1
|
Primrose JGB, Jain L, Alhilali M, Bolam SM, Monk AP, Munro JT, Dalbeth N, Poulsen RC. REST, RCOR1 and RCOR2 expression is reduced in osteoarthritic chondrocytes and contributes to increasing MMP13 and ADAMTS5 expression through upregulating HES1. Cell Signal 2023; 109:110800. [PMID: 37442513 DOI: 10.1016/j.cellsig.2023.110800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/15/2023]
Abstract
Expression of key transcriptional regulators is altered in chondrocytes in osteoarthritis (OA). This contributes to an increase in production of cartilage-catabolizing enzymes such as MMP13 and ADAMTS5. RCOR1 and RCOR2, binding partners for the transcriptional repressor REST, have previously been found to be downregulated in OA chondrocytes although their function in chondrocytes is unclear. HES1 is a known REST/RCOR1 target gene and HES1 has been shown to promote MMP13 and ADAMTS5 expression in murine OA chondrocytes. The purpose of this study was to determine whether reduced REST/RCOR levels leads to increased HES1 expression in human OA chondrocytes and whether HES1 also promotes ADAMTS5 and MMP13 expression in these cells. Chondrocytes were isolated from osteoarthritic and adjacent macroscopically normal cartilage obtained from patients undergoing total knee arthroplasty. RNA and protein levels of REST, RCOR1 and RCOR2 were lower, but levels of HES1 higher, in chondrocytes isolated from osteoarthritic compared to macroscopically normal cartilage. Over-expression of either REST, RCOR1 or RCOR2 resulted in reduced HES1 levels in OA chondrocytes whereas knockdown of REST, RCOR1 or RCOR2 led to increased HES1 expression in chondrocytes from macroscopically normal cartilage. In OA chondrocytes, ADAMTS5 and MMP13 expression were reduced following HES1 knockdown, but further enhanced following HES1 over-expression. Levels of phosphorylated CaMKII were higher in chondrocytes from OA cartilage consistent with previous findings that HES1 only promotes gene transcription in the presence of active CaMKII. These findings identify the REST/RCOR/HES1 pathway as a contributing factor leading to increased ADAMTS5 and MMP13 expression in OA chondrocytes.
Collapse
Affiliation(s)
| | - Lekha Jain
- Department of Pharmacology and Clinical Pharmacology, New Zealand
| | - Mariam Alhilali
- Department of Pharmacology and Clinical Pharmacology, New Zealand
| | - Scott M Bolam
- Department of Surgery, New Zealand; Department of Medicine, University of Auckland, Auckland, New Zealand
| | - A Paul Monk
- Auckland Bioengineering Institute, New Zealand
| | | | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Raewyn C Poulsen
- Department of Pharmacology and Clinical Pharmacology, New Zealand; Department of Medicine, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
2
|
Large 1p36 Deletions Affecting Arid1a Locus Facilitate Mycn-Driven Oncogenesis in Neuroblastoma. Cell Rep 2021; 30:454-464.e5. [PMID: 31940489 PMCID: PMC9022217 DOI: 10.1016/j.celrep.2019.12.048] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/23/2019] [Accepted: 12/13/2019] [Indexed: 01/02/2023] Open
Abstract
Loss of heterozygosity (LOH) at 1p36 occurs in multiple cancers, including neuroblastoma (NBL). MYCN amplification and 1p36 deletions tightly correlate with markers of tumor aggressiveness in NBL. Although distal 1p36 losses associate with single-copy MYCN tumors, larger deletions correlate with MYCN amplification, indicating two tumor suppressor regions in 1p36, only one of which facilitates MYCN oncogenesis. To better define this region, we genome-edited the syntenic 1p36 locus in primary mouse neural crest cells (NCCs), a putative NBL cell of origin. In in vitro cell transformation assays, we show that Chd5 loss confers most of the MYCN-independent tumor suppressor effects of 1p36 LOH. In contrast, MYCN-driven tumorigenesis selects for NCCs with Arid1a deletions from a pool of NCCs with randomly sized 1p36 deletions, establishing Arid1a as the MYCN-associated tumor suppressor. Our findings reveal that Arid1a loss collaborates with oncogenic MYCN and better define the tumor suppressor functions of 1p36 LOH in NBL.
Collapse
|
3
|
Baiula M, Caligiana A, Bedini A, Zhao J, Santino F, Cirillo M, Gentilucci L, Giacomini D, Spampinato S. Leukocyte Integrin Antagonists as a Novel Option to Treat Dry Age-Related Macular Degeneration. Front Pharmacol 2021; 11:617836. [PMID: 33584300 PMCID: PMC7878375 DOI: 10.3389/fphar.2020.617836] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/30/2020] [Indexed: 11/28/2022] Open
Abstract
Age-related macular degeneration (AMD) is a complex multifactorial degenerative disease that leads to irreversible blindness. AMD affects the macula, the central part of the retina responsible for sharp central vision. Retinal pigment epithelium (RPE) is the main cellular type affected in dry AMD. RPE cells form a monolayer between the choroid and the neuroretina and are in close functional relationship with photoreceptors; moreover, RPE cells are part of the blood retina barrier that is disrupted in ocular diseases such as AMD. During ocular inflammation lymphocytes and macrophages are recruited, contact RPE and produce pro-inflammatory cytokines, which play an important role in AMD pathogenesis. The interaction between RPE and immune cells is mediated by leukocyte integrins, heterodimeric transmembrane receptors, and adhesion molecules, including VCAM-1 and ICAM-1. Within this frame, this study aimed to characterize RPE-leukocytes interaction and to investigate any potentially beneficial effects induced by integrin antagonists (DS-70, MN27 and SR714), developed in previous studies. ARPE-19 cells were co-cultured for different incubation times with Jurkat cells and apoptosis and necrosis levels were analyzed by flow cytometry. Moreover, we measured the mRNA levels of the pro-inflammatory cytokine IL-1β and the expression of adhesion molecules VCAM-1 and ICAM-1. We found that RPE-lymphocyte interaction increased apoptosis and necrosis levels in RPE cells and the expression of IL-1β. This interaction was mediated by the binding of α4β1 and αLβ2 integrins to VCAM-1 and ICAM-1, respectively. The blockade of RPE-lymphocyte interaction with blocking antibodies highlighted the pivotal role played by integrins. Therefore, α4β1 and αLβ2 integrin antagonists were employed to disrupt RPE-lymphocyte crosstalk. Small molecule integrin antagonists proved to be effective in reducing RPE cell death and expression of IL-1β, demonstrating that integrin antagonists could protect RPE cells from detrimental effects induced by the interaction with immune cells recruited to the retina. Overall, the leukocyte integrin antagonists employed in the present study may represent a novel opportunity to develop new drugs to fight dry AMD.
Collapse
Affiliation(s)
- Monica Baiula
- Laboratory of Cellular and Molecular Pharmacology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Alberto Caligiana
- Laboratory of Cellular and Molecular Pharmacology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Andrea Bedini
- Laboratory of Cellular and Molecular Pharmacology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Junwei Zhao
- Department of Chemistry "G. Ciamician", University of Bologna, Bologna, Italy
| | - Federica Santino
- Department of Chemistry "G. Ciamician", University of Bologna, Bologna, Italy
| | - Martina Cirillo
- Laboratory of Design and Synthesis of Biologically Active Compounds, Department of Chemistry "G. Ciamician", University of Bologna, Bologna, Italy
| | - Luca Gentilucci
- Department of Chemistry "G. Ciamician", University of Bologna, Bologna, Italy
| | - Daria Giacomini
- Laboratory of Design and Synthesis of Biologically Active Compounds, Department of Chemistry "G. Ciamician", University of Bologna, Bologna, Italy
| | - Santi Spampinato
- Laboratory of Cellular and Molecular Pharmacology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,Specilization School of Hospital Pharmacy, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
4
|
Bedini A, Baiula M, Vincelli G, Formaggio F, Lombardi S, Caprini M, Spampinato S. Nociceptin/orphanin FQ antagonizes lipopolysaccharide-stimulated proliferation, migration and inflammatory signaling in human glioblastoma U87 cells. Biochem Pharmacol 2017; 140:89-104. [PMID: 28583844 DOI: 10.1016/j.bcp.2017.05.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 05/31/2017] [Indexed: 01/01/2023]
Abstract
Glioblastoma is among the most aggressive brain tumors and has an exceedingly poor prognosis. Recently, the importance of the tumor microenvironment in glioblastoma cell growth and progression has been emphasized. Toll-like receptor 4 (TLR4) recognizes bacterial lipopolysaccharide (LPS) and endogenous ligands originating from dying cells or the extracellular matrix involved in host defense and in inflammation. G-protein coupled receptors (GPCRs) have gained interest in anti-tumor drug discovery due to the role that they directly or indirectly play by transactivating other receptors, causing cell migration and proliferation. A proteomic analysis showed that the nociceptin receptor (NOPr) is among the GPCRs significantly expressed in glioblastoma cells, including U87 cells. We describe a novel role of the peptide nociceptin (N/OFQ), the endogenous ligand of the NOPr that counteracts cell migration, proliferation and increase in IL-1β mRNA elicited by LPS via TLR4 in U87 glioblastoma cells. Signaling pathways through which N/OFQ inhibits LPS-mediated cell migration and elevation of [Ca2+]i require β-arrestin 2 and are sensitive to TNFR-associated factor 6, c-Src and protein kinase C (PKC). LPS-induced cell proliferation and increase in IL-1β mRNA are counteracted by N/OFQ via β-arrestin 2, PKC and extracellular signal-regulated kinase 1/2; furthermore, the contributions of the transcription factors NF-kB and AP-1 were investigated. Independent of LPS, N/OFQ induces a significant increase in cell apoptosis. Contrary to what was observed in other cell models, a prolonged exposure to this endotoxin did not promote any tolerance of the cellular effects above described, including NOPr down-regulation while N/OFQ loses its inhibitory role.
Collapse
Affiliation(s)
- Andrea Bedini
- Department of Pharmacy and Biotechnology, University of Bologna, Irnerio 48, 40126 Bologna, Italy
| | - Monica Baiula
- Department of Pharmacy and Biotechnology, University of Bologna, Irnerio 48, 40126 Bologna, Italy
| | - Gabriele Vincelli
- Department of Pharmacy and Biotechnology, University of Bologna, Irnerio 48, 40126 Bologna, Italy
| | - Francesco Formaggio
- Department of Pharmacy and Biotechnology, University of Bologna, Irnerio 48, 40126 Bologna, Italy
| | - Sara Lombardi
- Department of Pharmacy and Biotechnology, University of Bologna, Irnerio 48, 40126 Bologna, Italy
| | - Marco Caprini
- Department of Pharmacy and Biotechnology, University of Bologna, Irnerio 48, 40126 Bologna, Italy
| | - Santi Spampinato
- Department of Pharmacy and Biotechnology, University of Bologna, Irnerio 48, 40126 Bologna, Italy.
| |
Collapse
|
5
|
Epigenomics of Neural Cells: REST-Induced Down- and Upregulation of Gene Expression in a Two-Clone PC12 Cell Model. BIOMED RESEARCH INTERNATIONAL 2015; 2015:202914. [PMID: 26413508 PMCID: PMC4564578 DOI: 10.1155/2015/202914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 07/16/2015] [Indexed: 01/02/2023]
Abstract
Cell epigenomics depends on the marks released by transcription factors operating via the assembly of complexes that induce focal changes of DNA and histone structure. Among these factors is REST, a repressor that, via its strong decrease, governs both neuronal and neural cell differentiation and specificity. REST operation on thousands of possible genes can occur directly or via indirect mechanisms including repression of other factors. In previous studies of gene down- and upregulation, processes had been only partially investigated in neural cells. PC12 are well-known neural cells sharing properties with neurons. In the widely used PC12 populations, low-REST cells coexist with few, spontaneous high-REST PC12 cells. High- and low-REST PC12 clones were employed to investigate the role and the mechanisms of the repressor action. Among 15,500 expressed genes we identified 1,770 target and nontarget, REST-dependent genes. Functionally, these genes were found to operate in many pathways, from synaptic function to extracellular matrix. Mechanistically, downregulated genes were predominantly repressed directly by REST; upregulated genes were mostly governed indirectly. Among other factors, Polycomb complexes cooperated with REST for downregulation, and Smad3 and Myod1 participated in upregulation. In conclusion, we have highlighted that PC12 clones are a useful model to investigate REST, opening opportunities to development of epigenomic investigation.
Collapse
|
6
|
Dothel G, Barbaro MR, Boudin H, Vasina V, Cremon C, Gargano L, Bellacosa L, De Giorgio R, Le Berre-Scoul C, Aubert P, Neunlist M, De Ponti F, Stanghellini V, Barbara G. Nerve fiber outgrowth is increased in the intestinal mucosa of patients with irritable bowel syndrome. Gastroenterology 2015; 148:1002-1011.e4. [PMID: 25655556 DOI: 10.1053/j.gastro.2015.01.042] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 01/24/2015] [Accepted: 01/27/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Mediators released by the intestinal mucosa of patients with irritable bowel syndrome (IBS) affect the function of enteric and extrinsic sensory nerves, which can contribute to the development of symptoms. Little is known about the effects of mucosal mediators on intestinal neuroplasticity. We investigated how these mediators affect the phenotypes of colonic mucosa nerve fibers, neuron differentiation, and fiber outgrowth. METHODS We analyzed mucosal biopsy samples collected from 101 patients with IBS and 23 asymptomatic healthy individuals (controls). We measured levels of neuronal-specific enolase, growth-associated protein 43, nerve growth factor (NGF), and tyrosine kinase receptor A (NTRK1) by immunohistochemistry and enzyme-linked immunosorbent assay. Primary rat enteric neurons and human SH-SY5Y cells were incubated with supernatants from the mucosal biopsies and analyzed by morphometric and polymerase chain reaction analyses. RESULTS Compared with mucosal tissues of controls, mucosa from patients with IBS had a significant increase in the area of lamina propria occupied by neuronal-specific enolase-positive (57.7% increase) and growth-associated protein 43-positive fibers (56.1% increase) and staining density of NGF (89.3% increase) (P < .05 for all). Levels of NGF protein were also increased in tissues from patients with IBS vs controls (18% increase; P = .16) along with levels of NTRK1 (64% increase; P < .05). Mucosal supernatants from tissues of patients with IBS induced higher levels of neuritogenesis in primary culture of enteric neurons, compared with controls, and more NGF-dependent neuronal sprouting in SH-SY5Y cells. CONCLUSIONS Nerve fiber density and sprouting, as well as expression of NGF and NTRK1, are significantly increased in mucosal tissues of patients with IBS. Mucosal mediators participate to these neuroplastic changes.
Collapse
Affiliation(s)
- Giovanni Dothel
- Department of Medical and Surgical Sciences and Center for Applied Biomedical Research (CRBA), University of Bologna, Italy
| | - Maria Raffaella Barbaro
- Department of Medical and Surgical Sciences and Center for Applied Biomedical Research (CRBA), University of Bologna, Italy
| | - Hélène Boudin
- Inserm, U913, Institut des Maladies de l'Appareil Digestif du CHU de Nantes; University of Nantes, Nantes, France
| | - Valentina Vasina
- Department of Medical and Surgical Sciences and Center for Applied Biomedical Research (CRBA), University of Bologna, Italy
| | - Cesare Cremon
- Department of Medical and Surgical Sciences and Center for Applied Biomedical Research (CRBA), University of Bologna, Italy
| | - Luciana Gargano
- Department of Medical and Surgical Sciences and Center for Applied Biomedical Research (CRBA), University of Bologna, Italy
| | - Lara Bellacosa
- Department of Medical and Surgical Sciences and Center for Applied Biomedical Research (CRBA), University of Bologna, Italy
| | - Roberto De Giorgio
- Department of Medical and Surgical Sciences and Center for Applied Biomedical Research (CRBA), University of Bologna, Italy
| | - Catherine Le Berre-Scoul
- Inserm, U913, Institut des Maladies de l'Appareil Digestif du CHU de Nantes; University of Nantes, Nantes, France
| | - Philippe Aubert
- Inserm, U913, Institut des Maladies de l'Appareil Digestif du CHU de Nantes; University of Nantes, Nantes, France
| | - Michel Neunlist
- Inserm, U913, Institut des Maladies de l'Appareil Digestif du CHU de Nantes; University of Nantes, Nantes, France
| | - Fabrizio De Ponti
- Department of Medical and Surgical Sciences and Center for Applied Biomedical Research (CRBA), University of Bologna, Italy
| | - Vincenzo Stanghellini
- Department of Medical and Surgical Sciences and Center for Applied Biomedical Research (CRBA), University of Bologna, Italy
| | - Giovanni Barbara
- Department of Medical and Surgical Sciences and Center for Applied Biomedical Research (CRBA), University of Bologna, Italy.
| |
Collapse
|
7
|
Li Z, Pei Q, Cao L, Xu L, Zhang B, Liu S. Propofol increases µ-opioid receptor expression in SH-SY5Y human neuroblastoma cells. Mol Med Rep 2012; 6:1333-6. [PMID: 22965315 DOI: 10.3892/mmr.2012.1073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 08/17/2012] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to explore the effect of propofol, a intravenous sedative-hypnotic agent used widely in inducing and maintaining anesthesia, on µ-opioid receptor (MOR) expression in a human neuronal cell line. SH-SY5Y human neuroblastoma cells were treated with various concentrations of propofol (1, 5, 10 or 20 µM) for different lengths of time (6, 12 or 24 h). Real-time quantitative RT-PCR showed that at a concentration range of 1-10 µM, propofol increased MOR mRNA levels in a statistically significant dose- and time-dependent manner within 12 h of treatment. Western blot analyses demonstrated that propofol treatment for 12 h dose-dependently increased the MOR protein levels. In the 12-h SH-SY5Y-treated cells, propofol dose-dependently increased MOR density (Bmax) in the cell membranes. In addition, in the presence of the transcription inhibitor actinomycin D (1 mg/ml), propofol (10 µM) had no significant effect on the MOR mRNA levels over time. The results suggested that propofol dose- and time-dependently enhances MOR expression in SH-SY5Y human neuroblastoma cells at the transcriptional level, leading to an increased density of ligand-binding MORs in the cell membranes. This study demonstrated for the first time a link between propofol and the opioid system, thereby providing new insights into propofol mechanism of action and potential for abuse.
Collapse
Affiliation(s)
- Zuojun Li
- Department of Pharmacy, Third Xiangya Hospital, Central South University, Changsha, P.R. China
| | | | | | | | | | | |
Collapse
|
8
|
Baiula M, Carbonari G, Dattoli SD, Calienni M, Bedini A, Spampinato S. REST is up-regulated by epidermal growth factor in HeLa cells and inhibits apoptosis by influencing histone H3 acetylation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:1252-63. [PMID: 22668508 DOI: 10.1016/j.bbamcr.2012.05.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 05/09/2012] [Accepted: 05/24/2012] [Indexed: 12/17/2022]
Abstract
REST (repressor element 1-silencing transcription factor) is a transcription factor that recruits histone deacetylases to silence gene transcription. REST appears to play a paradoxical role in cancer cells: it exhibits tumor suppressor activity or promotes tumorigenesis, depending upon the setting. The extracellular signaling molecules that control REST gene expression in cancer cells remain poorly understood. In this study, we report that REST expression in HeLa cells is elevated in cells exposed to epidermal growth factor or serum, whereas the rate of cell apoptosis is low. Apoptosis induced by serum withdrawal is significantly increased in HeLa cells treated with an antisense phosphorothioate oligodeoxynucleotide (AS ODN) capable of down-regulating REST expression, whereas in HeLa cells transfected with a REST expressing plasmid, REST overexpression reduces the marked apoptosis caused, in absence of serum, by exposure to an anti-Fas receptor antibody imitating the Fas ligand activity plus PD 98059, a blocker of extracellular signal-regulated kinase 1/2 activation. REST knockdown also reduces mRNA levels of the antiapoptotic protein Bcl-X(L) whereas in HeLa cells overexpressing REST, the reduction of Bcl-X(L) mRNA caused by the anti-Fas receptor antibody plus PD 98059 is significantly decreased. Finally, we report that acetylation of histone H3 is increased in HeLa cells exposed to AS ODN or anti-Fas receptor antibody, whereas it is reduced in cells transfected with the REST expressing plasmid. Our findings indicate that REST is a novel gene regulated by EGF in HeLa cells that potentially contributes to the modulation of apoptosis via epigenetic mechanisms.
Collapse
Affiliation(s)
- Monica Baiula
- Department of Pharmacology, University of Bologna, Bologna, Italy
| | | | | | | | | | | |
Collapse
|
9
|
Yanagita T, Satoh S, Uezono Y, Matsuo K, Nemoto T, Maruta T, Yoshikawa N, Iwakiri T, Minami K, Murakami M. Transcriptional up-regulation of cell surface Na V 1.7 sodium channels by insulin-like growth factor-1 via inhibition of glycogen synthase kinase-3β in adrenal chromaffin cells: enhancement of 22Na+ influx, 45Ca2+ influx and catecholamine secretion. Neuropharmacology 2011; 61:1265-74. [PMID: 21816165 DOI: 10.1016/j.neuropharm.2011.07.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 07/14/2011] [Accepted: 07/20/2011] [Indexed: 12/19/2022]
Abstract
Insulin-like growth factor-1 (IGF-1) plays important roles in the regulation of neuronal development. The electrical activity of Na(+) channels is crucial for the regulation of synaptic formation and maintenance/repair of neuronal circuits. Here, we examined the effects of chronic IGF-1 treatment on cell surface expression and function of Na(+) channels. In cultured bovine adrenal chromaffin cells expressing Na(V)1.7 isoform of voltage-dependent Na(+) channels, chronic IGF-1 treatment increased cell surface [(3)H]saxitoxin binding by 31%, without altering the Kd value. In cells treated with IGF-1, veratridine-induced (22)Na(+) influx, and subsequent (45)Ca(2+) influx and catecholamine secretion were augmented by 35%, 33%, 31%, respectively. Pharmacological properties of Na(+) channels characterized by neurotoxins were similar between nontreated and IGF-1-treated cells. IGF-1-induced up-regulation of [(3)H]saxitoxin binding was prevented by phosphatydil inositol-3 kinase inhibitors (LY204002 or wortmannin), or Akt inhibitor (Akt inhibitor IV). Glycogen synthase kinase-3 (GSK-3) inhibitors (LiCl, valproic acid, SB216763 or SB415286) also increased cell surface [(3)H]saxitoxin binding by ∼ 33%, whereas simultaneous treatment of IGF-1 with GSK-3 inhibitors did not produce additive increasing effect on [(3)H]saxitoxin binding. IGF-1 (100 nM) increased Ser(437)-phosphorylated Akt and Ser(9)-phosphorylated GSK-3β, and inhibited GSK-3β activity. Treatment with IGF-1, LiCl or SB216763 increased protein level of Na(+) channel α-subunit; it was prevented by cycloheximide. Either treatment increased α-subunit mRNA level by ∼ 48% and accelerated α-subunit gene transcription by ∼ 30% without altering α-subunit mRNA stability. Thus, inhibition of GSK-3β caused by IGF-1 up-regulates cell surface expression of functional Na(+) channels via acceleration of α-subunit gene transcription.
Collapse
Affiliation(s)
- Toshihiko Yanagita
- Department of Pharmacology, Miyazaki Medical College, University of Miyazaki, 5200 Kihara Kiyotake, Miyazaki 889-1692, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Bedini A, Baiula M, Carbonari G, Spampinato S. Transcription factor REST negatively influences the protein kinase C-dependent up-regulation of human mu-opioid receptor gene transcription. Neurochem Int 2009; 56:308-17. [PMID: 19913583 DOI: 10.1016/j.neuint.2009.10.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Revised: 10/20/2009] [Accepted: 10/29/2009] [Indexed: 11/15/2022]
Abstract
Mu-opioid receptor expression increases during neurogenesis, regulates the survival of maturing neurons and is implicated in ischemia-induced neuronal death. The repressor element 1 silencing transcription factor (REST), a regulator of a subset of genes in differentiating and post-mitotic neurons, is involved in its transcriptional repression. Extracellular signaling molecules and mechanisms that control the human mu-opioid receptor (hMOR) gene transcription are not clearly understood. We examined the role of protein kinase C (PKC) on hMOR transcription in a model of neuronal cells and in the context of the potential influence of REST. In native SH-SY5Y neuroblastoma cells, PKC activation with phorbol 12-myristate 13-acetate (PMA, 16 nM, 24h) down-regulated hMOR transcription and concomitantly elevated the REST binding activity to repressor element 1 of the hMOR promoter. In contrast, PMA activated hMOR gene transcription when REST expression was knocked down by an antisense strategy or by retinoic acid-induced cell differentiation. PMA acts through a PKC-dependent pathway requiring downstream MAP kinases and the transcription factor AP-1. In a series of hMOR-luciferase promoter/reporter constructs transfected into SH-SY5Y cells and PC12 cells, PMA up-regulated hMOR transcription in PC12 cells lacking REST, and in SH-SY5Y cells either transfected with constructs deficient in the REST DNA binding element or when REST was down-regulated in retinoic acid-differentiated cells. These findings help explain how hMOR transcription is regulated and may clarify its contribution to epigenetic modifications and reprogramming of differentiated neuronal cells exposed to PKC-activating agents.
Collapse
Affiliation(s)
- Andrea Bedini
- Department of Pharmacology, University of Bologna, Bologna, Italy
| | | | | | | |
Collapse
|
11
|
Schulte JH, Lim S, Schramm A, Friedrichs N, Koster J, Versteeg R, Ora I, Pajtler K, Klein-Hitpass L, Kuhfittig-Kulle S, Metzger E, Schüle R, Eggert A, Buettner R, Kirfel J. Lysine-specific demethylase 1 is strongly expressed in poorly differentiated neuroblastoma: implications for therapy. Cancer Res 2009; 69:2065-71. [PMID: 19223552 DOI: 10.1158/0008-5472.can-08-1735] [Citation(s) in RCA: 355] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Aberrant epigenetic changes in DNA methylation and histone acetylation are hallmarks of most cancers, whereas histone methylation was previously considered to be irreversible and less versatile. Recently, several histone demethylases were identified catalyzing the removal of methyl groups from histone H3 lysine residues and thereby influencing gene expression. Neuroblastomas continue to remain a clinical challenge despite advances in multimodal therapy. Here, we address the functional significance of the chromatin-modifying enzyme lysine-specific demethylase 1 (LSD1) in neuroblastoma. LSD1 expression correlated with adverse outcome and was inversely correlated with differentiation in neuroblastic tumors. Differentiation of neuroblastoma cells resulted in down-regulation of LSD1. Small interfering RNA-mediated knockdown of LSD1 decreased cellular growth, induced expression of differentiation-associated genes, and increased target gene-specific H3K4 methylation. Moreover, LSD1 inhibition using monoamine oxidase inhibitors resulted in an increase of global H3K4 methylation and growth inhibition of neuroblastoma cells in vitro. Finally, targeting LSD1 reduced neuroblastoma xenograft growth in vivo. Here, we provide the first evidence that a histone demethylase, LSD1, is involved in maintaining the undifferentiated, malignant phenotype of neuroblastoma cells. We show that inhibition of LSD1 reprograms the transcriptome of neuroblastoma cells and inhibits neuroblastoma xenograft growth. Our results suggest that targeting histone demethylases may provide a novel option for cancer therapy.
Collapse
Affiliation(s)
- Johannes H Schulte
- Department of Paediatric Oncology and Hematology, University Children's Hospital Essen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Nandrolone decreases mu opioid receptor expression in SH-SY5Y human neuroblastoma cells. Neuroreport 2008; 19:1131-5. [DOI: 10.1097/wnr.0b013e328305639d] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
13
|
Bedini A, Baiula M, Spampinato S. Transcriptional activation of human mu-opioid receptor gene by insulin-like growth factor-I in neuronal cells is modulated by the transcription factor REST. J Neurochem 2008; 105:2166-78. [DOI: 10.1111/j.1471-4159.2008.05303.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
14
|
Diss JKJ, Calissano M, Gascoyne D, Djamgoz MBA, Latchman DS. Identification and characterization of the promoter region of the Nav1.7 voltage-gated sodium channel gene (SCN9A). Mol Cell Neurosci 2007; 37:537-47. [PMID: 18249135 DOI: 10.1016/j.mcn.2007.12.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2007] [Revised: 11/15/2007] [Accepted: 12/06/2007] [Indexed: 12/19/2022] Open
Abstract
The Nav1.7 sodium channel plays an important role in pain and is also upregulated in prostate cancer. To investigate the mechanisms regulating physiological and pathophysiological Nav1.7 expression we identified the core promoter of this gene (SCN9A) in the human genome. In silico genomic analysis revealed a putative SCN9A 5' non-coding exon approximately 64,000 nucleotides from the translation start site, expression of which commenced at three very closely-positioned transcription initiation sites (TISs), as determined by 5' RACE experiments. The genomic region around these TISs possesses numerous core elements of a TATA-less promoter within a well-defined CpG island. Importantly, it acted as a promoter when inserted upstream of luciferase in a fusion construct. Moreover, the activity of the promoter-luciferase construct ostensibly paralleled endogenous Nav1.7 mRNA levels in vitro, with both increased in a quantitatively and qualitatively similar manner by numerous factors (including NGF, phorbol esters, retinoic acid, and Brn-3a transcription factor over-expression).
Collapse
Affiliation(s)
- James K J Diss
- Medical Molecular Biology Unit, Institute of Child Health, University College London, Guilford Street, London WC1N 1EH, UK.
| | | | | | | | | |
Collapse
|
15
|
Balasubramaniyan V, Boddeke E, Bakels R, Küst B, Kooistra S, Veneman A, Copray S. Effects of histone deacetylation inhibition on neuronal differentiation of embryonic mouse neural stem cells. Neuroscience 2006; 143:939-51. [PMID: 17084985 DOI: 10.1016/j.neuroscience.2006.08.082] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2006] [Revised: 08/18/2006] [Accepted: 08/24/2006] [Indexed: 01/07/2023]
Abstract
Neural stem cells (NSCs) are multipotent cells that have the capacity for self-renewal and for differentiation into the major cell types of the nervous system, i.e. neurons, astrocytes and oligodendrocytes. The molecular mechanisms regulating gene transcription resulting in NSC differentiation and cell lineage specification are slowly being unraveled. An important mechanism in transcriptional regulation is modulation of chromatin by histone acetylation and deacetylation, allowing or blocking the access of transcriptional factors to DNA sequences. The precise involvement of histone acetyltransferases and histone deacetylases (HDACs) in the differentiation of NSCs into mature functional neurons is still to be revealed. In this in vitro study we have investigated the effects of the HDAC inhibitor trichostatin A (TSA) on the differentiation pattern of embryonic mouse NSCs during culture in a minimal, serum-free medium, lacking any induction or growth factor. We demonstrated that under these basic conditions TSA treatment increased neuronal differentiation of the NSCs and decreased astrocyte differentiation. Most strikingly, electrophysiological recordings revealed that in our minimal culture system only TSA-treated NSC-derived neurons developed normal electrophysiological membrane properties characteristic for functional, i.e. excitable and firing, neurons. Furthermore, TSA-treated NSC-derived neurons were characterized by an increased elongation and arborization of the dendrites. Our study shows that chromatin structure modulation by HDACs plays an important role in the transcriptional regulation of the neuronal differentiation of embryonic NSCs particularly as far as the development of functional properties are concerned. Manipulation of HDAC activity may be an important tool to generate specific neuronal populations from NSCs for transplantation purposes.
Collapse
Affiliation(s)
- V Balasubramaniyan
- Department of Medical Physiology, University Medical Center Groningen, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
16
|
Ding Q, Vaynman S, Akhavan M, Ying Z, Gomez-Pinilla F. Insulin-like growth factor I interfaces with brain-derived neurotrophic factor-mediated synaptic plasticity to modulate aspects of exercise-induced cognitive function. Neuroscience 2006; 140:823-33. [PMID: 16650607 DOI: 10.1016/j.neuroscience.2006.02.084] [Citation(s) in RCA: 385] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2005] [Revised: 02/16/2006] [Accepted: 02/24/2006] [Indexed: 12/21/2022]
Abstract
The ability of exercise to benefit neuronal and cognitive plasticity is well recognized. This study reveals that the effects of exercise on brain neuronal and cognitive plasticity are in part modulated by a central source of insulin-like growth factor-I. Exercise selectively increased insulin-like growth factor-I expression without affecting insulin-like growth factor-II expression in the rat hippocampus. To determine the role that insulin-like growth factor-I holds in mediating exercise-induced neuronal and cognitive enhancement, a specific antibody against the insulin-like growth factor-I receptor was used to block the action of insulin-like growth factor-I in the hippocampus during a 5-day voluntary exercise period. A two-trial-per-day Morris water maze was performed for five consecutive days, succeeded by a probe trial 2 days later. Blocking hippocampal insulin-like growth factor-I receptors did not significantly attenuate the ability of exercise to enhance learning acquisition, but abolished the effect of exercise on augmenting recall. Blocking the insulin-like growth factor-I receptor significantly reversed the exercise-induced increase in the levels of brain-derived neurotrophic factor mRNA and protein and pro-brain-derived neurotrophic factor protein, suggesting that the effects of insulin-like growth factor-I may be partially accomplished by modulating the precursor to the mature brain-derived neurotrophic factor. A molecular analysis revealed that exercise significantly elevated proteins downstream to brain-derived neurotrophic factor activation important for synaptic function, i.e. synapsin I, and signal transduction cascades associated with memory processes, i.e. phosphorylated calcium/calmodulin protein kinase II and phosphorylated mitogen-activated protein kinase II. Blocking the insulin-like growth factor-I receptor abolished these exercise-induced increases. Our results illustrate a possible mechanism by which insulin-like growth factor-I interfaces with the brain-derived neurotrophic factor system to mediate exercise-induced synaptic and cognitive plasticity.
Collapse
Affiliation(s)
- Q Ding
- Department of Physiological Science, UCLA, 621 Charles E. Young Drive, Los Angeles, CA 90095, USA
| | | | | | | | | |
Collapse
|