1
|
De Paolis V, Paolillo N, Siri T, Grosso A, Lorello V, Spina C, Caporali G, La Regina F, Vignoli B, Giorgi C. An antisense-long-noncoding-RNA modulates p75 NTR expression levels during neuronal polarization. iScience 2025; 28:111566. [PMID: 39811648 PMCID: PMC11730960 DOI: 10.1016/j.isci.2024.111566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/05/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
Proper polarization of newly generated neurons is a critical process for neural network formation and brain development. The pan-neurotrophin p75NTR receptor plays a key role in this process localizing asymmetrically in one of the differentiating neurites and specifying its axonal identity in response to neurotrophins. During axonal specification, p75NTR levels are transiently modulated, yet the molecular mechanisms underlying this process are not known. Here, we identified a previously uncharacterized natural antisense transcript, AS-p75, encoded within the p75NGFR mouse gene. Using an in vitro model of polarizing murine neurons, we found that AS-p75 and p75NTR display divergent expression profiles and that p75NTR expression levels increase upon competition or depletion of AS-p75, indicating that AS-p75 is a negative regulator of p75NTR expression. Depletion of AS-p75 also results in altered p75NTR subcellular distribution and affects the polarization process. Overall, our data uncovered AS-p75 as a modulator of p75NTR expression, offering new insights into the regulation of this neurotrophin receptor during in vitro neuronal polarization.
Collapse
Affiliation(s)
- Veronica De Paolis
- Department of Experimental Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Viale Regina Elena 295, 00161 Rome, Italy
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Via Ercole Ramarini 32, 00015 Monterotondo, Italy
| | - Nicoletta Paolillo
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Viale Regina Elena 295, 00161 Rome, Italy
| | - Tiziano Siri
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Viale Regina Elena 295, 00161 Rome, Italy
- Department of Sciences, University of Roma Tre, Viale Guglielmo Marconi 446, 00146 Rome, Italy
- CERVO Brain Research Center, Quebec City, QC G1J 2G3, Canada
| | - Alessandra Grosso
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Viale Regina Elena 295, 00161 Rome, Italy
- Department of Biology and Biotechnology “Charles Darwin”, University of Rome “Sapienza”, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Veronica Lorello
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Viale Regina Elena 295, 00161 Rome, Italy
- Department of Biology and Biotechnology “Charles Darwin”, University of Rome “Sapienza”, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Cristina Spina
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Viale Regina Elena 295, 00161 Rome, Italy
- Department of Biology and Biotechnology “Charles Darwin”, University of Rome “Sapienza”, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Gabriele Caporali
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Viale Regina Elena 295, 00161 Rome, Italy
- Department of Biology and Biotechnology “Charles Darwin”, University of Rome “Sapienza”, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Federico La Regina
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Viale Regina Elena 295, 00161 Rome, Italy
| | - Beatrice Vignoli
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Viale Regina Elena 295, 00161 Rome, Italy
- Department of Cellular, Computational and Integrative Biology – CIBIO, University of Trento, Via Sommarive 9, 38123 Povo TN, Italy
| | - Corinna Giorgi
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Viale Regina Elena 295, 00161 Rome, Italy
- Institute of Molecular Biology and Pathology, National Research Council of Italy (IBPM-CNR), P.le Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
2
|
He Q, Li R, Zhong N, Ma J, Nie F, Zhang R. The role and molecular mechanisms of the early growth response 3 gene in schizophrenia. Am J Med Genet B Neuropsychiatr Genet 2024; 195:e32969. [PMID: 38327141 DOI: 10.1002/ajmg.b.32969] [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: 07/04/2023] [Revised: 01/15/2024] [Accepted: 01/22/2024] [Indexed: 02/09/2024]
Abstract
Schizophrenia is a chronic, debilitating mental illness caused by both genetic and environmental factors. Genetic factors play a major role in schizophrenia development. Early growth response 3 (EGR3) is a member of the EGR family, which is associated with schizophrenia. Accumulating studies have investigated the relationship between EGR3 and schizophrenia. However, the role of EGR3 in schizophrenia pathogenesis remains unclear. In the present review, we focus on the progress of research related to the role of EGR3 in schizophrenia, including association studies between EGR3 and schizophrenia, abnormal gene expressional analysis of EGR3 in schizophrenia, biological function studies of EGR3 in schizophrenia, the molecular regulatory mechanism of EGR3 and schizophrenia susceptibility candidate genes, and possible role of EGR3 in the immune system function in schizophrenia. In summary, EGR3 is a schizophrenia risk candidate factor and has comprehensive regulatory roles in schizophrenia pathogenesis. Further studies investigating the molecular mechanisms of EGR3 in schizophrenia are warranted for understanding the pathophysiology of this disorder as well as the development of new therapeutic strategies for the treatment and control of this disorder.
Collapse
Affiliation(s)
- Qi He
- School of Basic Medicine, Shaanxi Key Laboratory of Acupuncture and Medicine, Shannxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Ruochun Li
- Department of Medical Technology, Guiyang Healthcare Vocational University, Guiyang, Guizhou, China
| | - Nannan Zhong
- Department of Medical Technology, Guiyang Healthcare Vocational University, Guiyang, Guizhou, China
| | - Jie Ma
- Department of Electron Microscope, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Fayi Nie
- School of Basic Medicine, Shaanxi Key Laboratory of Acupuncture and Medicine, Shannxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Rui Zhang
- Department of Medical Technology, Guiyang Healthcare Vocational University, Guiyang, Guizhou, China
| |
Collapse
|
3
|
Xu M, Thottappillil N, Cherief M, Li Z, Zhu M, Xing X, Gomez-Salazar M, Mwirigi JM, Sankaranarayanan I, Tavares-Ferreira D, Zhang C, Wang XW, Archer M, Guan Y, Tower RJ, Cahan P, Price TJ, Clemens TL, James AW. Mapping Somatosensory Afferent Circuitry to Bone Identifies Neurotrophic Signals Required for Fracture Healing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597786. [PMID: 38895367 PMCID: PMC11185682 DOI: 10.1101/2024.06.06.597786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The profound pain accompanying bone fracture is mediated by somatosensory neurons, which also appear to be required to initiate bone regeneration following fracture. Surprisingly, the precise neuroanatomical circuitry mediating skeletal nociception and regeneration remains incompletely understood. Here, we characterized somatosensory dorsal root ganglia (DRG) afferent neurons innervating murine long bones before and after experimental long bone fracture in mice. Retrograde labeling of DRG neurons by an adeno-associated virus with peripheral nerve tropism showed AAV-tdT signal. Single cell transcriptomic profiling of 6,648 DRG neurons showed highest labeling across CGRP+ neuron clusters (6.9-17.2%) belonging to unmyelinated C fibers, thinly myelinated Aδ fibers and Aβ-Field LTMR (9.2%). Gene expression profiles of retrograde labeled DRG neurons over multiple timepoints following experimental stress fracture revealed dynamic changes in gene expression corresponding to the acute inflammatory ( S100a8 , S100a9 ) and mechanical force ( Piezo2 ). Reparative phase after fracture included morphogens such as Tgfb1, Fgf9 and Fgf18 . Two methods to surgically or genetically denervate fractured bones were used in combination with scRNA-seq to implicate defective mesenchymal cell proliferation and osteodifferentiation as underlying the poor bone repair capacity in the presence of attenuated innervation. Finally, multi-tissue scRNA-seq and interactome analyses implicated neuron-derived FGF9 as a potent regulator of fracture repair, a finding compatible with in vitro assessments of neuron-to-skeletal mesenchyme interactions.
Collapse
|
4
|
Martellucci S, Flütsch A, Carter M, Norimoto M, Pizzo D, Mantuano E, Sadri M, Wang Z, Chillin-Fuentes D, Rosenthal SB, Azmoon P, Gonias SL, Campana WM. Axon-derived PACSIN1 binds to the Schwann cell survival receptor, LRP1, and transactivates TrkC to promote gliatrophic activities. Glia 2024; 72:916-937. [PMID: 38372375 DOI: 10.1002/glia.24510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 01/12/2024] [Accepted: 01/19/2024] [Indexed: 02/20/2024]
Abstract
Schwann cells (SCs) undergo phenotypic transformation and then orchestrate nerve repair following PNS injury. The ligands and receptors that activate and sustain SC transformation remain incompletely understood. Proteins released by injured axons represent important candidates for activating the SC Repair Program. The low-density lipoprotein receptor-related protein-1 (LRP1) is acutely up-regulated in SCs in response to injury, activating c-Jun, and promoting SC survival. To identify novel LRP1 ligands released in PNS injury, we applied a discovery-based approach in which extracellular proteins in the injured nerve were captured using Fc-fusion proteins containing the ligand-binding motifs of LRP1 (CCR2 and CCR4). An intracellular neuron-specific protein, Protein Kinase C and Casein Kinase Substrate in Neurons (PACSIN1) was identified and validated as an LRP1 ligand. Recombinant PACSIN1 activated c-Jun and ERK1/2 in cultured SCs. Silencing Lrp1 or inhibiting the LRP1 cell-signaling co-receptor, the NMDA-R, blocked the effects of PACSIN1 on c-Jun and ERK1/2 phosphorylation. Intraneural injection of PACSIN1 into crush-injured sciatic nerves activated c-Jun in wild-type mice, but not in mice in which Lrp1 is conditionally deleted in SCs. Transcriptome profiling of SCs revealed that PACSIN1 mediates gene expression events consistent with transformation to the repair phenotype. PACSIN1 promoted SC migration and viability following the TNFα challenge. When Src family kinases were pharmacologically inhibited or the receptor tyrosine kinase, TrkC, was genetically silenced or pharmacologically inhibited, PACSIN1 failed to induce cell signaling and prevent SC death. Collectively, these studies demonstrate that PACSIN1 is a novel axon-derived LRP1 ligand that activates SC repair signaling by transactivating TrkC.
Collapse
Affiliation(s)
- Stefano Martellucci
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| | - Andreas Flütsch
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| | - Mark Carter
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| | - Masaki Norimoto
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| | - Donald Pizzo
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Elisabetta Mantuano
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Mahrou Sadri
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| | - Zixuan Wang
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| | - Daisy Chillin-Fuentes
- Center for Computational Biology & Bioinformatics, Altman Clinical & Translational Research Institute, University of California San Diego, La Jolla, California, USA
| | - Sara Brin Rosenthal
- Center for Computational Biology & Bioinformatics, Altman Clinical & Translational Research Institute, University of California San Diego, La Jolla, California, USA
| | - Pardis Azmoon
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Steven L Gonias
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Wendy M Campana
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
- Program in Neurosciences, University of California San Diego, La Jolla, California, USA
- Division of Research, San Diego VA Health Care System, San Diego, California, USA
| |
Collapse
|
5
|
Mirzahosseini G, Adam JM, Nasoohi S, El-Remessy AB, Ishrat T. Lost in Translation: Neurotrophins Biology and Function in the Neurovascular Unit. Neuroscientist 2023; 29:694-714. [PMID: 35769016 DOI: 10.1177/10738584221104982] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The neurovascular unit (NVU) refers to the functional building unit of the brain and the retina, where neurons, glia, and microvasculature orchestrate to meet the demand of the retina's and brain's function. Neurotrophins (NTs) are structural families of secreted proteins and are known for exerting neurotrophic effects on neuronal differentiation, survival, neurite outgrowth, synaptic formation, and plasticity. NTs include several molecules, such as nerve growth factor, brain-derived neurotrophic factor, NT-3, NT-4, and their precursors. Furthermore, NTs are involved in signaling pathways such as inflammation, apoptosis, and angiogenesis in a nonneuronal cell type. Interestingly, NTs and the precursors can bind and activate the p75 neurotrophin receptor (p75NTR) at low and high affinity. Mature NTs bind their cognate tropomyosin/tyrosine-regulated kinase receptors, crucial for maintenance and neuronal development in the brain and retina axis. Activation of p75NTR results in neuronal apoptosis and cell death, while tropomysin receptor kinase upregulation contributes to differentiation and cell growth. Recent findings indicate that modulation of NTs and their receptors contribute to neurovascular dysfunction in the NVU. Several chronic metabolic and acute ischemic diseases affect the NVU, including diabetic and ischemic retinopathy for the retina, as well as stroke, acute encephalitis, and traumatic brain injury for the brain. This work aims to review the current evidence through published literature studying the impact of NTs and their receptors, including the p75NTR receptor, on the injured and healthy brain-retina axis.
Collapse
Affiliation(s)
- Golnoush Mirzahosseini
- Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Justin Mark Adam
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sanaz Nasoohi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
6
|
Fading memories in aging and neurodegeneration: Is p75 neurotrophin receptor a culprit? Ageing Res Rev 2022; 75:101567. [PMID: 35051645 DOI: 10.1016/j.arr.2022.101567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/12/2021] [Accepted: 01/12/2022] [Indexed: 11/22/2022]
Abstract
Aging and age-related neurodegenerative diseases have become one of the major concerns in modern times as cognitive abilities tend to decline when we get older. It is well known that the main cause of this age-related cognitive deficit is due to aberrant changes in cellular, molecular circuitry and signaling pathways underlying synaptic plasticity and neuronal connections. The p75 neurotrophin receptor (p75NTR) is one of the important mediators regulating the fate of the neurons in the nervous system. Its importance in neuronal apoptosis is well documented. However, the mechanisms involving the regulation of p75NTR in synaptic plasticity and cognitive function remain obscure, although cognitive impairment has been associated with a higher expression of p75NTR in neurons. In this review, we discuss the current understanding of how neurons are influenced by p75NTR function to maintain normal neuronal synaptic strength and connectivity, particularly to support learning and memory in the hippocampus. We then discuss the age-associated alterations in neurophysiological mechanisms of synaptic plasticity and cognitive function. Furthermore, we also describe current evidence that has begun to elucidate how p75NTR regulates synaptic changes in aging and age-related neurodegenerative diseases, focusing on the hippocampus. Elucidating the role that p75NTR signaling plays in regulating synaptic plasticity will contribute to a better understanding of cognitive processes and pathological conditions. This will in turn provide novel approaches to improve therapies for the treatment of neurological diseases in which p75NTR dysfunction has been demonstrated.
Collapse
|
7
|
Vidal A, Redmer T. Tracking of Melanoma Cell Plasticity by Transcriptional Reporters. Int J Mol Sci 2022; 23:1199. [PMID: 35163127 PMCID: PMC8835814 DOI: 10.3390/ijms23031199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/12/2022] [Accepted: 01/18/2022] [Indexed: 02/05/2023] Open
Abstract
Clonal evolution and cellular plasticity are the genetic and non-genetic driving forces of tumor heterogeneity, which in turn determine tumor cell responses towards therapeutic drugs. Several lines of evidence suggest that therapeutic interventions foster the selection of drug-resistant neural crest stem-like cells (NCSCs) that establish minimal residual disease (MRD) in melanoma. Here, we establish a dual-reporter system, enabling the tracking of NGFR expression and mRNA stability and providing insights into the maintenance of NCSC states. We observed that a transcriptional reporter that contained a 1-kilobase fragment of the human NGFR promoter was activated only in a minor subset (0.72 ± 0.49%, range 0.3-1.5), and ~2-4% of A375 melanoma cells revealed stable NGFR mRNA. The combination of both reporters provides insights into phenotype switching and reveals that both cellular subsets gave rise to cellular heterogeneity. Moreover, whole transcriptome profiling and gene-set enrichment analysis (GSEA) of the minor cellular subset revealed hypoxia-associated programs that might serve as potential drivers of an in vitro switching of NGFR-associated phenotypes and relapse of post-BRAF inhibitor-treated tumors. Concordantly, we observed that the minor cellular subset increased in response to dabrafenib over time. In summary, our reporter-based approach provides insights into plasticity and identified a cellular subset that might be responsible for the establishment of MRD in melanoma.
Collapse
Affiliation(s)
- Anna Vidal
- Institute of Medical Biochemistry, University of Veterinary Medicine, 1210 Vienna, Austria;
| | - Torben Redmer
- Institute of Medical Biochemistry, University of Veterinary Medicine, 1210 Vienna, Austria;
- Unit of Laboratory Animal Pathology, Institute of Pathology, University of Veterinary Medicine, 1210 Vienna, Austria
| |
Collapse
|
8
|
P75 neurotrophin receptor controls subventricular zone neural stem cell migration after stroke. Cell Tissue Res 2021; 387:415-431. [PMID: 34698916 PMCID: PMC8975773 DOI: 10.1007/s00441-021-03539-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/04/2021] [Indexed: 12/23/2022]
Abstract
Stroke is the leading cause of adult disability. Endogenous neural stem/progenitor cells (NSPCs) originating from the subventricular zone (SVZ) contribute to the brain repair process. However, molecular mechanisms underlying CNS disease-induced SVZ NSPC-redirected migration to the lesion area are poorly understood. Here, we show that genetic depletion of the p75 neurotrophin receptor (p75NTR−/−) in mice reduced SVZ NSPC migration towards the lesion area after cortical injury and that p75NTR−/− NSPCs failed to migrate upon BDNF stimulation in vitro. Cortical injury rapidly increased p75NTR abundance in SVZ NSPCs via bone morphogenetic protein (BMP) receptor signaling. SVZ-derived p75NTR−/− NSPCs revealed an altered cytoskeletal network- and small GTPase family-related gene and protein expression. In accordance, BMP-treated non-migrating p75NTR−/− NSPCs revealed an altered morphology and α-tubulin expression compared to BMP-treated migrating wild-type NSPCs. We propose that BMP-induced p75NTR abundance in NSPCs is a regulator of SVZ NSPC migration to the lesion area via regulation of the cytoskeleton following cortical injury.
Collapse
|
9
|
Uddin MS, Mamun AA, Rahman MM, Jeandet P, Alexiou A, Behl T, Sarwar MS, Sobarzo-Sánchez E, Ashraf GM, Sayed AA, Albadrani GM, Peluso I, Abdel-Daim MM. Natural Products for Neurodegeneration: Regulating Neurotrophic Signals. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8820406. [PMID: 34239696 PMCID: PMC8241508 DOI: 10.1155/2021/8820406] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 05/20/2021] [Indexed: 12/12/2022]
Abstract
Neurodegenerative disorders (NDs) are heterogeneous groups of ailments typically characterized by progressive damage of the nervous system. Several drugs are used to treat NDs but they have only symptomatic benefits with various side effects. Numerous researches have been performed to prove the advantages of phytochemicals for the treatment of NDs. Furthermore, phytochemicals such as polyphenols might play a pivotal role in rescue from neurodegeneration due to their various effects as anti-inflammatory, antioxidative, and antiamyloidogenic agents by controlling apoptotic factors, neurotrophic factors (NTFs), free radical scavenging system, and mitochondrial stress. On the other hand, neurotrophins (NTs) including nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), NT4/5, and NT3 might have a crucial neuroprotective role, and their diminution triggers the development of the NDs. Polyphenols can interfere directly with intracellular signaling molecules to alter brain activity. Several natural products also improve the biosynthesis of endogenous genes encoding antiapoptotic Bcl-2 as well as NTFs such as glial cell and brain-derived NTFs. Various epidemiological studies have demonstrated that the initiation of these genes could play an essential role in the neuroprotective function of dietary compounds. Hence, targeting NTs might represent a promising approach for the management of NDs. In this review, we focus on the natural product-mediated neurotrophic signal-modulating cascades, which are involved in the neuroprotective effects.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Abdullah Al Mamun
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong
| | - Md Motiar Rahman
- Laboratory of Clinical Biochemistry and Nutritional Sciences (LCBNS), Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Philippe Jeandet
- Research Unit, Induced Resistance and Plant Bioprotection, USC INRAe 1488, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, P.O. Box 1039, CEDEX 2, 51687 Reims, France
| | - Athanasios Alexiou
- Novel Global Community Educational Foundation, 2770 Hebersham, Australia
- AFNP Med Austria, 1010 Wien, Austria
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Md Shahid Sarwar
- Department of Pharmacy, Noakhali Science and Technology University, Sonapur, Noakhali 3814, Bangladesh
| | - Eduardo Sobarzo-Sánchez
- Instituto de Investigación y Postgrado, Facultad de Ciencias de la Salud, Universidad Central de Chile, 8330507 Santiago, Chile
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amany A. Sayed
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Ghadeer M. Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| | - Ilaria Peluso
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), 00142 Rome, Italy
| | - Mohamed M. Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
10
|
Bao X, Song Y, Li T, Zhang S, Huang L, Zhang S, Cao J, Liu X, Zhang J. Comparative Transcriptome Profiling of Ovary Tissue between Black Muscovy Duck and White Muscovy Duck with High- and Low-Egg Production. Genes (Basel) 2020; 12:57. [PMID: 33396489 PMCID: PMC7824526 DOI: 10.3390/genes12010057] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 12/19/2022] Open
Abstract
The egg-laying rate is an important indicator for evaluating fertility of poultry. In order to better understand the laying mechanism of Muscovy ducks, gene expression profiles and pathways of ovarian tissues in high- and low-laying black (BH and BL) and white Muscovy ducks (WH and WL) during the peak production period were performed by using RNA-seq. The total number of reads produced for each ovarian sample ranged from 44,344,070 to 47,963,328. A total of 113, 619 and 87 differentially expressed genes (DEGs) were identified in BH-vs-WH, BL-vs-BH and BL-vs-WL, respectively. Among them, 54, 356 and 49 genes were up regulated and 59, 263 and 38 genes were down regulated. In addition, there were only 10 up-regulated genes in WL-vs-WH. In the comparison of DEGs in black and white Muscovy ducks, two co-expressed DEG genes were detected between BH-vs-WH and BL-vs-WL and seven DEGs were co-expressed between BL-vs-BH and WL-vs-WH. The RNA-Seq data were confirmed to be reliable by qPCR. Numerous DEGs known to be involved in ovarian development were identified, including TGFβ2, NGFR, CEBPD, CPEB2, POSTN, SMOC1, FGF18, EFNA5 and SDC4. Gene Ontology (GO) annotations indicated that DEGs related to ovarian development were mainly enriched in biological processes of "circadian sleep/wake cycle process," "negative regulation of transforming growth factor-β secretion," "positive regulation of calcium ion transport" in BH-vs-WH and "cell surface receptor signaling pathway," "Notch signaling pathway" and "calcium ion transport" in BL-vs-BH. Besides, "steroid biosynthetic process," "granulosa cell development" and "egg coat formation" were mainly enriched in BL-vs-WL and "reproduction," "MAPK cascade" and "mitotic cell cycle" were mainly enriched in WL-vs-WH. KEGG pathway analysis showed that the PI3K-Akt signaling pathway and ovarian steroidogenesis were the most enriched in Muscovy duck ovary transcriptome data. This work highlights potential genes and pathways that may affect ovarian development in Muscovy duck.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jianqin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China; (X.B.); (Y.S.); (T.L.); (S.Z.); (L.H.); (S.Z.); (J.C.); (X.L.)
| |
Collapse
|
11
|
Tsybko AS, Ilchibaeva TV, Filimonova EA, Eremin DV, Popova NK, Naumenko VS. The Chronic Treatment With 5-HT 2A Receptor Agonists Affects the Behavior and the BDNF System in Mice. Neurochem Res 2020; 45:3059-3075. [PMID: 33095437 DOI: 10.1007/s11064-020-03153-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/13/2020] [Accepted: 10/15/2020] [Indexed: 01/09/2023]
Abstract
Serotonin 5-HT2A receptors and the brain-derived neurotrophic factor (BDNF) are involved in the pathophysiology and treatment of many psychiatric diseases. However, the interaction between 5-HT2A and BDNF is still poorly understood. In the present paper, the effects of chronic treatment with mixed 5-HT2A/2C receptor agonist DOI, highly selective 5-HT2A agonists TCB-2 and 25CN-NBOH on behavior and the BDNF system have been investigated. Chronic treatment of males of C57Bl/6 mice with DOI, TCB-2 and 25CN-NBOH (1 mg/kg, i.p., 14 days) resulted in desensitization of 5-HT2A receptors. Treatment with 25CN-NBOH significantly increased startle amplitude. At the same time all used drugs failed to affect anxiety, exploratory and stereotyped behavior as well as spatial memory and learning. TCB-2 and 25CN-NBOH increased the BDNF mRNA level. All 5-HT2A agonists increased the proBDNF level but failed to alter the mature BDNF protein level. TrkB and p75NTR mRNA levels were affected by all utilized agonists. All drugs decreased the total level as well as membrane TrkB protein one indicating downregulation of TrkB receptors. All agonists decreased the membrane p75NTR protein level. Thus, we have shown for the first time that the chronic activation of the 5-HT2A receptor with agonists has affected the BDNF system almost on all levels-transcription, proBDNF production, TrkB and p75NTR receptors' level. The obtained data suggested possible suppression in BDNF-TrkB signaling under chronic treatment with 5-HT2A agonists.
Collapse
Affiliation(s)
- Anton S Tsybko
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of Russian Academy of Sciences, Prospekt Lavrentyeva, 10, 630090, Novosibirsk, Russia.
| | - Tatiana V Ilchibaeva
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of Russian Academy of Sciences, Prospekt Lavrentyeva, 10, 630090, Novosibirsk, Russia
| | - Elena A Filimonova
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of Russian Academy of Sciences, Prospekt Lavrentyeva, 10, 630090, Novosibirsk, Russia
| | - Dmitry V Eremin
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of Russian Academy of Sciences, Prospekt Lavrentyeva, 10, 630090, Novosibirsk, Russia
| | - Nina K Popova
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of Russian Academy of Sciences, Prospekt Lavrentyeva, 10, 630090, Novosibirsk, Russia
| | - Vladimir S Naumenko
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of Russian Academy of Sciences, Prospekt Lavrentyeva, 10, 630090, Novosibirsk, Russia
| |
Collapse
|
12
|
Nie F, Zhang Q, Ma J, Wang P, Gu R, Han J, Zhang R. Schizophrenia risk candidate EGR3 is a novel transcriptional regulator of RELN and regulates neurite outgrowth via the Reelin signal pathway in vitro. J Neurochem 2020; 157:1745-1758. [PMID: 33113163 DOI: 10.1111/jnc.15225] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/06/2020] [Accepted: 10/15/2020] [Indexed: 01/09/2023]
Abstract
Schizophrenia is a severe psychiatric disorder with a strong hereditary component that affects approximately 1% of the world's population. The disease is most likely caused by the altered expression of a number of genes that function at the level of biological pathways or gene networks. Transcription factors (TF) are indispensable regulators of gene expression. EGR3 is a TF associated with schizophrenia. In the current study, DNA microarray and ingenuity pathway analyses (IPA) demonstrated that EGR3 regulates Reelin signaling pathway in SH-SY5Y cells. ChIP and luciferase reporter studies confirmed that EGR3 directly binds to the promoter region of RELN thereby activating RELN expression. The expression of both EGR3 and RELN was decreased during neuronal differentiation induced by retinoic acid (RA) in SH-SY5Y cells, and EGR3 over-expression reduced neurite outgrowth which could be partially reversed by the knockdown of RELN. The expression levels of EGR3 and RELN in peripheral blood of subjects with schizophrenia were found to be down-regulated (compared with healthy controls), and were positively correlated. Furthermore, data mining from public databases revealed that the expression levels of EGR3 and RELN were presented a positive correlation in post-mortem brain tissue of subjects with schizophrenia. Taken together, this study suggests that EGR3 is a novel TF of the RELN gene and regulates neurite outgrowth via the Reelin signaling pathway. Our findings contribute to the understanding of the regulatory role of EGR3 in the pathophysiology and molecular mechanisms of schizophrenia, and potentially to the development of new therapies and diagnostic biomarkers for the disorder.
Collapse
Affiliation(s)
- Fayi Nie
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Qiaoxia Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Jie Ma
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.,Medical Research Center, Xi'an No. 3 Hospital, Xi'an, Shaanxi, China
| | - Pengjie Wang
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Ruiying Gu
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Jing Han
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Rui Zhang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
13
|
Baho E, Chattopadhyaya B, Lavertu-Jolin M, Mazziotti R, Awad PN, Chehrazi P, Groleau M, Jahannault-Talignani C, Vaucher E, Ango F, Pizzorusso T, Baroncelli L, Di Cristo G. p75 Neurotrophin Receptor Activation Regulates the Timing of the Maturation of Cortical Parvalbumin Interneuron Connectivity and Promotes Juvenile-like Plasticity in Adult Visual Cortex. J Neurosci 2019; 39:4489-4510. [PMID: 30936240 PMCID: PMC6554620 DOI: 10.1523/jneurosci.2881-18.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/22/2019] [Accepted: 03/14/2019] [Indexed: 01/09/2023] Open
Abstract
By virtue of their extensive axonal arborization and perisomatic synaptic targeting, cortical inhibitory parvalbumin (PV) cells strongly regulate principal cell output and plasticity and modulate experience-dependent refinement of cortical circuits during development. An interesting aspect of PV cell connectivity is its prolonged maturation time course, which is completed only by end of adolescence. The p75 neurotrophin receptor (p75NTR) regulates numerous cellular functions; however, its role on cortical circuit development and plasticity remains elusive, mainly because localizing p75NTR expression with cellular and temporal resolution has been challenging. By using RNAscope and a modified version of the proximity ligation assay, we found that p75NTR expression in PV cells decreases between the second and fourth postnatal week, at a time when PV cell synapse numbers increase dramatically. Conditional knockout of p75NTR in single PV neurons in vitro and in PV cell networks in vivo causes precocious formation of PV cell perisomatic innervation and perineural nets around PV cell somata, therefore suggesting that p75NTR expression modulates the timing of maturation of PV cell connectivity in the adolescent cortex. Remarkably, we found that PV cells still express p75NTR in adult mouse cortex of both sexes and that its activation is sufficient to destabilize PV cell connectivity and to restore cortical plasticity following monocular deprivation in vivo Together, our results show that p75NTR activation dynamically regulates PV cell connectivity, and represent a novel tool to foster brain plasticity in adults.SIGNIFICANCE STATEMENT In the cortex, inhibitory, GABA-releasing neurons control the output and plasticity of excitatory neurons. Within this diverse group, parvalbumin-expressing (PV) cells form the larger inhibitory system. PV cell connectivity develops slowly, reaching maturity only at the end of adolescence; however, the mechanisms controlling the timing of its maturation are not well understood. We discovered that the expression of the neurotrophin receptor p75NTR in PV cells inhibits the maturation of their connectivity in a cell-autonomous fashion, both in vitro and in vivo, and that p75NTR activation in adult PV cells promotes their remodeling and restores cortical plasticity. These results reveal a new p75NTR function in the regulation of the time course of PV cell maturation and in limiting cortical plasticity.
Collapse
Affiliation(s)
- Elie Baho
- Department of Neurosciences, Université de Montréal, Montréal, Québec H3T 1J4, Canada
- Centre de Recherche, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Québec H3T 1C5, Canada
| | - Bidisha Chattopadhyaya
- Department of Neurosciences, Université de Montréal, Montréal, Québec H3T 1J4, Canada
- Centre de Recherche, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Québec H3T 1C5, Canada
| | - Marisol Lavertu-Jolin
- Department of Neurosciences, Université de Montréal, Montréal, Québec H3T 1J4, Canada
- Centre de Recherche, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Québec H3T 1C5, Canada
| | - Raffaele Mazziotti
- Institute of Neuroscience Consiglio Nazionale delle Ricerche, 56124 Pisa, Italy
| | - Patricia N Awad
- Department of Neurosciences, Université de Montréal, Montréal, Québec H3T 1J4, Canada
- Centre de Recherche, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Québec H3T 1C5, Canada
| | - Pegah Chehrazi
- Department of Neurosciences, Université de Montréal, Montréal, Québec H3T 1J4, Canada
- Centre de Recherche, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Québec H3T 1C5, Canada
| | - Marianne Groleau
- École d'Optométrie, Université de Montréal, Montréal, Québec H3T 1P1, Canada
| | - Celine Jahannault-Talignani
- Institut de Génomique Fonctionnelle, université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé Et de la Recherche Médicale, 34090 Montpellier, France, and
| | - Elvire Vaucher
- École d'Optométrie, Université de Montréal, Montréal, Québec H3T 1P1, Canada
| | - Fabrice Ango
- Institut de Génomique Fonctionnelle, université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé Et de la Recherche Médicale, 34090 Montpellier, France, and
| | - Tommaso Pizzorusso
- Institute of Neuroscience Consiglio Nazionale delle Ricerche, 56124 Pisa, Italy
- Department of Neuroscience, Psychology, Drug Research and Child Health Neurofarba, University of Florence, 50139 Firenze, Italy
| | - Laura Baroncelli
- Institute of Neuroscience Consiglio Nazionale delle Ricerche, 56124 Pisa, Italy
| | - Graziella Di Cristo
- Department of Neurosciences, Université de Montréal, Montréal, Québec H3T 1J4, Canada,
- Centre de Recherche, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Québec H3T 1C5, Canada
| |
Collapse
|
14
|
A Role for Sensory end Organ-Derived Signals in Regulating Muscle Spindle Proprioceptor Phenotype. J Neurosci 2019; 39:4252-4267. [PMID: 30926747 DOI: 10.1523/jneurosci.2671-18.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/21/2019] [Accepted: 03/21/2019] [Indexed: 01/09/2023] Open
Abstract
Proprioceptive feedback from Group Ia/II muscle spindle afferents and Group Ib Golgi tendon afferents is critical for the normal execution of most motor tasks, yet how these distinct proprioceptor subtypes emerge during development remains poorly understood. Using molecular genetic approaches in mice of either sex, we identified 24 transcripts that have not previously been associated with a proprioceptor identity. Combinatorial expression analyses of these markers reveal at least three molecularly distinct proprioceptor subtypes. In addition, we find that 12 of these transcripts are expressed well after proprioceptors innervate their respective sensory receptors, and expression of three of these markers, including the heart development molecule Heg1, is significantly reduced in mice that lack muscle spindles. These data reveal Heg1 as a putative marker for proprioceptive muscle spindle afferents. Moreover, they suggest that the phenotypic specialization of functionally distinct proprioceptor subtypes depends, in part, on extrinsic sensory receptor organ-derived signals.SIGNIFICANCE STATEMENT Sensory feedback from muscle spindle (MS) and Golgi tendon organ (GTO) sensory end organs is critical for normal motor control, but how distinct MS and GTO afferent sensory neurons emerge during development remains poorly understood. Using (bulk) transcriptome analysis of genetically identified proprioceptors, this work reveals molecular markers for distinct proprioceptor subsets, including some that appear selectively expressed in MS afferents. Detailed analysis of the expression of these transcripts provides evidence that MS/GTO afferent subtype phenotypes may, at least in part, emerge through extrinsic, sensory end organ-derived signals.
Collapse
|
15
|
|
16
|
Sultan N, Amin LE, Zaher AR, Scheven BA, Grawish ME. Dental pulp stem cells: Novel cell-based and cell-free therapy for peripheral nerve repair. World J Stomatol 2019; 7:1-19. [DOI: 10.5321/wjs.v7.i1.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/15/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023] Open
Abstract
The regeneration of peripheral nerves comprises complicated steps involving a set of cellular and molecular events in distal nerve stumps with axonal sprouting and remyelination. Stem cell isolation and expansion for peripheral nerve repair (PNR) can be achieved using a wide diversity of prenatal and adult tissues, such as bone marrow or brain tissues. The ability to obtain stem cells for cell-based therapy (CBT) is limited due to donor site morbidity and the invasive nature of the harvesting process. Dental pulp stem cells (DPSCs) can be relatively and simply isolated from the dental pulps of permanent teeth, extracted for surgical or orthodontic reasons. DPSCs are of neural crest origin with an outstanding ability to differentiate into multiple cell lineages. They have better potential to differentiate into neural and glial cells than other stem cell sources through the expression and secretion of certain markers and a range of neurotropic factors; thus, they should be considered a good choice for PNR using CBT. In addition, these cells have paracrine effects through the secretion of neurotrophic growth factors and extracellular vesicles, which can enhance axonal growth and remyelination by decreasing the number of dying cells and activating local inhabitant stem cell populations, thereby revitalizing dormant or blocked cells, modulating the immune system and regulating inflammatory responses. The use of DPSC-derived secretomes holds great promise for controllable and manageable therapy for peripheral nerve injury. In this review, up-to-date information about the neurotrophic and neurogenic properties of DPSCs and their secretomes is provided.
Collapse
Affiliation(s)
- Nessma Sultan
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura 35516, Egypt
| | - Laila E Amin
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed R Zaher
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura 35516, Egypt
| | - Ben A Scheven
- School of Dentistry, Oral Biology, College of Medical and Dental Sciences, University of Birmingham, Birmingham B5 7EG, United Kingdom
| | - Mohammed E Grawish
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
17
|
Numakawa T, Odaka H, Adachi N, Chiba S, Ooshima Y, Matsuno H, Nakajima S, Yoshimura A, Fumimoto K, Hirai Y, Kunugi H. Basic fibroblast growth factor increased glucocorticoid receptors in cortical neurons through MAP kinase pathway. Neurochem Int 2018; 118:217-224. [PMID: 29958871 DOI: 10.1016/j.neuint.2018.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/30/2018] [Accepted: 06/25/2018] [Indexed: 01/09/2023]
Abstract
Prolonged and intense stress chronically increases blood concentration of glucocorticoids, which in turn causes downregulation of glucocorticoid receptor (GR) in the central nervous system (CNS). This process has been suggested to be involved in the pathogenesis of major depressive disorder (MDD). Here, we found that basic fibroblast growth factor (bFGF) increased the expression of GR in the rat cerebral cortex and cultured cortical neurons and restored the reduced GR expression caused by glucocorticoid exposure. Among intracellular signaling pathways stimulated by bFGF, extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) pathway was responsible for the upregulation of GR. The bFGF-induced GR was functional as a transcription factor to enhance transcription of a target gene. Because high stress augments bFGF levels in the brain, it is likely that bFGF plays a compensating role for reduced GR expression after stress and thus should be studied as a therapeutic target for the treatment of MDD.
Collapse
Affiliation(s)
- Tadahiro Numakawa
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Haruki Odaka
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan; Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Naoki Adachi
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Gakuen 2-1, Sanda City, Hyogo, 669-1337, Japan.
| | - Shuichi Chiba
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Yoshiko Ooshima
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Administrative Section of Radiation Protection, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Hitomi Matsuno
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Shingo Nakajima
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Endowed Research Division of Human Welfare Sciences, Ochanomizu University, Tokyo, Japan
| | - Aya Yoshimura
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Education and Research Facility of Animal Models for Human Diseases, Center for Research Promotion and Support, Fujita Health University, Aichi, Japan
| | - Kazuhiro Fumimoto
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Gakuen 2-1, Sanda City, Hyogo, 669-1337, Japan
| | - Yohei Hirai
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Gakuen 2-1, Sanda City, Hyogo, 669-1337, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan
| |
Collapse
|
18
|
Pfaffenseller B, Kapczinski F, Gallitano AL, Klamt F. EGR3 Immediate Early Gene and the Brain-Derived Neurotrophic Factor in Bipolar Disorder. Front Behav Neurosci 2018; 12:15. [PMID: 29459824 PMCID: PMC5807664 DOI: 10.3389/fnbeh.2018.00015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 01/17/2018] [Indexed: 01/09/2023] Open
Abstract
Bipolar disorder (BD) is a severe psychiatric illness with a consistent genetic influence, involving complex interactions between numerous genes and environmental factors. Immediate early genes (IEGs) are activated in the brain in response to environmental stimuli, such as stress. The potential to translate environmental stimuli into long-term changes in brain has led to increased interest in a potential role for these genes influencing risk for psychiatric disorders. Our recent finding using network-based approach has shown that the regulatory unit of early growth response gene 3 (EGR3) of IEGs family was robustly repressed in postmortem prefrontal cortex of BD patients. As a central transcription factor, EGR3 regulates an array of target genes that mediate critical neurobiological processes such as synaptic plasticity, memory and cognition. Considering that EGR3 expression is induced by brain-derived neurotrophic factor (BDNF) that has been consistently related to BD pathophysiology, we suggest a link between BDNF and EGR3 and their potential role in BD. A growing body of data from our group and others has shown that peripheral BDNF levels are reduced during mood episodes and also with illness progression. In this same vein, BDNF has been proposed as an important growth factor in the impaired cellular resilience related to BD. Taken together with the fact that EGR3 regulates the expression of the neurotrophin receptor p75NTR and may also indirectly induce BDNF expression, here we propose a feed-forward gene regulatory network involving EGR3 and BDNF and its potential role in BD.
Collapse
Affiliation(s)
- Bianca Pfaffenseller
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Flavio Kapczinski
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Amelia L Gallitano
- Department of Basic Medical Sciences, College of Medicine, University of Arizona, Phoenix, AZ, United States
| | - Fábio Klamt
- Laboratory of Cellular Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
19
|
Boerboom A, Reusch C, Pieltain A, Chariot A, Franzen R. KIAA1199: A novel regulator of MEK/ERK-induced Schwann cell dedifferentiation. Glia 2017; 65:1682-1696. [PMID: 28699206 DOI: 10.1002/glia.23188] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 12/14/2022]
Abstract
The molecular mechanisms that regulate Schwann cell (SC) plasticity and the role of the Nrg1/ErbB-induced MEK1/ERK1/2 signalling pathway in SC dedifferentiation or in myelination remain unclear. It is currently believed that different levels of MEK1/ERK1/2 activation define the state of SC differentiation. Thus, the identification of new regulators of MEK1/ERK1/2 signalling could help to decipher the context-specific aspects driving the effects of this pathway on SC plasticity. In this perspective, we have investigated the potential role of KIAA1199, a protein that promotes ErbB and MEK1/ERK1/2 signalling in cancer cells, in SC plasticity. We depleted KIAA1199 in the SC-derived MSC80 cell line with RNA-interference-based strategy and also generated Tamoxifen-inducible and conditional mouse models in which KIAA1199 is inactivated through homologous recombination, using the Cre-lox technology. We show that the invalidation of KIAA1199 in SC decreases the expression of cJun and other negative regulators of myelination and elevates Krox20, driving them towards a pro-myelinating phenotype. We further show that in dedifferentiation conditions, SC invalidated for KIAA1199 exhibit lower myelin clearance as well as increased myelination capacity. Finally, the Nrg1-induced activation of the MEK/ERK/1/2 pathway is severely reduced when KIAA1199 is absent, indicating that KIAA1199 promotes Nrg1-dependent MEK1 and ERK1/2 activation in SCs. In conclusion, this work identifies KIAA1199 as a novel regulator of MEK/ERK-induced SC dedifferentiation and contributes to a better understanding of the molecular control of SC dedifferentiation.
Collapse
Affiliation(s)
| | - Céline Reusch
- GIGA-Molecular Biology of Diseases, University of Liège, Belgium
| | | | - Alain Chariot
- GIGA-Molecular Biology of Diseases, University of Liège, Belgium.,Walloon Excellence in Lifesciences and Biotechnology (WELBIO), Wavre, Belgium
| | | |
Collapse
|
20
|
Boerboom A, Dion V, Chariot A, Franzen R. Molecular Mechanisms Involved in Schwann Cell Plasticity. Front Mol Neurosci 2017; 10:38. [PMID: 28261057 PMCID: PMC5314106 DOI: 10.3389/fnmol.2017.00038] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 01/31/2017] [Indexed: 01/09/2023] Open
Abstract
Schwann cell incredible plasticity is a hallmark of the utmost importance following nerve damage or in demyelinating neuropathies. After injury, Schwann cells undergo dedifferentiation before redifferentiating to promote nerve regeneration and complete functional recovery. This review updates and discusses the molecular mechanisms involved in the negative regulation of myelination as well as in the reprogramming of Schwann cells taking place early following nerve lesion to support repair. Significant advance has been made on signaling pathways and molecular components that regulate SC regenerative properties. These include for instance transcriptional regulators such as c-Jun or Notch, the MAPK and the Nrg1/ErbB2/3 pathways. This comprehensive overview ends with some therapeutical applications targeting factors that control Schwann cell plasticity and highlights the need to carefully modulate and balance this capacity to drive nerve repair.
Collapse
Affiliation(s)
| | - Valérie Dion
- GIGA-Neurosciences, University of Liège Liège, Belgium
| | - Alain Chariot
- GIGA-Molecular Biology of Diseases, University of LiègeLiège, Belgium; Walloon Excellence in Lifesciences and Biotechnology (WELBIO)Wavre, Belgium
| | | |
Collapse
|
21
|
Aghdaei FH, Soltani BM, Dokanehiifard S, Mowla SJ, Soleimani M. Overexpression of hsa-miR-939 follows by NGFR down-regulation and apoptosis reduction. J Biosci 2017; 42:23-30. [DOI: 10.1007/s12038-017-9669-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
22
|
Chakravarthy R, Mnich K, Gorman AM. Nerve growth factor (NGF)-mediated regulation of p75(NTR) expression contributes to chemotherapeutic resistance in triple negative breast cancer cells. Biochem Biophys Res Commun 2016; 478:1541-7. [PMID: 27577679 DOI: 10.1016/j.bbrc.2016.08.149] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 08/26/2016] [Indexed: 01/09/2023]
Abstract
Triple negative breast cancer [TNBC] cells are reported to secrete the neurotrophin nerve growth factor [NGF] and express its receptors, p75 neurotrophin receptor [p75(NTR)] and TrkA, leading to NGF-activated pro-survival autocrine signaling. This provides a rationale for NGF as a potential therapeutic target for TNBC. Here we show that exposure of TNBC cells to NGF leads to increased levels of p75(NTR), which was diminished by NGF-neutralizing antibody or NGF inhibitors [Ro 08-2750 and Y1086]. NGF-mediated increase in p75(NTR) levels were partly due to increased transcription and partly due to inhibition of proteolytic processing of p75(NTR). In contrast, proNGF caused a decrease in p75(NTR) levels. Functionally, NGF-induced increase in p75(NTR) caused a decrease in the sensitivity of TNBC cells to apoptosis induction. In contrast, knock-down of p75(NTR) using shRNA or small molecule inhibition of NGF-p75(NTR) interaction [using Ro 08-2750] sensitized TNBC cells to drug-induced apoptosis. In patient samples, the expression of NGF and NGFR [the p75(NTR) gene] mRNA are positively correlated in several subtypes of breast cancer, including basal-like breast cancer. Together these data suggest a positive feedback loop through which NGF-mediated upregulation of p75(NTR) can contribute to the chemo-resistance of TNBC cells.
Collapse
|
23
|
Pfaffenseller B, da Silva Magalhães PV, De Bastiani MA, Castro MAA, Gallitano AL, Kapczinski F, Klamt F. Differential expression of transcriptional regulatory units in the prefrontal cortex of patients with bipolar disorder: potential role of early growth response gene 3. Transl Psychiatry 2016; 6:e805. [PMID: 27163206 PMCID: PMC5070056 DOI: 10.1038/tp.2016.78] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 03/23/2016] [Indexed: 01/08/2023] Open
Abstract
Bipolar disorder (BD) is a severe mental illness with a strong genetic component. Despite its high degree of heritability, current genetic studies have failed to reveal individual loci of large effect size. In lieu of focusing on individual genes, we investigated regulatory units (regulons) in BD to identify candidate transcription factors (TFs) that regulate large groups of differentially expressed genes. Network-based approaches should elucidate the molecular pathways governing the pathophysiology of BD and reveal targets for potential therapeutic intervention. The data from a large-scale microarray study was used to reconstruct the transcriptional associations in the human prefrontal cortex, and results from two independent microarray data sets to obtain BD gene signatures. The regulatory network was derived by mapping the significant interactions between known TFs and all potential targets. Five regulons were identified in both transcriptional network models: early growth response 3 (EGR3), TSC22 domain family, member 4 (TSC22D4), interleukin enhancer-binding factor 2 (ILF2), Y-box binding protein 1 (YBX1) and MAP-kinase-activating death domain (MADD). With a high stringency threshold, the consensus across tests was achieved only for the EGR3 regulon. We identified EGR3 in the prefrontal cortex as a potential key target, robustly repressed in both BD signatures. Considering that EGR3 translates environmental stimuli into long-term changes in the brain, disruption in biological pathways involving EGR3 may induce an impaired response to stress and influence on risk for psychiatric disorders, particularly BD.
Collapse
Affiliation(s)
- B Pfaffenseller
- Bipolar Disorder Program, Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil,Laboratory of Cellular Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - P V da Silva Magalhães
- Bipolar Disorder Program, Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil,Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil,Department of Psychiatry, Universidade Federal do Rio Grande do Sul, 2350 Ramiro Barcelos Street, Porto Alegre 90035 903, Brazil. E-mail:
| | - M A De Bastiani
- Laboratory of Cellular Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - M A A Castro
- Bioinformatics and Systems Biology Laboratory, Federal University of Paraná, Polytechnic Center, Curitiba, Brazil
| | - A L Gallitano
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - F Kapczinski
- Bipolar Disorder Program, Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil,Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - F Klamt
- Laboratory of Cellular Biochemistry, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
24
|
Balakrishnan A, Stykel MG, Touahri Y, Stratton JA, Biernaskie J, Schuurmans C. Temporal Analysis of Gene Expression in the Murine Schwann Cell Lineage and the Acutely Injured Postnatal Nerve. PLoS One 2016; 11:e0153256. [PMID: 27058953 PMCID: PMC4826002 DOI: 10.1371/journal.pone.0153256] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 03/26/2016] [Indexed: 01/09/2023] Open
Abstract
Schwann cells (SCs) arise from neural crest cells (NCCs) that first give rise to SC precursors (SCPs), followed by immature SCs, pro-myelinating SCs, and finally, non-myelinating or myelinating SCs. After nerve injury, mature SCs ‘de-differentiate’, downregulating their myelination program while transiently re-activating early glial lineage genes. To better understand molecular parallels between developing and de-differentiated SCs, we characterized the expression profiles of a panel of 12 transcription factors from the onset of NCC migration through postnatal stages, as well as after acute nerve injury. Using Sox10 as a pan-glial marker in co-expression studies, the earliest transcription factors expressed in E9.0 Sox10+ NCCs were Sox9, Pax3, AP2α and Nfatc4. E10.5 Sox10+ NCCs coalescing in the dorsal root ganglia differed slightly, expressing Sox9, Pax3, AP2α and Etv5. E12.5 SCPs continued to express Sox10, Sox9, AP2α and Pax3, as well as initiating Sox2 and Egr1 expression. E14.5 immature SCs were similar to SCPs, except that they lost Pax3 expression. By E18.5, AP2α, Sox2 and Egr1 expression was turned off in the nerve, while Jun, Oct6 and Yy1 expression was initiated in pro-myelinating Sox9+/Sox10+ SCs. Early postnatal and adult SCs continued to express Sox9, Jun, Oct6 and Yy1 and initiated Nfatc4 and Egr2 expression. Notably, at all stages, expression of each marker was observed only in a subset of Sox10+ SCs, highlighting the heterogeneity of the SC pool. Following acute nerve injury, Egr1, Jun, Oct6, and Sox2 expression was upregulated, Egr2 expression was downregulated, while Sox9, Yy1, and Nfatc4 expression was maintained at similar frequencies. Notably, de-differentiated SCs in the injured nerve did not display a transcription factor profile corresponding to a specific stage in the SC lineage. Taken together, we demonstrate that uninjured and injured SCs are heterogeneous and distinct from one another, and de-differentiation recapitulates transcriptional aspects of several different embryonic stages.
Collapse
Affiliation(s)
- Anjali Balakrishnan
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Morgan G. Stykel
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, Alberta Children’s Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Yacine Touahri
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, Alberta Children’s Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Jo Anne Stratton
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, Alberta Children’s Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, Alberta Children’s Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Jeff Biernaskie
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, Alberta Children’s Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
- * E-mail: (CS); (JB)
| | - Carol Schuurmans
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
- * E-mail: (CS); (JB)
| |
Collapse
|
25
|
Egr3-dependent muscle spindle stretch receptor intrafusal muscle fiber differentiation and fusimotor innervation homeostasis. J Neurosci 2015; 35:5566-78. [PMID: 25855173 DOI: 10.1523/jneurosci.0241-15.2015] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Muscle stretch proprioceptors (muscle spindles) are required for stretch reflexes and locomotor control. Proprioception abnormalities are observed in many human neuropathies, but the mechanisms involved in establishing and maintaining muscle spindle innervation and function are still poorly understood. During skeletal muscle development, sensory (Ia-afferent) innervation induces contacted myotubes to transform into intrafusal muscle fibers that form the stretch receptor core. The transcriptional regulator Egr3 is induced in Ia-afferent contacted myotubes by Neuregulin1 (Nrg1)/ErbB receptor signaling and it has an essential role in spindle morphogenesis and function. Because Egr3 is widely expressed during development and has a pleiotropic function, whether Egr3 functions primarily in skeletal muscle, Ia-afferent neurons, or in Schwann cells that myelinate Ia-afferent axons remains unresolved. In the present studies, cell-specific ablation of Egr3 in mice showed that it has a skeletal muscle autonomous function in stretch receptor development. Moreover, using genetic tracing, we found that Ia-afferent contacted Egr3-deficient myotubes were induced in normal numbers, but their development was blocked to generate one to two shortened fibers that failed to express some characteristic myosin heavy chain (MyHC) proteins. These "spindle remnants" persisted into adulthood, remained innervated by Ia-afferents, and expressed neurotrophin3 (NT3), which is required for Ia-afferent neuron survival. However, they were not innervated by fusimotor axons and they did not express glial derived neurotrophic factor (GDNF), which is essential for fusimotor neuron survival. These results demonstrate that Egr3 has an essential role in regulating gene expression that promotes normal intrafusal muscle fiber differentiation and fusimotor innervation homeostasis.
Collapse
|
26
|
Schmid D, Zeis T, Sobrio M, Schaeren-Wiemers N. MAL overexpression leads to disturbed expression of genes that influence cytoskeletal organization and differentiation of Schwann cells. ASN Neuro 2014; 6:1759091414548916. [PMID: 25290060 PMCID: PMC4187015 DOI: 10.1177/1759091414548916] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In the developing peripheral nervous system, a coordinated reciprocal signaling between Schwann cells and axons is crucial for accurate myelination. The myelin and lymphocyte protein MAL is a component of lipid rafts that is important for targeting proteins and lipids to distinct domains. MAL overexpression impedes peripheral myelinogenesis, which is evident by a delayed onset of myelination and reduced expression of the myelin protein zero (Mpz/P0) and the low-affinity neurotrophin receptor p75(NTR). This study shows that MAL overexpression leads to a significant reduction of Mpz and p75(NTR) expression in primary mouse Schwann cell cultures, which was already evident before differentiation, implicating an effect of MAL in early Schwann cell development. Their transcription was robustly reduced, despite normal expression of essential transcription factors and receptors. Further, the cAMP response element-binding protein (CREB) and phosphoinositide 3-kinase signaling pathways important for Schwann cell differentiation were correctly induced, highlighting that other so far unknown rate limiting factors do exist. We identified novel genes expressed by Schwann cells in a MAL-dependent manner in vivo and in vitro. A number of those, including S100a4, RhoU and Krt23, are implicated in cytoskeletal organization and plasma membrane dynamics. We showed that S100a4 is predominantly expressed by nonmyelinating Schwann cells, whereas RhoU was localized within myelin membranes, and Krt23 was detected in nonmyelinating as well as in myelinating Schwann cells. Their differential expression during early peripheral nerve development further underlines their possible role in influencing Schwann cell differentiation and myelination.
Collapse
Affiliation(s)
- Daniela Schmid
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland
| | - Thomas Zeis
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland
| | - Monia Sobrio
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland
| | | |
Collapse
|
27
|
Jackson MZ, Gruner KA, Qin C, Tourtellotte WG. A neuron autonomous role for the familial dysautonomia gene ELP1 in sympathetic and sensory target tissue innervation. Development 2014; 141:2452-61. [PMID: 24917501 DOI: 10.1242/dev.107797] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Familial dysautonomia (FD) is characterized by severe and progressive sympathetic and sensory neuron loss caused by a highly conserved germline point mutation of the human ELP1/IKBKAP gene. Elp1 is a subunit of the hetero-hexameric transcriptional elongator complex, but how it functions in disease-vulnerable neurons is unknown. Conditional knockout mice were generated to characterize the role of Elp1 in migration, differentiation and survival of migratory neural crest (NC) progenitors that give rise to sympathetic and sensory neurons. Loss of Elp1 in NC progenitors did not impair their migration, proliferation or survival, but there was a significant impact on post-migratory sensory and sympathetic neuron survival and target tissue innervation. Ablation of Elp1 in post-migratory sympathetic neurons caused highly abnormal target tissue innervation that was correlated with abnormal neurite outgrowth/branching and abnormal cellular distribution of soluble tyrosinated α-tubulin in Elp1-deficient primary sympathetic and sensory neurons. These results indicate that neuron loss and physiologic impairment in FD is not a consequence of abnormal neuron progenitor migration, differentiation or survival. Rather, loss of Elp1 leads to neuron death as a consequence of failed target tissue innervation associated with impairments in cytoskeletal regulation.
Collapse
Affiliation(s)
- Marisa Z Jackson
- Department of Pathology (Division of Neuropathology), Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611, USA Northwestern University Integrated Neuroscience (NUIN) Program, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611, USA
| | - Katherine A Gruner
- Department of Pathology (Division of Neuropathology), Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611, USA
| | - Charles Qin
- Department of Pathology (Division of Neuropathology), Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611, USA
| | - Warren G Tourtellotte
- Department of Pathology (Division of Neuropathology), Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611, USA Northwestern University Integrated Neuroscience (NUIN) Program, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611, USA Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611, USA
| |
Collapse
|
28
|
Gross J, Olze H, Mazurek B. Differential expression of transcription factors and inflammation-, ROS-, and cell death-related genes in organotypic cultures in the modiolus, the organ of Corti and the stria vascularis of newborn rats. Cell Mol Neurobiol 2014; 34:523-38. [PMID: 24595552 PMCID: PMC11488946 DOI: 10.1007/s10571-014-0036-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 02/14/2014] [Indexed: 12/22/2022]
Abstract
Cells respond to injury and hypoxia by changing gene expression. To study how the main compartments of the cochlea, the stria vascularis (SV), the organ of Corti (OC), and the modiolus (MOD), respond to such stress, we analyzed the expression of selected genes using microarray analysis. Organotypic cultures of SV, OC, and MOD from newborn rats were used as an experimental model. In the present study, we compare the expression of a total of 50 genes involved in apoptosis and necrosis, reactive oxygen species (ROS) metabolism, inflammation as well as selected transcription factors (TF) and analyze their role for the different cell death patterns observed in the three regions. MOD, OC, and SV differ not only in their basal gene profiles but also in their ability to respond to injury and hypoxia. The results provide two coexpression clusters across the three regions, a Hif-1a coexpression cluster and a cluster around the cell death-associated transcripts Casp3, Capn1, Capn2, and Capns1. These clusters include the TF Jun, Bmyc, Nfyc, Foxd3, Hes1, the ROS-associated molecules Sod3, and Nos2, and the inflammatory chemokine Ccl20. The evidence of both clusters indicates the complex and regulated character of gene expression following injury and hypoxia across the three regions SV, OC, and MOD. The high vulnerability of spiral ganglion neurons in the MOD region, previously explained on the basis of the availability of neuro-trophic factors, is associated with the increased endogenous production of ROS and nitric oxide and inadequate activation of protective acting genes.
Collapse
Affiliation(s)
- Johann Gross
- Molecular Biology Research Laboratory, Department of Otorhinolaryngology, Charité-Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany,
| | | | | |
Collapse
|
29
|
Schmid D, Zeis T, Schaeren-Wiemers N. Transcriptional regulation induced by cAMP elevation in mouse Schwann cells. ASN Neuro 2014; 6:137-57. [PMID: 24641305 PMCID: PMC4834722 DOI: 10.1042/an20130031] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 01/16/2014] [Accepted: 02/05/2014] [Indexed: 12/23/2022] Open
Abstract
In peripheral nerves, Schwann cell development is regulated by a variety of signals. Some of the aspects of Schwann cell differentiation can be reproduced in vitro in response to forskolin, an adenylyl cyclase activator elevating intracellular cAMP levels. Herein, the effect of forskolin treatment was investigated by a comprehensive genome-wide expression study on primary mouse Schwann cell cultures. Additional to myelin-related genes, many so far unconsidered genes were ascertained to be modulated by forskolin. One of the strongest differentially regulated gene transcripts was the transcription factor Olig1 (oligodendrocyte transcription factor 1), whose mRNA expression levels were reduced in treated Schwann cells. Olig1 protein was localized in myelinating and nonmyelinating Schwann cells within the sciatic nerve as well as in primary Schwann cells, proposing it as a novel transcription factor of the Schwann cell lineage. Data analysis further revealed that a number of differentially expressed genes in forskolin-treated Schwann cells were associated with the ECM (extracellular matrix), underlining its importance during Schwann cell differentiation in vitro. Comparison of samples derived from postnatal sciatic nerves and from both treated and untreated Schwann cell cultures showed considerable differences in gene expression between in vivo and in vitro, allowing us to separate Schwann cell autonomous from tissue-related changes. The whole data set of the cell culture microarray study is provided to offer an interactive search tool for genes of interest.
Collapse
Key Words
- camp
- forskolin
- in vitro
- microarray
- schwann cell differentiation
- bmp, bone morphogenetic protein
- camp, cyclic adenosine monophosphate
- cns, central nervous system
- creb, camp-response-element-binding protein
- david, database for annotation, visualization and integrated discovery
- dgc, dystrophin–glycoprotein complex
- ecm, extracellular matrix
- fdr, false discovery rate
- go, gene ontology
- ipa, ingenuity pathway analysis
- mag, myelin-associated glycoprotein
- mapk, mitogen-activated protein kinase
- mbp, myelin basic protein
- mpz/p0, myelin protein zero
- nf-κb, nuclear factor κb
- olig1, oligodendrocyte transcription factor 1
- pca, principal component analysis
- pfa, paraformaldehyde
- pka, protein kinase a
- pns, peripheral nervous system
- qrt–pcr, quantitative rt–pcr
- s.d., standard deviation
Collapse
Affiliation(s)
- Daniela Schmid
- *Neurobiology, Department of Biomedicine, University Hospital Basel,
University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Thomas Zeis
- *Neurobiology, Department of Biomedicine, University Hospital Basel,
University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Nicole Schaeren-Wiemers
- *Neurobiology, Department of Biomedicine, University Hospital Basel,
University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| |
Collapse
|
30
|
Ma G, Song T, Chen M, Fu Y, Xu Y, Ma E, Wang W, Du J, Huang M. Hippocampal and thalamic neuronal metabolism in a putative rat model of schizophrenia. Neural Regen Res 2013; 8:2415-2423. [PMID: 25206551 PMCID: PMC4146111 DOI: 10.3969/j.issn.1673-5374.2013.26.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 07/01/2013] [Indexed: 12/17/2022] Open
Abstract
The transcription factor early growth response protein 3 (EGR3) is involved in schizophrenia. We developed a putative rat model of schizophrenia by transfecting lentiviral particles carrying the Egr3 gene into bilateral hippocampal dentate gyrus. We assessed spatial working memory using the Morris water maze test, and neuronal metabolite levels in bilateral hippocampus and thalamus were determined by 3.0 T proton magnetic resonance spectroscopy. Choline content was significantly greater in the hippocampus after transfection, while N-acetylaspartate and the ratio of N-acetylaspartate to creatine/phosphocreatine in the thalamus were lower than in controls. This study is the first to report evaluation of brain metabolites using 3.0 T proton magnetic resonance spectroscopy in rats transfected with Egr3, and reveals metabolic abnormalities in the hippocampus and thalamus in this putative model of schizophrenia.
Collapse
Affiliation(s)
- Guolin Ma
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Tianbin Song
- Department of Radiology, Beijing Shunyi Hospital, Beijing 101300, China
| | - Min Chen
- Department of Radiology, Beijing Hospital Affiliated to the Ministry of Public Health, Beijing 100730, China
| | - Yuan Fu
- Department of Radiology, Beijing Hospital Affiliated to the Ministry of Public Health, Beijing 100730, China
| | - Yong Xu
- Department of Mental Health, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Ensen Ma
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Wu Wang
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Jiang Du
- Department of Radiology, University of California, San Diego, CA 92103-8226, USA
| | - Mingxiong Huang
- Radiology Imaging Laboratory, Department of Radiology, University of California, San Diego, CA 92121, USA
| |
Collapse
|
31
|
Fang F, Shangguan AJ, Kelly K, Wei J, Gruner K, Ye B, Wang W, Bhattacharyya S, Hinchcliff ME, Tourtellotte WG, Varga J. Early growth response 3 (Egr-3) is induced by transforming growth factor-β and regulates fibrogenic responses. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1197-1208. [PMID: 23906810 DOI: 10.1016/j.ajpath.2013.06.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 06/01/2013] [Accepted: 06/19/2013] [Indexed: 01/09/2023]
Abstract
Members of the early growth response (Egr) gene family of transcription factors have nonredundant biological functions. Although Egr-3 is implicated primarily in neuromuscular development and immunity, its regulation and role in tissue repair and fibrosis has not been studied. We now show that in normal skin fibroblasts, Egr-3 was potently induced by transforming growth factor-β via canonical Smad3. Moreover, transient Egr-3 overexpression was sufficient to stimulate fibrotic gene expression, whereas deletion of Egr-3 resulted in substantially attenuated transforming growth factor-β responses. Genome-wide expression profiling in fibroblasts showed that genes associated with tissue remodeling and wound healing were prominently up-regulated by Egr-3. Notably, <5% of fibroblast genes regulated by Egr-1 or Egr-2 were found to be coregulated by Egr-3, revealing substantial functional divergence among these Egr family members. In a mouse model of scleroderma, development of dermal fibrosis was accompanied by accumulation of Egr-3-positive myofibroblasts in the lesional tissue. Moreover, skin biopsy samples from patients with scleroderma showed elevated Egr-3 levels in the dermis, and Egr-3 mRNA levels correlated with the extent of skin involvement. These results provide the first evidence that Egr-3, a functionally distinct member of the Egr family with potent effects on inflammation and immunity, is up-regulated in scleroderma and is necessary and sufficient for profibrotic responses, suggesting important and distinct roles in the pathogenesis of fibrosis.
Collapse
Affiliation(s)
- Feng Fang
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Anna J Shangguan
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Kathleen Kelly
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jun Wei
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Katherine Gruner
- Department of Pathology and Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Boping Ye
- College of Life and Science, China Pharmaceutical University, Nanjing, China
| | - Wenxia Wang
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Swati Bhattacharyya
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Monique E Hinchcliff
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Warren G Tourtellotte
- Department of Pathology and Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - John Varga
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
32
|
Baeza-Raja B, Eckel-Mahan K, Zhang L, Vagena E, Tsigelny IF, Sassone-Corsi P, Ptáček LJ, Akassoglou K. p75 neurotrophin receptor is a clock gene that regulates oscillatory components of circadian and metabolic networks. J Neurosci 2013; 33:10221-34. [PMID: 23785138 PMCID: PMC3685830 DOI: 10.1523/jneurosci.2757-12.2013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 04/01/2013] [Accepted: 05/01/2013] [Indexed: 01/09/2023] Open
Abstract
The p75 neurotrophin receptor (p75(NTR)) is a member of the tumor necrosis factor receptor superfamily with a widespread pattern of expression in tissues such as the brain, liver, lung, and muscle. The mechanisms that regulate p75(NTR) transcription in the nervous system and its expression in other tissues remain largely unknown. Here we show that p75(NTR) is an oscillating gene regulated by the helix-loop-helix transcription factors CLOCK and BMAL1. The p75(NTR) promoter contains evolutionarily conserved noncanonical E-box enhancers. Deletion mutagenesis of the p75(NTR)-luciferase reporter identified the -1039 conserved E-box necessary for the regulation of p75(NTR) by CLOCK and BMAL1. Accordingly, gel-shift assays confirmed the binding of CLOCK and BMAL1 to the p75(NTR-)1039 E-box. Studies in mice revealed that p75(NTR) transcription oscillates during dark and light cycles not only in the suprachiasmatic nucleus (SCN), but also in peripheral tissues including the liver. Oscillation of p75(NTR) is disrupted in Clock-deficient and mutant mice, is E-box dependent, and is in phase with clock genes, such as Per1 and Per2. Intriguingly, p75(NTR) is required for circadian clock oscillation, since loss of p75(NTR) alters the circadian oscillation of clock genes in the SCN, liver, and fibroblasts. Consistent with this, Per2::Luc/p75(NTR-/-) liver explants showed reduced circadian oscillation amplitude compared with those of Per2::Luc/p75(NTR+/+). Moreover, deletion of p75(NTR) also alters the circadian oscillation of glucose and lipid homeostasis genes. Overall, our findings reveal that the transcriptional activation of p75(NTR) is under circadian regulation in the nervous system and peripheral tissues, and plays an important role in the maintenance of clock and metabolic gene oscillation.
Collapse
Affiliation(s)
| | - Kristin Eckel-Mahan
- Center for Epigenetics and Metabolism, Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, and
| | | | | | - Igor F. Tsigelny
- San Diego Supercomputer Center and Department of Neurosciences, University of California, San Diego, La Jolla, California 92093
| | - Paolo Sassone-Corsi
- Center for Epigenetics and Metabolism, Department of Biological Chemistry, University of California, Irvine, Irvine, California 92697, and
| | - Louis J. Ptáček
- Department of Neurology, and
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, California 94158
| | | |
Collapse
|
33
|
Interplay between thyroxin, BDNF and GABA in injured neurons. Neuroscience 2013; 239:241-52. [DOI: 10.1016/j.neuroscience.2012.12.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Revised: 12/04/2012] [Accepted: 12/05/2012] [Indexed: 01/03/2023]
|
34
|
A sympathetic neuron autonomous role for Egr3-mediated gene regulation in dendrite morphogenesis and target tissue innervation. J Neurosci 2013; 33:4570-83. [PMID: 23467373 DOI: 10.1523/jneurosci.5481-12.2013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Egr3 is a nerve growth factor (NGF)-induced transcriptional regulator that is essential for normal sympathetic nervous system development. Mice lacking Egr3 in the germline have sympathetic target tissue innervation abnormalities and physiologic sympathetic dysfunction similar to humans with dysautonomia. However, since Egr3 is widely expressed and has pleiotropic function, it has not been clear whether it has a role within sympathetic neurons and if so, what target genes it regulates to facilitate target tissue innervation. Here, we show that Egr3 expression within sympathetic neurons is required for their normal innervation since isolated sympathetic neurons lacking Egr3 have neurite outgrowth abnormalities when treated with NGF and mice with sympathetic neuron-restricted Egr3 ablation have target tissue innervation abnormalities similar to mice lacking Egr3 in all tissues. Microarray analysis performed on sympathetic neurons identified many target genes deregulated in the absence of Egr3, with some of the most significantly deregulated genes having roles in axonogenesis, dendritogenesis, and axon guidance. Using a novel genetic technique to visualize axons and dendrites in a subpopulation of randomly labeled sympathetic neurons, we found that Egr3 has an essential role in regulating sympathetic neuron dendrite morphology and terminal axon branching, but not in regulating sympathetic axon guidance to their targets. Together, these results indicate that Egr3 has a sympathetic neuron autonomous role in sympathetic nervous system development that involves modulating downstream target genes affecting the outgrowth and branching of sympathetic neuron dendrites and axons.
Collapse
|
35
|
Shin YH, Lee SJ, Jung J. Extracellular ATP inhibits Schwann cell dedifferentiation and proliferation in an ex vivo model of Wallerian degeneration. Biochem Biophys Res Commun 2013; 430:852-7. [PMID: 23194661 DOI: 10.1016/j.bbrc.2012.11.057] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 11/14/2012] [Indexed: 12/31/2022]
Abstract
After nerve injury, Schwann cells proliferate and revert to a phenotype that supports nerve regeneration. This phenotype-changing process can be viewed as Schwann cell dedifferentiation. Here, we investigated the role of extracellular ATP in Schwann cell dedifferentiation and proliferation during Wallerian degeneration. Using several markers of Schwann cell dedifferentiation and proliferation in sciatic explants, we found that extracellular ATP inhibits Schwann cell dedifferentiation and proliferation during Wallerian degeneration. Furthermore, the blockage of lysosomal exocytosis in ATP-treated sciatic explants is sufficient to induce Schwann cell dedifferentiation. Together, these findings suggest that ATP-induced lysosomal exocytosis may be involved in Schwann cell dedifferentiation.
Collapse
Affiliation(s)
- Youn Ho Shin
- Department of Anatomy, College of Medicine, Kyung Hee University, Heogi-Dong 1, Dongdaemun-Gu, Seoul 130-701, Republic of Korea
| | | | | |
Collapse
|
36
|
Parsi S, Soltani BM, Hosseini E, Tousi SE, Mowla SJ. Experimental verification of a predicted intronic microRNA in human NGFR gene with a potential pro-apoptotic function. PLoS One 2012; 7:e35561. [PMID: 22558167 PMCID: PMC3338703 DOI: 10.1371/journal.pone.0035561] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 03/20/2012] [Indexed: 12/12/2022] Open
Abstract
Neurotrophins (NTs) are a family of secreted growth factor proteins primarily involved in the regulation of survival and appropriate development of neural cells, functioning by binding to their specific (TrkA, TtkB, and TrkC) and/or common NGFR receptor. NGFR is the common receptor of NTs, binding with low-affinity to all members of the family. Among different functions assigned to NGFR, it is also involved in apoptosis induction and tumorigenesis processes. Interestingly, some of the functions of NGFR appear to be ligand-independent, suggesting a probable involvement of non-coding RNA residing within the sequence of the gene. Here, we are reporting the existence of a conserved putative microRNA, named Hsa-mir-6165 [EBI accession#: FR873488]. Transfection of a DNA segment corresponding to the pre-mir-6165 sequence in Hela cell line caused the generation of mature exogenous mir-6165 (a ∼200,000 fold overexpression). Furthermore, using specific primers, we succeeded to detect the endogenous expression of mir-6165 in several glioma cell lines and glioma primary tumors known to express NGFR. Similar to the pro-apoptotic role of NGFR in some cell types, overexpression of pre-mir-6165 in U87 cell line resulted in an elevated rate of apoptosis. Moreover, coordinated with the increased level of mir-6165 in the transfected U87 cell line, two of its predicted target genes (Pkd1 and DAGLA) were significantly down-regulated. The latter findings suggest that some of the previously attributed functions of NGFR could be explained indirectly by co-transcription of mir-6165 in the cells.
Collapse
Affiliation(s)
- Sepideh Parsi
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bahram M. Soltani
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ebrahim Hosseini
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Samaneh E. Tousi
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed J. Mowla
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
37
|
Hossain S, de la Cruz-Morcillo MA, Sanchez-Prieto R, Almazan G. Mitogen-activated protein kinase p38 regulates krox-20 to direct schwann cell differentiation and peripheral myelination. Glia 2012; 60:1130-44. [DOI: 10.1002/glia.22340] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 03/16/2012] [Indexed: 12/24/2022]
|
38
|
Shulga A, Magalhães AC, Autio H, Plantman S, di Lieto A, Nykjær A, Carlstedt T, Risling M, Arumäe U, Castrén E, Rivera C. The loop diuretic bumetanide blocks posttraumatic p75NTR upregulation and rescues injured neurons. J Neurosci 2012; 32:1757-70. [PMID: 22302815 PMCID: PMC6703341 DOI: 10.1523/jneurosci.3282-11.2012] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 11/28/2011] [Accepted: 12/05/2011] [Indexed: 01/09/2023] Open
Abstract
Injured neurons become dependent on trophic factors for survival. However, application of trophic factors to the site of injury is technically extremely challenging. Novel approaches are needed to circumvent this problem. Here, we unravel the mechanism of the emergence of dependency of injured neurons on brain-derived neurotrophic factor (BDNF) for survival. Based on this mechanism, we propose the use of the diuretic bumetanide to prevent the requirement for BDNF and consequent neuronal death in the injured areas. Responses to the neurotransmitter GABA change from hyperpolarizing in intact neurons to depolarizing in injured neurons. We show in vivo in rats and ex vivo in mouse organotypic slice cultures that posttraumatic GABA(A)-mediated depolarization is a cause for the well known phenomenon of pathological upregulation of pan-neurotrophin receptor p75(NTR). The increase in intracellular Ca(2+) triggered by GABA-mediated depolarization activates ROCK (Rho kinase), which in turn leads to the upregulation of p75(NTR). We further show that high levels of p75(NTR) and its interaction with sortilin and proNGF set the dependency on BDNF for survival. Thus, application of bumetanide prevents p75(NTR) upregulation and neuronal death in the injured areas with reduced levels of endogenous BDNF.
Collapse
Affiliation(s)
- Anastasia Shulga
- Institute of Biotechnology
- Neuroscience Center, and
- Department of Neurological Sciences, University of Helsinki, FI-00014 Helsinki, Finland
| | | | | | - Stefan Plantman
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | - Anders Nykjær
- The Lundbeck Foundation Research Center MIND, Department of Medical Biochemistry, Aarhus University, DK-8000C Aarhus, Denmark
| | - Thomas Carlstedt
- Department of Hand Surgery, Karolinska Institutet, Södersjukhuset, SE-118 83 Stockholm, Sweden, and
| | - Mårten Risling
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | | | - Claudio Rivera
- Institute of Biotechnology
- Neuroscience Center, and
- Université de la Méditerranée, UMR S901 Aix-Marseille 2, Institut de Neurobiologie de la Méditerranée, 13009 Marseille, France
| |
Collapse
|
39
|
Guo AY, Sun J, Jia P, Zhao Z. A novel microRNA and transcription factor mediated regulatory network in schizophrenia. BMC SYSTEMS BIOLOGY 2010; 4:10. [PMID: 20156358 PMCID: PMC2834616 DOI: 10.1186/1752-0509-4-10] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 02/15/2010] [Indexed: 01/02/2023]
Abstract
Background Schizophrenia is a complex brain disorder with molecular mechanisms that have yet to be elucidated. Previous studies have suggested that changes in gene expression may play an important role in the etiology of schizophrenia, and that microRNAs (miRNAs) and transcription factors (TFs) are primary regulators of this gene expression. So far, several miRNA-TF mediated regulatory modules have been verified. We hypothesized that miRNAs and TFs might play combinatory regulatory roles for schizophrenia genes and, thus, explored miRNA-TF regulatory networks in schizophrenia. Results We identified 32 feed-forward loops (FFLs) among our compiled schizophrenia-related miRNAs, TFs and genes. Our evaluation revealed that these observed FFLs were significantly enriched in schizophrenia genes. By converging the FFLs and mutual feedback loops, we constructed a novel miRNA-TF regulatory network for schizophrenia. Our analysis revealed EGR3 and hsa-miR-195 were core regulators in this regulatory network. We next proposed a model highlighting EGR3 and miRNAs involved in signaling pathways and regulatory networks in the nervous system. Finally, we suggested several single nucleotide polymorphisms (SNPs) located on miRNAs, their target sites, and TFBSs, which may have an effect in schizophrenia gene regulation. Conclusions This study provides many insights on the regulatory mechanisms of genes involved in schizophrenia. It represents the first investigation of a miRNA-TF regulatory network for a complex disease, as demonstrated in schizophrenia.
Collapse
Affiliation(s)
- An-Yuan Guo
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | | |
Collapse
|
40
|
Lee HK, Shin YK, Jung J, Seo SY, Baek SY, Park HT. Proteasome inhibition suppresses Schwann cell dedifferentiation in vitro and in vivo. Glia 2010; 57:1825-34. [PMID: 19455715 DOI: 10.1002/glia.20894] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The ubiquitin-proteasome system (UPS), lysosomes, and autophagy are essential protein degradation systems for the regulation of a variety of cellular physiological events including the cellular response to injury. It has recently been reported that the UPS and autophagy mediate the axonal degeneration caused by traumatic insults and the retrieval of nerve growth factors. In the peripheral nerves, axonal degeneration after injury is accompanied by myelin degradation, which is tightly related to the reactive changes of Schwann cells called dedifferentiation. In this study, we examined the role of the UPS, lysosomal proteases, and autophagy in the early phase of Wallerian degeneration of injured peripheral nerves. We found that nerve injury induced an increase in the ubiquitin conjugation and lysosomal-associated membrane protein-1 expression within 1 day without any biochemical evidence for autophagy activation. Using an ex vivo explant culture of the sciatic nerve, we observed that inhibiting proteasomes or lysosomal serine proteases prevented myelin degradation, whereas this was not observed when inhibiting autophagy. Interestingly, proteasome inhibition, but not leupeptin, prevented Schwann cells from inducing dedifferentiation markers such as p75 nerve growth factor receptor and glial fibrillary acidic protein in vitro and in vivo. In addition, proteasome inhibitors induced cell cycle arrest and cellular process formation in cultured Schwann cells. Taken together, these findings indicate that the UPS plays a role in the phenotype changes of Schwann cells in response to nerve injury.
Collapse
Affiliation(s)
- Hyun Kyoung Lee
- Department of Physiology, Medical Science Research Institute, College of Medicine, Dong-A University, Seo-Gu, Busan, South Korea
| | | | | | | | | | | |
Collapse
|
41
|
Buser AM, Schmid D, Kern F, Erne B, Lazzati T, Schaeren-Wiemers N. The myelin protein MAL affects peripheral nerve myelination: a new player influencing p75 neurotrophin receptor expression. Eur J Neurosci 2009; 29:2276-90. [PMID: 19508690 DOI: 10.1111/j.1460-9568.2009.06785.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The myelin and lymphocyte protein (MAL) is a raft-associated membrane protein predominantly expressed by oligodendrocytes and Schwann cells. Here we show that MAL regulates myelination in the peripheral nervous system. In mice overexpressing MAL, myelination was retarded and fibers were hypomyelinated, whereas myelination in MAL knockout mice was accelerated. This was not due to impaired Schwann cell proliferation, differentiation or axonal sorting. We found that the expression level of p75 neurotrophin receptor mRNA and protein was strongly reduced in developing sciatic nerves in MAL-overexpressing mice. This reduction is well correlated with the observed alterations in myelination initiation, speed of myelination and alterations in Remak bundle development. Our results suggest a functional role for MAL in peripheral myelination by influencing the expression of membrane components that mediate axon-glia interaction during ensheathment and myelin wrapping.
Collapse
Affiliation(s)
- A M Buser
- Neurobiology, Department of Biomedicine and Neurology, University Hospital Basel, Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
42
|
Jessen KR, Mirsky R. Negative regulation of myelination: relevance for development, injury, and demyelinating disease. Glia 2009; 56:1552-1565. [PMID: 18803323 DOI: 10.1002/glia.20761] [Citation(s) in RCA: 386] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Dedifferentiation of myelinating Schwann cells is a key feature of nerve injury and demyelinating neuropathies. We review recent evidence that this dedifferentiation depends on activation of specific intracellular signaling molecules that drive the dedifferentiation program. In particular, we discuss the idea that Schwann cells contain negative transcriptional regulators of myelination that functionally complement positive regulators such as Krox-20, and that myelination is therefore determined by a balance between two opposing transcriptional programs. Negative transcriptional regulators should be expressed prior to myelination, downregulated as myelination starts but reactivated as Schwann cells dedifferentiate following injury. The clearest evidence for a factor that works in this way relates to c-Jun, while other factors may include Notch, Sox-2, Pax-3, Id2, Krox-24, and Egr-3. The role of cell-cell signals such as neuregulin-1 and cytoplasmic signaling pathways such as the extracellular-related kinase (ERK)1/2 pathway in promoting dedifferentiation of myelinating cells is also discussed. We also review evidence that neurotrophin 3 (NT3), purinergic signaling, and nitric oxide synthase are involved in suppressing myelination. The realization that myelination is subject to negative as well as positive controls contributes significantly to the understanding of Schwann cell plasticity. Negative regulators are likely to have a major role during injury, because they promote the transformation of damaged nerves to an environment that fosters neuronal survival and axonal regrowth. In neuropathies, however, activation of these pathways is likely to be harmful because they may be key contributors to demyelination, a situation which would open new routes for clinical intervention.
Collapse
Affiliation(s)
- Kristján R Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom.
| | | |
Collapse
|
43
|
Svaren J, Meijer D. The molecular machinery of myelin gene transcription in Schwann cells. Glia 2009; 56:1541-1551. [PMID: 18803322 DOI: 10.1002/glia.20767] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
During late fetal life, Schwann cells in the peripheral nerves singled out by the larger axons will transit through a promyelinating stage before exiting the cell cycle and initiating myelin formation. A network of extra- and intracellular signaling pathways, regulating a transcriptional program of cell differentiation, governs this progression of cellular changes, culminating in a highly differentiated cell. In this review, we focus on the roles of a number of transcription factors not only in myelination, during normal development, but also in demyelination, following nerve trauma. These factors include specification factors involved in early development of Schwann cells from neural crest (Sox10) as well as factors specifically required for transitions into the promyelinating and myelinating stages (Oct6/Scip and Krox20/Egr2). From this description, we can glean the first, still very incomplete, contours of a gene regulatory network that governs myelination and demyelination during development and regeneration.
Collapse
Affiliation(s)
- John Svaren
- Department of Comparative Biosciences, School of Veterinary Medicine and Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | |
Collapse
|
44
|
Eldredge LC, Gao XM, Quach DH, Li L, Han X, Lomasney J, Tourtellotte WG. Abnormal sympathetic nervous system development and physiological dysautonomia in Egr3-deficient mice. Development 2008; 135:2949-57. [PMID: 18653557 DOI: 10.1242/dev.023960] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Sympathetic nervous system development depends upon many factors that mediate neuron migration, differentiation and survival. Target tissue-derived nerve growth factor (NGF) signaling-induced gene expression is required for survival, differentiation and target tissue innervation of post-migratory sympathetic neurons. However, the transcriptional regulatory mechanisms mediated by NGF signaling are very poorly defined. Here, we identify Egr3, a member of the early growth response (Egr) family of transcriptional regulators, as having an important role in sympathetic nervous system development. Egr3 is regulated by NGF signaling and it is expressed in sympathetic neurons during development when they depend upon NGF for survival and target tissue innervation. Egr3-deficient mice have severe sympathetic target tissue innervation abnormalities and profound physiological dysautonomia. Unlike NGF, which is essential for sympathetic neuron survival and for axon branching within target tissues, Egr3 is required for normal terminal axon extension and branching, but not for neuron survival. The results indicate that Egr3 is a novel NGF signaling effector that regulates sympathetic neuron gene expression required for normal target tissue innervation and function. Egr3-deficient mice have a phenotype that is remarkably similar to humans with sympathetic nervous system disease, raising the possibility that it may have a role in some forms of human dysautonomia, most of which have no known cause.
Collapse
Affiliation(s)
- Laurie C Eldredge
- Department of Pathology, Northwestern University, Chicago, IL 60611, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Poirier R, Cheval H, Mailhes C, Garel S, Charnay P, Davis S, Laroche S. Distinct functions of egr gene family members in cognitive processes. Front Neurosci 2008; 2:47-55. [PMID: 18982106 PMCID: PMC2570062 DOI: 10.3389/neuro.01.002.2008] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Accepted: 05/12/2008] [Indexed: 12/11/2022] Open
Abstract
The different gene members of the Egr family of transcriptional regulators have often been considered to have related functions in brain, based on their co-expression in many cell-types and structures, the relatively high homology of the translated proteins and their ability to bind to the same consensus DNA binding sequence. Recent research, however, suggest this might not be the case. In this review, we focus on the current understanding of the functional roles of the different Egr family members in learning and memory. We briefly outline evidence from mutant mice that Egr1 is required specifically for the consolidation of long-term memory, while Egr3 is primarily essential for short-term memory. We also review our own recent findings from newly generated forebrain-specific conditional Egr2 mutant mice, which revealed that Egr2, as opposed to Egr1 and Egr3, is dispensable for several forms of learning and memory and on the contrary can act as an inhibitory constraint for certain cognitive functions. The studies reviewed here highlight the fact that Egr family members may have different, and in certain circumstances antagonistic functions in the adult brain.
Collapse
Affiliation(s)
- Roseline Poirier
- Univ. Paris Sud, Laboratoire de Neurobiologie de l'Apprentissage, de la Mémoire et de la Communication Orsay, France.
| | | | | | | | | | | | | |
Collapse
|
46
|
Jones EA, Jang SW, Mager GM, Chang LW, Srinivasan R, Gokey NG, Ward RM, Nagarajan R, Svaren J. Interactions of Sox10 and Egr2 in myelin gene regulation. NEURON GLIA BIOLOGY 2007; 3:377-87. [PMID: 18634568 PMCID: PMC2605513 DOI: 10.1017/s1740925x08000173] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Myelination in the PNS is accompanied by a large induction of the myelin protein zero (Mpz) gene to produce the most abundant component in peripheral myelin. Analyses of knockout mice have shown that the EGR2/Krox20 and SOX10 transcription factors are required for Mpz expression. Our recent work has shown that the dominant EGR2 mutations associated with human peripheral neuropathies cause disruption of EGR2/SOX10 synergy at specific sites, including a conserved enhancer element in the first intron of the Mpz gene. Further investigation of Egr2/Sox10 interactions reveals that activation of the Mpz intron element by Egr2 requires both Sox10-binding sites. In addition, both Egr1 and Egr3 cooperate with Sox10 to activate this element, which indicates that this capacity is conserved among Egr family members. Finally, a conserved composite structure of Egr2/Sox10-binding sites in the genes encoding Mpz, myelin-associated glycoprotein and myelin basic protein genes was used to screen for similar modules in other myelin genes, revealing a potential regulatory element in the periaxin gene. Overall, these results elucidate a working model for developmental regulation of Mpz expression, several facets of which extend to regulation of other peripheral myelin genes.
Collapse
Affiliation(s)
- Erin A Jones
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|