receptors in postmortem human brain as well. Further research is needed to understand the regional/cellular functional role of melatonin receptors in the regulation of dopaminergic behaviors, using models such as melatonin receptor knockout mice.
- Published
- 2005
- Full Text
- View/download PDF
32. Involvement of the pineal gland in diurnal cocaine reward in mice.
- Author
-
Kurtuncu M, Arslan AD, Akhisaroglu M, Manev H, and Uz T
- Subjects
- Animals, Behavior, Animal, Brain Chemistry drug effects, Brain Chemistry genetics, Cell Cycle Proteins, Circadian Rhythm physiology, Cocaine administration & dosage, Cocaine adverse effects, Cocaine-Related Disorders genetics, Cocaine-Related Disorders metabolism, Conditioning, Psychological drug effects, Gene Expression drug effects, Gene Expression genetics, Injections, Intraperitoneal, Melatonin pharmacology, Mice, Mice, Inbred C3H, Mice, Knockout, Nuclear Proteins drug effects, Nuclear Proteins genetics, Period Circadian Proteins, Pineal Gland metabolism, Pineal Gland surgery, Serotonin pharmacology, Circadian Rhythm drug effects, Cocaine-Related Disorders psychology, Pineal Gland physiology, Serotonin analogs & derivatives
- Abstract
Contribution of circadian mechanisms to the psychostimulant-induced behaviors has been suggested. The pineal gland is important component of circadian mechanisms. Using pinealectomized mice and sham-operated controls, we tested the contribution of pineal gland to the rewarding effects of cocaine in conditioned place preference test. Experiments were performed both during the day and at night. Controls with intact pineal glands demonstrated significant decrease in cocaine-induced conditioned place preference at night compared to daytime, whereas pinealectomized mice did not show any diurnal differences. Circadian mechanisms regulated by the pineal gland thus appear critically involved in cocaine-induced reward.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.