451
|
Alipanah L, Rohloff J, Winge P, Bones AM, Brembu T. Whole-cell response to nitrogen deprivation in the diatom Phaeodactylum tricornutum. JOURNAL OF EXPERIMENTAL BOTANY 2015; 66:6281-96. [PMID: 26163699 PMCID: PMC4588885 DOI: 10.1093/jxb/erv340] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Algal growth is strongly affected by nitrogen (N) availability. Diatoms, an ecologically important group of unicellular algae, have evolved several acclimation mechanisms to cope with N deprivation. In this study, we integrated physiological data with transcriptional and metabolite data to reveal molecular and metabolic modifications in N-deprived conditions in the marine diatom Phaeodactylum tricornutum. Physiological and metabolite measurements indicated that the photosynthetic capacity and chlorophyll content of the cells decreased, while neutral lipids increased in N-deprived cultures. Global gene expression analysis showed that P. tricornutum responded to N deprivation through an increase in N transport, assimilation, and utilization of organic N resources. Following N deprivation, reduced biosynthesis and increased recycling of N compounds like amino acids, proteins, and nucleic acids was observed at the transcript level. The majority of the genes associated with photosynthesis and chlorophyll biosynthesis were also repressed. Carbon metabolism was restructured through downregulation of the Calvin cycle and chrysolaminarin biosynthesis, and co-ordinated upregulation of glycolysis, the tricarboxylic acid cycle, and pyruvate metabolism, leading to funnelling of carbon sources to lipid metabolism. Finally, reallocation of membrane lipids and induction of de novo triacylglycerol biosynthesis directed cells to accumulation of neutral lipids.
Collapse
Affiliation(s)
- Leila Alipanah
- Department of Biology, Norwegian University of Science and Technology, N-7491 Trondheim, Norway
| | - Jens Rohloff
- Department of Biology, Norwegian University of Science and Technology, N-7491 Trondheim, Norway
| | - Per Winge
- Department of Biology, Norwegian University of Science and Technology, N-7491 Trondheim, Norway
| | - Atle M Bones
- Department of Biology, Norwegian University of Science and Technology, N-7491 Trondheim, Norway
| | - Tore Brembu
- Department of Biology, Norwegian University of Science and Technology, N-7491 Trondheim, Norway
| |
Collapse
|
452
|
de Almeida MR, de Bastiani D, Gaeta ML, de Araújo Mariath JE, de Costa F, Retallick J, Nolan L, Tai HH, Strömvik MV, Fett-Neto AG. Comparative transcriptional analysis provides new insights into the molecular basis of adventitious rooting recalcitrance in Eucalyptus. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2015; 239:155-65. [PMID: 26398800 DOI: 10.1016/j.plantsci.2015.07.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 07/16/2015] [Accepted: 07/26/2015] [Indexed: 05/21/2023]
Abstract
Adventitious rooting (AR) is essential in clonal propagation. Eucalyptus globulus is relevant for the cellulose industry due to its low lignin content. However, several useful clones are recalcitrant to AR, often requiring exogenous auxin, adding cost to clonal garden operations. In contrast, E. grandis is an easy-to-root species widely used in clonal forestry. Aiming at contributing to the elucidation of recalcitrance causes in E. globulus, we conducted a comparative analysis with these two species differing in rooting competence, combining gene expression and anatomical techniques. Recalcitrance in E. globulus is reversed by exposure to exogenous indole-3-acetic acid (IAA), which promotes important gene expression modifications in both species. The endogenous content of IAA was significantly higher in E. grandis than in E. globulus. The cambium zone was identified as an active area during AR, concentrating the first cell divisions. Immunolocalization assay showed auxin accumulation in cambium cells, further indicating the importance of this region for rooting. We then performed a cambium zone-specific gene expression analysis during AR using laser microdissection. The results indicated that the auxin-related genes TOPLESS and IAA12/BODENLOS and the cytokinin-related gene ARR1may act as negative regulators of AR, possibly contributing to the hard-to-root phenotype of E. globulus.
Collapse
Affiliation(s)
- Márcia Rodrigues de Almeida
- Plant Physiology Laboratory, Center for Biotechnology and Department of Botany, Federal University of Rio Grande do Sul, P.O. Box 15005, 91501-970 Porto Alegre, RS, Brazil; Plant Gene Regulation and Bioinformatics Laboratory, Department of Plant Science, McGill University, Ste. Anne de Bellevue, QC H9X3V9, Canada
| | - Daniela de Bastiani
- Plant Physiology Laboratory, Center for Biotechnology and Department of Botany, Federal University of Rio Grande do Sul, P.O. Box 15005, 91501-970 Porto Alegre, RS, Brazil
| | - Marcos Letaif Gaeta
- Plant Anatomy Laboratory, Department of Botany, Federal University of Rio Grande do Sul, 91501-970 Porto Alegre, RS, Brazil
| | | | - Fernanda de Costa
- Plant Physiology Laboratory, Center for Biotechnology and Department of Botany, Federal University of Rio Grande do Sul, P.O. Box 15005, 91501-970 Porto Alegre, RS, Brazil
| | - Jeffrey Retallick
- Potato Research Centre, Agriculture and Agri-Food Canada, PO Box 20280, Fredericton, NB E3B 4Z7, Canada
| | - Lana Nolan
- Potato Research Centre, Agriculture and Agri-Food Canada, PO Box 20280, Fredericton, NB E3B 4Z7, Canada
| | - Helen H Tai
- Potato Research Centre, Agriculture and Agri-Food Canada, PO Box 20280, Fredericton, NB E3B 4Z7, Canada
| | - Martina V Strömvik
- Plant Gene Regulation and Bioinformatics Laboratory, Department of Plant Science, McGill University, Ste. Anne de Bellevue, QC H9X3V9, Canada
| | - Arthur Germano Fett-Neto
- Plant Physiology Laboratory, Center for Biotechnology and Department of Botany, Federal University of Rio Grande do Sul, P.O. Box 15005, 91501-970 Porto Alegre, RS, Brazil.
| |
Collapse
|
453
|
Voets E, Marsman J, Demmers J, Beijersbergen R, Wolthuis R. The lethal response to Cdk1 inhibition depends on sister chromatid alignment errors generated by KIF4 and isoform 1 of PRC1. Sci Rep 2015; 5:14798. [PMID: 26423135 PMCID: PMC4589785 DOI: 10.1038/srep14798] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 08/24/2015] [Indexed: 12/18/2022] Open
Abstract
Cyclin-dependent kinase 1 (Cdk1) is absolutely essential for cell division. Complete ablation of Cdk1 precludes the entry of G2 phase cells into mitosis, and is early embryonic lethal in mice. Dampening Cdk1 activation, by reducing gene expression or upon treatment with cell-permeable Cdk1 inhibitors, is also detrimental for proliferating cells, but has been associated with defects in mitotic progression, and the formation of aneuploid daughter cells. Here, we used a large-scale RNAi screen to identify the human genes that critically determine the cellular toxicity of Cdk1 inhibition. We show that Cdk1 inhibition leads to fatal sister chromatid alignment errors and mitotic arrest in the spindle checkpoint. These problems start early in mitosis and are alleviated by depletion of isoform 1 of PRC1 (PRC1-1), by gene ablation of its binding partner KIF4, or by abrogation of KIF4 motor activity. Our results show that, normally, Cdk1 activity must rise above the level required for mitotic entry. This prevents KIF4-dependent PRC1-1 translocation to astral microtubule tips and safeguards proper chromosome congression. We conclude that cell death in response to Cdk1 inhibitors directly relates to chromosome alignment defects generated by insufficient repression of PRC1-1 and KIF4 during prometaphase.
Collapse
Affiliation(s)
- Erik Voets
- Division of Cell Biology I (B5) and Division of Molecular Carcinogenesis (B7), The Netherlands Cancer Insitute (NKI-AvL), Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Judith Marsman
- Division of Cell Biology I (B5) and Division of Molecular Carcinogenesis (B7), The Netherlands Cancer Insitute (NKI-AvL), Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Jeroen Demmers
- Proteomics Center, Erasmus University Medical Center, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | - Roderick Beijersbergen
- Division of Cell Biology I (B5) and Division of Molecular Carcinogenesis (B7), The Netherlands Cancer Insitute (NKI-AvL), Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Rob Wolthuis
- Division of Cell Biology I (B5) and Division of Molecular Carcinogenesis (B7), The Netherlands Cancer Insitute (NKI-AvL), Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.,Section of Oncogenetics, Department of Clinical Genetics and CCA/V-ICI Research Program Oncogenesis, VUmc Medical Faculty, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| |
Collapse
|
454
|
Barbosa C, Tan ZY, Wang R, Xie W, Strong JA, Patel RR, Vasko MR, Zhang JM, Cummins TR. Navβ4 regulates fast resurgent sodium currents and excitability in sensory neurons. Mol Pain 2015; 11:60. [PMID: 26408173 PMCID: PMC4582632 DOI: 10.1186/s12990-015-0063-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 09/10/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Increased electrical activity in peripheral sensory neurons including dorsal root ganglia (DRG) and trigeminal ganglia neurons is an important mechanism underlying pain. Voltage gated sodium channels (VGSC) contribute to the excitability of sensory neurons and are essential for the upstroke of action potentials. A unique type of VGSC current, resurgent current (INaR), generates an inward current at repolarizing voltages through an alternate mechanism of inactivation referred to as open-channel block. INaRs are proposed to enable high frequency firing and increased INaRs in sensory neurons are associated with pain pathologies. While Nav1.6 has been identified as the main carrier of fast INaR, our understanding of the mechanisms that contribute to INaR generation is limited. Specifically, the open-channel blocker in sensory neurons has not been identified. Previous studies suggest Navβ4 subunit mediates INaR in central nervous system neurons. The goal of this study was to determine whether Navβ4 regulates INaR in DRG sensory neurons. RESULTS Our immunocytochemistry studies show that Navβ4 expression is highly correlated with Nav1.6 expression predominantly in medium-large diameter rat DRG neurons. Navβ4 knockdown decreased endogenous fast INaR in medium-large diameter neurons as measured with whole-cell voltage clamp. Using a reduced expression system in DRG neurons, we isolated recombinant human Nav1.6 sodium currents in rat DRG neurons and found that overexpression of Navβ4 enhanced Nav1.6 INaR generation. By contrast neither overexpression of Navβ2 nor overexpression of a Navβ4-mutant, predicted to be an inactive form of Navβ4, enhanced Nav1.6 INaR generation. DRG neurons transfected with wild-type Navβ4 exhibited increased excitability with increases in both spontaneous activity and evoked activity. Thus, Navβ4 overexpression enhanced INaR and excitability, whereas knockdown or expression of mutant Navβ4 decreased INaR generation. CONCLUSION INaRs are associated with inherited and acquired pain disorders. However, our ability to selectively target and study this current has been hindered due to limited understanding of how it is generated in sensory neurons. This study identified Navβ4 as an important regulator of INaR and excitability in sensory neurons. As such, Navβ4 is a potential target for the manipulation of pain sensations.
Collapse
Affiliation(s)
- Cindy Barbosa
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, USA. .,Department of Pharmacology and Toxicology, Stark Neurosciences Research Institute, 320 West 25th Street, NB-414F, Indianapolis, IN, 46202-2266, USA.
| | - Zhi-Yong Tan
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, USA. .,Department of Pharmacology and Toxicology, Stark Neurosciences Research Institute, 320 West 25th Street, NB-414F, Indianapolis, IN, 46202-2266, USA.
| | - Ruizhong Wang
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, USA.
| | - Wenrui Xie
- Department of Anesthesiology, University of Cincinnati, Cincinnati, OH, USA.
| | - Judith A Strong
- Department of Anesthesiology, University of Cincinnati, Cincinnati, OH, USA.
| | - Reesha R Patel
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, USA. .,Department of Pharmacology and Toxicology, Stark Neurosciences Research Institute, 320 West 25th Street, NB-414F, Indianapolis, IN, 46202-2266, USA.
| | - Michael R Vasko
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, USA. .,Department of Pharmacology and Toxicology, Stark Neurosciences Research Institute, 320 West 25th Street, NB-414F, Indianapolis, IN, 46202-2266, USA.
| | - Jun-Ming Zhang
- Department of Anesthesiology, University of Cincinnati, Cincinnati, OH, USA.
| | - Theodore R Cummins
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, USA. .,Department of Pharmacology and Toxicology, Stark Neurosciences Research Institute, 320 West 25th Street, NB-414F, Indianapolis, IN, 46202-2266, USA.
| |
Collapse
|
455
|
Poli A, Ramazzotti G, Matteucci A, Manzoli L, Lonetti A, Suh PG, McCubrey JA, Cocco L. A novel DAG-dependent mechanism links PKCɑ and Cyclin B1 regulating cell cycle progression. Oncotarget 2015; 5:11526-40. [PMID: 25362646 PMCID: PMC4294327 DOI: 10.18632/oncotarget.2578] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 10/06/2014] [Indexed: 01/11/2023] Open
Abstract
Through the years, different studies showed the involvement of Protein Kinase C (PKC) in cell cycle control, in particular during G1/S transition. Little is known about their role at G2/M checkpoint. In this study, using K562 human erythroleukemia cell line, we found a novel and specific mechanism through which the conventional isoform PKC⍺ positively affects Cyclin B1 modulating G2/M progression of cell cycle. Since the kinase activity of this PKC isoform was not necessary in this process, we demonstrated that PKC⍺, physically interacting with Cyclin B1, avoided its degradation and stimulated its nuclear import at mitosis. Moreover, the process resulted to be strictly connected with the increase in nuclear diacylglycerol levels (DAG) at G2/M checkpoint, due to the activity of nuclear Phospholipase C β1 (PLCβ1), the only PLC isoform mainly localized in the nucleus of K562 cells. Taken together, our findings indicated a novel DAG dependent mechanism able to regulate the G2/M progression of the cell cycle.
Collapse
Affiliation(s)
- Alessandro Poli
- Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy
| | - Giulia Ramazzotti
- Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy
| | - Alessandro Matteucci
- CNR-National Research Council of Italy, Institute of Molecular Genetics, Bologna, Italy
| | - Lucia Manzoli
- Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy
| | - Annalisa Lonetti
- Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy
| | - Pann-Ghill Suh
- School of Nano-Biotechnology and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Lucio Cocco
- Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
456
|
Okada M, Ohtake F, Nishikawa H, Wu W, Saeki Y, Takana K, Ohta T. Liganded ERα Stimulates the E3 Ubiquitin Ligase Activity of UBE3C to Facilitate Cell Proliferation. Mol Endocrinol 2015; 29:1646-57. [PMID: 26389696 DOI: 10.1210/me.2015-1125] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Estrogen receptor (ER)α is a well-characterized ligand-dependent transcription factor. However, the global picture of its nongenomic functions remains to be illustrated. Here, we demonstrate a novel function of ERα during mitosis that facilitates estrogen-dependent cell proliferation. An E3 ubiquitin ligase, UBE3C, was identified in an ERα complex from estrogen-treated MCF-7 breast cancer cells arrested at mitosis. UBE3C interacts with ERα during mitosis in an estrogen-dependent manner. In vitro, estrogen dramatically stimulates the E3 activity of UBE3C in the presence of ERα. This effect was inhibited by the estrogen antagonist tamoxifen. Importantly, estrogen enhances the ubiquitination of cyclin B1 (CCNB1) and destabilizes CCNB1 during mitosis in a manner dependent on endogenous UBE3C. ERα, UBE3C, and CCNB1 colocalize in prophase nuclei and at metaphase spindles before CCNB1 is degraded in anaphase. Depletion of UBE3C attenuates estrogen-dependent cell proliferation without affecting the transactivation function of ERα. Collectively, these results demonstrate a novel ligand-dependent action of ERα that stimulates the activity of an E3 ligase. The mitotic role of estrogen may contribute to its effects on proliferation in addition to its roles in target gene expression.
Collapse
Affiliation(s)
- Maiko Okada
- Department of Translational Oncology (M.O., W.W., T.O.), Institute of Advanced Medical Science (H.N.), St. Marianna University Graduate School of Medicine, Kawasaki 216-8511, Japan; Institute of Molecular and Cellular Biosciences (M.O.), University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan; Division of Cellular and Molecular Toxicology (F.O.), Biological Safety Research Center, National Institute of Health Sciences, Setagaya-ku, Tokyo 158-8501, Japan; and Laboratory of Protein Metabolism (Y.S., K.T.), Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Fumiaki Ohtake
- Department of Translational Oncology (M.O., W.W., T.O.), Institute of Advanced Medical Science (H.N.), St. Marianna University Graduate School of Medicine, Kawasaki 216-8511, Japan; Institute of Molecular and Cellular Biosciences (M.O.), University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan; Division of Cellular and Molecular Toxicology (F.O.), Biological Safety Research Center, National Institute of Health Sciences, Setagaya-ku, Tokyo 158-8501, Japan; and Laboratory of Protein Metabolism (Y.S., K.T.), Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Hiroyuki Nishikawa
- Department of Translational Oncology (M.O., W.W., T.O.), Institute of Advanced Medical Science (H.N.), St. Marianna University Graduate School of Medicine, Kawasaki 216-8511, Japan; Institute of Molecular and Cellular Biosciences (M.O.), University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan; Division of Cellular and Molecular Toxicology (F.O.), Biological Safety Research Center, National Institute of Health Sciences, Setagaya-ku, Tokyo 158-8501, Japan; and Laboratory of Protein Metabolism (Y.S., K.T.), Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Wenwen Wu
- Department of Translational Oncology (M.O., W.W., T.O.), Institute of Advanced Medical Science (H.N.), St. Marianna University Graduate School of Medicine, Kawasaki 216-8511, Japan; Institute of Molecular and Cellular Biosciences (M.O.), University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan; Division of Cellular and Molecular Toxicology (F.O.), Biological Safety Research Center, National Institute of Health Sciences, Setagaya-ku, Tokyo 158-8501, Japan; and Laboratory of Protein Metabolism (Y.S., K.T.), Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Yasushi Saeki
- Department of Translational Oncology (M.O., W.W., T.O.), Institute of Advanced Medical Science (H.N.), St. Marianna University Graduate School of Medicine, Kawasaki 216-8511, Japan; Institute of Molecular and Cellular Biosciences (M.O.), University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan; Division of Cellular and Molecular Toxicology (F.O.), Biological Safety Research Center, National Institute of Health Sciences, Setagaya-ku, Tokyo 158-8501, Japan; and Laboratory of Protein Metabolism (Y.S., K.T.), Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Keiji Takana
- Department of Translational Oncology (M.O., W.W., T.O.), Institute of Advanced Medical Science (H.N.), St. Marianna University Graduate School of Medicine, Kawasaki 216-8511, Japan; Institute of Molecular and Cellular Biosciences (M.O.), University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan; Division of Cellular and Molecular Toxicology (F.O.), Biological Safety Research Center, National Institute of Health Sciences, Setagaya-ku, Tokyo 158-8501, Japan; and Laboratory of Protein Metabolism (Y.S., K.T.), Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Tomohiko Ohta
- Department of Translational Oncology (M.O., W.W., T.O.), Institute of Advanced Medical Science (H.N.), St. Marianna University Graduate School of Medicine, Kawasaki 216-8511, Japan; Institute of Molecular and Cellular Biosciences (M.O.), University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan; Division of Cellular and Molecular Toxicology (F.O.), Biological Safety Research Center, National Institute of Health Sciences, Setagaya-ku, Tokyo 158-8501, Japan; and Laboratory of Protein Metabolism (Y.S., K.T.), Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| |
Collapse
|
457
|
Inaba H, Sanghamitra NJM, Fujita K, Sho T, Kuchimaru T, Kitagawa S, Kizaka-Kondoh S, Ueno T. A metal carbonyl-protein needle composite designed for intracellular CO delivery to modulate NF-κB activity. MOLECULAR BIOSYSTEMS 2015; 11:3111-8. [PMID: 26360102 DOI: 10.1039/c5mb00327j] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Carbon monoxide (CO) has been recognized as a messenger for signal transduction in living cells and tissues. For intracellular CO delivery, several metal carbonyl complexes have been used as CO-releasing molecules (CO-RMs). To improve the properties of CO-RMs, such as the stability and the CO release rate, ligands and carriers of the metal complexes have been exploited. Here we report the development of an efficient intracellular CO delivery system using a protein scaffold. We used a protein needle reconstructed from gene product 5 of bacteriophage T4, which has high cellular permeability and stability. When ruthenium carbonyl complexes are conjugated to the needle using a His-tag triad at the C-terminus, the resulting composite has a significantly higher cellular uptake efficiency of Ru carbonyl and a 12-fold prolonged CO release rate relative to Ru(CO)3Cl(glycinate), a widely used CO-RM. We demonstrate that CO delivered by the composite activates the transcriptional factor nuclear factor-kappaB (NF-κB), which in turn leads to significant induction of expression of its target genes, HO1, NQO1, and IL6, through generation of reactive oxygen species (ROS). The signaling pathway is distinct from that of tumor necrosis factor (TNF)-α-induced activation of NF-κB. The protein needle-based CO-RM can be exploited to elucidate the biological functions of CO and used in the development of protein-based organometallic tools for modulation of cellular signaling.
Collapse
Affiliation(s)
- Hiroshi Inaba
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | | | | | | | | | | | | | | |
Collapse
|
458
|
Arden JD, Lavik KI, Rubinic KA, Chiaia N, Khuder SA, Howard MJ, Nestor-Kalinoski AL, Alberts AS, Eisenmann KM. Small-molecule agonists of mammalian Diaphanous-related (mDia) formins reveal an effective glioblastoma anti-invasion strategy. Mol Biol Cell 2015; 26:3704-18. [PMID: 26354425 PMCID: PMC4626057 DOI: 10.1091/mbc.e14-11-1502] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 09/04/2015] [Indexed: 12/26/2022] Open
Abstract
Formin agonists impede the most dangerous aspect of glioblastoma—tumor spread into surrounding healthy tissue. Formin activation impairs a novel aspect of the transformed cell and informs the development of antitumor strategies for a cancer needing a more effective therapy. The extensive invasive capacity of glioblastoma (GBM) makes it resistant to surgery, radiotherapy, and chemotherapy and thus makes it lethal. In vivo, GBM invasion is mediated by Rho GTPases through unidentified downstream effectors. Mammalian Diaphanous (mDia) family formins are Rho-directed effectors that regulate the F-actin cytoskeleton to support tumor cell motility. Historically, anti-invasion strategies focused upon mDia inhibition, whereas activation remained unexplored. The recent development of small molecules directly inhibiting or activating mDia-driven F-actin assembly that supports motility allows for exploration of their role in GBM. We used the formin inhibitor SMIFH2 and mDia agonists IMM-01/-02 and mDia2-DAD peptides, which disrupt autoinhibition, to examine the roles of mDia inactivation versus activation in GBM cell migration and invasion in vitro and in an ex vivo brain slice invasion model. Inhibiting mDia suppressed directional migration and spheroid invasion while preserving intrinsic random migration. mDia agonism abrogated both random intrinsic and directional migration and halted U87 spheroid invasion in ex vivo brain slices. Thus mDia agonism is a superior GBM anti-invasion strategy. We conclude that formin agonism impedes the most dangerous GBM component—tumor spread into surrounding healthy tissue. Formin activation impairs novel aspects of transformed cells and informs the development of anti-GBM invasion strategies.
Collapse
Affiliation(s)
- Jessica D Arden
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614
| | - Kari I Lavik
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614
| | - Kaitlin A Rubinic
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614
| | - Nicolas Chiaia
- Department of Neurosciences, University of Toledo Health Science Campus, Toledo, OH 43614
| | - Sadik A Khuder
- Departments of Medicine and Public Health and Homeland Security, University of Toledo Health Science Campus, Toledo, OH 43614
| | - Marthe J Howard
- Department of Neurosciences, University of Toledo Health Science Campus, Toledo, OH 43614
| | | | - Arthur S Alberts
- Laboratory of Cell Structure and Signal Integration, Van Andel Research Institute, Grand Rapids, MI 49503
| | - Kathryn M Eisenmann
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614 )
| |
Collapse
|
459
|
Nandi S, Sahana A, Mandal S, Sengupta A, Chatterjee A, Safin DA, Babashkina MG, Tumanov NA, Filinchuk Y, Das D. Hydrazine selective dual signaling chemodosimetric probe in physiological conditions and its application in live cells. Anal Chim Acta 2015; 893:84-90. [PMID: 26398426 DOI: 10.1016/j.aca.2015.08.041] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/16/2015] [Accepted: 08/24/2015] [Indexed: 02/05/2023]
Abstract
A rhodamine-cyanobenzene conjugate, (E)-4-((2-(3',6'-bis(diethylamino)-3-oxospiro[isoindoline-1,9'-xanthene]-2-yl)ethylimino)methyl)benzonitrile (1), which structure has been elucidated by single crystal X-ray diffraction, was synthesized for selective fluorescent "turn-on" and colorimetric recognition of hydrazine at physiological pH 7.4. It was established that 1 detects hydrazine up to 58 nM. The probe is useful for the detection of intracellular hydrazine in the human breast cancer cells MCF-7 using a fluorescence microscope. Spirolactam ring opening of 1, followed by its hydrolysis, was established as a probable mechanism for the selective sensing of hydrazine.
Collapse
Affiliation(s)
- Sandip Nandi
- Department of Chemistry, The University of Burdwan, Burdwan, 713104 West Bengal, India
| | - Animesh Sahana
- Department of Chemistry, The University of Burdwan, Burdwan, 713104 West Bengal, India
| | - Sandip Mandal
- Department of Chemistry, The University of Burdwan, Burdwan, 713104 West Bengal, India
| | - Archya Sengupta
- Department of Zoology, Visva Bharati University, Santiniketan, West Bengal, India
| | - Ansuman Chatterjee
- Department of Zoology, Visva Bharati University, Santiniketan, West Bengal, India
| | - Damir A Safin
- Institute of Condensed Matter and Nanosciences, Molecules, Solids and Reactivity (IMCN/MOST), Université catholique de Louvain, Place L. Pasteur 1, 1348 Louvain-la-Neuve, Belgium.
| | - Maria G Babashkina
- Institute of Condensed Matter and Nanosciences, Molecules, Solids and Reactivity (IMCN/MOST), Université catholique de Louvain, Place L. Pasteur 1, 1348 Louvain-la-Neuve, Belgium
| | - Nikolay A Tumanov
- Institute of Condensed Matter and Nanosciences, Molecules, Solids and Reactivity (IMCN/MOST), Université catholique de Louvain, Place L. Pasteur 1, 1348 Louvain-la-Neuve, Belgium
| | - Yaroslav Filinchuk
- Institute of Condensed Matter and Nanosciences, Molecules, Solids and Reactivity (IMCN/MOST), Université catholique de Louvain, Place L. Pasteur 1, 1348 Louvain-la-Neuve, Belgium
| | - Debasis Das
- Department of Chemistry, The University of Burdwan, Burdwan, 713104 West Bengal, India.
| |
Collapse
|
460
|
Ho CH, Hsu JL, Liu SP, Hsu LC, Chang WL, Chao CCK, Guh JH. Repurposing of phentolamine as a potential anticancer agent against human castration-resistant prostate cancer: A central role on microtubule stabilization and mitochondrial apoptosis pathway. Prostate 2015; 75:1454-66. [PMID: 26180030 DOI: 10.1002/pros.23033] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 05/15/2015] [Indexed: 12/29/2022]
Abstract
BACKGROUND Drug repurposing of phentolamine, an α-adrenoceptor antagonist, as an anticancer agent has been studied in human castration-resistant prostate cancer (CRPC). METHODS Cell proliferation was examined by sulforhodamine B and CFSE staining assays. Cell cycle progression and mitochondrial membrane potential (ΔΨm) were detected by flow cytometric analysis. Protein expression was detected by Western blotting. Effect on tubulin/microtubule was determined using confocal immunofluorescence microscopic examination, microtubule assembly detection, tubulin turbidity assay, and binding assay. Several assessments were used to characterize apoptotic signaling pathways and combinatory effect. RESULTS Phentolamine induced anti-proliferative effect in PC-3 and DU-145, two CRPC cell lines, and P-glycoprotein (P-gp) overexpressing cells. This effect was not significantly reduced in paclitaxel-resistant cells. Rhodamine 123 efflux assay showed that phentolamine was not a P-gp substrate. Phentolamine induced mitotic arrest of the cell cycle and formation of hyperdiploid cells, followed by an increase of apoptosis. Mitotic arrest was confirmed by cyclin B1 up-regulation, Cdk1 activation, and a dramatic increase of mitotic protein phosphorylation. Both in vitro and cellular identification demonstrated that phentolamine, similar to paclitaxel, induced tubulin polymerization and formation of multiple nuclei. Besides, it did not compete with paclitaxel binding on tubulin. Phentolamine induced the phosphorylation and degradation of Bcl-2 and Bcl-xL, two anti-apoptotic Bcl-2 family members, and the loss of ΔΨm indicating the induction of mitochondrial damage. It ultimately induced the activation of caspase-9, -8, and -3 and apoptotic cell death. Moreover, combination treatment with phentolamine and paclitaxel caused a synergistic apoptosis. CONCLUSIONS The data suggest that phentolamine is a potential anticancer agent. In contrast to a wide variety of microtubule disrupting agents, phentolamine induces microtubule assembly, leading to mitotic arrest of the cell cycle which "in turn" induces subsequent mitochondrial damage and activation of related apoptotic signaling pathways in CRPC cells. Furthermore, combination between phentolamine and paclitaxel induces a synergistic apoptotic cell death. Phentolamine has a simple chemical structure and is not a P-gp substrate. Optimization of phentolamine structure may also be a potential approach for further development.
Collapse
Affiliation(s)
- Chen-Hsun Ho
- Department of Urology, Taipei Medical University-Shuang Ho Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jui-Ling Hsu
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Shih-Ping Liu
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Urology, National Taiwan University Hospital, Taipei, Taiwan
| | - Lih-Ching Hsu
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Wei-Ling Chang
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
- Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Chuck C-K Chao
- Department of Biochemistry and Molecular Biology, Chang Gung University, Taoyuan, Taiwan
| | - Jih-Hwa Guh
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
461
|
Feng W, Cai D, Zhang B, Lou G, Zou X. Combination of HDAC inhibitor TSA and silibinin induces cell cycle arrest and apoptosis by targeting survivin and cyclinB1/Cdk1 in pancreatic cancer cells. Biomed Pharmacother 2015; 74:257-64. [PMID: 26349994 DOI: 10.1016/j.biopha.2015.08.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 08/04/2015] [Indexed: 12/12/2022] Open
Abstract
Histone deacetylases (HDAC) are involved in diverse biological processes and therefore emerge as potential targets for pancreatic cancer. Silibinin, an active component of silymarin, is known to inhibit growth of pancreatic cancer in vivo and in vitro. Herein, we examined the cytotoxic effects of TSA in combination with silibinin and investigated the possible mechanism in two pancreatic cancer cell lines (Panc1 and Capan2). Our study found that combination treatment of HDAC inhibitor and silibinin exerted additive growth inhibitory effect on pancreatic cancer cell. Annexin V-FITC/PI staining and flow cytometry analysis demonstrated that combination therapy induced G2/M cell cycle arrest and apoptosis in Panc1and Capan2 cells. The induction of apoptosis was further confirmed by evaluating the activation of caspases. Moreover, treatment with TSA and silibinin resulted in a profound reduction in the expression of cyclinA2, cyclinB1/Cdk1 and survivin. Taken together, our study might indicate that the novel combination of HDAC inhibitor and silibinin could offer therapeutic potential against pancreatic cancer.
Collapse
Affiliation(s)
- Wan Feng
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu Province, China
| | - Dawei Cai
- Medical school, Nanjing University, Nanjing, Jiangsu, China
| | - Bin Zhang
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu Province, China
| | - Guochun Lou
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu Province, China
| | - Xiaoping Zou
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu Province, China.
| |
Collapse
|
462
|
Tanenbaum ME, Stern-Ginossar N, Weissman JS, Vale RD. Regulation of mRNA translation during mitosis. eLife 2015; 4. [PMID: 26305499 PMCID: PMC4548207 DOI: 10.7554/elife.07957] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 08/02/2015] [Indexed: 12/21/2022] Open
Abstract
Passage through mitosis is driven by precisely-timed changes in transcriptional regulation and protein degradation. However, the importance of translational regulation during mitosis remains poorly understood. Here, using ribosome profiling, we find both a global translational repression and identified ∼200 mRNAs that undergo specific translational regulation at mitotic entry. In contrast, few changes in mRNA abundance are observed, indicating that regulation of translation is the primary mechanism of modulating protein expression during mitosis. Interestingly, 91% of the mRNAs that undergo gene-specific regulation in mitosis are translationally repressed, rather than activated. One of the most pronounced translationally-repressed genes is Emi1, an inhibitor of the anaphase promoting complex (APC) which is degraded during mitosis. We show that full APC activation requires translational repression of Emi1 in addition to its degradation. These results identify gene-specific translational repression as a means of controlling the mitotic proteome, which may complement post-translational mechanisms for inactivating protein function. DOI:http://dx.doi.org/10.7554/eLife.07957.001 The human body contains billions of cells, most of which formed via a process called mitosis in which a single cell divides to produce two new daughter cells. Actively dividing cells pass through a series of events (or phases) that are collectively known as the cell cycle. These phases allow the cell to grow in size, copy its genetic material, and then make preparations for cell division before taking the final decision to divide. Many proteins are involved in regulating the cell cycle and each protein has a particular role in specific phases. The levels of these proteins in cells may change during the cycle, which is often crucial to allow the cell to progress to the next phase. For example, cells need a group of proteins called the anaphase-promoting complex (or APC for short) to destroy other specific proteins at the end of mitosis. Another way in which the amount of protein in a cell can be adjusted is by controlling how much new protein is made during a process known as translation. During this process, a molecule called a messenger RNA (mRNA)—which contains information copied from a particular gene—is used as a template to assemble a new protein. However, it is not clear whether regulation of translation is involved in control of the cell division. Tanenbaum et al. now address this question using a technique called ribosome profiling to measure the translation of individual mRNA molecules. The experiments analysed the changes in protein production before, during and after mitosis. The overall level of translation of all the mRNAs was about 35% lower during mitosis. However, some mRNAs in particular experienced a very large reduction in the level of translation (between three- and ten-fold less than the levels before mitosis). One example of an mRNA whose translation is turned off in mitosis is the mRNA that makes a protein called Emi1. It is known from previous work that Emi1 inhibits the activity of the APC. Therefore, Emi1 needs to be inactivated in mitosis so that the APC can become active and promote progression to the next phase of the cell cycle. It was previously shown that Emi1 is destroyed during mitosis to allow the APC to operate. Tanenbaum et al. found that translation of the Emi1 mRNA must also be suppressed during mitosis in order to keep Emi1 protein levels very low and allow the APC to become fully active. These findings uncover a new role for the control of protein production in regulating the cell cycle. The next challenge will be to find out whether suppression of translation is also used in other biological systems where proteins need to be rapidly inactivated. DOI:http://dx.doi.org/10.7554/eLife.07957.002
Collapse
Affiliation(s)
- Marvin E Tanenbaum
- Department of Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Noam Stern-Ginossar
- Department of Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Jonathan S Weissman
- Department of Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| | - Ronald D Vale
- Department of Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
463
|
Effect of Fluoride-Modified Titanium Surface on Early Adhesion of Irradiated Osteoblasts. BIOMED RESEARCH INTERNATIONAL 2015; 2015:219752. [PMID: 26266253 PMCID: PMC4525467 DOI: 10.1155/2015/219752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/24/2015] [Indexed: 01/22/2023]
Abstract
Objective. The present study aimed to investigate the effect of fluoride-modified titanium surface on adhesion of irradiated osteoblasts. Materials and Methods. Fluoride-modified surface was obtained and the morphology, roughness, and chemical composition of the surface were evaluated by scanning electron microscopy, atomic force microscopy, and X-ray photoelectron spectroscopy, respectively. The adhesion of irradiated osteoblast-like cells, in terms of number, area, and fluorescence intensity on the titanium surface, was evaluated using immunofluorescence staining. Results. Numerous nanosize pits were seen only in the F-TiO surface. The pits were more remarkable and uniform on F-TiO surface than on TiO surface; however, the amplitude of peaks and bottoms on F-TiO surface appeared to be smaller than on TiO surface. The Sa value and Sdr percentage of TiO surfaces were significantly higher than those of F-TiO surface. The concentrations of main elements such as titanium, oxygen, and carbon were similar on both surfaces. The number of irradiated osteoblasts adhered on the control surface was larger than on fluoride-modified surface. Meanwhile, the cells on the fluoride-modified surface formed more actin filaments. Conclusions. The fluoride-modified titanium surface alters the adhesion of irradiated osteoblasts. Further studies are needed to investigate the proliferation, differentiation, maturation, gene expression, and cytokine production of irradiated osteoblasts on fluoride-modified titanium surface.
Collapse
|
464
|
Li L, Shi W, Wang Z, Gong Q, Ma H. Sensitive Fluorescence Probe with Long Analytical Wavelengths for γ-Glutamyl Transpeptidase Detection in Human Serum and Living Cells. Anal Chem 2015; 87:8353-9. [DOI: 10.1021/acs.analchem.5b01535] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Lihong Li
- Beijing National Laboratory
for Molecular Sciences, Key Laboratory of Analytical Chemistry for
Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Wen Shi
- Beijing National Laboratory
for Molecular Sciences, Key Laboratory of Analytical Chemistry for
Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Zhe Wang
- Beijing National Laboratory
for Molecular Sciences, Key Laboratory of Analytical Chemistry for
Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Qiuyu Gong
- Beijing National Laboratory
for Molecular Sciences, Key Laboratory of Analytical Chemistry for
Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Huimin Ma
- Beijing National Laboratory
for Molecular Sciences, Key Laboratory of Analytical Chemistry for
Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
465
|
Tan CSH, Ng YK, Ong WY. Epigenetic Regulation of Cytosolic Phospholipase A2 in SH-SY5Y Human Neuroblastoma Cells. Mol Neurobiol 2015; 53:3854-3872. [PMID: 26162318 DOI: 10.1007/s12035-015-9314-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 06/23/2015] [Indexed: 12/19/2022]
Abstract
Group IVA cytosolic phospholipase A2 (cPLA2 or PLA2G4A) is a key enzyme that contributes to inflammation via the generation of arachidonic acid and eicosanoids. While much is known about regulation of cPLA2 by posttranslational modification such as phosphorylation, little is known about its epigenetic regulation. In this study, treatment with histone deacetylase (HDAC) inhibitors, trichostatin A (TSA), valproic acid, tubacin and the class I HDAC inhibitor, MS-275, were found to increase cPLA2α messenger RNA (mRNA) expression in SH-SY5Y human neuroblastoma cells. Co-treatment of the histone acetyltransferase (HAT) inhibitor, anacardic acid, modulated upregulation of cPLA2α induced by TSA. Specific involvement of class I HDACs and HAT in cPLA2α regulation was further shown, and a Tip60-specific HAT inhibitor, NU9056, modulated the upregulation of cPLA2α induced by MS-275. In addition, co-treatment of with histone methyltransferase (HMT) inhibitor, 5'-deoxy-5'-methylthioadenosine (MTA) suppressed TSA-induced cPLA2α upregulation. The above changes in cPLA2 mRNA expression were reflected at the protein level by Western blots and immunocytochemistry. Chromatin immunoprecipitation (ChIP) showed TSA increased binding of trimethylated H3K4 to the proximal promoter region of the cPLA2α gene. Cell injury after TSA treatment as indicated by lactate dehydrogenase (LDH) release was modulated by anacardic acid, and a role of cPLA2 in mediating TSA-induced injury shown, after co-incubation with the cPLA2 selective inhibitor, arachidonoyl trifluoromethyl ketone (AACOCF3). Together, results indicate epigenetic regulation of cPLA2 and the potential of such regulation for treatment of chronic inflammation.
Collapse
Affiliation(s)
- Charlene Siew-Hon Tan
- Department of Anatomy, National University of Singapore, Singapore, 119260, Singapore
| | - Yee-Kong Ng
- Department of Anatomy, National University of Singapore, Singapore, 119260, Singapore
| | - Wei-Yi Ong
- Department of Anatomy, National University of Singapore, Singapore, 119260, Singapore. .,Neurobiology and Ageing Research Programme, National University of Singapore, Singapore, 119260, Singapore.
| |
Collapse
|
466
|
Activation and function of murine primary microglia in the absence of the prion protein. J Neuroimmunol 2015; 286:25-32. [PMID: 26298321 DOI: 10.1016/j.jneuroim.2015.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 07/02/2015] [Accepted: 07/04/2015] [Indexed: 11/21/2022]
Abstract
The prion protein (PrP(C)) is predominantly expressed in the nervous and immune systems and is involved in relevant cell signaling. Microglia participate in neuroimmune interactions, and their regulatory mechanisms are critical for both health and disease. Despite recent reports with a microglial cell line, little is known about the relevance of PrP(C) in brain microglia. We investigated the role of PrP(C) in mouse primary microglia, and found no differences between wild type and Prnp-null cells in cell morphology or the expression of a microglial marker. Translocation of NF-κB to the nucleus also did not differ, nor did cytokine production. The levels of iNOS were also similar and, finally, microglia of either genotype showed no differences in either rates of phagocytosis or migration, even following activation. Thus, functional roles of PrP(C) in primary microglial cells are - if present - much more subtle than in transformed microglial cell lines.
Collapse
|
467
|
Wainwright EN, Wilhelm D, Combes AN, Little MH, Koopman P. ROBO2 restricts the nephrogenic field and regulates Wolffian duct-nephrogenic cord separation. Dev Biol 2015; 404:88-102. [PMID: 26116176 DOI: 10.1016/j.ydbio.2015.05.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/28/2015] [Accepted: 05/30/2015] [Indexed: 01/03/2023]
Abstract
ROBO2 plays a key role in regulating ureteric bud (UB) formation in the embryo, with mutations in humans and mice leading to supernumerary kidneys. Previous studies have established that the number and position of UB outgrowths is determined by the domain of metanephric mesenchymal Gdnf expression, which is expanded anteriorly in Robo2 mouse mutants. To clarify how this phenotype arises, we used high-resolution 3D imaging to reveal an increase in the number of nephrogenic cord cells, leading to extension of the metanephric mesenchyme field in Robo2-null mouse embryos. Ex vivo experiments suggested a dependence of this effect on proliferative signals from the Wolffian duct. Loss of Robo2 resulted in a failure of the normal separation of the mesenchyme from the Wolffian duct/ureteric epithelium, suggesting that aberrant juxtaposition of these two compartments in Robo2-null mice exposes the mesenchyme to abnormally high levels of proliferative stimuli. Our data suggest a new model in which SLIT-ROBO signalling acts not by attenuating Gdnf expression or activity, but instead by limiting epithelial/mesenchymal interactions in the nascent metanephros and restricting the extent of the nephrogenic field. These insights illuminate the aetiology of multiplex kidney formation in human individuals with ROBO2 mutations.
Collapse
Affiliation(s)
- Elanor N Wainwright
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Dagmar Wilhelm
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Alexander N Combes
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Melissa H Little
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
468
|
Ren Y, Suzuki H, Jagarlamudi K, Golnoski K, McGuire M, Lopes R, Pachnis V, Rajkovic A. Lhx8 regulates primordial follicle activation and postnatal folliculogenesis. BMC Biol 2015; 13:39. [PMID: 26076587 PMCID: PMC4487509 DOI: 10.1186/s12915-015-0151-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 06/10/2015] [Indexed: 12/23/2022] Open
Abstract
Background The early stages of ovarian follicle formation—beginning with the breakdown of germ cell cysts and continuing with the formation of primordial follicles and transition to primary and secondary follicles—are critical in determining reproductive life span and fertility. Previously, we discovered that global knockouts of germ cell-specific transcriptional co-regulators Sohlh1, Sohlh2, Lhx8, and Nobox, cause rapid oocyte loss and ovarian failure. Also factors such as Nobox and Sohlh1 are associated with human premature ovarian failure. In this study, we developed a conditional knockout of Lhx8 to study oocyte-specific pathways in postnatal folliculogenesis. Results The conditional deficiency of Lhx8 in the oocytes of primordial follicles leads to massive primordial oocyte activation, in part, by indirectly interacting with the PI3K-AKT pathway, as shown by synergistic effects on FOXO3 nucleocytoplasmic translocation and rpS6 activation. However, LHX8 does not directly regulate members of the PI3K-AKT pathway; instead, we show that LHX8 represses Lin28a expression, a known regulator of mammalian metabolism and of the AKT/mTOR pathway. LHX8 can bind to the Lin28a promoter, and the depletion of Lin28a in Lhx8-deficient oocytes partially suppresses primordial oocyte activation. Moreover, unlike the PI3K-AKT pathway, LHX8 is critical beyond primordial follicle activation, and blocks the primary to secondary follicle transition. Conclusions Our results indicate that the LHX8-LIN28A pathway is essential in the earliest stages of primordial follicle activation, and LHX8 is an important oocyte-specific transcription factor in the ovary for regulating postnatal folliculogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s12915-015-0151-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yu Ren
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Hitomi Suzuki
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Department of Experimental Animal Model for Human Disease, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, 113-8510, Japan.
| | - Krishna Jagarlamudi
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Kayla Golnoski
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Megan McGuire
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Rita Lopes
- Division of Molecular Neurobiology, MRC National Institute of Medical Research, London, NW7 1AA, UK.
| | - Vassilis Pachnis
- Division of Molecular Neurobiology, MRC National Institute of Medical Research, London, NW7 1AA, UK.
| | - Aleksandar Rajkovic
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
469
|
Madeira MH, Elvas F, Boia R, Gonçalves FQ, Cunha RA, Ambrósio AF, Santiago AR. Adenosine A2AR blockade prevents neuroinflammation-induced death of retinal ganglion cells caused by elevated pressure. J Neuroinflammation 2015; 12:115. [PMID: 26054642 PMCID: PMC4465153 DOI: 10.1186/s12974-015-0333-5] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/27/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Elevated intraocular pressure (IOP) is a major risk factor for glaucoma, a degenerative disease characterized by the loss of retinal ganglion cells (RGCs). There is clinical and experimental evidence that neuroinflammation is involved in the pathogenesis of glaucoma. Since the blockade of adenosine A2A receptor (A2AR) confers robust neuroprotection and controls microglia reactivity in the brain, we now investigated the ability of A2AR blockade to control the reactivity of microglia and neuroinflammation as well as RGC loss in retinal organotypic cultures exposed to elevated hydrostatic pressure (EHP) or lipopolysaccharide (LPS). METHODS Retinal organotypic cultures were either incubated with LPS (3 μg/mL), to elicit a pro-inflammatory response, or exposed to EHP (+70 mmHg), to mimic increased IOP, for 4 or 24 h, in the presence or absence of the A2AR antagonist SCH 58261 (50 nM). A2AR expression, microglial reactivity and neuroinflammatory response were evaluated by immunohistochemistry, quantitative PCR (qPCR) and enzyme-linked immunosorbent assay (ELISA). RGC loss was assessed by immunohistochemistry. In order to investigate the contribution of pro-inflammatory mediators to RGC loss, the organotypic retinal cultures were incubated with rabbit anti-tumour necrosis factor (TNF) (2 μg/mL) and goat anti-interleukin-1β (IL-1β) (1 μg/mL) antibodies. RESULTS We report that the A2AR antagonist (SCH 58261) prevented microglia reactivity, increase in pro-inflammatory mediators as well as RGC loss upon exposure to either LPS or EHP. Additionally, neutralization of TNF and IL-1β prevented RGC loss induced by LPS or EHP. CONCLUSIONS This work demonstrates that A2AR blockade confers neuroprotection to RGCs by controlling microglia-mediated retinal neuroinflammation and prompts the hypothesis that A2AR antagonists may be a novel therapeutic option to manage glaucomatous disorders.
Collapse
Affiliation(s)
- Maria H Madeira
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3004-548, Coimbra, Portugal. .,CNC.IBILI, University of Coimbra, 3004-517, Coimbra, Portugal.
| | - Filipe Elvas
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3004-548, Coimbra, Portugal.
| | - Raquel Boia
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3004-548, Coimbra, Portugal. .,CNC.IBILI, University of Coimbra, 3004-517, Coimbra, Portugal.
| | - Francisco Q Gonçalves
- CNC.IBILI, University of Coimbra, 3004-517, Coimbra, Portugal. .,CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal.
| | - Rodrigo A Cunha
- CNC.IBILI, University of Coimbra, 3004-517, Coimbra, Portugal. .,CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal. .,Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal.
| | - António Francisco Ambrósio
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3004-548, Coimbra, Portugal. .,CNC.IBILI, University of Coimbra, 3004-517, Coimbra, Portugal. .,Association for Innovation and Biomedical Research on Light (AIBILI), 3000-548, Coimbra, Portugal.
| | - Ana Raquel Santiago
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3004-548, Coimbra, Portugal. .,CNC.IBILI, University of Coimbra, 3004-517, Coimbra, Portugal. .,Association for Innovation and Biomedical Research on Light (AIBILI), 3000-548, Coimbra, Portugal. .,Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal. .,IBILI, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3004-548, Coimbra, Portugal.
| |
Collapse
|
470
|
Baffet AD, Hu DJ, Vallee RB. Cdk1 Activates Pre-mitotic Nuclear Envelope Dynein Recruitment and Apical Nuclear Migration in Neural Stem Cells. Dev Cell 2015; 33:703-16. [PMID: 26051540 DOI: 10.1016/j.devcel.2015.04.022] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 02/20/2015] [Accepted: 04/24/2015] [Indexed: 10/23/2022]
Abstract
Dynein recruitment to the nuclear envelope is required for pre-mitotic nucleus-centrosome interactions in nonneuronal cells and for apical nuclear migration in neural stem cells. In each case, dynein is recruited to the nuclear envelope (NE) specifically during G2 via two nuclear pore-mediated mechanisms involving RanBP2-BicD2 and Nup133-CENP-F. The mechanisms responsible for cell-cycle control of this behavior are unknown. We now find that Cdk1 serves as a direct master controller for NE dynein recruitment in neural stem cells and HeLa cells. Cdk1 phosphorylates conserved sites within RanBP2 and activates BicD2 binding and early dynein recruitment. Late recruitment is triggered by a Cdk1-induced export of CENP-F from the nucleus. Forced NE targeting of BicD2 overrides Cdk1 inhibition, fully rescuing dynein recruitment and nuclear migration in neural stem cells. These results reveal how NE dynein recruitment is cell-cycle regulated and identify the trigger mechanism for apical nuclear migration in the brain.
Collapse
Affiliation(s)
- Alexandre D Baffet
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA.
| | - Daniel J Hu
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Richard B Vallee
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
471
|
Voit R, Seiler J, Grummt I. Cooperative Action of Cdk1/cyclin B and SIRT1 Is Required for Mitotic Repression of rRNA Synthesis. PLoS Genet 2015; 11:e1005246. [PMID: 26023773 PMCID: PMC4449194 DOI: 10.1371/journal.pgen.1005246] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 04/27/2015] [Indexed: 12/29/2022] Open
Abstract
Mitotic repression of rRNA synthesis requires inactivation of the RNA polymerase I (Pol I)-specific transcription factor SL1 by Cdk1/cyclin B-dependent phosphorylation of TAFI110 (TBP-associated factor 110) at a single threonine residue (T852). Upon exit from mitosis, T852 is dephosphorylated by Cdc14B, which is sequestered in nucleoli during interphase and is activated upon release from nucleoli at prometaphase. Mitotic repression of Pol I transcription correlates with transient nucleolar enrichment of the NAD+-dependent deacetylase SIRT1, which deacetylates another subunit of SL1, TAFI68. Hypoacetylation of TAFI68 destabilizes SL1 binding to the rDNA promoter, thereby impairing transcription complex assembly. Inhibition of SIRT1 activity alleviates mitotic repression of Pol I transcription if phosphorylation of TAFI110 is prevented. The results demonstrate that reversible phosphorylation of TAFI110 and acetylation of TAFI68 are key modifications that regulate SL1 activity and mediate fluctuations of pre-rRNA synthesis during cell cycle progression. In metazoans, transcription is arrested during mitosis. Previous studies have established that mitotic repression of cellular transcription is mediated by Cdk1/cyclin B-dependent phosphorylation of basal transcription factors that nucleate transcription complex formation. Repression of rDNA transcription at the onset of mitosis is brought about by inactivation of the TBP-containing transcription factor SL1 by Cdk1/cyclin B-dependent phosphorylation of the TAFI110 subunit, which impairs the interaction with UBF and the assembly of pre-initiation complexes. Here we show that hCdc14B, the phosphatase that regulates Cdk1/cyclin B activity and progression through mitosis, promotes reactivation of rDNA transcription by dephosphorylating TAFI110. In addition, the NAD+-dependent deacetylase SIRT1 becomes transiently enriched in nucleoli at the onset of mitosis. SIRT1 deacetylates TAFI68, another subunit of SL1, hypoacetylation of TAFI68 destabilizing SL1 binding to the rDNA promoter and impairing transcription complex assembly. The results reveal that modulation of SL1 activity by reversible acetylation of TAFI68 and phosphorylation of TAFI110 are key modifications that mediate oscillation of rDNA transcription during cell cycle progression.
Collapse
Affiliation(s)
- Renate Voit
- Division of Molecular Biology of the Cell II, German Cancer Research Center, DKFZ-ZMBH Alliance, Heidelberg, Germany
- * E-mail:
| | - Jeanette Seiler
- Division of Molecular Biology of the Cell II, German Cancer Research Center, DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Ingrid Grummt
- Division of Molecular Biology of the Cell II, German Cancer Research Center, DKFZ-ZMBH Alliance, Heidelberg, Germany
| |
Collapse
|
472
|
Gaillard D, Xu M, Liu F, Millar SE, Barlow LA. β-Catenin Signaling Biases Multipotent Lingual Epithelial Progenitors to Differentiate and Acquire Specific Taste Cell Fates. PLoS Genet 2015; 11:e1005208. [PMID: 26020789 PMCID: PMC4447363 DOI: 10.1371/journal.pgen.1005208] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 04/13/2015] [Indexed: 11/29/2022] Open
Abstract
Continuous taste bud cell renewal is essential to maintain taste function in adults; however, the molecular mechanisms that regulate taste cell turnover are unknown. Using inducible Cre-lox technology, we show that activation of β-catenin signaling in multipotent lingual epithelial progenitors outside of taste buds diverts daughter cells from a general epithelial to a taste bud fate. Moreover, while taste buds comprise 3 morphological types, β-catenin activation drives overproduction of primarily glial-like Type I taste cells in both anterior fungiform (FF) and posterior circumvallate (CV) taste buds, with a small increase in Type II receptor cells for sweet, bitter and umami, but does not alter Type III sour detector cells. Beta-catenin activation in post-mitotic taste bud precursors likewise regulates cell differentiation; forced activation of β-catenin in these Shh+ cells promotes Type I cell fate in both FF and CV taste buds, but likely does so non-cell autonomously. Our data are consistent with a model where β-catenin signaling levels within lingual epithelial progenitors dictate cell fate prior to or during entry of new cells into taste buds; high signaling induces Type I cells, intermediate levels drive Type II cell differentiation, while low levels may drive differentiation of Type III cells. Taste is a fundamental sense that helps the body determine whether food can be ingested. Taste dysfunction can be a side effect of cancer therapies, can result from an alteration of the renewal capacities of the taste buds, and is often associated with psychological distress and malnutrition. Thus, understanding how taste cells renew throughout adult life, i.e. how newly born cells replace old cells as they die, is essential to find potential therapeutic targets to improve taste sensitivity in patients suffering taste dysfunction. Here we show that a specific molecular pathway, Wnt/β-catenin signaling, controls renewal of taste cells by regulating separate stages of taste cell turnover. We show that activating this pathway directs the newly born cells to become primarily a specific taste cell type whose role is to support the other taste cells and help them work efficiently.
Collapse
Affiliation(s)
- Dany Gaillard
- Department of Cell & Developmental Biology, and the Rocky Mountain Taste & Smell Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Mingang Xu
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Cell & Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Fei Liu
- Institute for Regenerative Medicine at Scott & White Hospital, Texas A&M University System Health Science Center, Temple, Texas, United States of America
| | - Sarah E. Millar
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Linda A. Barlow
- Department of Cell & Developmental Biology, and the Rocky Mountain Taste & Smell Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
473
|
Hallam D, Wan T, Saretzki G. Dietary restriction mitigates age-related accumulation of DNA damage, but not all changes in mouse corneal epithelium. Exp Gerontol 2015; 67:72-9. [PMID: 25937374 DOI: 10.1016/j.exger.2015.04.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 04/28/2015] [Accepted: 04/29/2015] [Indexed: 12/18/2022]
Abstract
The cornea protects the anterior eye and accounts for two thirds of the eyes refractive capacity. The homeostasis of corneal epithelium is thought to be maintained by putative stem cells residing in the epithelial basal layer. As a tissue constantly exposed to environmental stress, the cornea is hypothesised to accumulate persistent DNA damage events with time in stem cell populations. Recently, telomere associated DNA damage foci (TAFs) have been suggested as a marker for persistent DNA damage which can be used to detect senescent cells during ageing. Dietary restriction (DR) is the only known non-genetic intervention that is able to increase both life and health span among various animal species. The aim of this study was to analyse changes in corneal properties with age and under 16 months of DR. We employed immunofluorescence staining for ɣH2A.X together with telomere fluorescence in situ hybridisation (immuno-FISH) on mouse corneas from young, old ad libitum (AL) fed as well as dietary restricted (DR) mice. Our data show that the central corneas of old mice had significantly more general and telomere-associated DNA damage compared to young mice while DR treatment was able to reduce the amount of DNA damage significantly. We also found that the thickness of the peripheral region of the cornea, where the putative stem cells may reside, decreased with age regardless of whether the animals underwent DR treatment or not. Number of bullae, which indicates corneal edema, accumulated in old corneas in the central area and DR treatment mitigated the formation of these bullae. Moreover, the protein levels of the stem cell marker TAp63 decreased with age only in the central but not the peripheral region of the cornea. This finding suggests that epithelial progenitors might be better maintained in the peripheral than the central cornea during ageing. Together with the finding that the peripheral corneal showed no increase in DNA damage during age, we speculate that in mice, like humans, the putative stem cells reside in the peripheral cornea.
Collapse
Affiliation(s)
- Dean Hallam
- Institute for Cell and Molecular Bioscience, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle University Newcastle upon Tyne, UK
| | - Tengfei Wan
- Institute for Cell and Molecular Bioscience, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle University Newcastle upon Tyne, UK
| | - Gabriele Saretzki
- Institute for Cell and Molecular Bioscience, Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle University Newcastle upon Tyne, UK.
| |
Collapse
|
474
|
Lehrberg J, Gardiner DM. Regulation of Axolotl (Ambystoma mexicanum) Limb Blastema Cell Proliferation by Nerves and BMP2 in Organotypic Slice Culture. PLoS One 2015; 10:e0123186. [PMID: 25923915 PMCID: PMC4414535 DOI: 10.1371/journal.pone.0123186] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 03/02/2015] [Indexed: 11/18/2022] Open
Abstract
We have modified and optimized the technique of organotypic slice culture in order to study the mechanisms regulating growth and pattern formation in regenerating axolotl limb blastemas. Blastema cells maintain many of the behaviors that are characteristic of blastemas in vivo when cultured as slices in vitro, including rates of proliferation that are comparable to what has been reported in vivo. Because the blastema slices can be cultured in basal medium without fetal bovine serum, it was possible to test the response of blastema cells to signaling molecules present in serum, as well as those produced by nerves. We also were able to investigate the response of blastema cells to experimentally regulated changes in BMP signaling. Blastema cells responded to all of these signals by increasing the rate of proliferation and the level of expression of the blastema marker gene, Prrx-1. The organotypic slice culture model provides the opportunity to identify and characterize the spatial and temporal co-regulation of pathways in order to induce and enhance a regenerative response.
Collapse
Affiliation(s)
- Jeffrey Lehrberg
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California, United States of America
| | - David M. Gardiner
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California, United States of America
- * E-mail:
| |
Collapse
|
475
|
CDK1 structures reveal conserved and unique features of the essential cell cycle CDK. Nat Commun 2015; 6:6769. [PMID: 25864384 PMCID: PMC4413027 DOI: 10.1038/ncomms7769] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 02/26/2015] [Indexed: 12/23/2022] Open
Abstract
CDK1 is the only essential cell cycle CDK in human cells and is required for successful completion of M-phase. It is the founding member of the CDK family and is conserved across all eukaryotes. Here we report the crystal structures of complexes of CDK1–Cks1 and CDK1–cyclin B–Cks2. These structures confirm the conserved nature of the inactive monomeric CDK fold and its ability to be remodelled by cyclin binding. Relative to CDK2–cyclin A, CDK1–cyclin B is less thermally stable, has a smaller interfacial surface, is more susceptible to activation segment dephosphorylation and shows differences in the substrate sequence features that determine activity. Both CDK1 and CDK2 are potential cancer targets for which selective compounds are required. We also describe the first structure of CDK1 bound to a potent ATP-competitive inhibitor and identify aspects of CDK1 structure and plasticity that might be exploited to develop CDK1-selective inhibitors. Cyclin-dependent kinases are the principal drivers of cell cycle progression. Here the authors present several crystal structures of Cdk1 in complex with cyclin B and/or the assembly factors Cks1/2 and a small molecule inhibitor to reveal key features of this essential mitotic kinase.
Collapse
|
476
|
Robellet X, Thattikota Y, Wang F, Wee TL, Pascariu M, Shankar S, Bonneil É, Brown CM, D'Amours D. A high-sensitivity phospho-switch triggered by Cdk1 governs chromosome morphogenesis during cell division. Genes Dev 2015; 29:426-39. [PMID: 25691469 PMCID: PMC4335297 DOI: 10.1101/gad.253294.114] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The initiation of chromosome morphogenesis marks the beginning of mitosis in eukaryotic cells. Robellet et al. found that multisite phosphorylation of the chromatin-binding sensor Smc4 integrates the activation state of Cdk1 with the dynamic binding of the condensation machinery to chromatin. Abrogation of this event leads to chromosome segregation defects and lethality, while moderate reduction reveals the existence of a novel chromatin transition state specific to mitosis, the intertwist configuration. The initiation of chromosome morphogenesis marks the beginning of mitosis in all eukaryotic cells. Although many effectors of chromatin compaction have been reported, the nature and design of the essential trigger for global chromosome assembly remain unknown. Here we reveal the identity of the core mechanism responsible for chromosome morphogenesis in early mitosis. We show that the unique sensitivity of the chromosome condensation machinery for the kinase activity of Cdk1 acts as a major driving force for the compaction of chromatin at mitotic entry. This sensitivity is imparted by multisite phosphorylation of a conserved chromatin-binding sensor, the Smc4 protein. The multisite phosphorylation of this sensor integrates the activation state of Cdk1 with the dynamic binding of the condensation machinery to chromatin. Abrogation of this event leads to chromosome segregation defects and lethality, while moderate reduction reveals the existence of a novel chromatin transition state specific to mitosis, the intertwist configuration. Collectively, our results identify the mechanistic basis governing chromosome morphogenesis in early mitosis and how distinct chromatin compaction states can be established via specific thresholds of Cdk1 kinase activity.
Collapse
Affiliation(s)
- Xavier Robellet
- Institute for Research in Immunology and Cancer (IRIC), Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, Quebec H3C 3J7, Canada
| | - Yogitha Thattikota
- Institute for Research in Immunology and Cancer (IRIC), Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, Quebec H3C 3J7, Canada
| | - Fang Wang
- Institute for Research in Immunology and Cancer (IRIC), Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, Quebec H3C 3J7, Canada
| | - Tse-Luen Wee
- Advanced BioImaging Facility (ABIF), Department of Physiology, McGill University, Montréal, Quebec H3G 0B1, Canada
| | - Mirela Pascariu
- Institute for Research in Immunology and Cancer (IRIC), Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, Quebec H3C 3J7, Canada
| | - Sahana Shankar
- Institute for Research in Immunology and Cancer (IRIC), Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, Quebec H3C 3J7, Canada
| | - Éric Bonneil
- Institute for Research in Immunology and Cancer (IRIC)
| | - Claire M Brown
- Advanced BioImaging Facility (ABIF), Department of Physiology, McGill University, Montréal, Quebec H3G 0B1, Canada
| | - Damien D'Amours
- Institute for Research in Immunology and Cancer (IRIC), Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, Quebec H3C 3J7, Canada
| |
Collapse
|
477
|
Expression of CDK1(Tyr15), pCDK1(Thr161), Cyclin B1 (total) and pCyclin B1(Ser126) in vulvar squamous cell carcinoma and their relations with clinicopatological features and prognosis. PLoS One 2015; 10:e0121398. [PMID: 25849598 PMCID: PMC4388712 DOI: 10.1371/journal.pone.0121398] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 01/31/2015] [Indexed: 01/30/2023] Open
Abstract
Cyclin B1-CDK1 complex plays an important role in the regulation of cell cycle. Activation of Cyclin B1 and CDK1 and the formation of the complex in G2/M are under multiple regulations involving many regulators such as isoforms of 14-3-3 and CDC25 and Wee1. Abnormal expression of Cyclin B1 and CDK1 has been detected in various tumors. However, to our knowledge no previous study has investigated Cyclin B1 and CDK1 in vulvar cancer. Therefore, we evaluated the statuses of CDK1Tyr15, pCDK1Thr161, Cyclin B1 (total) and pCyclin B1Ser126 in 297 cases of vulvar squamous cell carcinomas by immunohistochemistry. Statistical analyses were performed to explore their clinicopathological and prognostic values. In at least 25% of tumor cases high expression of CDK1Tyr15, pCDK1Thr161, Cyclin B1 (total) and pCyclin B1Ser126 was observed, compared to the low levels in normal vulvar squamous epithelium. Elevated levels of CDK1Tyr15, pCDK1Thr161, Cyclin B1 (total) and pCyclin B1Ser126 were correlated with advanced tumor behaviors and aggressive features. Although CDK1Tyr15, pCDK1Thr161, Cyclin B1 (total) and pCyclin B1Ser126 could not be identified as prognostic factors, combinations of (pCDK1Thr161 C+N + 14-3-3σN), (pCDK1Thr161 C+N + 14-3-3ηC), (pCDK1Thr161 C+N + Wee1C) and (pCDK1Thr161 C+N + 14-3-3σN + 14-3-3ηC + Wee1C) were correlated with disease-specific survival (p = 0.036, p = 0.029, p = 0.042 and p = 0.007, respectively) in univariate analysis. The independent prognostic significance of (pCDK1Thr161 C+N + 14-3-3σN + 14-3-3ηC + Wee1C) was confirmed by multivariate analysis. In conclusion, CDK1Tyr15, pCDK1Thr161, Cyclin B1 (total) and pCyclin B1Ser126 may be involved in progression of vulvar squamous cell carcinoma. The combination of pCDK1Thr161, 14-3-3σ, 14-3-3η and Wee1 was a statistically independent prognostic factor.
Collapse
|
478
|
He XB, Kim M, Kim SY, Yi SH, Rhee YH, Kim T, Lee EH, Park CH, Dixit S, Harrison FE, Lee SH. Vitamin C facilitates dopamine neuron differentiation in fetal midbrain through TET1- and JMJD3-dependent epigenetic control manner. Stem Cells 2015; 33:1320-32. [PMID: 25535150 PMCID: PMC4435601 DOI: 10.1002/stem.1932] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/06/2014] [Accepted: 12/04/2014] [Indexed: 12/11/2022]
Abstract
Intracellular Vitamin C (VC) is maintained at high levels in the developing brain by the activity of sodium-dependent VC transporter 2 (Svct2), suggesting specific VC functions in brain development. A role of VC as a cofactor for Fe(II)-2-oxoglutarate-dependent dioxygenases has recently been suggested. We show that VC supplementation in neural stem cell cultures derived from embryonic midbrains greatly enhanced differentiation toward midbrain-type dopamine (mDA) neurons, the neuronal subtype associated with Parkinson's disease. VC induced gain of 5-hydroxymethylcytosine (5hmC) and loss of H3K27m3 in DA phenotype gene promoters, which are catalyzed by Tet1 and Jmjd3, respectively. Consequently, VC enhanced DA phenotype gene transcriptions in the progenitors by Nurr1, a transcription factor critical for mDA neuron development, to be more accessible to the gene promoters. Further mechanism studies including Tet1 and Jmjd3 knockdown/inhibition experiments revealed that both the 5hmC and H3K27m3 changes, specifically in the progenitor cells, are indispensible for the VC-mediated mDA neuron differentiation. We finally show that in Svct2 knockout mouse embryos, mDA neuron formation in the developing midbrain decreased along with the 5hmC/H3k27m3 changes. These findings together indicate an epigenetic role of VC in midbrain DA neuron development.
Collapse
Affiliation(s)
- Xi-Biao He
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 133-791, Korea
| | - Mirang Kim
- Biomedical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 305-806, Korea
| | - Seon-Young Kim
- Biomedical Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 305-806, Korea
| | - Sang-Hoon Yi
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 133-791, Korea
| | - Yong-Hee Rhee
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 133-791, Korea
| | - Taeho Kim
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 133-791, Korea
| | - Eun-Hye Lee
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 133-791, Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 133-791, Korea
| | - Chang-Hwan Park
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 133-791, Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 133-791, Korea
| | - Shilpy Dixit
- Division of Diabetes, Endocrinology & Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee, U.S.A
| | - Fiona E. Harrison
- Division of Diabetes, Endocrinology & Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee, U.S.A
| | - Sang-Hun Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 133-791, Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 133-791, Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 133-791, Korea
| |
Collapse
|
479
|
Tang Y, Liu L, Sheng M, Xiong K, Huang L, Gao Q, Wei J, Wu T, Yang S, Liu H, Mu Y, Li K. Wip1 knockout inhibits the proliferation and enhances the migration of bone marrow mesenchymal stem cells. Exp Cell Res 2015; 334:310-22. [PMID: 25839408 DOI: 10.1016/j.yexcr.2015.03.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/12/2015] [Accepted: 03/21/2015] [Indexed: 10/23/2022]
Abstract
Mesenchymal stem cells (MSCs), a unique population of multipotent adult progenitor cells originally found in bone marrow (BM), are extremely useful for multifunctional therapeutic approaches. However, the growth arrest and premature senescence of MSCs in vitro prevent the in-depth characterization of these cells. In addition, the regulatory factors involved in MSCs migration remain largely unknown. Given that protein phosphorylation is associated with the processes of MSCs proliferation and migration, we focused on wild-type p53-inducible phosphatase-1 (Wip1), a well-studied modulator of phosphorylation, in this study. Our results showed that Wip1 knockout significantly inhibited MSCs proliferation and induced G2-phase cell-cycle arrest by reducing cyclinB1 expression. Compared with WT-MSCs, Wip1(-/-) MSCs displayed premature growth arrest after six passages in culture. Transwell and scratch assays revealed that Wip1(-/-) MSCs migrate more effectively than WT-MSCs. Moreover, the enhanced migratory response of Wip1(-/-) MSCs may be attributed to increases in the induction of Rac1-GTP activity, the pAKT/AKT ratio, the rearrangement of filamentous-actin (f-actin), and filopodia formation. Based on these results, we then examined the effect of treatment with a PI3K/AKT and Rac1 inhibitor, both of which impaired the migratory activity of MSCs. Therefore, we propose that the PI3K/AKT/Rac1 signaling axis mediates the Wip1 knockout-induced migration of MSCs. Our findings indicate that the principal function of Wip1 in MSCs transformation is the maintenance of proliferative capacity. Nevertheless, knocking out Wip1 increases the migratory capacity of MSCs. This dual effect of Wip1 provides the potential for purposeful routing of MSCs.
Collapse
Affiliation(s)
- Yiting Tang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; State Key Laboratory of Animal Nutrition and Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lan Liu
- State Key Laboratory of Animal Nutrition and Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ming Sheng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Kai Xiong
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, Grønnegårdsvej 7, 1870 Frederiksberg C, Denmark
| | - Lei Huang
- State Key Laboratory of Animal Nutrition and Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qian Gao
- State Key Laboratory of Animal Nutrition and Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jingliang Wei
- State Key Laboratory of Animal Nutrition and Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Tianwen Wu
- State Key Laboratory of Animal Nutrition and Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Shulin Yang
- State Key Laboratory of Animal Nutrition and Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Honglin Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Yulian Mu
- State Key Laboratory of Animal Nutrition and Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Kui Li
- State Key Laboratory of Animal Nutrition and Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
480
|
Ishihara K, Nguyen PA, Wühr M, Groen AC, Field CM, Mitchison TJ. Organization of early frog embryos by chemical waves emanating from centrosomes. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0454. [PMID: 25047608 DOI: 10.1098/rstb.2013.0454] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The large cells in early vertebrate development face an extreme physical challenge in organizing their cytoplasm. For example, amphibian embryos have to divide cytoplasm that spans hundreds of micrometres every 30 min according to a precise geometry, a remarkable accomplishment given the extreme difference between molecular and cellular scales in this system. How do the biochemical reactions occurring at the molecular scale lead to this emergent behaviour of the cell as a whole? Based on recent findings, we propose that the centrosome plays a crucial role by initiating two autocatalytic reactions that travel across the large cytoplasm as chemical waves. Waves of mitotic entry and exit propagate out from centrosomes using the Cdk1 oscillator to coordinate the timing of cell division. Waves of microtubule-stimulated microtubule nucleation propagate out to assemble large asters that position spindles for the following mitosis and establish cleavage plane geometry. By initiating these chemical waves, the centrosome rapidly organizes the large cytoplasm during the short embryonic cell cycle, which would be impossible using more conventional mechanisms such as diffusion or nucleation by structural templating. Large embryo cells provide valuable insights to how cells control chemical waves, which may be a general principle for cytoplasmic organization.
Collapse
Affiliation(s)
- Keisuke Ishihara
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA Marine Biological Laboratory, Woods Hole, MA, USA
| | - Phuong A Nguyen
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA Marine Biological Laboratory, Woods Hole, MA, USA
| | - Martin Wühr
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA Marine Biological Laboratory, Woods Hole, MA, USA
| | - Aaron C Groen
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA Marine Biological Laboratory, Woods Hole, MA, USA
| | - Christine M Field
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA Marine Biological Laboratory, Woods Hole, MA, USA
| | - Timothy J Mitchison
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA Marine Biological Laboratory, Woods Hole, MA, USA
| |
Collapse
|
481
|
Arquint C, Gabryjonczyk AM, Nigg EA. Centrosomes as signalling centres. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0464. [PMID: 25047618 DOI: 10.1098/rstb.2013.0464] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Centrosomes-as well as the related spindle pole bodies (SPBs) of yeast-have been extensively studied from the perspective of their microtubule-organizing roles. Moreover, the biogenesis and duplication of these organelles have been the subject of much attention, and the importance of centrosomes and the centriole-ciliary apparatus for human disease is well recognized. Much less developed is our understanding of another facet of centrosomes and SPBs, namely their possible role as signalling centres. Yet, many signalling components, including kinases and phosphatases, have been associated with centrosomes and spindle poles, giving rise to the hypothesis that these organelles might serve as hubs for the integration and coordination of signalling pathways. In this review, we discuss a number of selected studies that bear on this notion. We cover different processes (cell cycle control, development, DNA damage response) and organisms (yeast, invertebrates and vertebrates), but have made no attempt to be comprehensive. This field is still young and although the concept of centrosomes and SPBs as signalling centres is attractive, it remains primarily a concept-in need of further scrutiny. We hope that this review will stimulate thought and experimentation.
Collapse
Affiliation(s)
- Christian Arquint
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | | | - Erich A Nigg
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| |
Collapse
|
482
|
Andrzejewska Z, Névo N, Thomas L, Bailleux A, Chauvet V, Benmerah A, Antignac C. Lysosomal Targeting of Cystinosin Requires AP-3. Traffic 2015; 16:712-26. [PMID: 25753619 DOI: 10.1111/tra.12277] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 02/20/2015] [Accepted: 02/23/2015] [Indexed: 01/28/2023]
Abstract
Cystinosin is a lysosomal cystine transporter defective in cystinosis, an autosomal recessive lysosomal storage disorder. It is composed of seven transmembrane (TM) domains and contains two lysosomal targeting motifs: a tyrosine-based signal (GYDQL) in its C-terminal tail and a non-classical motif in its fifth inter-TM loop. Using the yeast two-hybrid system, we showed that the GYDQL motif specifically interacted with the μ subunit of the adaptor protein complex 3 (AP-3). Moreover, cell surface biotinylation and total internal reflection fluorescence microscopy revealed that cystinosin was partially mislocalized to the plasma membrane (PM) in AP-3-depleted cells. We generated a chimeric CD63 protein to specifically analyze the function of the GYDQL motif. This chimeric protein was targeted to lysosomes in a manner similar to cystinosin and was partially mislocalized to the PM in AP-3 knockdown cells where it also accumulated in the trans-Golgi network and early endosomes. Together with the fact that the surface levels of cystinosin and of the CD63-GYDQL chimeric protein were not increased when clathrin-mediated endocytosis was impaired, our data show that the tyrosine-based motif of cystinosin is a 'strong' AP-3 interacting motif responsible for lysosomal targeting of cystinosin by a direct intracellular pathway.
Collapse
Affiliation(s)
- Zuzanna Andrzejewska
- Inserm U1163, Laboratory of Hereditary Kidney Diseases, Paris, France.,Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Nathalie Névo
- Inserm U1163, Laboratory of Hereditary Kidney Diseases, Paris, France.,Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Lucie Thomas
- Inserm U1163, Laboratory of Hereditary Kidney Diseases, Paris, France.,Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Anne Bailleux
- Inserm U1163, Laboratory of Hereditary Kidney Diseases, Paris, France.,Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Véronique Chauvet
- Inserm U1163, Laboratory of Hereditary Kidney Diseases, Paris, France.,Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Alexandre Benmerah
- Inserm U1163, Laboratory of Hereditary Kidney Diseases, Paris, France.,Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Corinne Antignac
- Inserm U1163, Laboratory of Hereditary Kidney Diseases, Paris, France.,Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Department of Genetics, Necker Hospital, Paris,, France
| |
Collapse
|
483
|
Talaber G, Tuckermann JP, Okret S. ACTH controls thymocyte homeostasis independent of glucocorticoids. FASEB J 2015; 29:2526-34. [PMID: 25733567 DOI: 10.1096/fj.14-268508] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/09/2015] [Indexed: 12/22/2022]
Abstract
It has been known for decades that lowering the circulating glucocorticoid (GC) concentration as in Addison's disease or after removing the adrenals results in thymus enlargement, largely due to thymocyte expansion. This has been attributed to the loss of the proapoptotic effects on thymocytes by adrenal GCs. Here, we test this concept and report that ACTH directly controls thymic growth post-adrenalectomy (ADX) independent of the proapoptotic effect of GCs. This was supported by the finding that ADX caused thymus enlargement and a 1.7-fold (P < 0.001) increase in thymocyte number in GR(LckCre) mice resistant to GC-induced thymocyte apoptosis, similar to the increase seen in wild-type mice (2.2-fold; P < 0.01). We show by immunostaining that melanocortin receptor subtype 2, which selectively binds ACTH, is partly expressed on the thymic epithelium. Furthermore, ACTH in comparison to vehicle induced a 2.0-fold (P < 0.01) increase in fetal thymic organ culture thymocyte numbers in vitro and enhanced 2.2-fold (P < 0.05) the expression of delta-like ligand 4, a factor that supports T-cell development. Additionally, adrenalectomized GR(LckCre) mice treated with ACTH under conditions that repressed endogenous ACTH secretion showed increased thymocyte cellularity (1.9-fold; P < 0.01) and splenic naive T-cell numbers (2.5-fold; P < 0.001) compared to when treated with PBS. Altogether, our results show that ACTH directly controls thymocyte homeostasis independent of GCs. These results revise the old paradigm behind compensatory thymus growth following ADX, now demonstrating that ACTH has a central role in regulating thymocyte expansion when systemic GC concentration is low.
Collapse
Affiliation(s)
- Gergely Talaber
- *Department of Biosciences and Nutrition, Karolinska Institutet, Novum, Huddinge, Sweden; and Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Jan Peter Tuckermann
- *Department of Biosciences and Nutrition, Karolinska Institutet, Novum, Huddinge, Sweden; and Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Sam Okret
- *Department of Biosciences and Nutrition, Karolinska Institutet, Novum, Huddinge, Sweden; and Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| |
Collapse
|
484
|
Chang WL, Yu CC, Chen CS, Guh JH. Tubulin-binding agents down-regulate matrix metalloproteinase-2 and -9 in human hormone-refractory prostate cancer cells – a critical role of Cdk1 in mitotic entry. Biochem Pharmacol 2015; 94:12-21. [PMID: 25615907 DOI: 10.1016/j.bcp.2015.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 01/10/2015] [Accepted: 01/13/2015] [Indexed: 12/14/2022]
Abstract
Tubulin is an important target for anticancer therapy. Taxanes and vinca alkaloids are two groups of tubulin-binding agents in cancer chemotherapy. Besides tubulin binding, these groups of agents can also down-regulate protein levels of matrix metalloproteinase (MMP)-2 and -9, two important cancer-associated zinc-dependent endopeptidases in invasion and metastasis. However, the mechanism of action waits to be explored. In this study, protein levels but not mRNA expressions of MMP-2 and -9 were down-regulated by paclitaxel (a microtubule-stabilization agent), vincristine and evodiamine (two tubulin-depolymerization agents). These agents induced an increase of protein expression of cyclin B1, MPM2 (mitosis-specific phosphoprotein) and polo-like kinase (PLK) 1 phosphorylation. The data showed a negative relationship between the levels of mitotic proteins and MMP-2 and -9 expressions. MG132 (a specific cell-permeable proteasome inhibitor) blocked mitotic entry and arrested cell cycle at G2 phase, preventing down-regulation of MMP-2 and -9. Cell cycle synchronization experiments by thymidine block or nocodazole treatment showed that mitotic exit inhibited the down-regulation of MMP-2 and -9, confirming negative relationship between cell mitosis and protein levels of MMP-2 and -9 expressions. Cyclin-dependent kinase (Cdk) 1 is a key kinase in mitotic entry. Knockdown of Cdk1 almost completely inhibited the down-regulation of MMP-2 and -9 induced by tubulin-binding agents. In conclusion, the data suggest that mitotic entry and Cdk1 plays a central role in down-regulation of MMP-2 and -9 protein expressions. Tubulin-binding agents cause mitotic arrest and Cdk1 activation, which may contribute largely to the down-regulation of both MMP-2 and -9 expressions.
Collapse
Affiliation(s)
- Wei-Ling Chang
- School of Pharmacy, National Taiwan University, No. 1, Sect. 1, Jen-Ai Rd, Taipei 100, Taiwan; The Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Chia-Chun Yu
- School of Pharmacy, National Taiwan University, No. 1, Sect. 1, Jen-Ai Rd, Taipei 100, Taiwan
| | - Ching-Shih Chen
- The Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA.
| | - Jih-Hwa Guh
- School of Pharmacy, National Taiwan University, No. 1, Sect. 1, Jen-Ai Rd, Taipei 100, Taiwan.
| |
Collapse
|
485
|
Alkan Z, Duong FL, Hawkes WC. Selenoprotein W controls epidermal growth factor receptor surface expression, activation and degradation via receptor ubiquitination. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1087-95. [PMID: 25721765 DOI: 10.1016/j.bbamcr.2015.02.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 02/02/2015] [Accepted: 02/16/2015] [Indexed: 12/22/2022]
Abstract
Epidermal growth factor (EGF) receptor (EGFR) is the founding member of the ErbB family of growth factor receptors that modulate a complex network of intracellular signaling pathways controlling growth, proliferation, differentiation, and motility. Selenoprotein W (SEPW1) is a highly conserved, diet-regulated 9kDa thioredoxin-like protein required for normal cell cycle progression. We report here that SEPW1 is required for EGF-induced EGFR activation and that it functions by suppressing EGFR ubiquitination and receptor degradation. SEPW1 depletion inhibited EGF-dependent cell cycle entry in breast and prostate epithelial cells. In prostate cells, SEPW1 depletion decreased EGFR auto-phosphorylation, while SEPW1 overexpression increased EGFR auto-phosphorylation. SEPW1 depletion increased the rate of EGFR degradation, which decreased total and surface EGFR and suppressed EGF-dependent EGFR endocytosis, EGFR dimer formation, and activation of EGF-dependent pathways. EGFR ubiquitination was increased in SEPW1-depleted cells--in agreement with the increased rate of EGFR degradation, and suggests that SEPW1 suppresses EGFR ubiquitination. Ubiquitination-directed lysozomal degradation controls post-translational EGFR expression and is dysregulated in many cancers. Thus, suppression of EGFR ubiquitination by SEPW1 may be related to the putative increase in cancer risk associated with high selenium intakes. Knowledge of the mechanisms underlying SEPW1's regulation of EGFR ubiquitination may reveal new opportunities for nutritional cancer prevention or cancer drug development.
Collapse
Affiliation(s)
- Zeynep Alkan
- USDA-ARS Western Human Nutrition Research Center, 430 West Health Sciences Drive, University of California, Davis, CA 95616, USA.
| | - Frank L Duong
- Cedars Sinai Medical Center, Department of Medicine, 8750 Beverly Boulevard, Atrium 103, West Hollywood, CA 90048, USA
| | - Wayne C Hawkes
- USDA-ARS Western Human Nutrition Research Center, 430 West Health Sciences Drive, University of California, Davis, CA 95616, USA
| |
Collapse
|
486
|
Activation status of human microglia is dependent on lesion formation stage and remyelination in multiple sclerosis. J Neuropathol Exp Neurol 2015; 74:48-63. [PMID: 25470347 DOI: 10.1097/nen.0000000000000149] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Similar to macrophages, microglia adopt diverse activation states and contribute to repair and tissue damage in multiple sclerosis. Using reverse transcription-quantitative polymerase chain reaction and immunohistochemistry, we show that in vitro M1-polarized (proinflammatory) human adult microglia express the distinctive markers CD74, CD40, CD86, and CCR7, whereas M2 (anti-inflammatory) microglia express mannose receptor and the anti-inflammatory cytokine CCL22. The expression of these markers was assessed in clusters of activated microglia in normal-appearing white matter (preactive lesions) and areas of remyelination, representing reparative multiple sclerosis lesions. We show that activated microglia in preactive and remyelinating lesions express CD74, CD40, CD86, and the M2 markers CCL22 and CD209, but not mannose receptor. To examine whether this intermediate microglia profile is static or dynamic and thus susceptible to changes in the microenvironment, we polarized microglia into M1 or M2 phenotype in vitro and then subsequently treated them with the opposing polarization regimen. These studies revealed that expression of CD40, CXCL10, and mannose receptor is dynamic and that microglia, like macrophages, can switch between M1 and M2 phenotypic profiles. Taken together, our data define the differential activation states of microglia during lesion development in multiple sclerosis-affected CNS tissues and underscore the plasticity of human adult microglia in vitro.
Collapse
|
487
|
Abstract
In this article, we will discuss the biochemistry of mitosis in eukaryotic cells. We will focus on conserved principles that, importantly, are adapted to the biology of the organism. It is vital to bear in mind that the structural requirements for division in a rapidly dividing syncytial Drosophila embryo, for example, are markedly different from those in a unicellular yeast cell. Nevertheless, division in both systems is driven by conserved modules of antagonistic protein kinases and phosphatases, underpinned by ubiquitin-mediated proteolysis, which create molecular switches to drive each stage of division forward. These conserved control modules combine with the self-organizing properties of the subcellular architecture to meet the specific needs of the cell. Our discussion will draw on discoveries in several model systems that have been important in the long history of research on mitosis, and we will try to point out those principles that appear to apply to all cells, compared with those in which the biochemistry has been specifically adapted in a particular organism.
Collapse
Affiliation(s)
- Samuel Wieser
- The Gurdon Institute, Cambridge CB2 1QN, United Kingdom
| | | |
Collapse
|
488
|
Sá SI, Fonseca BM, Teixeira N, Madeira MD. Estrogen receptors α and β have different roles in the induction and trafficking of progesterone receptors in hypothalamic ventromedial neurons. FEBS J 2015; 282:1126-36. [PMID: 25612677 DOI: 10.1111/febs.13207] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 11/21/2014] [Accepted: 01/16/2015] [Indexed: 12/21/2022]
Abstract
Progesterone receptor (PR) activation in the ventrolateral division of the hypothalamic ventromedial nucleus (VMNvl) is essential for promoting female sexual behavior. Estrogen receptor (ER) α, in contrast to ERβ, has been implicated in the induction of PRs. The simultaneous activation of ERα and ERβ, although not increasing the number of PR-immunoreactive neurons in the VMNvl, facilitates lordosis, which suggests that ERβ and/or the ERα-ERβ interaction might play a role in PR dynamics and/or PR expression by individual neurons. To address this question, we used western blot and immunohistochemical studies to determine the amounts and subcellular distributions of both PR isoforms in VMNvl neurons of ovariectomized rats injected with estradiol benzoate or with specific agonists of ERα and ERβ, alone or in association. The present data show that ERα activation does not change PR expression in individual neurons, but increases the number of PRs in the VMNvl, because it increases the number of neurons expressing PRs. Conversely, ERβ activation does not change the total number of PRs in the VMNvl, but increases the labeling intensity of the perikaryal cytoplasm, which suggests that it promotes the transport of PRs from neurites into cell bodies. In addition, the simultaneous activation of ERα and ERβ increases the expression of PRs by individual neurons and, consequently, increases the total number of PRs in the VMNvl. Our findings reveal that individual and simultaneous activation of ERα and ERβ have different effects on the levels and subcellular location of PRs in VMNvl neurons.
Collapse
Affiliation(s)
- Susana I Sá
- Department of Anatomy, Faculty of Medicine, University of Porto, Portugal; Center of Experimental Morphology (CME), Faculty of Medicine, University of Porto, Portugal; Center for Health Technology and Services Research (CINTESIS), Faculty of Medicine, University of Porto, Portugal
| | | | | | | |
Collapse
|
489
|
Wang X, Burghardt RC, Romero JJ, Hansen TR, Wu G, Bazer FW. Functional roles of arginine during the peri-implantation period of pregnancy. III. Arginine stimulates proliferation and interferon tau production by ovine trophectoderm cells via nitric oxide and polyamine-TSC2-MTOR signaling pathways. Biol Reprod 2015; 92:75. [PMID: 25653279 DOI: 10.1095/biolreprod.114.125989] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
In mammal species, arginine is a multifunctional amino acid required for survival, growth, and development of conceptuses (embryo/fetus and associated extraembryonic membranes) during the peri-implantation period of pregnancy. However, functional roles of arginine with respect to it being a substrate for production of nitric oxide (NO) and polyamines on trophectoderm cell proliferation and function remain largely unknown. To systematically assess roles of arginine in conceptus development and its effect on interferon tau (IFNT) production for pregnancy recognition signaling in ruminants, an established ovine trophectoderm (oTr1) cell line isolated from Day-15 ovine conceptuses were used to determine their response to arginine, putrescine, and NO donors, as well as their associated inhibitors. Arginine at physiological concentration (0.2 mM) stimulated maximum oTr cell proliferation (increased 2.0-fold at 48 h and 2.6-fold at 96 h; P < 0.05), stimulated IFNT production (IFNT/cell increased 3.1-fold; P < 0.05), and increased total protein per cell by more than 1.5-fold (P < 0.05). It also increased phosphorylated tuberous sclerosis protein (p-TSC2) and phosphorylated mechanistic target of rapamycin (MTOR) abundance by more than 2.7- and 4.3-fold (P < 0.0001) after long-term incubation, respectively. When Nω-nitro-L-arginine methyl ester hydrochloride (L-NAME; NO synthase inhibitor), DL-α-difluoromethylornithine hydrochloride hydrate (DFMO; ornithine decarboxylase inhibitor), and the combination (L-NAME + DFMO) were added, the effects of arginine on cell proliferation was reduced by 10.7%, 16.1%, and 22.3% (P < 0.05) at 48 h, and 15.3%, 27.2%, and 39.1% (P < 0.05) at 96 h of incubation, respectively, but values remained 1.5-fold higher (P < 0.05) than for the arginine-free control, which suggests that arginine, per se, serves as a growth factor. Both putrescine and NO stimulate cell proliferation via activation of the TSC2-MTOR signaling cascade, whereas only putrescine increased IFNT production. Collectively, our results indicate that arginine is essential for oTr1 cell proliferation and IFNT production via the NO/polyamine-TSC2-MTOR signaling pathways, particularly the pathway involving polyamine biosynthesis.
Collapse
Affiliation(s)
- Xiaoqiu Wang
- Center for Animal Biotechnology and Genomics, Texas A&M University, College Station, Texas Department of Animal Science, Texas A&M University, College Station, Texas
| | - Robert C Burghardt
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - Jared J Romero
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Thomas R Hansen
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Guoyao Wu
- Center for Animal Biotechnology and Genomics, Texas A&M University, College Station, Texas Department of Animal Science, Texas A&M University, College Station, Texas
| | - Fuller W Bazer
- Center for Animal Biotechnology and Genomics, Texas A&M University, College Station, Texas Department of Animal Science, Texas A&M University, College Station, Texas
| |
Collapse
|
490
|
Abstract
The centrosome was discovered in the late 19th century when mitosis was first described. Long recognized as a key organelle of the spindle pole, its core component, the centriole, was realized more than 50 or so years later also to comprise the basal body of the cilium. Here, we chart the more recent acquisition of a molecular understanding of centrosome structure and function. The strategies for gaining such knowledge were quickly developed in the yeasts to decipher the structure and function of their distinctive spindle pole bodies. Only within the past decade have studies with model eukaryotes and cultured cells brought a similar degree of sophistication to our understanding of the centrosome duplication cycle and the multiple roles of this organelle and its component parts in cell division and signaling. Now as we begin to understand these functions in the context of development, the way is being opened up for studies of the roles of centrosomes in human disease.
Collapse
Affiliation(s)
- Jingyan Fu
- Cancer Research UK Cell Cycle Genetics Group, Department of Genetics, University of Cambridge, Cambridge CB2 3EH, United Kingdom
| | - Iain M Hagan
- Cancer Research UK Manchester Institute, University of Manchester, Withington, Manchester M20 4BX, United Kingdom
| | - David M Glover
- Cancer Research UK Cell Cycle Genetics Group, Department of Genetics, University of Cambridge, Cambridge CB2 3EH, United Kingdom
| |
Collapse
|
491
|
Asghar U, Witkiewicz AK, Turner NC, Knudsen ES. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat Rev Drug Discov 2015; 14:130-46. [PMID: 25633797 PMCID: PMC4480421 DOI: 10.1038/nrd4504] [Citation(s) in RCA: 1305] [Impact Index Per Article: 130.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cancer represents a pathological manifestation of uncontrolled cell division; therefore, it has long been anticipated that our understanding of the basic principles of cell cycle control would result in effective cancer therapies. In particular, cyclin-dependent kinases (CDKs) that promote transition through the cell cycle were expected to be key therapeutic targets because many tumorigenic events ultimately drive proliferation by impinging on CDK4 or CDK6 complexes in the G1 phase of the cell cycle. Moreover, perturbations in chromosomal stability and aspects of S phase and G2/M control mediated by CDK2 and CDK1 are pivotal tumorigenic events. Translating this knowledge into successful clinical development of CDK inhibitors has historically been challenging, and numerous CDK inhibitors have demonstrated disappointing results in clinical trials. Here, we review the biology of CDKs, the rationale for therapeutically targeting discrete kinase complexes and historical clinical results of CDK inhibitors. We also discuss how CDK inhibitors with high selectivity (particularly for both CDK4 and CDK6), in combination with patient stratification, have resulted in more substantial clinical activity.
Collapse
Affiliation(s)
- Uzma Asghar
- Breakthrough Breast Cancer Research Centre, Chester Beatty Laboratories, Institute of Cancer Research, London, SW3 6JB, UK
| | - Agnieszka K Witkiewicz
- Simmons Cancer Center and Department of Pathology, University of Texas Southwestern, Dallas, USA
| | - Nicholas C Turner
- Institute of Cancer Research and Royal Marsden NHS Foundation Trust Breast Cancer Unit, London, SW3 6JJ, UK
| | - Erik S Knudsen
- Simmons Cancer Center and Department of Pathology, University of Texas Southwestern, Dallas, USA
| |
Collapse
|
492
|
Wu Y, Zhuo X, Dai Z, Guo X, Wang Y, Zhang C, Lai L. Modeling the mitotic regulatory network identifies highly efficient anti-cancer drug combinations. MOLECULAR BIOSYSTEMS 2015; 11:497-505. [PMID: 25418836 DOI: 10.1039/c4mb00610k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Targeting mitotic regulation is recognized as an important strategy for cancer therapy. Aurora A/B kinase and polo-like kinase 1 (PLK1) are the key mitotic regulators, and many inhibitors have been developed. Combinations of these inhibitors are anticipated to be more effective therapeutics compared with single-inhibitor treatments; however, a systematic analysis of the combined effects is lacking. Here, we constructed the first mammalian cell mitotic regulation network model, which spans from mitotic entry to anaphase initiation, and contains all key mitotic kinase targets. The combined effects of different kinase inhibitors and microtubule inhibitors were systematically explored. Simultaneous inhibition of Aurora B and PLK1 strongly induces polyploidy. Microtubule inhibitor dosage can be significantly reduced when combined with a PLK1 inhibitor. The efficacy of these inhibitor combinations was validated by our experimental results. The mitotic regulatory network model provides a platform to study the complex interactions during mitosis, enables identification of mitotic regulators, and determines targets for drug discovery research. The suggested use of combining microtubule inhibitors with PLK1 inhibitors is anticipated to enhance microtubule-inhibitor tolerance in a wide range of patients.
Collapse
Affiliation(s)
- Yiran Wu
- BNLMS, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, and Peking-Tsinghua Center for Life Sciences at College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | | | | | | | | | | | | |
Collapse
|
493
|
Cdk1-dependent mitotic enrichment of cortical myosin II promotes cell rounding against confinement. Nat Cell Biol 2015; 17:148-59. [DOI: 10.1038/ncb3098] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/17/2014] [Indexed: 12/16/2022]
|
494
|
Extracellular dopamine and alterations on dopamine transporter are related to reserpine toxicity in Caenorhabditis elegans. Arch Toxicol 2015; 90:633-45. [PMID: 25579234 DOI: 10.1007/s00204-015-1451-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 01/06/2015] [Indexed: 10/24/2022]
Abstract
Reserpine is used as an animal model of parkinsonism. We hypothesized that the involuntary movements induced by reserpine in rodents are induced by dopaminergic toxicity caused by extracellular dopamine accumulation. The present study tested the effects of reserpine on the dopaminergic system in Caenorhabditis elegans. Reserpine was toxic to worms (decreased the survival, food intake, development and changed egg laying and defecation cycles). In addition, reserpine increased the worms' locomotor rate on food and decreased dopamine levels. Morphological evaluations of dopaminergic CEP neurons confirmed neurodegeneration characterized by decreased fluorescence intensity and the number of worms with intact CEP neurons, and increased number of shrunken somas per worm. These effects were unrelated to reserpine's effect on decreased expression of the dopamine transporter, dat-1. Interestingly, the locomotor rate on food and the neurodegenerative parameters fully recovered to basal conditions upon reserpine withdrawal. Furthermore, reserpine decreased survival in vesicular monoamine transporter and dat-1 loss-of-function mutant worms. In addition, worms pre-exposed to dopamine followed by exposure to reserpine had decreased survival. Reserpine activated gst-4, which controls a phase II detoxification enzymes downstream of nuclear factor (erythroid-derived-2)-like 2. Our findings establish that the dopamine transporter, dat-1, plays an important role in reserpine toxicity, likely by increasing extracellular dopamine concentrations.
Collapse
|
495
|
Lei Q, Zhang L, Xia Y, Ye T, Yang F, Zhu Y, Song X, Wang N, Xu Y, Liu X, Yu L. A novel benzothiazole derivative SKLB826 inhibits human hepatocellular carcinoma growth via inducing G2/M phase arrest and apoptosis. RSC Adv 2015. [DOI: 10.1039/c5ra05387k] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hepatocellular carcinoma is the fifth most common cancer and durable responses in conventional treatments are limited so researchers have been devoted to developing new anti-HCC agents.
Collapse
Affiliation(s)
- Qian Lei
- State Key Laboratory of Biotherapy and Cancer Center
- West China Hospital
- Sichuan University, and Collaborative Innovation Center for Biothrapy
- Chengdu 610041
- China
| | - Lidan Zhang
- School of Chemical Engineering
- Sichuan University
- Chengdu 610041
- China
| | - Yong Xia
- State Key Laboratory of Biotherapy and Cancer Center
- West China Hospital
- Sichuan University, and Collaborative Innovation Center for Biothrapy
- Chengdu 610041
- China
| | - Tinghong Ye
- State Key Laboratory of Biotherapy and Cancer Center
- West China Hospital
- Sichuan University, and Collaborative Innovation Center for Biothrapy
- Chengdu 610041
- China
| | - Fangfang Yang
- State Key Laboratory of Biotherapy and Cancer Center
- West China Hospital
- Sichuan University, and Collaborative Innovation Center for Biothrapy
- Chengdu 610041
- China
| | - Yongxia Zhu
- State Key Laboratory of Biotherapy and Cancer Center
- West China Hospital
- Sichuan University, and Collaborative Innovation Center for Biothrapy
- Chengdu 610041
- China
| | - Xuejiao Song
- State Key Laboratory of Biotherapy and Cancer Center
- West China Hospital
- Sichuan University, and Collaborative Innovation Center for Biothrapy
- Chengdu 610041
- China
| | - Ningyu Wang
- State Key Laboratory of Biotherapy and Cancer Center
- West China Hospital
- Sichuan University, and Collaborative Innovation Center for Biothrapy
- Chengdu 610041
- China
| | - Ying Xu
- State Key Laboratory of Biotherapy and Cancer Center
- West China Hospital
- Sichuan University, and Collaborative Innovation Center for Biothrapy
- Chengdu 610041
- China
| | - Xiaowei Liu
- State Key Laboratory of Biotherapy and Cancer Center
- West China Hospital
- Sichuan University, and Collaborative Innovation Center for Biothrapy
- Chengdu 610041
- China
| | - Luoting Yu
- State Key Laboratory of Biotherapy and Cancer Center
- West China Hospital
- Sichuan University, and Collaborative Innovation Center for Biothrapy
- Chengdu 610041
- China
| |
Collapse
|
496
|
Lan QY, Liu QL, Cai J, Liu AW. 3-cinnamoyl-4-hydroxy-6-methyl-2H-pyran-2-one (CHP) inhibits human ovarian cell proliferation by inducing apoptosis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:155-163. [PMID: 25755702 PMCID: PMC4348880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 12/24/2014] [Indexed: 06/04/2023]
Abstract
Coumarins induce apoptosis by activating mitochondrial pathway and caspase-3-dependent apoptotic pathway. In the present study, we first time investigated the effect of 3-cinnamoyl-4-hydroxy-6-methyl-2H-pyran-2-one (CHP) on induction of apoptosis in human ovarian carcinoma cells. The data from MTT assay revealed a significant inhibitory effect on cell viability at 30 (87%) and 50 μM (74%) concentration of CHP in OVCAR-3 and OVCAR-420 cells, respectively after 72 h. Apoptosis analysis using annexin V/PI double staining followed by flow cytometry showed 59 and 52% binding to annexin V-FITC in OVCAR-3 and OVCAR-420 cells respectively. propidium iodide (PI) staining and flow cytometry examination indicated a significant increase in percentage of cells in G2/M phase after treatment with CHP compared to DMSO control group. Reactive oxygen species (ROS) assay kit showed increase in levels of ROS. We used rhodamine-123 (Rh-123) staining and flow cytometry assay to determine changes in mitochondrial membrane potential (ΛΨm). The results revealed that CHP significantly decreased MMP to 85.65 ± 1.2443% & 49.78 ± 1.6554% at 10 and 30 μM respectively in OVCAR-3 compared to 95.97 ± 2.1243% in control group. Western blot analysis clearly indicated a significant increase in the expression of Caspase-3, Bax, and release of Cytochrome c and decrease in Bcl-2, CDK1 and Cyclin B1 expression on treatment with CHP. Therefore, CHP may become a potential candidate for the treatment of human ovarian cancers.
Collapse
Affiliation(s)
- Qiong-Yu Lan
- Department of Oncology, The Second Affiliated Hospital of Nanchang University Nanchang 330006, P. R. China
| | - Que-Ling Liu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University Nanchang 330006, P. R. China
| | - Jing Cai
- Department of Oncology, The Second Affiliated Hospital of Nanchang University Nanchang 330006, P. R. China
| | - An-Wen Liu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University Nanchang 330006, P. R. China
| |
Collapse
|
497
|
Wang X, Johnson GA, Burghardt RC, Wu G, Bazer FW. Uterine histotroph and conceptus development. I. cooperative effects of arginine and secreted phosphoprotein 1 on proliferation of ovine trophectoderm cells via activation of the PDK1-Akt/PKB-TSC2-MTORC1 signaling cascade. Biol Reprod 2014; 92:51. [PMID: 25550342 DOI: 10.1095/biolreprod.114.125971] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The greatest limitation to reproductive performance in most mammals, including humans, is embryonic mortality, which, in general, claims 20%-40% of the embryos during the peri-implantation period of pregnancy. Both arginine and secreted phosphoprotein 1 (SPP1) are multifunctional molecules that increase significantly in ovine uterine histotroph during early pregnancy. However, little is known about the relationship and underlying mechanisms for synergistic effects of arginine and SPP1, if any, on conceptus (embryo/fetus and associated extraembryonic membranes) development. Therefore, we conducted in vitro experiments using our established ovine trophectoderm cell line (oTr1) isolated from Day 15 ovine conceptuses to determine their proliferative response to individual and synergistic effects of arginine and recombinant SPP1 (rSPP1) that contains an RGD binding sequence. At physiological concentrations, arginine (0.2 mM) stimulated oTr1 cell proliferation 1.7-fold (P < 0.05) at 48 h, whereas rSPP1 (10 ng/ml) had no such effect. However, an additive effect on oTr1 cell proliferation was induced by combination of arginine and SPP1 as compared to the control (2.1-fold increase; P < 0.01), arginine alone (1.3-fold increase; P < 0.05), and rSPP1 alone (1.5-fold increase; P < 0.01). This additive effect was mediated through cooperative activation of the PDK1-Akt/PKB-TSC2-MTORC1 cell signaling cascade. Collectively, results suggest that arginine and SPP1 in histotroph act cooperatively to enhance survival, growth, and development of ovine conceptuses.
Collapse
Affiliation(s)
- Xiaoqiu Wang
- Center for Animal Biotechnology and Genomics, Texas A&M University, College Station, Texas Department of Animal Science, Texas A&M University, College Station, Texas
| | - Greg A Johnson
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - Robert C Burghardt
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - Guoyao Wu
- Center for Animal Biotechnology and Genomics, Texas A&M University, College Station, Texas Department of Animal Science, Texas A&M University, College Station, Texas
| | - Fuller W Bazer
- Center for Animal Biotechnology and Genomics, Texas A&M University, College Station, Texas Department of Animal Science, Texas A&M University, College Station, Texas
| |
Collapse
|
498
|
Li PH, Cao WJ, Mao LL, Huang H, Zheng JN, Pei DS. p42.3 promotes cell proliferation and invasion in human Renal-Cell Carcinoma. Int J Clin Exp Med 2014; 7:4959-4966. [PMID: 25663993 PMCID: PMC4307440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 12/08/2014] [Indexed: 06/04/2023]
Abstract
p42.3 is a tumor-specific gene and found to be over-expressed in many tumor cell lines and primary tumor tissues. It plays a significant role in neoplastic transformation and tumor progression. To date, the association between p42.3 and Renal-Cell Carcinoma (RCC) has not been reported. This study investigated the biological effects and mechanisms of p42.3 in RCC progression. In this study, we found that p42.3 is overexpressed in various kinds of RCC cells, and knockdown of p42.3 dramatically reduced cell proliferation and invasion in vitro. Our studies revealed that overexpression of p42.3 accelerates the epithelial-mesenchymal transition (EMT) progression and induces RCC cells proliferation and invasion. Further studies show that p42.3 may involve in activation of β-catenin and participate in RCC cell invasion. Combined, these data indicate that p42.3 contributes to promoting RCC cells proliferation and invasion through accelerates the EMT progression and β-catenin activation.
Collapse
Affiliation(s)
- Pei-Hua Li
- Department of Otorhinolaryngology, Affiliated Hospital of Xuzhou Medical CollegeXuzhou 221002, China
| | - Wen-Jia Cao
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical CollegeXuzhou 221002, China
| | - Lin-Lin Mao
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical CollegeXuzhou 221002, China
| | - Hui Huang
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical CollegeXuzhou 221002, China
| | - Jun-Nian Zheng
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical CollegeXuzhou 221002, China
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical CollegeXuzhou 221002, China
| | - Dong-Sheng Pei
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical CollegeXuzhou 221002, China
| |
Collapse
|
499
|
Abstract
PURPOSE To quantify the amount of drug loss from cadaveric human eyes, which are injected via the pars plana with a known volume of dye at variable intraocular pressures. METHODS Eight cadaver eyes were divided into 2 intraocular pressure groups: normal (15 mmHg; 4 eyes) or high (30 mmHg; 4 eyes). Each eye was injected with 50 μL of hematoxylin dye, and the subsequent reflux was immediately collected on a Schirmer's test strip. The test strip was scanned and digitally analyzed to determine the area of saturation and total color intensity present. Using a previously established equation, total volume of reflux and the amount of dye within that reflux were calculated. RESULTS The average total volume of refluxed fluid was 1.68 μL (median, 0.62 μL), with a range of 0 μL to 8.05 μL. The average volume of refluxed dye was 0.37 μL (median, 0.08 μL), with a range of 0 μL to 2.15 μL. On average, only 0.74% of the original 50 μL of injected dye was lost (median, 0.15%), with a range from 0% to 4.30%. CONCLUSION Although the presence of subconjunctival bleb formation after intravitreal injection may be a concern to the clinician, data from the present study shows that only a very small amount of the injected therapeutic agent is lost in the reflux.
Collapse
|
500
|
Seyedpour SM, Pachenari M, Janmaleki M, Alizadeh M, Hosseinkhani H. Effects of an antimitotic drug on mechanical behaviours of the cytoskeleton in distinct grades of colon cancer cells. J Biomech 2014; 48:1172-8. [PMID: 25678199 DOI: 10.1016/j.jbiomech.2014.11.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 11/17/2014] [Accepted: 11/20/2014] [Indexed: 11/28/2022]
Abstract
Biomechanical behaviours of cells change during cancer progression due to alterations in the main cytoskeletal proteins. Microtubules play a vital role in mitosis and in supporting the integrity of the cell due to their ability to withstand high compressive loads. Accordingly, microtubule-targeting agents (MTAs) have become one of the most promising classes of drugs in cancer therapy. This study evaluated changes in visco-elastic parameters induced by an appropriate concentration of an antimitotic drug in two different grades of colon cancer cells. Actin microfilaments and microtubules contents in the cells were evaluated by Western blot analysis and fluorescence intensity calculation. Micropipette aspiration experiments showed that the MTA had distinct mechanical effects on different cell lines. The more aggressive the cells, the greater the reduction in elasticity and viscosity. Invasive cells had a higher initial instantaneous Young's modulus than primary cells, but this reduced to approximately one half of the values for primary cells after 48 h of drug treatment. A considerable association was seen between the changes in mechanical properties and the microtubule to F-actin microfilament content ratio, which decreased with MTA treatment.
Collapse
Affiliation(s)
- S M Seyedpour
- Department of Mechanical Engineering, Iran University of Science and Technology, Tehran, Iran; Medical Nanotechnology and Tissue Engineering Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - M Pachenari
- Department of Mechanical Engineering, Iran University of Science and Technology, Tehran, Iran; Medical Nanotechnology and Tissue Engineering Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - M Janmaleki
- Medical Nanotechnology and Tissue Engineering Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - M Alizadeh
- Department of Mechanical Engineering, Iran University of Science and Technology, Tehran, Iran
| | - H Hosseinkhani
- Graduate Institute of Biomedical Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| |
Collapse
|