1
|
Song QX, Zhang YY, Li YL, Liu F, Liu YJ, Li YK, Li CJ, Zhou C, Shen JF. The crucial role of NR2A mediating the activation of satellite glial cells in the trigeminal ganglion contributes to orofacial inflammatory pain during TMJ inflammation. Neuropharmacology 2024; 261:110173. [PMID: 39357737 DOI: 10.1016/j.neuropharm.2024.110173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/31/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Temporomandibular joint inflammatory diseases are a significant subtype of temporomandibular disorders (TMD) characterized by inflammatory pain in the orofacial area. The N-methyl-D-aspartate receptor (NMDAR), specifically the NR2A subtype, was crucial in neuropathic pain. However, the exact role of NR2A in inflammatory pain in the TMJ and the molecular and cellular mechanisms mediating peripheral sensitization in the trigeminal ganglion (TG) remain unclear. This study utilized male and female mice to induce the TMJOA model by injecting Complete Freund's adjuvant (CFA) into the TMJ and achieve conditional knockout (CKO) of NR2A in the TG using Cre/Loxp technology. The Von-Frey filament test results showed that CFA-induced orofacial pain with reduced mechanical withdrawal threshold (MWT), which was not developed in NR2A CKO mice. Additionally, the up-regulation of interleukin (IL)-1β, IL-6, and nerve growth factor (NGF) in the TG induced by CFA did not occur by NR2A deficiency. In vitro, NMDA activated satellite glial cells (SGCs) with high expression of glial fibrillary acidic protein (GFAP), and both NMDA and LPS led to increased IL-1β, IL-6, and NGF in SGCs. NR2A deficiency reduced these stimulating effects of NMDA and LPS. The regulation of IL-1β involved the p38, Protein Kinase A (PKA), and Protein Kinase C (PKC) pathways, while IL-6 signaling relied on PKA and PKC pathways. NGF regulation was primarily through the p38 pathway. This study highlighted NR2A's crucial role in the TG peripheral sensitization during TMJ inflammation by mediating ILs and NGF, suggesting potential targets for orofacial inflammatory pain management.
Collapse
Affiliation(s)
- Qin-Xuan Song
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease & West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yan-Yan Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease & West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yue-Ling Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease & West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Fei Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease & West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Ya-Jing Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease & West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yi-Ke Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease & West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Chun-Jie Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease & West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Jie-Fei Shen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Disease & West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Morfin C, Sebastian A, Wilson SP, Amiri B, Murugesh DK, Hum NR, Christiansen BA, Loots GG. Mef2c regulates bone mass through Sost-dependent and -independent mechanisms. Bone 2024; 179:116976. [PMID: 38042445 DOI: 10.1016/j.bone.2023.116976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 12/04/2023]
Abstract
Mef2c is a transcription factor that mediates key cellular behaviors that promote endochondral ossification and bone formation. Previously, Mef2c has been shown to regulate Sost transcription via its osteocyte-specific enhancer, ECR5, and conditional deletions of Mef2cfl/fl with either Col1-Cre or Dmp1-Cre produced generalized high bone mass (HBM) consistent with Van Buchem Disease phenotypes. However, Sost-/-; Mef2cfl/fl; Dmp1-Cre mice produced a significantly higher bone mass phenotype that Sost-/- alone suggesting that Mef2c modulates bone mass through additional mechanisms, independent of Sost. To identify new Mef2c transcriptional targets important in bone metabolism, we profiled gene expression by single-cell RNA sequencing in subpopulations of cells isolated from Mef2cfl/fl; Dmp1-Cre and Mef2cfl/fl; Bglap-Cre femurs, both strains exhibiting similar high bone mass phenotypes. However, we found Mef2cfl/fl; Bglap-Cre to also display a growth plate defect characterized by an expansion of several osteoprogenitor subpopulations. Differential gene expression analysis identified a total of 96 up- and 2434 down- regulated genes in Mef2cfl/fl; Bglap-Cre and 176 up- and 1041 down- regulated genes in Mef2cfl/fl; Dmp1-Cre bone cell subpopulations compared to wildtype mice. Mef2c deletion affected the transcriptomes across several cell types including mesenchymal progenitors (MP), osteoprogenitors (OSP), osteoblast (OB), and osteocyte (OCY) subpopulations. Several energy metabolism genes such as Uqcrb, Ndufv2, Ndufs3, Ndufa13, Ndufb9, Ndufb5, Cox6a1, Cox5a, Atp5o, Atp5g2, Atp5b, Atp5 were significantly down regulated in Mef2c-deficient OBs and OCYs, in both strains. Binding motif analysis of promoter regions of differentially expressed genes identified Mef2c binding in Bone Sialoprotein (BSP/Ibsp), a gene known to cause increased trabecular BV/TV in the femurs of Ibsp-/- mice. Immunohistochemical analysis confirmed the absence of Ibsp protein in OBs and OCYs. These findings suggests that the HBM in Sost-/-; Mef2cfl/fl; Dmp1-Cre is caused by a multitude of transcriptional changes in genes that regulate bone formation, two of which are Sost and Ibsp.
Collapse
Affiliation(s)
- Cesar Morfin
- School of Natural Sciences, University of California, Merced, CA, United States; Physical and Life Sciences Directorate, Lawrence Livermore, National Laboratories, Livermore, CA, United States; Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA, United States
| | - Aimy Sebastian
- Physical and Life Sciences Directorate, Lawrence Livermore, National Laboratories, Livermore, CA, United States
| | - Stephen P Wilson
- Physical and Life Sciences Directorate, Lawrence Livermore, National Laboratories, Livermore, CA, United States
| | - Beheshta Amiri
- Physical and Life Sciences Directorate, Lawrence Livermore, National Laboratories, Livermore, CA, United States
| | - Deepa K Murugesh
- Physical and Life Sciences Directorate, Lawrence Livermore, National Laboratories, Livermore, CA, United States
| | - Nicholas R Hum
- Physical and Life Sciences Directorate, Lawrence Livermore, National Laboratories, Livermore, CA, United States
| | - Blaine A Christiansen
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA, United States
| | - Gabriela G Loots
- School of Natural Sciences, University of California, Merced, CA, United States; Physical and Life Sciences Directorate, Lawrence Livermore, National Laboratories, Livermore, CA, United States; Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA, United States.
| |
Collapse
|
3
|
Català-Solsona J, Lituma PJ, Lutzu S, Siedlecki-Wullich D, Fábregas-Ordoñez C, Miñano-Molina AJ, Saura CA, Castillo PE, Rodriguez-Álvarez J. Activity-Dependent Nr4a2 Induction Modulates Synaptic Expression of AMPA Receptors and Plasticity via a Ca 2+/CRTC1/CREB Pathway. J Neurosci 2023; 43:3028-3041. [PMID: 36931707 PMCID: PMC10146469 DOI: 10.1523/jneurosci.1341-22.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/19/2023] Open
Abstract
Transcription factors have a pivotal role in synaptic plasticity and the associated modification of neuronal networks required for memory formation and consolidation. The nuclear receptors subfamily 4 group A (Nr4a) have emerged as possible modulators of hippocampal synaptic plasticity and cognitive functions. However, the molecular and cellular mechanisms underlying Nr4a2-mediated hippocampal synaptic plasticity are not completely known. Here, we report that neuronal activity enhances Nr4a2 expression and function in cultured mouse hippocampal neurons (both sexes) by an ionotropic glutamate receptor/Ca2+/cAMP response element-binding protein/CREB-regulated transcription factor 1 (iGluR/Ca2+/CREB/CRTC1) pathway. Nr4a2 activation mediates BDNF production and increases expression of iGluRs, thereby affecting LTD at CA3-CA1 synapses in acute mouse hippocampal slices (both sexes). Together, our results indicate that the iGluR/Ca2+/CREB/CRTC1 pathway mediates activity-dependent expression of Nr4a2, which is involved in glutamatergic synaptic plasticity by increasing BDNF and synaptic GluA1-AMPARs. Therefore, Nr4a2 activation could be a therapeutic approach for brain disorders associated with dysregulated synaptic plasticity.SIGNIFICANCE STATEMENT A major factor that regulates fast excitatory synaptic transmission and plasticity is the modulation of synaptic AMPARs. However, despite decades of research, the underlying mechanisms of this modulation remain poorly understood. Our study identified a molecular pathway that links neuronal activity with AMPAR modulation and hippocampal synaptic plasticity through the activation of Nr4a2, a member of the nuclear receptor subfamily 4. Since several compounds have been described to activate Nr4a2, our study not only provides mechanistic insights into the molecular pathways related to hippocampal synaptic plasticity and learning, but also identifies Nr4a2 as a potential therapeutic target for pathologic conditions associated with dysregulation of glutamatergic synaptic function.
Collapse
Affiliation(s)
- Judit Català-Solsona
- Institut de Neurociències and Departamento Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, 08193, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, 28031, Spain
| | - Pablo J Lituma
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, New York 10461
| | - Stefano Lutzu
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, New York 10461
| | - Dolores Siedlecki-Wullich
- Institut de Neurociències and Departamento Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, 08193, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, 28031, Spain
| | - Cristina Fábregas-Ordoñez
- Institut de Neurociències and Departamento Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, 08193, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, 28031, Spain
| | - Alfredo J Miñano-Molina
- Institut de Neurociències and Departamento Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, 08193, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, 28031, Spain
| | - Carlos A Saura
- Institut de Neurociències and Departamento Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, 08193, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, 28031, Spain
| | - Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, New York 10461
- Department of Psychiatry & Behavioral Sciences, Albert Einstein College of Medicine, New York, New York 10461
| | - José Rodriguez-Álvarez
- Institut de Neurociències and Departamento Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, 08193, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, 28031, Spain
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, New York 10461
| |
Collapse
|
4
|
Hammond-Weinberger DR, Wang Y, Glavis-Bloom A, Spitzer NC. Mechanism for neurotransmitter-receptor matching. Proc Natl Acad Sci U S A 2020; 117:4368-4374. [PMID: 32041885 PMCID: PMC7049162 DOI: 10.1073/pnas.1916600117] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Synaptic communication requires the expression of functional postsynaptic receptors that match the presynaptically released neurotransmitter. The ability of neurons to switch the transmitter they release is increasingly well documented, and these switches require changes in the postsynaptic receptor population. Although the activity-dependent molecular mechanism of neurotransmitter switching is increasingly well understood, the basis of specification of postsynaptic neurotransmitter receptors matching the newly expressed transmitter is unknown. Using a functional assay, we show that sustained application of glutamate to embryonic vertebrate skeletal muscle cells cultured before innervation is necessary and sufficient to up-regulate ionotropic glutamate receptors from a pool of different receptors expressed at low levels. Up-regulation of these ionotropic receptors is independent of signaling by metabotropic glutamate receptors. Both imaging of glutamate-induced calcium elevations and Western blots reveal ionotropic glutamate receptor expression prior to immunocytochemical detection. Sustained application of glutamate to skeletal myotomes in vivo is necessary and sufficient for up-regulation of membrane expression of the GluN1 NMDA receptor subunit. Pharmacological antagonists and morpholinos implicate p38 and Jun kinases and MEF2C in the signal cascade leading to ionotropic glutamate receptor expression. The results suggest a mechanism by which neuronal release of transmitter up-regulates postsynaptic expression of appropriate transmitter receptors following neurotransmitter switching and may contribute to the proper expression of receptors at the time of initial innervation.
Collapse
Affiliation(s)
- Dena R Hammond-Weinberger
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093-0357;
| | - Yunxin Wang
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093-0357
| | - Alex Glavis-Bloom
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093-0357
| | - Nicholas C Spitzer
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093-0357;
- Center for Neural Circuits and Behavior, Kavli Institute for Brain and Mind, University of California San Diego, La Jolla, CA 92161
| |
Collapse
|
5
|
Tejeda GS, Esteban‐Ortega GM, San Antonio E, Vidaurre ÓG, Díaz‐Guerra M. Prevention of excitotoxicity-induced processing of BDNF receptor TrkB-FL leads to stroke neuroprotection. EMBO Mol Med 2019; 11:e9950. [PMID: 31273936 PMCID: PMC6609917 DOI: 10.15252/emmm.201809950] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 05/02/2019] [Accepted: 05/03/2019] [Indexed: 12/14/2022] Open
Abstract
Neuroprotective strategies aimed to pharmacologically treat stroke, a prominent cause of death, disability, and dementia, have remained elusive. A promising approach is restriction of excitotoxic neuronal death in the infarct penumbra through enhancement of survival pathways initiated by brain-derived neurotrophic factor (BDNF). However, boosting of neurotrophic signaling after ischemia is challenged by downregulation of BDNF high-affinity receptor, full-length tropomyosin-related kinase B (TrkB-FL), due to calpain-degradation, and, secondarily, regulated intramembrane proteolysis. Here, we have designed a blood-brain barrier (BBB) permeable peptide containing TrkB-FL sequences (TFL457 ) which prevents receptor disappearance from the neuronal surface, early induced after excitotoxicity. In this way, TFL457 interferes TrkB-FL cleavage by both proteolytic systems and increases neuronal viability via a PLCγ-dependent mechanism. By preserving downstream CREB and MEF2 promoter activities, TFL457 initiates a feedback mechanism favoring increased levels in excitotoxic neurons of critical prosurvival mRNAs and proteins. This neuroprotective peptide could be highly relevant for stroke therapy since, in a mouse ischemia model, it counteracts TrkB-FL downregulation in the infarcted brain, efficiently decreases infarct size, and improves neurological outcome.
Collapse
Affiliation(s)
- Gonzalo S Tejeda
- Instituto de Investigaciones Biomédicas “Alberto Sols”Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de Madrid (CSIC‐UAM)MadridSpain
- Present address:
Gardiner LaboratoryInstitute of Cardiovascular and Medical SciencesCollege of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Gema M Esteban‐Ortega
- Instituto de Investigaciones Biomédicas “Alberto Sols”Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de Madrid (CSIC‐UAM)MadridSpain
| | - Esther San Antonio
- Instituto de Investigaciones Biomédicas “Alberto Sols”Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de Madrid (CSIC‐UAM)MadridSpain
| | - Óscar G Vidaurre
- Instituto de Investigaciones Biomédicas “Alberto Sols”Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de Madrid (CSIC‐UAM)MadridSpain
| | - Margarita Díaz‐Guerra
- Instituto de Investigaciones Biomédicas “Alberto Sols”Consejo Superior de Investigaciones Científicas‐Universidad Autónoma de Madrid (CSIC‐UAM)MadridSpain
| |
Collapse
|
6
|
Abstract
Gene expression is controlled by sequence-specific transcription factors (TFs), which bind to regulatory sequences in DNA. The degree to which the arrangement of motif sites within regulatory elements determines their function remains unclear. Here, we show that the positional distribution of TF motif sites within nucleosome-depleted regions of DNA fall into six distinct classes. These patterns are highly consistent across cell types and bring together factors that have similar functional and binding properties. Furthermore, the position of motif sites appears to be related to their known functions. Our results suggest that TFs play distinct roles in forming a functional enhancer, facilitated by their position within a regulatory sequence. Gene expression is controlled by sequence-specific transcription factors (TFs), which bind to regulatory sequences in DNA. TF binding occurs in nucleosome-depleted regions of DNA (NDRs), which generally encompass regions with lengths similar to those protected by nucleosomes. However, less is known about where within these regions specific TFs tend to be found. Here, we characterize the positional bias of inferred binding sites for 103 TFs within ∼500,000 NDRs across 47 cell types. We find that distinct classes of TFs display different binding preferences: Some tend to have binding sites toward the edges, some toward the center, and some at other positions within the NDR. These patterns are highly consistent across cell types, suggesting that they may reflect TF-specific intrinsic structural or functional characteristics. In particular, TF classes with binding sites at NDR edges are enriched for those known to interact with histones and chromatin remodelers, whereas TFs with central enrichment interact with other TFs and cofactors such as p300. Our results suggest distinct regiospecific binding patterns and functions of TF classes within enhancers.
Collapse
|
7
|
Memantine, an NMDA Receptor Antagonist, Prevents Thyroxin-induced Hypertension, but Not Cardiac Remodeling. J Cardiovasc Pharmacol 2018; 70:305-313. [PMID: 29112047 DOI: 10.1097/fjc.0000000000000521] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Stimulation of glutamatergic tone has been causally linked to myocardial pathogenesis and amplified systemic blood pressure (BP). Memantine, a noncompetitive N-methyl-D-aspartate glutamatergic receptor (NMDA-R) antagonist, has been proposed to be an active cardioprotective drug. However, the efficacy of memantine and subsequently the possible involvement of the NMDA-R in the thyroxin (T4)-induced cardiovascular complications have never been investigated. We examined the effect of memantine (30 mg·kg·d) on the T4 (500 μg·kg·d)-provoked increase in mouse BP, cardiac hypertrophy indicated by enlarged overall myocardial mass, and reformed reactions of the contractile myocardium both in vivo and ex vivo after 2 weeks of treatment. Memantine alone did not result in any cardiovascular pathology in mice. Instead, memantine significantly prevented the T4-triggered systemic hypertension. But, it did not reverse cardiac hypertrophy, coupled in vivo left ventricular dysfunction (LV) or ex vivo right ventricular (RV) papillary muscle contractile alterations of the T4-treated mice. Our results openly direct the cardiovascular safety and tolerability of memantine therapy. Yet, extra research is necessary to endorse these prospective advantageous outcomes. Also, we believe that this is the first study to inspect the possible role of NMDA-R in the T4-stimulated cardiovascular disorders and concluded that NMDA-R could play a key role in the T4-induced hypertension.
Collapse
|
8
|
Tu S, Akhtar MW, Escorihuela RM, Amador-Arjona A, Swarup V, Parker J, Zaremba JD, Holland T, Bansal N, Holohan DR, Lopez K, Ryan SD, Chan SF, Yan L, Zhang X, Huang X, Sultan A, McKercher SR, Ambasudhan R, Xu H, Wang Y, Geschwind DH, Roberts AJ, Terskikh AV, Rissman RA, Masliah E, Lipton SA, Nakanishi N. NitroSynapsin therapy for a mouse MEF2C haploinsufficiency model of human autism. Nat Commun 2017; 8:1488. [PMID: 29133852 PMCID: PMC5684358 DOI: 10.1038/s41467-017-01563-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 09/27/2017] [Indexed: 01/07/2023] Open
Abstract
Transcription factor MEF2C regulates multiple genes linked to autism spectrum disorder (ASD), and human MEF2C haploinsufficiency results in ASD, intellectual disability, and epilepsy. However, molecular mechanisms underlying MEF2C haploinsufficiency syndrome remain poorly understood. Here we report that Mef2c +/-(Mef2c-het) mice exhibit behavioral deficits resembling those of human patients. Gene expression analyses on brains from these mice show changes in genes associated with neurogenesis, synapse formation, and neuronal cell death. Accordingly, Mef2c-het mice exhibit decreased neurogenesis, enhanced neuronal apoptosis, and an increased ratio of excitatory to inhibitory (E/I) neurotransmission. Importantly, neurobehavioral deficits, E/I imbalance, and histological damage are all ameliorated by treatment with NitroSynapsin, a new dual-action compound related to the FDA-approved drug memantine, representing an uncompetitive/fast off-rate antagonist of NMDA-type glutamate receptors. These results suggest that MEF2C haploinsufficiency leads to abnormal brain development, E/I imbalance, and neurobehavioral dysfunction, which may be mitigated by pharmacological intervention.
Collapse
Affiliation(s)
- Shichun Tu
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, 92121, USA.
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA.
| | - Mohd Waseem Akhtar
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, 92121, USA
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Rosa Maria Escorihuela
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
- Departament de PsiquiatriaiMedicina Legal, Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Alejandro Amador-Arjona
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Vivek Swarup
- Center for Autism Research and Treatment (CART), University of California, Los Angeles, CA, 90095, USA
| | - James Parker
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, 92121, USA
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Jeffrey D Zaremba
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Timothy Holland
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Neha Bansal
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Daniel R Holohan
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Kevin Lopez
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, 92121, USA
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Scott D Ryan
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Shing Fai Chan
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Li Yan
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Xiaofei Zhang
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Xiayu Huang
- Bioinformatics Core Facility, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Abdullah Sultan
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, 92121, USA
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Scott R McKercher
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, 92121, USA
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
- Department of Molecular Medicine and Neuroscience, Neuroscience Translational Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Rajesh Ambasudhan
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, 92121, USA
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Huaxi Xu
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Yuqiang Wang
- Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Daniel H Geschwind
- Center for Autism Research and Treatment (CART), University of California, Los Angeles, CA, 90095, USA
| | - Amanda J Roberts
- Department of Neuroscience and Mouse Behavioral Assessment Core, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Alexey V Terskikh
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA, 92161, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
- National Institute on Aging, NIH, Bethesda, MD, 20892, USA
| | - Stuart A Lipton
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, 92121, USA.
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA.
- Department of Molecular Medicine and Neuroscience, Neuroscience Translational Center, The Scripps Research Institute, La Jolla, CA, 92037, USA.
- Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA.
| | - Nobuki Nakanishi
- Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA, 92121, USA.
- Neuroscience and Aging Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
9
|
Dong C, Yang XZ, Zhang CY, Liu YY, Zhou RB, Cheng QD, Yan EK, Yin DC. Myocyte enhancer factor 2C and its directly-interacting proteins: A review. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017; 126:22-30. [DOI: 10.1016/j.pbiomolbio.2017.02.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 11/24/2016] [Accepted: 02/01/2017] [Indexed: 11/27/2022]
|
10
|
Latchney SE, Jiang Y, Petrik DP, Eisch AJ, Hsieh J. Inducible knockout of Mef2a, -c, and -d from nestin-expressing stem/progenitor cells and their progeny unexpectedly uncouples neurogenesis and dendritogenesis in vivo. FASEB J 2015; 29:5059-71. [PMID: 26286136 DOI: 10.1096/fj.15-275651] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 08/13/2015] [Indexed: 12/11/2022]
Abstract
Myocyte enhancer factor (Mef)-2 transcription factors are implicated in activity-dependent neuronal processes during development, but the role of MEF2 in neural stem/progenitor cells (NSPCs) in the adult brain is unknown. We used a transgenic mouse in which Mef2a, -c, and -d were inducibly deleted in adult nestin-expressing NSPCs and their progeny. Recombined cells in the hippocampal granule cell layer were visualized and quantified by yellow fluorescent protein (YFP) expression. In control mice, postmitotic neurons expressed Mef2a, -c, and -d, whereas type 1 stem cells and proliferating progenitors did not. Based on this expression, we hypothesized that Mef2a, -c, and -d deletion in adult nestin-expressing NSPCs and their progeny would result in fewer mature neurons. Control mice revealed an increase in YFP(+) neurons and dendrite formation over time. Contrary to our hypothesis, inducible Mef2 KO mice also displayed an increase in YFP(+) neurons over time-but with significantly stunted dendrites-suggesting an uncoupling of neuron survival and dendritogenesis. We also found non-cell-autonomous effects after Mef2a, -c, and -d deletion. These in vivo findings indicate a surprising functional role for Mef2a, -c, and -d in cell- and non-cell-autonomous control of adult hippocampal neurogenesis that is distinct from its role during development.
Collapse
Affiliation(s)
- Sarah E Latchney
- *Department of Psychiatry and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yindi Jiang
- *Department of Psychiatry and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - David P Petrik
- *Department of Psychiatry and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Amelia J Eisch
- *Department of Psychiatry and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jenny Hsieh
- *Department of Psychiatry and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
11
|
Higa KK, Ji B, Buell MR, Risbrough VB, Powell SB, Young JW, Geyer MA, Zhou X. Restoration of Sp4 in Forebrain GABAergic Neurons Rescues Hypersensitivity to Ketamine in Sp4 Hypomorphic Mice. Int J Neuropsychopharmacol 2015; 18:pyv063. [PMID: 26037489 PMCID: PMC4756721 DOI: 10.1093/ijnp/pyv063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/29/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Ketamine produces schizophrenia-like behavioral phenotypes in healthy people. Prolonged ketamine effects and exacerbation of symptoms after the administration of ketamine have been observed in patients with schizophrenia. More recently, ketamine has been used as a potent antidepressant to treat patients with major depression. The genes and neurons that regulate behavioral responses to ketamine, however, remain poorly understood. Sp4 is a transcription factor for which gene expression is restricted to neuronal cells in the brain. Our previous studies demonstrated that Sp4 hypomorphic mice display several behavioral phenotypes relevant to psychiatric disorders, consistent with human SP4 gene associations with schizophrenia, bipolar disorder, and major depression. Among those behavioral phenotypes, hypersensitivity to ketamine-induced hyperlocomotion has been observed in Sp4 hypomorphic mice. METHODS In the present study, we used the Cre-LoxP system to restore Sp4 gene expression, specifically in either forebrain excitatory or GABAergic inhibitory neurons in Sp4 hypomorphic mice. Mouse behavioral phenotypes related to psychiatric disorders were examined in these distinct rescue mice. RESULTS Restoration of Sp4 in forebrain excitatory neurons did not rescue deficient sensorimotor gating nor ketamine-induced hyperlocomotion. Restoration of Sp4 in forebrain GABAergic neurons, however, rescued ketamine-induced hyperlocomotion, but did not rescue deficient sensorimotor gating. CONCLUSIONS Our studies suggest that the Sp4 gene in forebrain GABAergic neurons regulates ketamine-induced hyperlocomotion.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xianjin Zhou
- Department of Psychiatry, University of California San Diego, La Jolla, CA (Ms Higa, Drs Ji, Risbrough, Powell, Young, Geyer, and Zhou, and Ms Buell); Research Service, VA San Diego Healthcare System, La Jolla, CA (Drs Risbrough, Powell, Young, Geyer, and Zhou, and Ms Buell); Neurosciences Graduate Program, University of California San Diego, La Jolla, CA (Ms Higa).
| |
Collapse
|
12
|
Okamoto SI, Nakamura T, Cieplak P, Chan SF, Kalashnikova E, Liao L, Saleem S, Han X, Clemente A, Nutter A, Sances S, Brechtel C, Haus D, Haun F, Sanz-Blasco S, Huang X, Li H, Zaremba JD, Cui J, Gu Z, Nikzad R, Harrop A, McKercher SR, Godzik A, Yates JR, Lipton SA. S-nitrosylation-mediated redox transcriptional switch modulates neurogenesis and neuronal cell death. Cell Rep 2014; 8:217-28. [PMID: 25001280 DOI: 10.1016/j.celrep.2014.06.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 04/17/2014] [Accepted: 06/03/2014] [Indexed: 11/24/2022] Open
Abstract
Redox-mediated posttranslational modifications represent a molecular switch that controls major mechanisms of cell function. Nitric oxide (NO) can mediate redox reactions via S-nitrosylation, representing transfer of an NO group to a critical protein thiol. NO is known to modulate neurogenesis and neuronal survival in various brain regions in disparate neurodegenerative conditions. However, a unifying molecular mechanism linking these phenomena remains unknown. Here, we report that S-nitrosylation of myocyte enhancer factor 2 (MEF2) transcription factors acts as a redox switch to inhibit both neurogenesis and neuronal survival. Structure-based analysis reveals that MEF2 dimerization creates a pocket, facilitating S-nitrosylation at an evolutionally conserved cysteine residue in the DNA binding domain. S-Nitrosylation disrupts MEF2-DNA binding and transcriptional activity, leading to impaired neurogenesis and survival in vitro and in vivo. Our data define a molecular switch whereby redox-mediated posttranslational modification controls both neurogenesis and neurodegeneration via a single transcriptional signaling cascade.
Collapse
Affiliation(s)
- Shu-Ichi Okamoto
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Tomohiro Nakamura
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Piotr Cieplak
- Bioinformatics and Systems Biology Program, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Shing Fai Chan
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Evgenia Kalashnikova
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Lujian Liao
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Sofiyan Saleem
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Xuemei Han
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Arjay Clemente
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Anthony Nutter
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Sam Sances
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Christopher Brechtel
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Daniel Haus
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Florian Haun
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Sara Sanz-Blasco
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Xiayu Huang
- Bioinformatics and Systems Biology Program, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Hao Li
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jeffrey D Zaremba
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jiankun Cui
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Zezong Gu
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Rana Nikzad
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Anne Harrop
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Scott R McKercher
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Adam Godzik
- Bioinformatics and Systems Biology Program, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - John R Yates
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Stuart A Lipton
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
13
|
Abstract
High levels of homocysteine (Hcy), known as hyperhomocysteinmia (HHcy), are correlated with an increase in extracellular matrix remodelling (ECM) via the matrix metalloproteinases (MMPs) and plasminogen/plasmin system. This results in an increase deposition of collagen that leads to endothelial-myocyte (EM) and myocyte-myocyte (MM) uncoupling; the physiological consequences are a plethora of cardiovascular pathologies. Homocysteine-induced increase in intracellular and mitochondrial Ca(2+) plays an important role in increasing reactive oxygen species (ROS) within mitochondria and instigating mitophagy within the cell. This occurs via several Hcy-mitigated processes: agonizing N-methyl-d-aspartate receptor-1 (NMDA-R1), decreasing expression of peroxisome proliferator activator receptor (PPAR) [thereby increasing oxidation], impairing Ca(2+) handling via Na(+)/Ca(2+) exchanger (NCX1) and Sarco endoplasmic reticulum Ca(2+) ATPase (SERCA-2a). The end result is an increase in ROS that directly or indirectly lead to MMP activation within mitochondria or the cytoplasm. Hcy induces a mitochondrial permeability transition that allows MMPs to be released from mitochondria thereby metabolizing matrix and impairing cardiac function. Further work remains to be elucidated concerning the specific mitochondrial mitophagic mechanisms under which matrix metabolism and remodelling occurs. Moreover, the therapeutic implications of NMDA and PPAR ligands are some promise to patient.
Collapse
Affiliation(s)
- Thomas P Vacek
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | | | | |
Collapse
|
14
|
Bian Y, Zhou W, Zhao Y, Li X, Geng W, Hao R, Yang Q, Huang W. High-dose siRNAs upregulate mouse Eri-1 at both transcription and posttranscription levels. PLoS One 2011; 6:e26466. [PMID: 22039495 PMCID: PMC3198429 DOI: 10.1371/journal.pone.0026466] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 09/27/2011] [Indexed: 01/05/2023] Open
Abstract
The eri-1 gene encodes a 3′ exonuclease that can negatively regulate RNA interference via siRNase activity. High-dose siRNAs (hd-siRNAs) can enhance Eri-1 expression, which in return degrade siRNAs and greatly reduces RNAi efficiency. Here we report that hd-siRNAs induce mouse Eri-1 (meri-1) expression through the recruitment of Sp1, Ets-1, and STAT3 to the meri-1 promoter and the formation of an Sp1-Ets-1-STAT3 complex. In addition, hd-siRNAs also abolish the 3′ untranslated region (UTR) mediated posttranscriptional repression of meri-1. Our findings demonstrate the molecular mechanism underlying the upregulation of meri-1 by hd-siRNA.
Collapse
Affiliation(s)
- Yingnan Bian
- Department of Biochemistry, School of Life Science, Fudan University, Shanghai, China
| | - Wei Zhou
- Department of Biochemistry, School of Life Science, Fudan University, Shanghai, China
| | - Yingchun Zhao
- Department of Biochemistry, School of Life Science, Fudan University, Shanghai, China
| | - Xiaoping Li
- Department of Biochemistry, School of Life Science, Fudan University, Shanghai, China
| | - Wei Geng
- Department of Biochemistry, School of Life Science, Fudan University, Shanghai, China
| | - Ruixin Hao
- Department of Biochemistry, School of Life Science, Fudan University, Shanghai, China
| | - Qing Yang
- Department of Biochemistry, School of Life Science, Fudan University, Shanghai, China
| | - Weida Huang
- Department of Biochemistry, School of Life Science, Fudan University, Shanghai, China
- Laboratory for Synthetic Biology, Centers for Nano-Medicine, Shanghai Advanced Research Institute, Chinese Academy Sciences, Pudong, Shanghai, China
- * E-mail:
| |
Collapse
|
15
|
Azim S, Banday AR, Tabish M. Alternatively spliced three novel transcripts of gria1 in the cerebellum and cortex of mouse brain. Neurochem Res 2011; 37:193-201. [PMID: 21927928 DOI: 10.1007/s11064-011-0599-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 08/30/2011] [Accepted: 09/08/2011] [Indexed: 12/01/2022]
Abstract
Glutamate receptor type 1 (GluR1) subunit of α-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptors plays an important role in the expression of long-term potentiation and memory formation. GluR1 is encoded by gria1 gene containing 16 exons and 15 introns in mouse. Previous studies have reported two alternatively spliced variants of this subunit. These flip and flop variants differ enormously in their properties as well as expression. In our studies, we report the presence of three new transcripts of this gene present in the cerebellum and cortex of mouse brain produced by alternative splicing at 5' end. Four new exons are reported; N1 is located in 5' untranslated region, N2 is located in the 1st intronic region while N3 and N4 are located in the 2nd intronic region. The properties of these new exons encoding N-terminal variants are highly diverse. N1, N3 and N4 are coding while N2 is a non-coding exon and results in a truncated transcript. The existence of N2 exon containing transcript is further supported by the presence of an Expressed Sequence Tag from the database. The translated amino acid sequences of these transcripts differ in the presence of signal peptide as well as in their phosphorylation and acetylation pattern. The differences in their properties might be involved in receptor modulation.
Collapse
Affiliation(s)
- Shafquat Azim
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, UP 202002, India
| | | | | |
Collapse
|
16
|
|
17
|
Pinacho R, Villalmanzo N, Lalonde J, Haro JM, Meana JJ, Gill G, Ramos B. The transcription factor SP4 is reduced in postmortem cerebellum of bipolar disorder subjects: control by depolarization and lithium. Bipolar Disord 2011; 13:474-85. [PMID: 22017217 PMCID: PMC3202296 DOI: 10.1111/j.1399-5618.2011.00941.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Regulation of gene expression is important for the development and function of the nervous system. However, the transcriptional programs altered in psychiatric diseases are not completely characterized. Human gene association studies and analysis of mutant mice suggest that the transcription factor specificity protein 4 (SP4) may be implicated in the pathophysiology of psychiatric diseases. We hypothesized that SP4 levels may be altered in the brain of bipolar disorder (BD) subjects and regulated by neuronal activity and drug treatment. METHODS We analyzed messenger RNA (mRNA) and protein levels of SP4 and SP1 in the postmortem prefrontal cortex and cerebellum of BD subjects (n = 10) and controls (n = 10). We also examined regulation of SP4 mRNA and protein levels by neuronal activity and lithium in rat cerebellar granule neurons. RESULTS We report a reduction of SP4 and SP1 proteins, but not mRNA levels, in the cerebellum of BD subjects. SP4 protein and mRNA levels were also reduced in the prefrontal cortex. Moreover, we found in rat cerebellar granule neurons that under non-depolarizing conditions SP4, but not SP1, was polyubiquitinated and degraded by the proteasome while lithium stabilized SP4 protein. CONCLUSIONS Our study provides the first evidence of altered SP4 protein in the cerebellum and prefrontal cortex in BD subjects supporting a possible role of transcription factor SP4 in the pathogenesis of the disease. In addition, our finding that SP4 stability is regulated by depolarization and lithium provides a pathway through which neuronal activity and lithium could control gene expression suggesting that normalization of SP4 levels could contribute to treatment of affective disorders.
Collapse
Affiliation(s)
- Raquel Pinacho
- Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain
| | - Nuria Villalmanzo
- Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain
| | - Jasmin Lalonde
- Department of Anatomy and Cellular Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Josep Maria Haro
- Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain
| | - J Javier Meana
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Bizkaia,CIBERSAM, Bizkaia, Spain
| | - Grace Gill
- Department of Anatomy and Cellular Biology, Tufts University School of Medicine, Boston, MA, USA,Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Belén Ramos
- Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain,Department of Pathology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
18
|
Traynelis SF, Wollmuth LP, McBain CJ, Menniti FS, Vance KM, Ogden KK, Hansen KB, Yuan H, Myers SJ, Dingledine R. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol Rev 2010; 62:405-96. [PMID: 20716669 PMCID: PMC2964903 DOI: 10.1124/pr.109.002451] [Citation(s) in RCA: 2622] [Impact Index Per Article: 187.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The mammalian ionotropic glutamate receptor family encodes 18 gene products that coassemble to form ligand-gated ion channels containing an agonist recognition site, a transmembrane ion permeation pathway, and gating elements that couple agonist-induced conformational changes to the opening or closing of the permeation pore. Glutamate receptors mediate fast excitatory synaptic transmission in the central nervous system and are localized on neuronal and non-neuronal cells. These receptors regulate a broad spectrum of processes in the brain, spinal cord, retina, and peripheral nervous system. Glutamate receptors are postulated to play important roles in numerous neurological diseases and have attracted intense scrutiny. The description of glutamate receptor structure, including its transmembrane elements, reveals a complex assembly of multiple semiautonomous extracellular domains linked to a pore-forming element with striking resemblance to an inverted potassium channel. In this review we discuss International Union of Basic and Clinical Pharmacology glutamate receptor nomenclature, structure, assembly, accessory subunits, interacting proteins, gene expression and translation, post-translational modifications, agonist and antagonist pharmacology, allosteric modulation, mechanisms of gating and permeation, roles in normal physiological function, as well as the potential therapeutic use of pharmacological agents acting at glutamate receptors.
Collapse
Affiliation(s)
- Stephen F Traynelis
- Department of Pharmacology, Emory University School of Medicine, Rollins Research Center, 1510 Clifton Road, Atlanta, GA 30322-3090, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Hepatitis B virus (HBV) is tightly controlled by a number of noncytotoxic mechanisms. This control occurs within the host hepatocyte at different steps of the HBV replication cycle. HBV persists by establishing a nuclear minichromosome, HBV cccDNA, serving as a transcription template for the viral pregenome and viral mRNAs. Nucleoside/nucleotide analogues widely used for antiviral therapy as well as most antiviral cytokines act at steps after transcription of HBV RNAs and thus can control virus replication but do not directly affect its gene expression. Control of HBV at the level of transcription in contrast is able to restrict both, HBV replication and gene expression. In the review, we focus on how HBV is controlled at the level of transcription. We discuss how the composition of transcription factors determines HBV gene expression and replication and how this may be influenced by antivirally active substances, e.g. the cytokine IL-6 or helioxanthin analogues, or by the differentiation state of the hepatocyte.
Collapse
Affiliation(s)
- M Quasdorff
- Department of Gastroenterology and Hepatology, University Hospital Cologne, Germany
| | | |
Collapse
|
20
|
Shen YC, Liao DL, Lu CL, Chen JY, Liou YJ, Chen TT, Chen CH. Resequencing of the vesicular glutamate transporter 2 gene (VGLUT2) reveals some rare genetic variants that may increase the genetic burden in schizophrenia. Schizophr Res 2010; 121:179-86. [PMID: 20541370 DOI: 10.1016/j.schres.2010.05.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 04/01/2010] [Accepted: 05/16/2010] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Vesicular glutamate transporters (VGLUT1-3) package glutamate into vesicles in the presynaptic terminal and regulate the release of glutamate. In mesencephalic dopamine neuron culture, the majority of isolated dopamine neurons express VGLUT2, but not VGLUT1 or 3, have been demonstrated. As related to the dysregulated glutamatergic hypothesis of schizophrenia, the gene encoding VGLUT2 is the most plausible candidate involved in the pathogenesis of this illness. METHODS We searched for genetic variants in the promoter region and 12 exons (including UTR ends) of the VGLUT2 gene using direct sequencing in a sample of Han Chinese schizophrenic patients (n=375) and non-psychotic controls (n=366) from Taiwan, and conducted a case-control association study. RESULTS We identified 8 common SNPs in the VGLUT2 gene. SNP and haplotype-based analyses showed no association with schizophrenia. Besides, we identified 9 rare variants in 13 out of 375 patients, including 3 variants located at the promoter region, 2 synonymous variants located at protein coding regions, and 4 variants located at UTR ends. No rare variants were found in the control subjects. Collectively, these rare variants were significantly overrepresented in the patient group (3.5% versus 0, p value of Fisher's exact test=2.3x10(-5)), suggesting they may contribute to the pathogenesis of schizophrenia. CONCLUSION Although the functional significance of these rare variants remains to be characterized, our study may lend support to the multiple rare mutations hypothesis of schizophrenia, and may provide genetic clues to indicate the involvement of the glutamate transmission pathway in the pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Yu-Chih Shen
- Department of Psychiatry, Tzu Chi General Hospital and University, Hualien, Taiwan
| | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Hepatitis B virus (HBV) is tightly controlled by a number of noncytotoxic mechanisms. This control occurs within the host hepatocyte at different steps of the HBV replication cycle. HBV persists by establishing a nuclear minichromosome, HBV cccDNA, serving as a transcription template for the viral pregenome and viral mRNAs. Nucleoside/nucleotide analogues widely used for antiviral therapy as well as most antiviral cytokines act at steps after transcription of HBV RNAs and thus can control virus replication but do not directly affect its gene expression. Control of HBV at the level of transcription in contrast is able to restrict both, HBV replication and gene expression. In the review, we focus on how HBV is controlled at the level of transcription. We discuss how the composition of transcription factors determines HBV gene expression and replication and how this may be influenced by antivirally active substances, e.g. the cytokine IL-6 or helioxanthin analogues, or by the differentiation state of the hepatocyte.
Collapse
Affiliation(s)
- M Quasdorff
- Department of Gastroenterology and Hepatology, University Hospital Cologne, Germany
| | | |
Collapse
|
22
|
Hota SK, Hota KB, Prasad D, Ilavazhagan G, Singh SB. Oxidative-stress-induced alterations in Sp factors mediate transcriptional regulation of the NR1 subunit in hippocampus during hypoxia. Free Radic Biol Med 2010; 49:178-91. [PMID: 20381604 DOI: 10.1016/j.freeradbiomed.2010.03.027] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 03/16/2010] [Accepted: 03/30/2010] [Indexed: 11/23/2022]
Abstract
Ascent to high altitude is associated with tissue hypoxia resulting from the decrease in partial pressure of atmospheric oxygen. The hippocampus, in particular, is highly vulnerable to hypoxic insult, which at least in part can be attributed to the occurrence of glutamate excitotoxicity. Although this excitotoxic damage is often related to increased NMDA receptor activation and subsequent calcium-mediated free radical generation, the mechanisms involving the transcriptional regulation of NMDA receptor subunit expression by hypoxic stress remains to be explored. Our study reveals a novel mechanism for the regulation of expression of the NR1 subunit of NMDA receptors by the Sp family of transcription factors through an oxidative-stress-mediated mechanism that also involves the molecular chaperone Hsp90. The findings not only show the occurrence of lipid peroxidation and DNA damage in hippocampal cells exposed to hypoxia but also reveal a calcium-independent mechanism of selective oxidation and degradation of Sp3 by the 20S proteasome. This along with increased DNA binding activity of Sp1 leads to NR1 upregulation in the hippocampus during hypoxic stress. The study therefore provides evidence for free radical-mediated regulation of gene expression in hypoxia and the scope of the use of antioxidants in preventing excitotoxic neuronal damage during hypoxia.
Collapse
Affiliation(s)
- Sunil Kumar Hota
- Defence Institute of High Altitude Research, Leh, Ladakh, Jammu and Kashmir, India
| | | | | | | | | |
Collapse
|
23
|
Tyagi N, Vacek JC, Givvimani S, Sen U, Tyagi SC. Cardiac specific deletion of N-methyl-d-aspartate receptor 1 ameliorates mtMMP-9 mediated autophagy/mitophagy in hyperhomocysteinemia. J Recept Signal Transduct Res 2010; 30:78-87. [PMID: 20170426 DOI: 10.3109/10799891003614808] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Autophagy is an important process in the pathogenesis of cardiovascular diseases; however, the proximal triggers for mitochondrial autophagy were unknown. The N-methyl-d-aspartate receptor 1 (NMDA-R1) is a receptor for homocysteine (Hcy) and plays a key role in cardiac dysfunction. Cardiac-specific deletion of NMDA-R1 has been shown to ameliorate Hcy-induced myocyte contractility. Hcy activates mitochondrial matrix metalloproteinase-9 (mtMMP-9) and induces translocation of connexin-43 (Cxn-43) to the mitochondria (mtCxn-43). We sought to show cardiac-specific deletion of NMDA-R1 mitigates Hcy-induced mtCxn-43 translocation, mtMMP-9-mediated mtCxn-43 degradation, leading to mitophagy, in part, by decreasing mitochondrial permeability (MPT). Cardiac-specific knockout (KO) of NAMDA-R1 was generated using the cre/lox approach. The myocyte mitochondria were isolated from wild type (WT), WT + Hcy (1.8 g of DL-Hcy/L in the drinking water for 6 weeks), NMDA-R1 KO + Hcy, and NR1(fl/fl)/Cre (NR1(fl/fl)) genetic control mice. Mitochondrial respiratory capacity and MPT were measured by fluorescence-dye methods. The mitochondrial superoxide and peroxinitrite levels were detected by confocal microscopy using Mito-SOX and dihydrorhodamine-123. The mtMMP-9 activity and expression were detected by zymography and RT-PCR analyses. The mtCxn-43 translocation was detected by confocal microscopy. The degradation of mtCxn-43 and LC3-I/II (a marker of autophagy) were detected by Western blot. These results suggested that Hcy enhanced intramitochondrial nitrosative stress in myocytes. There was a robust increase in mtMMP-9 activity. An increase in translocation and degradation of mtCxn-43 was also noted. These increases led to mitophagy. The effects were ameliorated by cardiac-specific deletion of NMDA-R1. We concluded that HHcy increased mitochondrial nitrosative stress, thereby activating mtMMP-9 and inciting the degradation of mtCxn-43. This led to mitophagy, in part, by activating NMDA-R1. The findings of this study will lead to therapeutic ramifications for mitigating cardiovascular diseases by inhibiting the mitochondrial mitophagy and NMDA-R1 receptor.
Collapse
Affiliation(s)
- Neetu Tyagi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, KY 40292, USA
| | | | | | | | | |
Collapse
|
24
|
Seki M, Soussou W, Manabe SI, Lipton SA. Protection of retinal ganglion cells by caspase substrate-binding peptide IQACRG from N-methyl-D-aspartate receptor-mediated excitotoxicity. Invest Ophthalmol Vis Sci 2009; 51:1198-207. [PMID: 19815732 DOI: 10.1167/iovs.09-4102] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
PURPOSE This study investigated whether the enzymatically inactive caspase mimetic IQACRG protects rat retinal ganglion cells (RGCs) from excitotoxic insults. Minimally invasive delivery of the peptide to the retina was explored, and the mechanisms of neuroprotection were elucidated. METHODS IQACRG was linked to penetratin (P-IQACRG) to facilitate cellular uptake. RGC labeling by biotinylated-P-IQACRG delivered via intravitreal or subconjunctival injection was demonstrated by avidin-biotin chemistry. The authors used histologic and electrophysiological measures to evaluate the neuroprotective potential of P-IQACRG against RGC death induced by N-methyl-D-aspartate (NMDA) in vitro and in vivo. In addition, they monitored activity of an enzyme that is downstream of caspase-1, matrix metalloproteinase-9 (MMP-9), and protein levels of the caspase-3/7 substrate, myocyte enhancer factor 2C (MEF2C), to determine the effectiveness of IQACRG in blocking excessive caspase activity. RESULTS IQACRG significantly reduced NMDA-induced RGC death in culture and in vivo. Ex vivo electrophysiological recording of the retina on multielectrode arrays demonstrated functional rescue of RGCs by IQACRG. The authors also found that delivery of IQACRG to the retina inhibited NMDA-triggered MMP-9 activity and prevented cleavage of MEF2C protein that would otherwise have been engendered by caspase activation preceding RGC death. Strikingly, subconjunctival injection of P-IQACRG was very effective in preventing NMDA-induced RGC death in vivo. CONCLUSIONS These data demonstrate that IQACRG protects RGCs from excitotoxicity in vitro and in vivo. The positive results with subconjunctival administration of P-IQACRG suggest that in the future this treatment may be useful clinically in diseases such as glaucoma and retinal ischemia.
Collapse
Affiliation(s)
- Masaaki Seki
- Del E. Webb Center for Neuroscience, Aging, and Stem Cell Research, Burnham Institute for Medical Research, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
25
|
Coupling of energy metabolism and synaptic transmission at the transcriptional level: role of nuclear respiratory factor 1 in regulating both cytochrome c oxidase and NMDA glutamate receptor subunit genes. J Neurosci 2009; 29:483-92. [PMID: 19144849 DOI: 10.1523/jneurosci.3704-08.2009] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neuronal activity and energy metabolism are tightly coupled processes. Regions high in neuronal activity, especially of the glutamatergic type, have high levels of cytochrome c oxidase (COX). Perturbations in neuronal activity affect the expressions of COX and glutamatergic NMDA receptor subunit 1 (NR1). The present study sought to test our hypothesis that the coupling extends to the transcriptional level, whereby NR1 and possibly other NR subunits and COX are coregulated by the same transcription factor, nuclear respiratory factor 1 (NRF-1), which regulates all COX subunit genes. By means of multiple approaches, including in silico analysis, electrophoretic mobility shift and supershift assays, in vivo chromatin immunoprecipitation, promoter mutations, and real-time quantitative PCR, NRF-1 was found to functionally bind to the promoters of Grin 1 (NR1), Grin 2b (NR2b) and COX subunit genes, but not of Grin2a and Grin3a genes. These transcripts were upregulated by KCl and downregulated by tetrodotoxin (TTX) in cultured primary neurons. However, silencing of NRF-1 with small interference RNA blocked the upregulation of Grin1, Grin2b, and COX induced by KCl, and overexpression of NRF-1 rescued these transcripts that were suppressed by TTX. NRF-1 binding sites on Grin1 and Grin2b genes are also highly conserved among mice, rats, and humans. Thus, NRF-1 is an essential transcription factor critical in the coregulation of NR1, NR2b, and COX, and coupling exists at the transcriptional level to ensure coordinated expressions of proteins important for synaptic transmission and energy metabolism.
Collapse
|
26
|
Moshal KS, Kumar M, Tyagi N, Mishra PK, Metreveli N, Rodriguez WE, Tyagi SC. Restoration of contractility in hyperhomocysteinemia by cardiac-specific deletion of NMDA-R1. Am J Physiol Heart Circ Physiol 2009; 296:H887-92. [PMID: 19181966 DOI: 10.1152/ajpheart.00750.2008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Homocysteine (HCY) activated mitochondrial matrix metalloproteinase-9 and led to cardiomyocyte dysfunction, in part, by inducing mitochondrial permeability (MPT). Treatment with MK-801 [N-methyl-d-aspartate (NMDA) receptor antagonist] ameliorated the HCY-induced decrease in myocyte contractility. However, the role of cardiomyocyte NMDA-receptor 1 (R1) activation in hyperhomocysteinemia (HHCY) leading to myocyte dysfunction was not well understood. We tested the hypothesis that the cardiac-specific deletion of NMDA-R1 mitigated the HCY-induced decrease in myocyte contraction, in part, by decreasing nitric oxide (NO). Cardiomyocyte-specific knockout of NMDA-R1 was generated using cre/lox technology. NMDA-R1 expression was detected by Western blot and confocal microscopy. MPT was determined using a spectrophotometer. Myocyte contractility and calcium transients were studied using the IonOptix video-edge detection system and fura 2-AM loading. We observed that HHCY induced NO production by agonizing NMDA-R1. HHCY induced the MPT by agonizing NMDA-R1. HHCY caused a decrease in myocyte contractile performance, maximal rate of contraction and relaxation, and prolonged the time to 90% peak shortening and 90% relaxation by agonizing NMDA-R1. HHCY decreased contraction amplitude with the increase in calcium concentration. The recovery of calcium transient was prolonged in HHCY mouse myocyte by agonizing NMDA-R1. It was suggested that HHCY increased mitochondrial NO levels and induced MPT, leading to the decline in myocyte mechanical function by agonizing NMDA-R1.
Collapse
Affiliation(s)
- Karni S Moshal
- Dept. of Physiology and Biophysics, 500 S. Preston St., HSC Bldg. A-1115, Univ. of Louisville, Louisville, KY 40202, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Myocyte enhancer factor 2C as a neurogenic and antiapoptotic transcription factor in murine embryonic stem cells. J Neurosci 2008; 28:6557-68. [PMID: 18579729 DOI: 10.1523/jneurosci.0134-08.2008] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cell-based therapies require a reliable source of cells that can be easily grown, undergo directed differentiation, and remain viable after transplantation. Here, we generated stably transformed murine ES (embryonic stem) cells that express a constitutively active form of myocyte enhancer factor 2C (MEF2CA). MEF2C has been implicated as a calcium-dependent transcription factor that enhances survival and affects synapse formation of neurons as well as differentiation of cardiomyocytes. We now report that expression of MEF2CA, both in vitro and in vivo, under regulation of the nestin enhancer effectively produces "neuronal" progenitor cells that differentiate into a virtually pure population of neurons. Histological, electrophysiological, and behavioral analyses demonstrate that MEF2C-directed neuronal progenitor cells transplanted into a mouse model of cerebral ischemia can successfully differentiate into functioning neurons and ameliorate stroke-induced behavioral deficits.
Collapse
|
28
|
Watanabe K, Kanno T, Oshima T, Miwa H, Tashiro C, Nishizaki T. Vagotomy upregulates expression of the N-methyl-D-aspartate receptor NR2D subunit in the stomach. J Gastroenterol 2008; 43:322-6. [PMID: 18592148 DOI: 10.1007/s00535-008-2163-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Accepted: 01/21/2008] [Indexed: 02/04/2023]
Abstract
BACKGROUND N-methyl-D-aspartate (NMDA) receptor is the major excitatory neurotransmitter receptor in the central nervous system. Recent evidence has pointed to expression of NMDA receptor in peripheral non-neuronal tissues and organs; however, little is known about the expression of the receptor in the stomach. The present study identified what types of NMDA receptor subunits are expressed in the rat stomach and examined whether vagotomy affects their expression. METHODS To see the expression and distribution of the mRNAs and proteins for the NMDA receptor subunits, real-time reverse transcription-polymerase chain reaction, Western blotting, in situ hybridization, and immunohistochemistry were carried out on rat stomach with and without vagotomy. RESULTS Of the NMDA receptor subunits, NR1, NR2 (2A, 2B, 2C, 2D), and NR3 (3A, 3B), all NR subunit mRNAs except for the NR2B subunit mRNA were expressed in the intact rat stomach, with huge expression of NR2D mRNA. Expression of NR2D subunit mRNA and protein significantly increased after vagotomy. Increased expression was found in mucosal cells, the myenteric plexus, and the outer membrane of the stomach with vagotomy. CONCLUSIONS Vagotomy upregulates expression of the NR2D subunit in the stomach and therefore, the NR2D subunit may be implicated in gastric motility.
Collapse
Affiliation(s)
- Kanako Watanabe
- Department of Physiology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya 663-8501, Japan
| | | | | | | | | | | |
Collapse
|
29
|
Small-molecule activation of neuronal cell fate. Nat Chem Biol 2008; 4:408-10. [PMID: 18552832 DOI: 10.1038/nchembio.95] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Accepted: 05/16/2008] [Indexed: 11/08/2022]
Abstract
We probed an epigenetic regulatory path from small molecule to neuronal gene activation. Isoxazole small molecules triggered robust neuronal differentiation in adult neural stem cells, rapidly signaling to the neuronal genome via Ca(2+) influx. Ca(2+)-activated CaMK phosphorylated and mediated nuclear export of the MEF2 regulator HDAC5, thereby de-repressing neuronal genes. These results provide new tools to explore the epigenetic signaling circuitry specifying neuronal cell fate and new leads for neuro-regenerative drugs.
Collapse
|
30
|
Multisignal regulation of the rat NMDA1 receptor subunit gene — A pivotal role of glucocorticoid-dependent transcription. Life Sci 2008; 82:1137-41. [DOI: 10.1016/j.lfs.2008.03.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2007] [Revised: 03/10/2008] [Accepted: 03/18/2008] [Indexed: 11/15/2022]
|
31
|
Majdzadeh N, Morrison BE, D'Mello SR. Class IIA HDACs in the regulation of neurodegeneration. FRONT BIOSCI-LANDMRK 2008; 13:1072-82. [PMID: 17981613 DOI: 10.2741/2745] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Neurodegenerative diseases affect millions of patients annually and are a significant burden on the health care systems around the world. While there are symptomatic remedies for patients suffering from various neurodegenerative diseases, there are no cures as of today. Cell death by apoptosis is a common hallmark of neurodegeneration. Therefore, deciphering the molecular pathways regulating this process is of significant value to scientists' endeavor to understand neurodegenerative disorders. Efforts along these lines have uncovered a number of molecular pathways that regulate neuronal apoptosis. Recently, a family of proteins known as histone deacetylases (HDACs) has been linked to regulation of cell survival as well as death. The focus of this review is to summarize our current understanding of the role of HDACs and in particular a subgroup of proteins in this family classified as class IIa HDACs in the regulation of neuronal cell death. It is apparent based on the information presented in this review that although very similar in their primary sequence, members of this family of proteins often have distinct roles in orchestrating apoptotic cell death in the brain.
Collapse
Affiliation(s)
- Nazanin Majdzadeh
- University of Texas at Dallas, Department of Molecular and Cell Biology, Richardson, Texas 75080, USA
| | | | | |
Collapse
|
32
|
Laenen K, Haegeman G, Vanhoenacker P. Structure of the human 5-HT7 receptor gene and characterization of its promoter region. Gene 2007; 391:252-63. [PMID: 17321075 DOI: 10.1016/j.gene.2007.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2007] [Revised: 01/08/2007] [Accepted: 01/08/2007] [Indexed: 10/23/2022]
Abstract
The molecular mechanism regulating serotonin 5-HT(7) receptor expression is still unclear. In this study we provide evidence that transcription of the 5-HT(7) gene is at least partly regulated by Sp1 and Sp3. We isolated and sequenced >3000 bp of the upstream sequences and identified by RACE a number of transcriptional initiation sites over a region of 300 bp upstream of the coding region. This region has a high GC content, but contains no obvious TATA or a CAAT box. Besides a Sp1/Sp3 consensus motif, regulatory elements for AP2, Egr-1 and MAZ are also present. Transient transfection assays using deletion variants indicated that the GC-rich region is essential for full promoter activity. The role of Sp1 in this was confirmed by transient overexpression of both wild type Sp1 or dominant-negative forms. By gel shift and supershift analyses, targeting the Sp1 consensus sequence and the GC-rich region just upstream of the transcription initiation sites, binding of Sp1 and Sp3 was demonstrated. Both in vitro as well as in vivo experiments, using a cell line which endogenously expresses the 5-HT(7) receptor, indicated that mithramycin A, an inhibitor of Sp1/3 transcription factor binding, was able to inhibit 5-HT(7) promoter activity. Taken together these results support the essential role of Sp factors in regulating 5-HT(7) promoter activity.
Collapse
Affiliation(s)
- Koen Laenen
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Department of Molecular Biology, Ghent University, UGent, K.L. Ledeganckstraat 35, B-9000 Gent, Belgium
| | | | | |
Collapse
|
33
|
Zanzouri M, Lauritzen I, Lazdunski M, Patel A. The background K+ channel TASK-3 is regulated at both the transcriptional and post-transcriptional levels. Biochem Biophys Res Commun 2006; 348:1350-7. [PMID: 16925981 DOI: 10.1016/j.bbrc.2006.07.194] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2006] [Accepted: 07/28/2006] [Indexed: 10/24/2022]
Abstract
The K(+) channel TASK-3 is highly expressed in cerebellar granule neurons where it encodes the K(+) current IKso. Besides the role of TASK-3 in controlling cellular excitability and shaping neuronal responses, it has recently been proposed to contribute to the development and maturation of neurons in the cerebellum. K(+) dependent apoptosis and tumorigenesis have also been attributed to TASK-3 over-expression. Transcription of TASK-3 is strongly dependent on depolarization-induced Ca(2+)-entry. To understand the mechanisms involved in TASK-3 regulation, we have characterized a minimal promoter which specifically expresses in cellular backgrounds expressing endogenous TASK-3. Moreover, we have cloned and characterized the 5' and 3' untranslated regions of TASK-3. Both regions contribute to inhibit expression of a reporter gene. Given the direct consequence of membrane potential on TASK-3 expression, this is an important first step towards the understanding of the complex regulation of this gene.
Collapse
Affiliation(s)
- Marc Zanzouri
- Institut de Pharmacologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, UMR 6097, Université de Nice-Sophia Antipolis, 660 Route des Lucioles, Valbonne 06560, France
| | | | | | | |
Collapse
|
34
|
Rosenberger D, Moshal KS, Kartha GK, Tyagi N, Sen U, Lominadze D, Maldonado C, Roberts AM, Tyagi SC. Arrhythmia and neuronal/endothelial myocyte uncoupling in hyperhomocysteinemia. Arch Physiol Biochem 2006; 112:219-27. [PMID: 17178594 PMCID: PMC3182485 DOI: 10.1080/13813450601093443] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Elevated levels of homocysteine (Hcy) known as hyperhomocysteinemia (HHcy) are associated with arrhythmogenesis and sudden cardiac death (SCD). Hcy decreases constitutive neuronal and endothelial nitric oxide (NO), and cardiac diastolic relaxation. Hcy increases the iNOS/NO, peroxynitrite, mitochondrial NADPH oxidase, and suppresses superoxide dismutase (SOD) and redoxins. Hcy activates matrix metalloproteinase (MMP), disrupts connexin-43 and increases collagen/elastin ratio. The disruption of connexin-43 and accumulation of collagen (fibrosis) disrupt the normal pattern of cardiac conduction and attenuate NO transport from endothelium to myocyte (E-M) causing E-M uncoupling, leading to a pro-arrhythmic environment. The goal of this review is to elaborate the mechanism of Hcy-mediated iNOS/NO in E-M uncoupling and SCD. It is known that Hcy creates arrhythmogenic substrates (i.e. increase in collagen/elastin ratio and disruption in connexin-43) and exacerbates heart failure during chronic volume overload. Also, Hcy behaves as an agonist to N-methyl-D-aspartate (NMDA, an excitatory neurotransmitter) receptor-1, and blockade of NMDA-R1 reduces the increase in heart rate-evoked by NMDA-analog and reduces SCD. This review suggest that Hcy increases iNOS/NO, superoxide, metalloproteinase activity, and disrupts connexin-43, exacerbates endothelial-myocyte uncoupling and cardiac failure secondary to inducing NMDA-R1.
Collapse
Affiliation(s)
- Dorothea Rosenberger
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Tsang SWY, Pomakian J, Marshall GA, Vinters HV, Cummings JL, Chen CPLH, Wong PTH, Lai MKP. Disrupted muscarinic M1 receptor signaling correlates with loss of protein kinase C activity and glutamatergic deficit in Alzheimer's disease. Neurobiol Aging 2006; 28:1381-7. [PMID: 16828202 DOI: 10.1016/j.neurobiolaging.2006.06.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2006] [Revised: 04/08/2006] [Accepted: 06/02/2006] [Indexed: 11/21/2022]
Abstract
There are few studies on the clinical and neurochemical correlates of postsynaptic cholinergic dysfunction in Alzheimer's disease (AD). We have previously found that attenuation of guanine nucleotide-binding (G-) protein coupling to muscarinic M(1) receptors in the neocortex was associated with dementia severity. The present study aims to study whether this loss of M(1)/G-protein coupling is related to alterations in signaling kinases and NMDA receptors. Postmortem frontal cortices of 22 AD subjects and 12 elderly controls were obtained to measure M(1) receptors, M(1)/G-protein coupling, NMDA receptors as well as protein kinase C (PKC) and Src kinase activities. We found that the extent of M(1)/G-protein coupling loss was correlated with reductions in PKC activity and NMDA receptor density. In contrast, Src kinase activity was neither altered nor associated with M(1)/G-protein coupling. Given the well established roles of neuronal PKC signaling and NMDA receptor function in cognitive processes, our results lend further insight into the mechanisms by which postsynaptic cholinergic dysfunction may underlie the cognitive features of AD, and suggest alternative therapeutic targets to cholinergic replacement.
Collapse
Affiliation(s)
- Shirley W Y Tsang
- Dementia Research Laboratory, Department of Clinical Research, Singapore General Hospital, Singapore
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Qiu Z, Norflus F, Singh B, Swindell MK, Buzescu R, Bejarano M, Chopra R, Zucker B, Benn CL, DiRocco DP, Cha JHJ, Ferrante RJ, Hersch SM. Sp1 Is Up-regulated in Cellular and Transgenic Models of Huntington Disease, and Its Reduction Is Neuroprotective. J Biol Chem 2006; 281:16672-80. [PMID: 16595660 DOI: 10.1074/jbc.m511648200] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Interactions between mutant huntingtin (Htt) and a variety of transcription factors including specificity proteins (Sp) have been suggested as a central mechanism in Huntington disease (HD). However, the transcriptional activity induced by Htt in neurons that triggers neuronal death has yet to be fully elucidated. In the current study, we characterized the relationship of Sp1 to Htt protein aggregation and neuronal cell death. We found increased levels of Sp1 in neuronal-like PC12 cells expressing mutant Htt, primary striatal neurons, and brain tissue of HD transgenic mice. Sp1 levels were also elevated when 3-nitropropionate (3-NP) was used to induce cell death in PC12 cells. To assess the effects of knocking down Sp1 in HD pathology, we used Sp1 siRNA, a heterozygous Sp1 knock-out mouse, and mithramycin A, a DNA-intercalating agent that inhibits Sp1 function. The three approaches consistently yielded reduced levels of Sp1 which ameliorated toxicity caused by either mutant Htt or 3-NP. In addition, when HD mice were crossed with Sp1 heterozygous knock-out mice, the resulting offspring did not experience the loss of dopamine D2 receptor mRNA characteristic of HD mice, and survived longer than their HD counterparts. Our data suggest that enhancement of transcription factor Sp1 contributes to the pathology of HD and demonstrates that its suppression is beneficial.
Collapse
Affiliation(s)
- Zhihua Qiu
- Massachusetts General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, 114 16th Street, Charlestown, MA 02129, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Papadodima O, Sergaki M, Hurel C, Mamalaki A, Matsas R. Characterization of the BM88 promoter and identification of an 88 bp fragment sufficient to drive neurone-specific expression. J Neurochem 2005; 95:146-59. [PMID: 16181419 DOI: 10.1111/j.1471-4159.2005.03350.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BM88 is a neurone-specific protein implicated in cell cycle exit and differentiation of neuronal precursors. It is widely expressed in terminally differentiated neurones but also in neuronal progenitors, albeit in lower levels. Thus BM88 expression shows a tight correlation with the progression of progenitor cells towards neuronal differentiation. Here we report the genomic organization and proximal promoter characterization of the human and mouse BM88 genes. Both promoters lie in a CpG island, are TATA-less and have multiple transcription start sites. Deletion analysis performed on the human BM88 gene revealed an 88 bp minimal promoter fragment that is preferentially active in neural cells. Importantly, this minimal promoter is sufficient to confer specific transcriptional activity in primary neurones, but not in glial cells. Within the promoter region there are four functional Sp1-binding sites. Simultaneous mutations to all four Sp1 sites results in complete loss of promoter activity. Transactivation experiments revealed that Sp1 directly activates the BM88 promoter while activation also occurs in the presence of neurogenin-1. Characterization of the promoter elements that control neurone-specific and developmental expression of BM88 should contribute to the elucidation of the transcriptional networks that regulate the transition from a proliferative neural progenitor to a post-mitotic neurone.
Collapse
Affiliation(s)
- Olga Papadodima
- Department of Biochemistry, Hellenic Pasteur Institute, 11521 Athens, Greece
| | | | | | | | | |
Collapse
|
38
|
Zarain-Herzberg A, Lee-Rivera I, Rodríguez G, López-Colomé AM. Cloning and characterization of the chick NMDA receptor subunit-1 gene. ACTA ACUST UNITED AC 2005; 137:235-51. [PMID: 15950782 DOI: 10.1016/j.molbrainres.2005.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2004] [Revised: 02/22/2005] [Accepted: 03/13/2005] [Indexed: 10/25/2022]
Abstract
The N-methyl-D-aspartate family of glutamate receptors (NMDARs) are tetrameric cation channels including NR1, NR2, and possibly NR3 subunits. The physiological properties of the receptor are directly related to the subunit composition of the oligomer. Whereas NR1 is essential for the formation of functional channels, NR2 and NR3 play a modulatory role. This work reports, for the first time, the cloning of a non-mammalian NR1 gene, including the 5'-regulatory region. The chick gene spans 31 kb of genomic DNA sequence composed of 22 exons interrupted by 21 introns. The exon/intron organization and the deduced amino acid sequence up to the end of exon 19 showed 85% homology to mammalian NR1 cloned genes. Significant differences from mammals were found at the C-terminal region which in the chick gene, includes a novel exon (exon 20) previously identified at the mRNA level in the chick retina. The basal promoter activity was shown to reside within the proximal 377 bp of 5'-regulatory region. The transcriptional activity of the 5'-flanking region of the chick NR1 gene was shown to be higher in neuronally-differentiated PC12 cells and in chick retinal neurons, than in non-differentiated PC12 cells and Müller glia. Comparison of the 5'-flanking region of chick NR1 gene with mammalian NR1 genes suggests that, in spite of significant differences in the nucleotide sequence, they share common DNA binding sites such as RE1, SP1, AP2, CREB, NFkappaB, and MEF2; therefore, some of the molecular mechanisms involved in transcriptional regulation of NR1 gene expression could be conserved among species.
Collapse
|
39
|
Qiang M, Rani CSS, Ticku MK. Neuron-restrictive silencer factor regulates the N-methyl-D-aspartate receptor 2B subunit gene in basal and ethanol-induced gene expression in fetal cortical neurons. Mol Pharmacol 2005; 67:2115-25. [PMID: 15755907 DOI: 10.1124/mol.104.010751] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neuron-restrictive silencer factor (NRSF) is a transcriptional repressor of multiple neuronal genes. This study addressed the role of NRSF in N-methyl-D-aspartate (NMDA) receptor NR2B promoter activity and the molecular mechanisms of ethanol-induced NR2B up-regulation in fetal cortical neurons. The 5'-flanking region of the NR2B gene contains five NRSE-like elements. Functional analysis of the upstream regions of the NR2B gene by transient transfection of neurons revealed that neuron-restrictive silencer element (NRSE) motifs located between base pair -1407 and -2741 represses transcription of the gene. Analysis by electrophoretic mobility shift assay and reporter gene assay identified NRSE2 and 3 as responsible for repressing NR2B gene transcription. The identity of NRSF as the functional binding factor is suggested by the specific binding of in vitro synthesized NRSF or cell lysate to the labeled probes and the specific antibody-induced supershift. Furthermore, whereas mutations of NRSE2 and 3 motifs increased the promoter activity, overexpression of NRSF reduced it significantly. The pattern of NRSF expression during development was investigated and demonstrated that the highest expression is on embryonic day 14 with moderate expression on postnatal day 0, reflecting a possible role of NRSF as a regulator during development. Treatment of cultured cortical neurons with 100 mM ethanol for 5 days caused a significant decrease in the NRSF mRNA and protein levels, NRSF/NRSE binding activity, and an increase in the promoter activity. Therefore, our studies suggest that NRSF is a negative regulator of NR2B expression and may contribute to the ethanol-induced up-regulation of the NR2B gene in fetal cortical neurons.
Collapse
Affiliation(s)
- Mei Qiang
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA.
| | | | | |
Collapse
|
40
|
Mejía-Guerra MK, Lareo LR. In SilicoIdentification of Regulatory Elements of GRIN1 Genes. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2005; 9:106-15. [PMID: 15805781 DOI: 10.1089/omi.2005.9.106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The ionotropic receptor of glutamate activated by N-methyl-D-aspartate (iGluR-NMDA) is a multiheteromeric complex constituted by at least three different types of subunits, encoded by seven different genes. The subunits of iGluR-NMDA have a complex system of regulation of their gene expression. Their expression is specific for each type of neural cell, as well as for the age of the organism. Moreover, there are reports that iGluR-NMDA expression is species-specific. Even though this macromolecular complex is very important in physiology and pathology of the central nervous system, knowledge to date about the regulatory elements controlling expression is scarce. We present the results of an in silico prediction of potential regulatory elements, some of which coincide with the few known experimentally. We also present the important differences regarding the presence and the localization of the regulatory elements among human, rat, and mouse species.
Collapse
Affiliation(s)
- María Katherine Mejía-Guerra
- Department of Nutrition and Biochemistry, Computational and Structural Biochemistry and Bioinformatics, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | |
Collapse
|
41
|
Abstract
The myocyte enhancer factor 2 (MEF2) transcription factors were originally identified, as their family name implies, on the basis of their role in muscle differentiation. Expression of the four MEF2 proteins, however, is not restricted to contractile tissue. While it has been known for more than a decade that MEF2s are abundantly expressed in neurons, their contributions to the development and function of the nervous system are only now being elucidated. Interestingly, the emerging mechanisms regulating MEF2 in neurons have significant parallels with the regulatory mechanisms in muscle, despite the quite distinct identities of these two electrically excitable tissues. The goal of this chapter is to provide an introduction to those regulatory mechanisms and their consequences for brain development. As such, we first provide an overview of MEF2 itself and its expression within the central nervous system. The second part of this chapter describes the signaling molecules that regulate MEF2 transcriptional activity and their contributions to MEF2 function. The third part of this chapter discusses the role of MEF2 proteins in the developing nervous system and compares the analogous functions of this protein family in muscle and brain.
Collapse
Affiliation(s)
- Aryaman K Shalizi
- Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
42
|
Wang X, Tang X, Gong X, Albanis E, Friedman SL, Mao Z. Regulation of hepatic stellate cell activation and growth by transcription factor myocyte enhancer factor 2. Gastroenterology 2004; 127:1174-88. [PMID: 15480995 DOI: 10.1053/j.gastro.2004.07.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Hepatic stellate cells (HSCs) undergo activation during the development of liver fibrosis. Transcriptional regulation plays a key role in this process. We studied the role of transcription factor myocyte enhancer factor 2 (MEF2) during HSC activation. METHODS Culture of HSCs isolated from rat liver on plastic dishes and HSC-T6 on a basement membrane-like matrix were used as models of HSC activation and deactivation, respectively. The expression and activity of MEF2 were correlated with HSC activation. The roles of MEF2 during HSC activation were assessed in vitro and in vivo by animal models of fibrosis. RESULTS Early induction of MEF2 messenger RNA and protein accompanied culture-induced HSC activation. This was associated with enhanced MEF2 DNA binding and transactivation activity. p38 mitogen-activated protein kinase but not extracellular signal-regulated kinase pathway was required for increased MEF2 activity during HSC activation. Increased MEF2 protein also correlated with fibrosis in vivo. Reversal of HSC activation was paralleled by a marked decrease in MEF2 protein and activity. Functionally, enhancing MEF2 significantly increased the expression of alpha-smooth muscle actin (alpha-SMA), activated collagen I promoter activity, and stimulated HSC proliferation. MEF2 interference RNA significantly inhibited expression of alpha-SMA, collagen alpha1(I), and proliferating cell nuclear antigen. CONCLUSIONS The studies provide the first evidence for the presence of MEF2 in the liver and show that MEF2 regulates multiple aspects of HSC activation. These studies show a novel role of MEF2 as a key nuclear mediator that may participate in the pathologic process of liver fibrogenesis in vivo.
Collapse
Affiliation(s)
- Xuemin Wang
- Liver Research Center, Department of Medicine, Rhode Island Hospital and Brown University School of Medicine, Providence 02903, USA
| | | | | | | | | | | |
Collapse
|
43
|
Rodova M, Brownback K, Werle MJ. Okadaic acid augments utrophin in myogenic cells. Neurosci Lett 2004; 363:163-7. [PMID: 15172107 DOI: 10.1016/j.neulet.2004.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2004] [Revised: 03/05/2004] [Accepted: 04/01/2004] [Indexed: 11/21/2022]
Abstract
Duchenne muscular dystrophy is a fatal childhood disease caused by mutations that abolish the expression of dystrophin in muscle. Utrophin is a paralogue of dystrophin and can functionally replace it in skeletal muscle. A potential therapeutic approach is to increase utrophin levels in muscle. One way to achieve this aim is to increase the expression of the utrophin gene at a transcriptional level via promoter activation. In this study, we have shown that utrophin A mRNA levels can be induced by okadaic acid in murine myogenic C2C12 cells. We have found that a utrophin A promoter reporter can be induced by Sp1 in C2C12 myoblasts, but not in myotubes. This activation can be enhanced by okadaic acid treatment. Our data suggest that this induction is due to Sp1 phosphorylation during myogenesis and thus, utrophin expression in muscle could be regulated by treatment with phosphatase inhibitors. Control of utrophin promoter activation could then be used to increase the expression of utrophin, and thus ameliorate the symptoms of Duchenne muscular dystrophy.
Collapse
MESH Headings
- Animals
- Base Sequence/genetics
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Cell Line
- Cytoskeletal Proteins/genetics
- Enzyme Inhibitors/pharmacology
- Enzyme Inhibitors/therapeutic use
- Gene Expression Regulation, Developmental/drug effects
- Gene Expression Regulation, Developmental/genetics
- Humans
- Membrane Proteins/genetics
- Mice
- Molecular Sequence Data
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/therapy
- Myoblasts, Skeletal/drug effects
- Myoblasts, Skeletal/metabolism
- Okadaic Acid/pharmacology
- Phosphoric Monoester Hydrolases/antagonists & inhibitors
- Phosphoric Monoester Hydrolases/metabolism
- Phosphorylation
- Promoter Regions, Genetic/genetics
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Sp1 Transcription Factor/genetics
- Sp1 Transcription Factor/metabolism
- Sp1 Transcription Factor/pharmacology
- Up-Regulation/drug effects
- Up-Regulation/genetics
- Utrophin
Collapse
Affiliation(s)
- Marianna Rodova
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | | |
Collapse
|
44
|
Leysen I, Van der Gucht E, Eysel UT, Huybrechts R, Vandesande F, Arckens L. Time-dependent changes in the expression of the MEF2 transcription factor family during topographic map reorganization in mammalian visual cortex. Eur J Neurosci 2004; 20:769-80. [PMID: 15255987 DOI: 10.1111/j.1460-9568.2004.03535.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Removal of retinal input from a restricted region of adult mammalian visual cortex leads to a substantial reorganization of the retinotopy within the lesion projection zone (LPZ) of primary visual cortex (area 17). Little is known about the molecular mechanisms underlying such cortical plasticity. We investigated whether small but homonymous central retinal lesions induced differences in gene expression patterns between central area 17, the LPZ, vs. peripheral area 17 of the adult cat. Systematic differential mRNA display screening revealed higher levels for the mRNA encoding the transcription factor MEF2A in the LPZ. Semi-quantitative PCR confirmed this dependency of mef2A mRNA expression on visual eccentricity in area 17 of animals with retinal lesions in contrast to normal animals. Western blotting experiments extended these data to the protein level and to two other members of the MEF2 transcription factor family, i.e. MEF2C and MEF2D. Quantitative analysis of the Western blotting experiments further revealed a post-lesion survival time-dependent change in expression for all three MEF2 family members. The lesion effect was maximal at 3 days and 1 month post-lesion, but only minor at 2 weeks post-lesion. Interestingly, complete removal of retinal input from area 17 by surgery did not significantly alter the expression of the MEF2 transcription factors, excluding a definite correlation between neuronal activity and MEF2A expression levels. MEF2A immunocytochemistry confirmed both qualitatively and quantitatively the Western blotting observations in all animal models. Together, our findings identified a brain plasticity-related expression pattern for the MEF2 transcription factor family in adult mammalian neocortex.
Collapse
Affiliation(s)
- Inge Leysen
- Laboratory of Neuroplasticity and Neuroproteomics, Katholieke Universiteit Leuven, Naamsesstraat 59, B-3000 Leuven, Belgium
| | | | | | | | | | | |
Collapse
|
45
|
Ma L, Song L, Radoi GE, Harrison NL. Transcriptional regulation of the mouse gene encoding the alpha-4 subunit of the GABAA receptor. J Biol Chem 2004; 279:40451-61. [PMID: 15265862 DOI: 10.1074/jbc.m406827200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The gamma-aminobutyric acid type A receptors (GABAA-Rs) mediate fast inhibitory synaptic transmission in the brain. The alpha4 subunit of the GABAA-R confers distinct pharmacological properties on the receptor and its expression pattern exhibits plasticity in response to physiological and pharmacological stimuli, including withdrawal from progesterone and alcohol. We have analyzed the promoter region of the mouse GABRA4 gene that encodes the alpha4 subunit and found that the promoter has multiple transcriptional initiation sites and lacks a TATA box. The minimal promoter for GABRA4 spans the region between -444 to -19 bp relative to the coding ATG and shows high activity in cultured mouse cortical neurons. Both Sp3 and Sp4 transcription factors can interact with the two Sp1 binding sites within the minimal promoter and are critical for maximal activity of the promoter in neurons.
Collapse
Affiliation(s)
- Limei Ma
- Department of Anesthesiology, Weill Medical College, Cornell University, New York, New York 10021, USA
| | | | | | | |
Collapse
|
46
|
Liu A, Hoffman PW, Lu W, Bai G. NF-kappaB site interacts with Sp factors and up-regulates the NR1 promoter during neuronal differentiation. J Biol Chem 2004; 279:17449-58. [PMID: 14970236 DOI: 10.1074/jbc.m311267200] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The NR1 gene undergoes induction in neurogenesis mainly via promoter de-repression, and up-regulation during neuronal differentiation by undefined mechanism(s). Here, we show that in the distal region the NR1 promoter has an active NF-kappaB site sharing the consensus with the immunoglobulin (Ig)/human immunodeficiency virus NF-kappaB site. Mutation of this site significantly reduced NR1 promoter up-regulation during neuronal differentiation of P19 cells. Electrophoretic mobility shift assays revealed that P19 nuclei constitutively contained p50 and that neuronal differentiation not only increased nuclear p50 but also induced p65 nuclear translocation. Responding to this change was an up-regulation of NF-kappaB-dependent promoter activity. However, inhibition of NF-kappaB nuclear translocation by an IkappaBalpha super-repressor or decoy DNA only moderately inhibited NR1 promoter up-regulation. Interestingly, the NR1 NF-kappaB site strongly interacted with Sp3/Sp1, instead of NF-kappaB factors, in P19 nuclear extracts. This interaction was reduced for Sp3 following neuronal differentiation, accompanied by dynamic expression of Sp factors. Cotransfection of Sp factors (Sp1, 3, or 4) upregulated the NR1 NF-kappaB site dramatically in differentiated neurons, but only moderately in undifferentiated P19 cells. This up-regulation was strong for Sp1 in differentiated cells and for Sp3 in undifferentiated cells. Chromatin-immunoprecipitation assays further demonstrated that Sp1 and Sp3 interacted with the NR1 NF-kappaB site in situ, and Sp3 lost its interaction after neuronal differentiation. We conclude that the NF-kappaB site positively regulates the NR1 promoter during neuronal differentiation via interacting mainly with Sp factors and neuronal differentiation reduces the effect of Sp3 factor on this site.
Collapse
Affiliation(s)
- Anguo Liu
- Department of Biomedical Sciences, Dental School, Program in Neuroscience, and Program in Cellular and Molecular Biology, University of Maryland, Baltimore, Maryland 21201, USA
| | | | | | | |
Collapse
|
47
|
Abstract
The cis-element profile (or cis-profile) of a gene refers to the collection of transcription factor binding sites (TFBS) regulating the transcription of the gene. Underlying the various published studies that attempt to discover cis-elements in the vicinity of co-expressed genes via pattern detection algorithms, there is an implicit assumption that a correlation exists between co-expressed genes and their cis-profiles. In this study, we show that the cis-similarity, defined as the proportion of shared TFBS between two cis-element profiles, is higher for functionally linked interacting proteins as well as for members of a signal transduction pathway. A similar analysis of the enzymes catalyzing the conversion of adjacent substrates to products in a collection of metabolic pathways, did not reveal higher cis-similarity. The analysis is based on three distinct sources of publicly available data, namely, 1) the BIND database of interacting proteins, 2) known interactions in NMDAR protein complex, 3) the apoptosis pathway and nine pathways related to metabolism of cofactors and vitamins all from KEGG. Additionally, we analyze the cis-element profiles of all the genes in the glutamate receptor (GR) sub-complex of NMDAR complex to detect a set of cis-elements that occur adjacent to a majority of the genes. We show that most of the corresponding transcription factors are known to be involved in GR regulation by comparing our findings with the published biomedical literature. In addition, we were able to detect transcripts whose gene products associate with GR by searching for transcripts that share the same regulatory signals as those detected for GR. This suggests a novel computational methodology for constructing high-order gene regulatory models and detecting co-regulated gene products.
Collapse
Affiliation(s)
- Sridhar Hannenhalli
- Informatics Research, Celera Genomics, 45 West Gude Drive, Rockville, Maryland 20850, USA.
| | | |
Collapse
|
48
|
Safe S, Kim K. Nuclear receptor-mediated transactivation through interaction with Sp proteins. PROGRESS IN NUCLEIC ACID RESEARCH AND MOLECULAR BIOLOGY 2004; 77:1-36. [PMID: 15196889 DOI: 10.1016/s0079-6603(04)77001-4] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-4466, USA
| | | |
Collapse
|
49
|
Histone deacetylase inhibition by sodium butyrate chemotherapy ameliorates the neurodegenerative phenotype in Huntington's disease mice. J Neurosci 2003. [PMID: 14561870 DOI: 10.1523/jneurosci.23-28-09418.2003] [Citation(s) in RCA: 470] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The precise cause of neuronal death in Huntington's disease (HD) is unknown. Although no single specific protein-protein interaction of mutant huntingtin has emerged as the pathologic trigger, transcriptional dysfunction may contribute to the neurodegeneration observed in HD. Pharmacological treatment using the histone deacetylase inhibitor sodium butyrate to modulate transcription significantly extended survival in a dose-dependent manner, improved body weight and motor performance, and delayed the neuropathological sequelae in the R6/2 transgenic mouse model of HD. Sodium butyrate also increased histone and Specificity protein-1 acetylation and protected against 3-nitropropionic acid neurotoxicity. Microarray analysis showed increased expression of alpha- and beta-globins and MAP kinase phosphatase-1 in sodium butyrate-treated R6/2 mice, indicative of improved oxidative phosphorylation and transcriptional regulation. These findings strengthen the hypothesis that transcriptional dysfunction plays a role in the pathogenesis of HD and suggest that therapies aimed at modulating transcription may target early pathological events and provide clinical benefits to HD patients.
Collapse
|
50
|
Linseman DA, Bartley CM, Le SS, Laessig TA, Bouchard RJ, Meintzer MK, Li M, Heidenreich KA. Inactivation of the myocyte enhancer factor-2 repressor histone deacetylase-5 by endogenous Ca(2+) //calmodulin-dependent kinase II promotes depolarization-mediated cerebellar granule neuron survival. J Biol Chem 2003; 278:41472-81. [PMID: 12896970 DOI: 10.1074/jbc.m307245200] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cerebellar granule neuron (CGN) survival depends on activity of the myocyte enhancer factor-2 (MEF2) transcription factors. Neuronal MEF2 activity is regulated by depolarization via a mechanism that is presently unclear. Here, we show that depolarization-mediated MEF2 activity and CGN survival are compromised by overexpression of the MEF2 repressor histone deacetylase-5 (HDAC5). Furthermore, removal of depolarization induced rapid cytoplasm-to-nuclear translocation of endogenous HDAC5. This effect was mimicked by addition of the calcium/calmodulin-dependent kinase (CaMK) inhibitor KN93 to depolarizing medium. Removal of depolarization or KN93 addition resulted in dephosphorylation of HDAC5 and its co-precipitation with MEF2D. HDAC5 nuclear translocation triggered by KN93 induced a marked loss of MEF2 activity and subsequent apoptosis. To selectively decrease CaMKII, CGNs were incubated with an antisense oligonucleotide to CaMKIIalpha. This antisense decreased CaMKIIalpha expression and induced nuclear shuttling of HDAC5 in CGNs maintained in depolarizing medium. Selectivity of the CaMKIIalpha antisense was demonstrated by its lack of effect on CaMKIV-mediated CREB phosphorylation. Finally, antisense to CaMKIIalpha induced caspase-3 activation and apoptosis, whereas a missense control oligonucleotide had no effect on CGN survival. These results indicate that depolarization-mediated calcium influx acts through CaMKII to inhibit HDAC5, thereby sustaining high MEF2 activity in CGNs maintained under depolarizing conditions.
Collapse
Affiliation(s)
- Daniel A Linseman
- Department of Pharmacology, University of Colorado Health Sciences Center and the Denver Veterans Affairs Medical Center, Denver, Colorado 80262, USA
| | | | | | | | | | | | | | | |
Collapse
|