1
|
Devkota P, Danzi MC, Lemmon VP, Bixby JL, Wuchty S. Computational Identification of Kinases That Control Axon Growth in Mouse. SLAS DISCOVERY 2020; 25:792-800. [PMID: 32613890 DOI: 10.1177/2472555220930697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The determination of signaling pathways and transcriptional networks that control various biological processes is a major challenge from both basic science and translational medicine perspectives. Because such analysis can point to critical disease driver nodes to target for therapeutic purposes, we combined data from phenotypic screening experiments and gene expression studies of mouse neurons to determine information flow through a molecular interaction network using a network propagation approach. We hypothesized that differences in information flow between control and injured conditions prioritize relevant driver nodes that cause this state change. Identifying paths likely taken from potential source nodes to a set of transcription factors (TFs), called sinks, we found that kinases are enriched among source genes sending significantly different amounts of information to TFs in an axonal injury model. Additionally, TFs found to be differentially active during axon growth were enriched in the set of sink genes that received significantly altered amounts of information from source genes. Notably, such enrichment levels hold even when restricting the set of source genes to only those kinases observed to support or hamper neurite growth. That way, we found a set of 71 source genes that send significantly different levels of information to axon growth-relevant TFs. We analyzed their information flow changes in response to axonal injury and their influences on TFs predicted to facilitate or antagonize axon growth. Finally, we drew a network diagram of the interactions and changes in information flow between these source genes and their axon growth-relevant sink TFs.
Collapse
Affiliation(s)
- Prajwal Devkota
- Department of Computer Science, University of Miami, Miami, FL, USA
| | - Matt C Danzi
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Vance P Lemmon
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA.,Miami Institute of Data Science and Computing, University of Miami, Miami, FL, USA.,Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - John L Bixby
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA.,Miami Institute of Data Science and Computing, University of Miami, Miami, FL, USA.,Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Stefan Wuchty
- Department of Computer Science, University of Miami, Miami, FL, USA.,Miami Institute of Data Science and Computing, University of Miami, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Biology, University of Miami, Miami, FL, USA
| |
Collapse
|
2
|
Zhan H, Wang Y, Yu S, Cai G, Zeng Y, Ma J, Liu W, Wu W. Upregulation of Mlxipl induced by cJun in the spinal dorsal horn after peripheral nerve injury counteracts mechanical allodynia by inhibiting neuroinflammation. Aging (Albany NY) 2020; 12:11004-11024. [PMID: 32518215 PMCID: PMC7346034 DOI: 10.18632/aging.103313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/28/2020] [Indexed: 12/18/2022]
Abstract
Mlxipl regulates glucose metabolism, lipogenesis and tumorigenesis and has a wide-ranging impact on human health and disease. However, the role of Mlxipl in neuropathic pain remains unknown. In this study, we found that Mlxipl was increased in the ipsilateral L4-L6 spinal dorsal horn after Spared Nerve Injury surgery. Knockdown of Mlxipl in the ipsilateral L4-L6 spinal dorsal horn by intraspinal microinjection aggravated Spared Nerve Injury-induced mechanical allodynia and inflammation in the spinal dorsal horn, on the contrary, overexpression of Mlxipl inhibited mechanical allodynia and inflammation. Subsequently, the rat Mlxipl promoter was analyzed using bioinformatics methods to predict the upstream transcription factor cJun. Luciferase assays and ChIP-qPCR confirmed that cJun bound to the promoter of Mlxipl and enhanced its expression. Finally, we demonstrated that Mlxipl inhibited the inflammatory responses of lipopolysaccharide-induced microglia and that Mlxipl was regulated by the transcription factor cJun. These findings suggested that cJun-induced Mlxipl upregulation in the spinal dorsal horn after peripheral nerve injury provided a protective mechanism for the development and progression of neuropathic pain by inhibiting microglial-derived neuroinflammation. Targeting Mlxipl in the spinal dorsal horn might represent an effective strategy for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Hongrui Zhan
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.,Department of Rehabilitation, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong Province, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai 519000, Guangdong Province, China
| | - Yaping Wang
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Shi Yu
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Guiyuan Cai
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yanyan Zeng
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Junqin Ma
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Wei Liu
- Department of Rehabilitation, Shenzhen University General Hospital, Shenzhen 518055, China
| | - Wen Wu
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| |
Collapse
|
3
|
Herdegen T. REVIEW ■ : Jun, Fos, and CREB/ATF Transcription Factors in the Brain: Control of Gene Expression under Normal and Pathophysiological Conditions. Neuroscientist 2016. [DOI: 10.1177/107385849600200310] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The expression and activation of transcription factors and the control of gene transcription in the nervous system is a recent and rapidly expanding field in neurosciences. This research area may provide insights concerning the information transfer that arises from postsynaptic potentials or ligand-coupling of membrane receptors and terminates in gene expression. Visualization of both de novo synthesis of inducible transcription factors (ITFs) and phosphorylation of preexisting transcription factors have been used to mark neurons, pathways, and networks excited by various stimuli. This article summarizes basics of the transcription process and the complex functions of Jun, Fos, and CREB/ATF proteins, as well as the use of ITFs as experimental instruments in neurophysiology and neurobiology. The major focus is on the alterations in ITF expression following acute or chronic pathophysiological stimuli as mirrors of alterations in neuronal programs underlying adaptation, dysfunctions, or the development of diseases affecting the nervous system. NEUROSCIENTIST 2:153-161, 1996
Collapse
|
4
|
Morgan JI, Curran T. Review : The Immediate-Early Gene Response and Neuronal Death and Regeneration. Neuroscientist 2016. [DOI: 10.1177/107385849500100203] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The induction of the cellular immediate-early gene (cIEG) response is the earliest known transcriptional con sequence of neuronal excitation. It is believed that the products of the cIEGs (such as c- fos and c- jun) serve to bring about persistent changes in neuronal phenotype by altering gene expression and modifying signal transduction pathways. This has led, on the one hand, to the burgeoning use of Fos immunohistochemistry as a surrogate form of neuronal activity mapping and, on the other hand, to a quest for the biochemical functions of individual cIEG products in the nervous system. Recent studies of neuronal death and regen eration have contributed substantially to our vision of the molecular and biological characteristics of the cIEG response. Indeed, they have challenged some of its long-held tenets. Therefore, we will use these results to illustrate our most contemporary view of the cIEG response in the nervous system. The Neuroscientist 1:68- 75,1995
Collapse
Affiliation(s)
- James I. Morgan
- Roche Institute of Molecular Biology Roche Research
Center Nutley, New Jersey
| | - Tom Curran
- Roche Institute of Molecular Biology Roche Research
Center Nutley, New Jersey
| |
Collapse
|
5
|
Sanna MD, Ghelardini C, Galeotti N. Blockade of the spinal BDNF-activated JNK pathway prevents the development of antiretroviral-induced neuropathic pain. Neuropharmacology 2016; 105:543-552. [PMID: 26898292 DOI: 10.1016/j.neuropharm.2016.02.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/22/2016] [Accepted: 02/15/2016] [Indexed: 12/26/2022]
Abstract
UNLABELLED Although antiretroviral agents have been used successfully in suppressing viral production, they have also been associated with a number of side effects. The antiretroviral toxic neuropathy induces debilitating and extremely difficult to treat pain syndromes that often lead to discontinuation of antiretroviral therapy. Due to the critical need for the identification of novel therapeutic targets to improve antiretroviral neuropathic pain management, we investigated the role of the JNK signalling pathway in the mechanism of antiretroviral painful neuropathy. Mice were exposed to zalcitabine (2',3'-dideoxycytidine, ddC) and stavudine (2',3'-didehydro-3'-deoxythymidine, d4T) that induced a persistent mechanical allodynia and a transient cold allodynia. Treatment with the JNK blocker SP600125 before antiretroviral administration abolished mechanical hypersensitivity with no effect on thermal response. A robust spinal JNK overphosphorylation was observed on post-injection day 1 and 3, along with a JNK-dependent increase in p-c-Jun and ATF3 protein levels. Co-immunoprecipitation experiments showed the presence of a heterodimeric complex between ATF3 and c-Jun indicating that these transcription factors can act together to regulate transcription through heterodimerization. A rise in BDNF and caspase-3 protein levels was detected on day 1 and BDNF sequestration prevented both caspase-3 and p-JNK increase. These data suggest that BDNF plays a role in the early stages of ddC-induced allodynia by promoting apoptotic events and the activation of a hypernociceptive JNK-mediated pathway. We illustrated the activation of a BDNF-mediated JNK pathway involved in the early events responsible for the promotion of neuropathic pain, leading to a better knowledge of the mechanisms involved in the antiretroviral neuropathy. SUMMARY JNK blockade prevents antiretroviral-induced pain hypersensitivity. This may represent a potential prophylactic treatment of neuropathic pain to improve antiretroviral tolerability.
Collapse
Affiliation(s)
- Maria Domenica Sanna
- Laboratory of Neuropsychopharmacology, Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Florence, Italy
| | - Carla Ghelardini
- Laboratory of Neuropsychopharmacology, Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Florence, Italy
| | - Nicoletta Galeotti
- Laboratory of Neuropsychopharmacology, Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Florence, Italy.
| |
Collapse
|
6
|
Ma TC, Willis DE. What makes a RAG regeneration associated? Front Mol Neurosci 2015; 8:43. [PMID: 26300725 PMCID: PMC4528284 DOI: 10.3389/fnmol.2015.00043] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 07/24/2015] [Indexed: 12/31/2022] Open
Abstract
Regenerative failure remains a significant barrier for functional recovery after central nervous system (CNS) injury. As such, understanding the physiological processes that regulate axon regeneration is a central focus of regenerative medicine. Studying the gene transcription responses to axon injury of regeneration competent neurons, such as those of the peripheral nervous system (PNS), has provided insight into the genes associated with regeneration. Though several individual “regeneration-associated genes” (RAGs) have been identified from these studies, the response to injury likely regulates the expression of functionally coordinated and complementary gene groups. For instance, successful regeneration would require the induction of genes that drive the intrinsic growth capacity of neurons, while simultaneously downregulating the genes that convey environmental inhibitory cues. Thus, this view emphasizes the transcriptional regulation of gene “programs” that contribute to the overall goal of axonal regeneration. Here, we review the known RAGs, focusing on how their transcriptional regulation can reveal the underlying gene programs that drive a regenerative phenotype. Finally, we will discuss paradigms under which we can determine whether these genes are injury-associated, or indeed necessary for regeneration.
Collapse
Affiliation(s)
- Thong C Ma
- Department of Neurology, Columbia University New York, NY, USA
| | - Dianna E Willis
- Brain Mind Research Institute, Weill Cornell Medical College New York, NY, USA ; Burke-Cornell Medical Research Institute White Plains, NY, USA
| |
Collapse
|
7
|
Cheng X, Fu R, Gao M, Liu S, Li YQ, Song FH, Bruce I, Zhou LH, Wu W. Intrathecal application of short interfering RNA knocks down c-jun expression and augments spinal motoneuron death after root avulsion in adult rats. Neuroscience 2013; 241:268-79. [DOI: 10.1016/j.neuroscience.2013.03.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 03/07/2013] [Accepted: 03/07/2013] [Indexed: 12/21/2022]
|
8
|
Li Z, Du B, Li S, Lv X, Zhou S, Yu Y, Wang W, Zheng Z. Cloning and characterization of an apolipoprotein C2 promoter in the mouse central nervous system. Neural Regen Res 2013; 8:156-61. [PMID: 25206486 PMCID: PMC4107507 DOI: 10.3969/j.issn.1673-5374.2013.02.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2012] [Accepted: 07/03/2012] [Indexed: 11/24/2022] Open
Abstract
Apolipoprotein C2 is an important member of the apolipoprotein C family, and is a potent activator of lipoprotein lipase. In the central nervous system, apolipoprotein C2 plays an important role in the catabolism of triglyceride-rich lipoproteins. Studies into the exact regulatory mechanism of mouse apolipoprotein C2 expression have not been reported. In this study, seven luciferase expression vectors, which contained potential mouse apolipoprotein C2 gene promoters, were constructed and co-transfected with pRL-TK into HEK293T cells to investigate apolipoprotein C2 promoter activity. Luciferase assays indicated that the apolipoprotein C2 promoter region was mainly located in the +104 bp to +470 bp region. The activity of the different lengths of apolipoprotein C2 promoter region varied. This staggered negative-positive-negative arrangement indicates the complex regulation of apolipoprotein C2 expression and provides important clues for elucidating the regulatory mechanism of apolipoprotein C2 gene transcription.
Collapse
Affiliation(s)
- Zhaoyang Li
- Key Laboratory of Transgenetic Animal Research, Department of Laboratory Animal, China Medical University, Shenyang 110001, Liaoning Province, China
| | - Bing Du
- Key Laboratory of Transgenetic Animal Research, Department of Laboratory Animal, China Medical University, Shenyang 110001, Liaoning Province, China
| | - Shengyang Li
- Key Laboratory of Transgenetic Animal Research, Department of Laboratory Animal, China Medical University, Shenyang 110001, Liaoning Province, China ; College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, Liaoning Province, China
| | - Xiangchuan Lv
- Key Laboratory of Transgenetic Animal Research, Department of Laboratory Animal, China Medical University, Shenyang 110001, Liaoning Province, China
| | - Shenglai Zhou
- Key Laboratory of Transgenetic Animal Research, Department of Laboratory Animal, China Medical University, Shenyang 110001, Liaoning Province, China
| | - Yang Yu
- Key Laboratory of Transgenetic Animal Research, Department of Laboratory Animal, China Medical University, Shenyang 110001, Liaoning Province, China
| | - Wei Wang
- Key Laboratory of Transgenetic Animal Research, Department of Laboratory Animal, China Medical University, Shenyang 110001, Liaoning Province, China
| | - Zhihong Zheng
- Key Laboratory of Transgenetic Animal Research, Department of Laboratory Animal, China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
9
|
Specificity of peripheral nerve regeneration: interactions at the axon level. Prog Neurobiol 2012; 98:16-37. [PMID: 22609046 DOI: 10.1016/j.pneurobio.2012.05.005] [Citation(s) in RCA: 289] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 04/12/2012] [Accepted: 05/08/2012] [Indexed: 12/13/2022]
Abstract
Peripheral nerves injuries result in paralysis, anesthesia and lack of autonomic control of the affected body areas. After injury, axons distal to the lesion are disconnected from the neuronal body and degenerate, leading to denervation of the peripheral organs. Wallerian degeneration creates a microenvironment distal to the injury site that supports axonal regrowth, while the neuron body changes in phenotype to promote axonal regeneration. The significance of axonal regeneration is to replace the degenerated distal nerve segment, and achieve reinnervation of target organs and restitution of their functions. However, axonal regeneration does not always allows for adequate functional recovery, so that after a peripheral nerve injury, patients do not recover normal motor control and fine sensibility. The lack of specificity of nerve regeneration, in terms of motor and sensory axons regrowth, pathfinding and target reinnervation, is one the main shortcomings for recovery. Key factors for successful axonal regeneration include the intrinsic changes that neurons suffer to switch their transmitter state to a pro-regenerative state and the environment that the axons find distal to the lesion site. The molecular mechanisms implicated in axonal regeneration and pathfinding after injury are complex, and take into account the cross-talk between axons and glial cells, neurotrophic factors, extracellular matrix molecules and their receptors. The aim of this review is to look at those interactions, trying to understand if some of these molecular factors are specific for motor and sensory neuron growth, and provide the basic knowledge for potential strategies to enhance and guide axonal regeneration and reinnervation of adequate target organs.
Collapse
|
10
|
Ruff CA, Staak N, Patodia S, Kaswich M, Rocha-Ferreira E, Da Costa C, Brecht S, Makwana M, Fontana X, Hristova M, Rumajogee P, Galiano M, Bohatschek M, Herdegen T, Behrens A, Raivich G. Neuronal c-Jun is required for successful axonal regeneration, but the effects of phosphorylation of its N-terminus are moderate. J Neurochem 2012; 121:607-18. [PMID: 22372722 PMCID: PMC4491308 DOI: 10.1111/j.1471-4159.2012.07706.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 02/05/2012] [Accepted: 02/21/2012] [Indexed: 12/18/2022]
Abstract
Although neural c-Jun is essential for successful peripheral nerve regeneration, the cellular basis of this effect and the impact of c-Jun activation are incompletely understood. In the current study, we explored the effects of neuron-selective c-Jun deletion, substitution of serine 63 and 73 phosphoacceptor sites with non-phosphorylatable alanine, and deletion of Jun N-terminal kinases 1, 2 and 3 in mouse facial nerve regeneration. Removal of the floxed c-jun gene in facial motoneurons using cre recombinase under control of a neuron-specific synapsin promoter (junΔS) abolished basal and injury-induced neuronal c-Jun immunoreactivity, as well as most of the molecular responses following facial axotomy. Absence of neuronal Jun reduced the speed of axonal regeneration following crush, and prevented most cut axons from reconnecting to their target, significantly reducing functional recovery. Despite blocking cell death, this was associated with a large number of shrunken neurons. Finally, junΔS mutants also had diminished astrocyte and microglial activation and T-cell influx, suggesting that these non-neuronal responses depend on the release of Jun-dependent signals from neighboring injured motoneurons. The effects of substituting serine 63 and 73 phosphoacceptor sites (junAA), or of global deletion of individual kinases responsible for N-terminal c-Jun phosphorylation were mild. junAA mutants showed decrease in neuronal cell size, a moderate reduction in post-axotomy CD44 levels and slightly increased astrogliosis. Deletion of Jun N-terminal kinase (JNK)1 or JNK3 showed delayed functional recovery; deletion of JNK3 also interfered with T-cell influx, and reduced CD44 levels. Deletion of JNK2 had no effect. Thus, neuronal c-Jun is needed in regeneration, but JNK phosphorylation of the N-terminus mostly appears to not be required for its function.
Collapse
Affiliation(s)
- Crystal A Ruff
- Perinatal Brain Repair Group, Inst Women's Health, University College London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Moore DL, Goldberg JL. Multiple transcription factor families regulate axon growth and regeneration. Dev Neurobiol 2012; 71:1186-211. [PMID: 21674813 DOI: 10.1002/dneu.20934] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Understanding axon regenerative failure remains a major goal in neuroscience, and reversing this failure remains a major goal for clinical neurology. Although an inhibitory central nervous system environment clearly plays a role, focus on molecular pathways within neurons has begun to yield fruitful insights. Initial steps forward investigated the receptors and signaling pathways immediately downstream of environmental cues, but recent work has also shed light on transcriptional control mechanisms that regulate intrinsic axon growth ability, presumably through whole cassettes of gene target regulation. Here we will discuss transcription factors that regulate neurite growth in vitro and in vivo, including p53, SnoN, E47, cAMP-responsive element binding protein (CREB), signal transducer and activator of transcription 3 (STAT3), nuclear factor of activated T cell (NFAT), c-Jun activating transcription factor 3 (ATF3), sex determining region Ybox containing gene 11 (Sox11), nuclear factor κ-light chain enhancer of activated B cells (NFκB), and Krüppel-like factors (KLFs). Revealing the similarities and differences among the functions of these transcription factors may further our understanding of the mechanisms of transcriptional regulation in axon growth and regeneration.
Collapse
Affiliation(s)
- Darcie L Moore
- Bascom Palmer Eye Institute and the Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Florida, USA
| | | |
Collapse
|
12
|
Patodia S, Raivich G. Role of transcription factors in peripheral nerve regeneration. Front Mol Neurosci 2012; 5:8. [PMID: 22363260 PMCID: PMC3277281 DOI: 10.3389/fnmol.2012.00008] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2011] [Accepted: 01/24/2012] [Indexed: 11/13/2022] Open
Abstract
Following axotomy, the activation of multiple intracellular signaling cascades causes the expression of a cocktail of regeneration-associated transcription factors which interact with each other to determine the fate of the injured neurons. The nerve injury response is channeled through manifold and parallel pathways, integrating diverse inputs, and controlling a complex transcriptional output. Transcription factors form a vital link in the chain of regeneration, converting injury-induced stress signals into downstream protein expression via gene regulation. They can regulate the intrinsic ability of axons to grow, by controlling expression of whole cassettes of gene targets. In this review, we have investigated the functional roles of a number of different transcription factors - c-Jun, activating transcription factor 3, cAMP response element binding protein, signal transducer, and activator of transcription-3, CCAAT/enhancer binding proteins β and δ, Oct-6, Sox11, p53, nuclear factor kappa-light-chain-enhancer of activated B cell, and ELK3 - in peripheral nerve regeneration. Studies involving use of conditional mutants, microarrays, promoter region mapping, and different injury paradigms, have enabled us to understand their distinct as well as overlapping roles in achieving anatomical and functional regeneration after peripheral nerve injury.
Collapse
Affiliation(s)
- Smriti Patodia
- Centre for Perinatal Brain Protection and Repair, University College London London, UK
| | | |
Collapse
|
13
|
Tedeschi A. Tuning the orchestra: transcriptional pathways controlling axon regeneration. Front Mol Neurosci 2012; 4:60. [PMID: 22294979 PMCID: PMC3257844 DOI: 10.3389/fnmol.2011.00060] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 12/23/2011] [Indexed: 12/13/2022] Open
Abstract
Trauma in the adult mammalian central nervous system leads to irreversible structural and functional impairment due to failed regeneration attempts. In contrast, neurons in the peripheral nervous system exhibit a greater regenerative ability. It has been proposed that an orchestrated sequence of transcriptional events controlling the expression of specific sets of genes may be the underlying basis of an early cell-autonomous regenerative response. Understanding whether transcriptional fine tuning, in parallel with strategies aimed at counteracting extrinsic impediments promotes axon re-growth following central nervous system injuries represents an exciting challenge for future studies. Transcriptional pathways controlling axon regeneration are presented and discussed in this review.
Collapse
Affiliation(s)
- Andrea Tedeschi
- Department of Neurology, F. M. Kirby Neurobiology Center, Children's Hospital Boston Boston, MA, USA
| |
Collapse
|
14
|
Verhaagen J, Van Kesteren RE, Bossers KAM, Macgillavry HD, Mason MR, Smit AB. Molecular target discovery for neural repair in the functional genomics era. HANDBOOK OF CLINICAL NEUROLOGY 2012; 109:595-616. [PMID: 23098739 DOI: 10.1016/b978-0-444-52137-8.00037-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
A comprehensive understanding of the molecular pathways activated by traumatic neural injury is of major importance for the development of treatments for spinal cord injury (SCI). High-throughput gene expression profiling is a powerful approach to reveal genome-wide changes in gene expression during a specific biological process. Microarray analysis of injured nerves or neurons would ideally generate new hypotheses concerning the progression or deregulation of injury- and repair-related biological processes, such as neural scar formation and axon regeneration. These hypotheses should subsequently be tested experimentally and would eventually provide the molecular substrates for the development of novel therapeutics. Over the last decade, this approach has elucidated numerous extrinsic (mostly neural scar-associated) as well as neuron-intrinsic genes that are regulated following an injury. To date, the main challenge is to translate the observed injury-induced gene expression changes into a mechanistic framework to understand their functional implications. To achieve this, research on neural repair will have to adopt the conceptual advances and analytical tools provided by the functional genomics and systems biology revolution. Based on progress made in bioinformatics, high-throughput and high-content functional cellular screening, and in vivo gene transfer technology, we propose a multistep "roadmap" that provides an integrated strategy for molecular target discovery for repair of the injured spinal cord.
Collapse
Affiliation(s)
- Joost Verhaagen
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
15
|
van Kesteren RE, Mason MRJ, Macgillavry HD, Smit AB, Verhaagen J. A gene network perspective on axonal regeneration. Front Mol Neurosci 2011; 4:46. [PMID: 22125511 PMCID: PMC3222109 DOI: 10.3389/fnmol.2011.00046] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 11/02/2011] [Indexed: 01/12/2023] Open
Abstract
The regenerative capacity of injured neurons in the central nervous system is limited due to the absence of a robust neuron-intrinsic injury-induced gene response that supports axon regeneration. In peripheral neurons axotomy induces a large cohort of regeneration-associated genes (RAGs). The forced expression of some of these RAGs in injured neurons has some beneficial effect on axon regeneration, but the reported effects are rather small. Transcription factors (TFs) provide a promising class of RAGs. TFs are hubs in the regeneration-associated gene network, and potentially control the coordinate expression of many RAGs simultaneously. Here we discuss the use of combined experimental and computational methods to identify novel regeneration-associated TFs with a key role in initiating and maintaining the RAG-response in injured neurons. We propose that a relatively small number of hub TFs with multiple functional connections in the RAG network might provide attractive new targets for gene-based and/or pharmacological approaches to promote axon regeneration in the central nervous system.
Collapse
Affiliation(s)
- Ronald E van Kesteren
- Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University Amsterdam, Netherlands
| | | | | | | | | |
Collapse
|
16
|
Jandke A, Da Costa C, Sancho R, Nye E, Spencer-Dene B, Behrens A. The F-box protein Fbw7 is required for cerebellar development. Dev Biol 2011; 358:201-12. [PMID: 21827743 DOI: 10.1016/j.ydbio.2011.07.030] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 07/20/2011] [Accepted: 07/21/2011] [Indexed: 12/15/2022]
Abstract
The F-box protein Fbw7 (also known as Fbxw7, hCdc4 and Sel-10) functions as a substrate recognition component of a SCF-type E3 ubiquitin ligase. SCF(Fbw7) facilitates polyubiquitination and subsequent degradation of various proteins such as Notch, cyclin E, c-Myc and c-Jun. Fbw7 is highly expressed in the nervous system and controls neural stem cell differentiation and apoptosis via Notch and c-Jun during embryonic development (Hoeck et al., 2010). Fbw7 deletion in the neural lineage is perinatal lethal and thus prohibits studying the role of Fbw7 in the adult nervous system. fbw7 mRNA is highly expressed in the postnatal brain and to gain insights into the function of Fbw7 in postnatal neurogenesis we analysed Fbw7 function in the cerebellum. We generated conditional Fbw7-knockout mice (fbw7(∆Cb)) by inactivating Fbw7 specifically in the cerebellar anlage. This resulted in decreased cerebellar size, reduced Purkinje cell number and defects in axonal arborisation. Moreover, Fbw7-deficient cerebella showed supranumeral fissures and aberrant progenitor cell migration. Protein levels of the Fbw7 substrates Notch1 and N-terminally phosphorylated c-Jun were upregulated in fbw7(∆Cb) mice. Concomitant deletion of c-Jun, and also the junAA knock-in mutation which specifically abrogates c-Jun N-terminal phosphorylation, rescued Purkinje cell numbers and arborisation in the fbw7(∆Cb) background. Taken together these data demonstrate that Fbw7 is essential during cerebellar development, and identify N-terminally phosphorylated c-Jun as an important substrate of SCF(Fbw7) during neurogenesis.
Collapse
Affiliation(s)
- Anett Jandke
- Mammalian Genetics Laboratory, Cancer Research UK, London Research Institute, 44, Lincoln's Inn Fields, London, WC2A3LY, UK
| | | | | | | | | | | |
Collapse
|
17
|
Wang LL, Zhao XC, Yan LF, Wang YQ, Cheng X, Fu R, Zhou LH. C-jun phosphorylation contributes to down regulation of neuronal nitric oxide synthase protein and motoneurons death in injured spinal cords following root-avulsion of the brachial plexus. Neuroscience 2011; 189:397-407. [PMID: 21596101 DOI: 10.1016/j.neuroscience.2011.04.070] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 04/11/2011] [Accepted: 04/30/2011] [Indexed: 01/11/2023]
Abstract
Previous studies have shown that c-jun and neuronal nitric oxide synthase (nNOS) are both induced in injured motoneurons, but their roles in motoneuron death remain unclear. We hypothesized that nNOS might be the downstream effector of c-jun N-terminal kinase (JNK)/c-jun in avulsion-induced motoneuron death. Here, we found that brachial root-avulsion induced a temporary increase in JNK activity and three- and four-fold increases in phospho-c-jun and c-jun, respectively; however, brachial root-avulsion caused a decrease in nNOS protein expression from 4 h to 14 days post-injury. At 14 days post-injury, almost all nNOS-positive motoneurons were co-localized with phospho-c-jun-positive motoneurons in ipsilateral ventral horns. The JNK inhibitor SP600125, applied immediately post-injury, resulted in an upregulation of nNOS protein both in injured spinal cords and motoneurons and caused a slight alleviation of motoneuron death by inhibiting c-jun phosphorylation at 14 days post-injury. Our results demonstrated that the JNK/c-jun signal transduction pathway is involved in root-avulsion. The inhibition of c-jun phosphorylation prevents nNOS levels from dropping below baseline levels in the spinal cord and partially alleviates motoneuron death following root-avulsion. Therefore, inhibiting c-jun phosphorylation or up-regulating the nNOS protein in injured spinal cords at the early stage might be used in the future as the molecular-target strategies to prevent the motoneurons degeneration in root-avulsion.
Collapse
Affiliation(s)
- L-L Wang
- Zhong Shan School of Medicine, Sun Yat-Sen University, No. 74 Zhongshan Road 2, Guangzhou 510080, PR China
| | | | | | | | | | | | | |
Collapse
|
18
|
Raivich G. Transcribing the path to neurological recovery-From early signals through transcription factors to downstream effectors of successful regeneration. Ann Anat 2011; 193:248-58. [PMID: 21501955 DOI: 10.1016/j.aanat.2011.01.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 01/16/2011] [Accepted: 01/19/2011] [Indexed: 11/29/2022]
Abstract
The peripheral nervous system is known to regenerate comparatively well and this ability is mirrored in the de novo expression or upregulation of a wide variety of molecules involved in axonal outgrowth starting with transcription factors, but also including growth-stimulating substances, guidance and cell adhesion molecules, intracellular signaling enzymes and proteins involved in regulating cell-surface cytoskeletal interactions. Recent studies using pharmacological agents, and global as well as neuron-selective gene inactivation techniques have shed light on those endogenous molecules that play a non-redundant role in mediating regenerative axonal outgrowth in vivo. The aim of the current review is to sketch the sequence of molecular events from early sensors of injury to transcription factors to downstream effectors that cooperate in successful regeneration and functional recovery.
Collapse
Affiliation(s)
- Gennadij Raivich
- Perinatal Brain Repair Group, Department of Obstetrics and Gynaecology, University College London, 86-96 Chenies Mews, London, UK.
| |
Collapse
|
19
|
Peddie CJ, Keast JR. Pelvic Nerve Injury Causes a Rapid Decrease in Expression of Choline Acetyltransferase and Upregulation of c-Jun and ATF-3 in a Distinct Population of Sacral Preganglionic Neurons. Front Neurosci 2011; 5:6. [PMID: 21283532 PMCID: PMC3031092 DOI: 10.3389/fnins.2011.00006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 01/07/2011] [Indexed: 11/13/2022] Open
Abstract
Autonomic regulation of the urogenital organs is impaired by injuries sustained during pelvic surgery or compression of lumbosacral spinal nerves (e.g., cauda equina syndrome). To understand the impact of injury on both sympathetic and parasympathetic components of this nerve supply, we performed an experimental surgical and immunohistochemical study on adult male rats, where the structure of this complex part of the nervous system has been well defined. We performed unilateral transection of pelvic or hypogastric nerves and analyzed relevant regions of lumbar and sacral spinal cord, up to 4 weeks after injury. Expression of c-Jun, the neuronal injury marker activating transcription factor-3 (ATF-3), and choline acetyltransferase (ChAT) were examined. We found little evidence for chemical or structural changes in substantial numbers of functionally related but uninjured spinal neurons (e.g., in sacral preganglionic neurons after hypogastric nerve injury), failing to support the concept of compensatory events. The effects of injury were greatest in sacral cord, ipsilateral to pelvic nerve transection. Here, around half of all preganglionic neurons expressed c-Jun within 1 week of injury, and substantial ATF-3 expression also occurred, especially in neurons with complete loss of ChAT-immunoreactivity. There did not appear to be any death of retrogradely labeled neurons, in contrast to axotomy studies performed on other regions of spinal cord or sacral ventral root avulsion models. Each of the effects we observed occurred in only a subpopulation of preganglionic neurons at that spinal level, raising the possibility that distinct functional subgroups have different susceptibility to trauma-induced degeneration and potentially different regenerative abilities. Identification of the cellular basis of these differences may provide insights into organ-specific strategies for attenuating degeneration or promoting regeneration of these circuits after trauma.
Collapse
Affiliation(s)
- Christopher J Peddie
- Pain Management Research Institute and Kolling Institute of Medical Research, University of Sydney at Royal North Shore Hospital St Leonards, NSW, Australia
| | | |
Collapse
|
20
|
Smith RP, Lerch-Haner JK, Pardinas JR, Buchser WJ, Bixby JL, Lemmon VP. Transcriptional profiling of intrinsic PNS factors in the postnatal mouse. Mol Cell Neurosci 2010; 46:32-44. [PMID: 20696251 DOI: 10.1016/j.mcn.2010.07.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Revised: 07/22/2010] [Accepted: 07/27/2010] [Indexed: 11/25/2022] Open
Abstract
Neurons in the peripheral nervous system (PNS) display a higher capacity to regenerate after injury than those in the central nervous system, suggesting cell specific transcriptional modules underlying axon growth and inhibition. We report a systems biology based search for PNS specific transcription factors (TFs). Messenger RNAs enriched in dorsal root ganglion (DRG) neurons compared to cerebellar granule neurons (CGNs) were identified using subtractive hybridization and DNA microarray approaches. Network and transcription factor binding site enrichment analyses were used to further identify TFs that may be differentially active. Combining these techniques, we identified 32 TFs likely to be enriched and/or active in the PNS. Twenty-five of these TFs were then tested for an ability to promote CNS neurite outgrowth in an overexpression screen. Real-time PCR and immunohistochemical studies confirmed that one representative TF, STAT3, is intrinsic to PNS neurons, and that constitutively active STAT3 is sufficient to promote CGN neurite outgrowth.
Collapse
Affiliation(s)
- Robin P Smith
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, Miller School of Medicine, University of Miami, 1400 NW 12th Ave, Miami, FL 33136, USA
| | | | | | | | | | | |
Collapse
|
21
|
Ryu V, Gallaher Z, Czaja K. Plasticity of nodose ganglion neurons after capsaicin- and vagotomy-induced nerve damage in adult rats. Neuroscience 2010; 167:1227-38. [PMID: 20197082 DOI: 10.1016/j.neuroscience.2010.02.049] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Revised: 02/19/2010] [Accepted: 02/19/2010] [Indexed: 11/25/2022]
Abstract
Previous reports show that vagal afferent innervation of the stomach eventually regenerates from surviving nodose ganglion (NG) neurons after subdiaphragmatic vagotomy. Systemic capsaicin treatment destroys gastric vagal afferent neurons expressing vanilloid receptor 1 (VR1). However, it is not known whether gastric innervation lost after neuronal destruction can be restored. Here, we report that capsaicin-induced damage of NG neurons innervating the stomach in adult rats is followed by restoration of vagal afferent projections. Specifically, we compared measures of neuronal plasticity in NG and vagi after subdiaphragmatic vagotomy or capsaicin treatment. The numbers of VR1-immunoreactive neurons projecting to the stomach were significantly reduced 10 days after either capsaicin treatment or vagotomy. However, the VR1-immunoreactive afferent innervation of the stomach was restored to levels exceeding those of vagotomized rats by 37 days after capsaicin, whereas neither total afferent innervation nor VR1-immunoreactive innervation reached control levels, even by 67 days after vagotomy. Capsaicin treatment significantly increased NG neuronal nitric oxide synthase (nNOS) immunoreactivity at 10 days after capsaicin, and this increase was sustained for the duration of the study, indicating higher nNOS demand in restoration of vagal projections. Vagotomy was associated with a much smaller increase in the number of nNOS-immunoreactive NG neurons, detectable only at 10 days after surgery. The number of nNOS-immunopositive gastric-projecting neurons was dramatically reduced 10 days after either capsaicin treatment or vagotomy but returned to the control level in both groups at 67 days. We found a significantly higher number of growth cones in capsaicin-treated animals compared with controls. Capsaicin significantly increased the number of nNOS-immunopositive and nNOS-immunonegative growth cones in NG at all time points. Vagotomy did not increase the number of nNOS(-) growth cones in NG. We conclude that capsaicin treatment may result in more significant restorative capacities than vagotomy, mainly because of sprouting of capsaicin-insensitive nerve fibers.
Collapse
Affiliation(s)
- V Ryu
- Department of Veterinary and Comparative Anatomy, Pharmacology, and Physiology, College of Veterinary Medicine, Washington State University, Pullman, WA 99163-6520, USA
| | | | | |
Collapse
|
22
|
Richardson PM, Miao T, Wu D, Zhang Y, Yeh J, Bo X. RESPONSES OF THE NERVE CELL BODY TO AXOTOMY. Neurosurgery 2009; 65:A74-9. [DOI: 10.1227/01.neu.0000352378.26755.c3] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Abstract
OBJECTIVE
Peripheral nerve injury causes retrograde changes in the damaged neurons, which are beneficial to axonal regeneration. Better understanding of the mechanisms of induction and mediation of these conditioning responses would help to design strategies to invoke stronger regenerative responses in neurons in situations when these responses are inadequate.
METHODS
Relevant literature is reviewed.
RESULTS
Experimental preparations that measure the influence of peripheral axotomy on regeneration in the central axons of primary sensory neurons are useful to examine mechanisms of conditioning neurons. Despite 4 decades of speculation, the nature of the damage signals from injured nerves that initiate axonal signals to the nerve cell body remains elusive. Members of the family of neuropoietic cytokines are clearly implicated, but what induces them is unknown. Multiple changes in gene regulation in axotomized neurons have been described, and dozens of growth-associated genes have been identified: neurotrophic factors, transcription factors, molecules participating in axonal transport, and molecules active in the growth cone. The mechanisms of interaction of a few regeneration-associated molecules with the signaling cascades that lead to actin and tubulin remodeling at the growth cone are understood in some detail. In animals, viral gene therapy to deliver regeneration-associated genes to neurons or other local measures to induce these genes can improve regeneration. A few pharmacological agents, administered systemically, have small beneficial effects on axonal regeneration.
CONCLUSION
Advances in laboratory research have provided knowledge of cell body responses to axotomy with clinical relevance.
Collapse
Affiliation(s)
- Peter M. Richardson
- Department of Neurosurgery, Queen Mary, University of London, The Royal London Hospital, London, England
| | - Tizong Miao
- Neuroscience Centre, Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry, Queen Mary, University of London, London, England
| | - Dongsheng Wu
- Neuroscience Centre, Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry, Queen Mary, University of London, London, England
| | - Yi Zhang
- Neuroscience Centre, Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry, Queen Mary, University of London, London, England
| | - John Yeh
- Department of Neurosurgery, Queen Mary, University of London, The Royal London Hospital, London, England
| | - Xuenong Bo
- Neuroscience Centre, Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry, Queen Mary, University of London, London, England
| |
Collapse
|
23
|
Itoh A, Horiuchi M, Bannerman P, Pleasure D, Itoh T. Impaired regenerative response of primary sensory neurons in ZPK/DLK gene-trap mice. Biochem Biophys Res Commun 2009; 383:258-62. [PMID: 19358824 DOI: 10.1016/j.bbrc.2009.04.009] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Accepted: 04/02/2009] [Indexed: 12/13/2022]
Abstract
Rapid and persistent activation of c-JUN is necessary for axonal regeneration after nerve injury, although upstream molecular events leading to c-JUN activation remain largely unknown. ZPK/DLK/MAP3K12 activates the c-Jun N-terminal kinase pathway at an apical level. We investigated axonal regeneration of the dorsal root ganglion (DRG) neurons of homozygous ZPK/DLK gene-trap mice. In vitro neurite extension assays using DRG explants from 14day-old mice revealed that neurite growth rates of the ZPK/DLK gene-trap DRG explants were reduced compared to those of the wild-type DRG explants. Three ZPK/DLK gene-trap mice which survived into adulthood were subjected to sciatic nerve axotomy. At 24h after axotomy, phosphorylated c-JUN-positive DRG neurons were significantly less frequent in ZPK/DLK gene-trap mice than in wild-type mice. These results indicate that ZPK/DLK is involved in regenerative responses of mammalian DRG neurons to axonal injury through activation of c-JUN.
Collapse
Affiliation(s)
- Aki Itoh
- Department of Neurology, School of Medicine, University of California, Davis, 601A Shriners Hospitals for Children Northern California, Sacramento, CA 95817-2215, USA
| | | | | | | | | |
Collapse
|
24
|
Abstract
Transcription is a molecular requisite for long-term synaptic plasticity and long-term memory formation. Thus, in the last several years, one main interest of molecular neuroscience has been the identification of families of transcription factors that are involved in both of these processes. Transcription is a highly regulated process that involves the combined interaction and function of chromatin and many other proteins, some of which are essential for the basal process of transcription, while others control the selective activation or repression of specific genes. These regulated interactions ultimately allow a sophisticated response to multiple environmental conditions, as well as control of spatial and temporal differences in gene expression. Evidence based on correlative changes in expression, genetic mutations, and targeted molecular inhibition of gene expression have shed light on the function of transcription in both synaptic plasticity and memory formation. This review provides a brief overview of experimental work showing that several families of transcription factors, including CREB, C/EBP, Egr, AP-1, and Rel, have essential functions in both processes. The results of this work suggest that patterns of transcription regulation represent the molecular signatures of long-term synaptic changes and memory formation.
Collapse
Affiliation(s)
- Cristina M Alberini
- Department of Neuroscience, Mount Sinai School of Medicine, New York, NY 10029, USA.
| |
Collapse
|
25
|
Raivich G. c-Jun expression, activation and function in neural cell death, inflammation and repair. J Neurochem 2008; 107:898-906. [PMID: 18793328 DOI: 10.1111/j.1471-4159.2008.05684.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Up-regulation of c-Jun is a common event in the developing, adult as well as in injured nervous system that serves as a model of transcriptional control of brain function. Functional studies employing in vivo strategies using gene deletion, targeted expression of dominant negative isoforms and pharmacological inhibitors all suggest a three pronged role of c-Jun action, exercising control over neural cell death and degeneration, in gliosis and inflammation as well as in plasticity and repair. In vitro, structural and molecular studies reveal several non-overlapping activation cascades via N-terminal c-Jun phosphorylation at serine 63 and 73 (Ser63, Ser73), and threonine 91 and 93 (Thr91, Thr93) residues, the dephosphorylation at Thr239, the p300-mediated lysine acetylation of the near C-terminal region (Lys268, Lys271, Lys 273), as well as the Jun-independent activities of the Jun N-terminal family of serine/threonine kinases, that regulate the different and disparate cellular responses. A better understanding of these non-overlapping roles in vivo could considerably increase the potential of pharmacological agents to improve neurological outcome following trauma, neonatal encephalopathy and stroke, as well as in neurodegenerative disease.
Collapse
Affiliation(s)
- Gennadij Raivich
- Department of Obstetrics and Gynaecology, Perinatal Brain Repair Group, EGA Institute of Women's Health, London, UK.
| |
Collapse
|
26
|
Raivich G, Makwana M. The making of successful axonal regeneration: Genes, molecules and signal transduction pathways. ACTA ACUST UNITED AC 2007; 53:287-311. [PMID: 17079020 DOI: 10.1016/j.brainresrev.2006.09.005] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2006] [Revised: 09/12/2006] [Accepted: 09/18/2006] [Indexed: 12/16/2022]
Abstract
Unlike its central counterpart, the peripheral nervous system is well known for its comparatively good potential for regeneration following nerve fiber injury. This ability is mirrored by the de novo expression or upregulation of a wide variety of molecules including transcription factors, growth-stimulating substances, cell adhesion molecules, intracellular signaling enzymes and proteins involved in regulating cell-surface cytoskeletal interactions, that promote neurite outgrowth in cultured neurons. However, their role in vivo is less known. Recent studies using neutralizing antibodies, gene inactivation and overexpression techniques have started to shed light on those endogenous molecules that play a key role in axonal outgrowth and the process of successful functional repair in the injured nervous system. The aim of the current review is to provide a summary on this rapidly growing field and the experimental techniques used to define the specific effects of candidate signaling molecules on axonal regeneration in vivo.
Collapse
Affiliation(s)
- Gennadij Raivich
- Perinatal Brain Repair Group, Department of Obstetrics and Gynaecology, University College London, 86-96 Chenies Mews, London, UK.
| | | |
Collapse
|
27
|
Waetzig V, Zhao Y, Herdegen T. The bright side of JNKs-Multitalented mediators in neuronal sprouting, brain development and nerve fiber regeneration. Prog Neurobiol 2006; 80:84-97. [PMID: 17045385 DOI: 10.1016/j.pneurobio.2006.08.002] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2006] [Revised: 08/25/2006] [Accepted: 08/30/2006] [Indexed: 12/11/2022]
Abstract
The c-Jun N-terminal kinases (JNKs) are important regulators of physiological and pathological processes in the central and peripheral nervous system. In general, JNKs are considered as mediators of neuronal degeneration in response to stress and injury. However, recent data have provided substantial evidence that JNKs are also essential for physiological and regenerative signalling in neurons. This review summarizes the importance of JNKs for neurite formation and outgrowth, brain development, dendritic architecture and regeneration of nerve fibers after injury. We discuss putative mechanisms which control the bipartite actions of individual JNK isoforms for neuronal death and repair after nerve fiber injury with a particular focus on the role of the transcription factor c-Jun.
Collapse
Affiliation(s)
- Vicki Waetzig
- Institute of Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Hospitalstrasse 4, 24105 Kiel, Germany
| | | | | |
Collapse
|
28
|
Schulte T, Brecht S, Herdegen T, Illert M, Mehdorn HM, Hamel W. Induction of immediate early gene expression by high-frequency stimulation of the subthalamic nucleus in rats. Neuroscience 2006; 138:1377-85. [PMID: 16460881 DOI: 10.1016/j.neuroscience.2005.12.034] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2005] [Revised: 11/28/2005] [Accepted: 12/01/2005] [Indexed: 11/16/2022]
Abstract
Deep brain stimulation is associated with delayed improvement of parkinsonian symptoms, such as hypokinesia with subthalamic nucleus stimulation, or dystonia with globus pallidus internus stimulation. The latency observed is better explained by molecular alterations than immediate electrophysiological processes, and clinical improvement may involve adaptive gene expression. Here, we have studied immediate early gene expression as fast molecular response to subthalamic nucleus stimulation. Bipolar electrodes were implanted bilaterally into the subthalamic nucleus of anesthetized male Wistar rats. High-frequency stimulation (130 Hz or 80 Hz, 60 micros, 300 microA) or low-frequency stimulation (5 Hz, 60 micros, 300 microA) was performed with the right electrode for 15, 60, 120, and 240 min whereas the silent left electrode served as negative control. Brains were fixed by transcardial perfusion and frozen sections were stained with polyclonal antibodies directed against three immediate early gene-encoded proteins, c-Fos, c-Jun, and Krox-24 (NGFI-A, Egr-1, Zif268, Tis8, Zenk). After 120 and 240 h, c-Fos immunoreactivity was strongly upregulated in subthalamic nucleus neurons on the stimulated site. In contrast, no c-Fos immunoreactivity was detected on the non-stimulated site except for single positive cells located in close proximity to the electrode tracks. Furthermore, c-Fos immunoreactivity was induced in subthalamic nucleus projection areas, such as primary and secondary motor cortex, primary somatosensory and insular cortex, lateral and medial globus pallidus, suprageniculate thalamic nucleus, pontine nuclei, medial geniculate nucleus, and substantia nigra. Similarly, c-Jun and Krox-24 were induced at the site of stimulation and in projection areas following high-frequency subthalamic nucleus stimulation. Whereas high frequency stimulation with 80 Hz was similarly effective none of the three immediate early gene-encoded proteins was induced with low-frequency stimulation (5 Hz) for 4 h. This is in accordance with the therapeutic effects of deep brain stimulation which are only elicited with high frequency stimulation. Our data provide evidence that immediate early gene expression in the subthalamic nucleus is rapidly and substantially induced by high-frequency stimulation. The induction of immediate early genes in projection sites suggests ipsilateral transsynaptic modulation of neuronal activity.
Collapse
Affiliation(s)
- T Schulte
- Department of Neurosurgery, Universityhospital Schleswig-Holstein/Campus Kiel, Germany
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
Peripheral nerve injury is normally followed by a robust regenerative response. Here we describe the early changes associated with injury from the initial rise in intracellular calcium and the subsequent activation of transcription factors and cytokines leading to an inflammatory reaction, and the expression of growth factors, cytokines, neuropeptides, and other secreted molecules involved in cell-to-cell communication promoting regeneration and neurite outgrowth. The aim of this review is to summarize the molecular mechanisms that play a part in executing successful regeneration.
Collapse
Affiliation(s)
- Milan Makwana
- Centre for Perinatal Brain Protection & Repair, Department of Obstetrics and Gynaecology, University College London, UK
| | | |
Collapse
|
30
|
|
31
|
Lindwall C, Dahlin L, Lundborg G, Kanje M. Inhibition of c-Jun phosphorylation reduces axonal outgrowth of adult rat nodose ganglia and dorsal root ganglia sensory neurons. Mol Cell Neurosci 2004; 27:267-79. [PMID: 15519242 DOI: 10.1016/j.mcn.2004.07.001] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2004] [Revised: 05/28/2004] [Accepted: 07/02/2004] [Indexed: 10/26/2022] Open
Abstract
The role of c-Jun activation for survival and regeneration of sensory neurons is unclear. Here we report that c-Jun N-terminal kinase (JNK)-mediated c-Jun activation is important for axonal outgrowth of sensory neurons in rat nodose and dorsal root ganglia (DRG). Peripheral severance of the vagus or the sciatic nerve resulted in a massive and rapid, but transient increase of the activated JNK (p-JNK) in neuronal nuclei, followed by c-Jun phosphorylation and activating transcription factor-3 (ATF3) induction. JNK inhibition by the selective JNK inhibitors SP600125 and (D)-JNKI1 did not affect neuronal survival in explanted or dissociated ganglia, but dramatically reduced axonal outgrowth, c-Jun activation, and ATF3 induction. Using retrograde labeling, we demonstrated that activated c-Jun (p-c-Jun) and ATF3 were associated with regenerative neurons. Taken together, our results suggest that JNK-mediated c-Jun activation is one of the first cell body reactions in response to nerve injury and that this activation and subsequent ATF3 induction are associated with axonal outgrowth.
Collapse
Affiliation(s)
- Charlotta Lindwall
- Department of Cell and Organism Biology, Lund University, SE-223 62 Lund, Sweden.
| | | | | | | |
Collapse
|
32
|
Phillips RJ, Baronowsky EA, Powley TL. Long-term regeneration of abdominal vagus: efferents fail while afferents succeed. J Comp Neurol 2003; 455:222-37. [PMID: 12454987 DOI: 10.1002/cne.10470] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Vagal afferents regenerate, by 18 weeks after subdiaphragmatic transection, to reinnervate the gut and to differentiate into the two types of terminals normally found in the smooth muscle wall of the gastrointestinal (GI) tract (Phillips et al. [2000] J Comp Neurol. 421:325-346). Regeneration, however, is neither complete nor entirely accurate by 18 weeks. Moreover, the capacity of the vagal efferents to reinnervate the GI tract under comparable conditions has not been evaluated. Therefore, to determine whether a more extended postaxotomy survival interval would (1). result in more extensive reinnervation of smooth muscle, (2). facilitate correction of the inaccuracies of the regenerated axons and terminals, and (3). yield motor as well as sensory reinnervation of GI targets, Sprague-Dawley rats received either complete subdiaphragmatic vagotomies (n = 18) or sham surgeries (n = 12). Physiological endpoints that might normalize as vagal elements regenerated, including body weight, daily food intake, size of first daily meal, and metabolic efficiency, were monitored. At 45 weeks after the vagotomies, the animals were randomly assigned to afferent (wheat germ agglutinin-horseradish peroxidase) or efferent (cholera toxin subunit B-horseradish peroxidase) mapping conditions, and labeled axons and terminals in the stomach and first 8 cm of the small intestine were inventoried in whole-mounts. Afferent regeneration was more extensive at 45 weeks than previously observed at 18 weeks after surgery; however, the amount of GI innervation was still not comparable to the intact pattern of the sham rats. Furthermore, abnormal patterns of sensory organization occurred throughout the reinnervated field, with small bundles of axons forming complex tangles and some individual axons terminating in ectopic locations. The presence of growth cone profiles suggested that vagal reorganization was ongoing even 45 weeks after surgery. In contrast to this relatively extensive, albeit incomplete, sensory reinnervation of the gut, motor fibers had failed to reinnervate the GI tract. Thus, dramatic differences exist in the regenerative capacities of the sensory and motor arms of the vagus under the same surgical and maintenance conditions. Furthermore, the functional measures of disordered energy regulation did not normalize over the 45 weeks during which afferent but not efferent innervation was restored.
Collapse
Affiliation(s)
- Robert J Phillips
- Purdue University, Department of Psychological Sciences, West Lafayette, Indiana 47907, USA.
| | | | | |
Collapse
|
33
|
Ji J, Dheen ST, Tay SSW. Molecular analysis of the vagal motoneuronal degeneration after right vagotomy. J Neurosci Res 2002; 69:406-17. [PMID: 12125081 DOI: 10.1002/jnr.10300] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The aim of this study was to investigate the vagal motoneuronal degeneration after right vagotomy using in situ hybridization, RT-PCR, and immunohistochemistry methods. The morphology of the vagal motoneurons in dorsal motor nucleus of the vagus nerve (DMV) and nucleus of ambiguus (NA) after right vagotomy was examined by using Nissl staing and TUNEL. The expression of inducible nitric oxide synthase (iNOS), bcl-2, bax, and caspase-3 in DMV and NA of rats after right vagotomy was studied. Additionally, the involvement of the N-methyl-D-aspartate (NMDA) receptor-calcium-neuronal nitric oxide synthase (nNOS) pathway in the vagal motoneuronal degeneration was addressed by double-immunolabeling analysis of nNOS with NMDAR1 and calbindin D28K in right-vagotomized rats. The neurons in right DMV and NA displayed a darkly stained, shrunken morphology at 1 day and 5 days following right vagotomy as shown by Nissl staining. Quantitative analysis revealed that, at 1 day and 5 days following right vagotomy, the number of neurons in right DMV, but not NA, was significantly reduced in comparison with that of control rats. Occasional TUNEL-positive neurons were detected in right DMV of rat at 1 day after right vagotomy. The expression of iNOS protein and mRNA was absent in DMV and NA of control rats. However, the iNOS mRNA expression was induced bilaterally in DMV and NA at 1 day postoperation and continued to be up-regulated until 5 days after vagotomy as shown by in situ hybridization. Immunohistochemistry analysis also showed the increased expression of iNOS in bilateral DMV and NA of vagotomized rats. RT-PCR analysis revealed the enhanced bcl-2 and reduced bax mRNA levels and subsequent up-regulation of both bcl-2 and bax mRNA in right sides of the vagotomized brainstems at 1 day and 5 days postoperation, respectively. In situ hybridization analysis confirmed the up-regulation of bcl-2 and bax mRNA in right DMV and NA of the rats at 5 days following operation. Immunohistochemistry analysis showed up-regulated Bcl-2 immunoreactivity and undetectable changes in Bax immunoreactivity in DMV and NA of rats at 1 day after vagotomy, whereas enhancement of both Bcl-2 and Bax immunoreactivity was observed at 5 days postoperation. In addition, the caspase-3 mRNA level was elevated ipsilaterally in DMV and NA at 1 day and 5 days following right vagotomy. Double-immunofluorescence analysis showed complete colocalization of nNOS with NMDAR1 and with calbindin in ipsilateral DMV and NA at 10 days following right vagotomy. This study suggests that the signal pathway for NMDAR1-calcium-nNOS and the up-regulation of iNOS in DMV and NA may be involved in the vagal motor neurodgeneration after right vagotomy. Furthermore, our results imply that the apoptosis pathway mediated by Bcl-2, Bax, and caspase-3 may be activated in vagal motoneurons after right vagotomy.
Collapse
Affiliation(s)
- Junfeng Ji
- Department of Anatomy, National University of Singapore, Singapore
| | | | | |
Collapse
|
34
|
Säljö A, Bao F, Jingshan S, Hamberger A, Hansson HA, Haglid KG. Exposure to short-lasting impulse noise causes neuronal c-Jun expression and induction of apoptosis in the adult rat brain. J Neurotrauma 2002; 19:985-91. [PMID: 12225658 DOI: 10.1089/089771502320317131] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Exposure to impulse noise, above a certain intensity, is harmful to auditory function. Effects of impulse noise on the central nervous system (CNS) are largely unexplored, and there is little information on critical threshold values and time factors. We have recently shown that neurofilament proteins are affected in the cerebral cortex and the hippocampus. Now we show that impulse noise induces expression of the immediate early gene c-Jun products, proposed to play a role in the initiation of neuronal death, and apoptosis as revealed by TUNEL staining. Rat brains were investigated immunohistochemically 2 h to 21 days after exposure to impulse noise of 198 dB or 202 dB. c-Jun was expressed in neuronal perikarya in layers II-VI of the temporal cortex, the cingulate and the piriform cortices at 2 h to 21 days after both exposure levels. Granule neurons of the dentate gyrus and the CA1-3 in the hippocampus pyramidal neurons were similarly affected. The elevated expression of c-Jun products remained high at all postexposure times. TUNEL staining was positive among the same nerve cell populations 6 h after exposure and persisted even at 7 days at both exposure levels.
Collapse
Affiliation(s)
- Annette Säljö
- Department of Anatomy and Cell Biology, Göteborg University, Göteborg, Sweden.
| | | | | | | | | | | |
Collapse
|
35
|
Groneberg DA, Wiegand S, Dinh QT, Peiser C, Springer J, Fischer A. Expression of immediate early genes in sensory ganglia. Neurochem Res 2001; 26:1113-7. [PMID: 11700953 DOI: 10.1023/a:1012366721845] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
C-Jun and c-Fos belong to the family of immediate early genes. Apart from their role as transcription factors, a basal expression was shown for them in central nervous system tissues. The expression of c-Jun and c-Fos in sensory ganglia of guinea pig, rat and murine sensory ganglia was examined under normal, unstimulated conditions by quantitative double-immunohistochemistry. 4.6 +/- 2.8% of neuron-specific protein gene-product 9.5 -positive cells in nodose ganglia, 51.6 +/- 2.1% in jugular ganglia, 46.4 +/- 3.0% in trigeminal ganglia and 42.5 +/- 1.3% of cervical dorsal root ganglia neurons were positive for c-Jun in the guinea pig (less than 1% for c-Fos). In rat and mouse, less than 1% of the sensory neurons exhibited c-Jun and c-Fos-immunoreactivity. The high basal expression of c-Jun in guinea pig sensory neurons suggests that in this species the presence of c-Jun does not only depend on specific stimulation and is not exclusively associated with neuronal plasticity of gene expression and functional changes.
Collapse
Affiliation(s)
- D A Groneberg
- Division of Allergy Research, Biomedical Research Center, Charité Medical School, Humboldt University of Berlin, Germany.
| | | | | | | | | | | |
Collapse
|
36
|
Lewis MW, Travagli RA. Effects of substance P on identified neurons of the rat dorsal motor nucleus of the vagus. Am J Physiol Gastrointest Liver Physiol 2001; 281:G164-72. [PMID: 11408269 PMCID: PMC3062486 DOI: 10.1152/ajpgi.2001.281.1.g164] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Previous evidence suggests that substance P (SP) activates subpopulations of neurons within the dorsal motor nucleus of the vagus (DMV). In this study we aimed at identifying these subpopulations in relation to their gastrointestinal projection organs or vagal branches and characterizing pharmacologically the SP response. Using whole cell patch-clamp recordings from identified gastrointestinal-projecting vagal motoneurons, we found that SP induced an inward current in all neuronal groups except for cecum-projecting cells. The lowest percentage of SP-responding neurons was found in fundus-projecting cells, where SP also had a concentration-response curve that was shifted to the left (P < 0.05). Independently from the projections, the SP response was reduced by sendide and MEN 10,376 and mimicked by a combination of [Sar(9)-Met(O(2))(11)]SP and alpha-neurokinin. SP and alpha-neurokinin also increased the frequency, but not the amplitude, of postsynaptic currents. In conclusion, we demonstrated that SP induces both pre- and postsynaptic effects on DMV neurons via activation of neurokinin NK(1) and NK(2) receptors. The magnitude of the SP response was correlated to the peripheral target organ.
Collapse
Affiliation(s)
- M W Lewis
- Division of Gastroenterology and Department of Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | |
Collapse
|
37
|
Galiano M, Liu ZQ, Kalla R, Bohatschek M, Koppius A, Gschwendtner A, Xu S, Werner A, Kloss CU, Jones LL, Bluethmann H, Raivich G. Interleukin-6 (IL6) and cellular response to facial nerve injury: effects on lymphocyte recruitment, early microglial activation and axonal outgrowth in IL6-deficient mice. Eur J Neurosci 2001; 14:327-41. [PMID: 11553283 DOI: 10.1046/j.0953-816x.2001.01647.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Nerve injury triggers numerous changes in the injured neurons and surrounding non-neuronal cells. Of particular interest are molecular signals that play a role in the overall orchestration of this multifaceted cellular response. Here we investigated the function of interleukin-6 (IL6), a multifunctional neurotrophin and cytokine rapidly expressed in the injured nervous system, using the facial axotomy model in IL6-deficient mice and wild-type controls. Transgenic deletion of IL6 caused a massive decrease in the recruitment of CD3-positive T-lymphocytes and early microglial activation during the first 4 days after injury in the axotomized facial nucleus. This was accompanied by a more moderate reduction in peripheral regeneration at day 4, lymphocyte recruitment (day 14) and enhanced perikaryal sprouting (day 14). Motoneuron cell death, phagocytosis by microglial cells and recruitment of granulocytes and macrophages into injured peripheral nerve were not affected. In summary, IL6 lead to a variety of effects on the cellular response to neural trauma. However, the particularly strong actions on lymphocytes and microglia suggest that this cytokine plays a central role in the initiation of immune surveillance in the injured central nervous system.
Collapse
Affiliation(s)
- M Galiano
- Department of Neuromorphology, Max-Planck Institute for Neurobiology, Am Klopferspitz 18A, D-82152 Martinsried, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Manabe T, Kuramoto N, Nakamichi N, Aramachi K, Baba K, Hirai T, Yoneyama M, Yoneda Y. Degradation of c-Fos protein expressed by N-methyl-D-aspartic acid in nuclear fractions of murine hippocampus. Brain Res 2001; 905:34-43. [PMID: 11423077 DOI: 10.1016/s0006-8993(01)02464-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
In both nuclear and cytosolic fractions of murine hippocampus, constitutive expression was seen with Fra-2 protein, but not with other Fos family members tested including c-Fos, Fos-B and Fra-1 proteins. Fos-B protein was only detected in nuclear fractions. The systemic administration of N-methyl-D-aspartic acid (NMDA) induced marked and transient expression of c-Fos protein, but not other family members, in both hippocampal fractions 2 h later. In vitro incubation at 30 degrees C led to more rapid degradation of inducible c-Fos protein than constitutive Fra-2 protein in nuclear fractions obtained 2 h after the administration of NMDA, without significantly affecting that of both member proteins in cytosolic fractions. The addition of phosphatase inhibitors significantly delayed the initial degradation rate of inducible c-Fos protein, with concomitant facilitation of that of constitutive Fra-2 protein, in nuclear fractions. The addition of protease inhibitors also delayed the initial degradation of constitutive Fra-2 protein, without markedly altering that of inducible c-Fos protein, in nuclear fractions. Immunoprecipitation analysis revealed that NMDA induced phosphorylation of c-Fos protein on tyrosine residues in nuclear fractions to a lesser extent than that on serine residues 2 h after administration. These results suggest that NMDA signals may be propagated to the nucleus to induce both expression and degradation of c-Fos protein through a molecular mechanism associated with phosphorylation on serine and/or tyrosine residues in murine hippocampus.
Collapse
Affiliation(s)
- T Manabe
- Department of Pharmacology, Setsunan University, Hirakata, Osaka 573-0101, Japan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Kalla R, Liu Z, Xu S, Koppius A, Imai Y, Kloss CU, Kohsaka S, Gschwendtner A, Möller JC, Werner A, Raivich G. Microglia and the early phase of immune surveillance in the axotomized facial motor nucleus: Impaired microglial activation and lymphocyte recruitment but no effect on neuronal survival or axonal regeneration in macrophage-colony stimulating factor-defici. J Comp Neurol 2001. [DOI: 10.1002/cne.1060] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
40
|
Casanovas A, Ribera J, Hager G, Kreutzberg GW, Esquerda JE. c-Jun regulation in rat neonatal motoneurons postaxotomy. J Neurosci Res 2001; 63:469-79. [PMID: 11241582 DOI: 10.1002/jnr.1041] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Motoneurons respond to peripheral nerve transection by either regenerative or degenerative events depending on their state of maturation. Since the expression of c-Jun has been involved in the early signalling of the regenerative process that follows nerve transection in adults, we have investigated c-Jun on rat neonatal axotomized motoneurons during the period in which neuronal death is induced. Changes in levels of c-Jun protein and its mRNA were determined by means of quantitative immunocytochemistry and in situ hybridization. Three hours after nerve transection performed on postnatal day (P)3, c-Jun protein and mRNA is induced in axotomized spinal cord motoneurons, and high levels were reached between 1 and 10 days after. This response is associated with a detectable c-Jun activation by phosphorylation on serine 63. No changes were found in the levels of activating transcription factor -2. Most of dying motoneurons were not labelled by either a specific c-Jun antibody or a c-jun mRNA probe. However, dying motoneurons were specifically stained by a polyclonal anti c-Jun antibody, indicating that some c-Jun antibodies react with unknown epitopes, probably distinct from c-Jun p39, that are specifically associated with apoptosis.
Collapse
Affiliation(s)
- A Casanovas
- Unitat de Neurobiologia Cellular, Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina, Universitat de Lleida, Spain
| | | | | | | | | |
Collapse
|
41
|
Runnerstam M, Bao F, Huang Y, Shi J, Gutierrez E, Hamberger A, Hansson HA, Viano D, Haglid K. A new model for diffuse brain injury by rotational acceleration: II. Effects on extracellular glutamate, intracranial pressure, and neuronal apoptosis. J Neurotrauma 2001; 18:259-73. [PMID: 11284547 DOI: 10.1089/08977150151070892] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The aim of this study is to monitor excitatory amino acids (EAAs) in the extracellular fluids of the brain and to characterize regional neuronal damage in a new experimental model for brain injury, in which rabbits were exposed to 180-260 krad/s2 rotational head acceleration. This loading causes extensive subarachnoid hemorrhage, focal tissue bleeding, reactive astrocytosis, and axonal damage. Animals were monitored for intracranial pressure (ICP) and for amino acids in the extracellular fluids. Immunohistochemistry was used to study expression of the gene c-Jun and apoptosis with the terminal deoxynucleotidyl transferase nick-end labeling (TUNEL) technique. Extracellular glutamate, glycine, and taurine increased significantly in the hippocampus within a few hours and remained high after 24 h. Neuronal nuclei in the granule layers of the hippocampus and cerebellum were positive for c-Jun after 24 h. Little immunoreactivity was detected in the cerebral cortex. c-Jun-positive neuronal perikarya and processes were found in granule and pyramidal CA4 layers of the hippocampus and among the Purkinje cells of the cerebellum. Also some microglial cells stained positively for c-Jun. TUNEL reactivity was most intense at 10 days after trauma and was extensive in neurons of the cerebral cortex, hippocampus, and cerebellum. The initial response of the brain after rotational head injury involves brain edema after 24 h and an excitotoxic neuronal microenvironment in the first hour, which leads to extensive delayed neuronal cell death by apoptosis necrosis in the cerebral cortex, hippocampus and cerebellum.
Collapse
Affiliation(s)
- M Runnerstam
- Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Edelstein K, Beaulé C, D'Abramo R, Amir S. Expression profiles of JunB and c-Fos proteins in the rat circadian system. Brain Res 2000; 870:54-65. [PMID: 10869501 DOI: 10.1016/s0006-8993(00)02401-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The immediate-early genes c-Fos and JunB are implicated in light signaling within the suprachiasmatic nucleus (SCN), the mammalian circadian clock. Light induces phase-dependent expression of c-Fos and JunB within the retinorecipient SCN. In the absence of light, rhythmic expression of these genes has been observed in the dorsal region of the SCN with peak expression observed near dawn. The present study examined the pattern of induction of c-Fos and JunB in the SCN and intergeniculate leaflet (IGL) of rats housed in constant conditions, under light-dark cycles, or in dark-adapted light-stimulated animals. In contrast with previous studies, no evidence of expression of c-Fos and JunB was observed within the dorsal SCN, regardless of circadian time. Strain differences could account for the absence of rhythmic JunB expression, whereas the use of a monoclonal anti-c-Fos antibody in the present study may account for the absence of dorsal SCN c-Fos staining. The profile of light-induced c-Fos and JunB expression was consistent with previous observations. In the SCN, light-induced expression of c-Fos and JunB was phase dependent, whereas in the IGL light-induced both c-Fos and JunB regardless of circadian time.
Collapse
Affiliation(s)
- K Edelstein
- Center for Studies in Behavioral Neurobiology, Concordia University, 1455 de Maisonneuve Blvd. West, Montréal, Québec, Canada H3G 1M8
| | | | | | | |
Collapse
|
43
|
Schmitt AB, Breuer S, Voell M, Schwaiger FW, Spitzer C, Pech K, Brook GA, Noth J, Kreutzberg GW, Nacimiento W. GAP-43 (B-50) and C-Jun are up-regulated in axotomized neurons of Clarke's nucleus after spinal cord injury in the adult rat. Neurobiol Dis 1999; 6:122-30. [PMID: 10343327 DOI: 10.1006/nbdi.1998.0231] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The growth-associated protein GAP-43 (B-50) and the transcription factor C-Jun are involved in regeneration of the injured nervous system. In this study, we investigated the possibility of the induction of GAP-43 and C-Jun in axotomized neurons of Clarke's nucleus (CN) in adult rats, of which a large population undergoes degeneration several weeks after a low thoracic lateral funiculotomy of the spinal cord. In situ hybridization and immunohistochemistry revealed a transient up-regulation of GAP-43 mRNA, C-Jun protein, and its activated, phosphorylated form, peaking around 7 days after injury in preferentially large diameter CN-neurons ipsilateral and caudal to the lesion. Our results document that some populations of axotomized central nervous system neurons, similar to axotomized regenerating neurons of the peripheral nervous system, can up-regulate GAP-43 and C-Jun, even if they are destined to degenerate. This might reflect a transient regenerative capacity, which fails over time.
Collapse
Affiliation(s)
- A B Schmitt
- Department of Neurology, Aachen University Medical School, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Gonz�lez-Hern�ndez T, Rustioni A. Nitric oxide synthase and growth-associated protein are coexpressed in primary sensory neurons after peripheral injury. J Comp Neurol 1999. [DOI: 10.1002/(sici)1096-9861(19990201)404:1<64::aid-cne5>3.0.co;2-m] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
45
|
Herdegen T, Leah JD. Inducible and constitutive transcription factors in the mammalian nervous system: control of gene expression by Jun, Fos and Krox, and CREB/ATF proteins. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 1998; 28:370-490. [PMID: 9858769 DOI: 10.1016/s0165-0173(98)00018-6] [Citation(s) in RCA: 1061] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This article reviews findings up to the end of 1997 about the inducible transcription factors (ITFs) c-Jun, JunB, JunD, c-Fos, FosB, Fra-1, Fra-2, Krox-20 (Egr-2) and Krox-24 (NGFI-A, Egr-1, Zif268); and the constitutive transcription factors (CTFs) CREB, CREM, ATF-2 and SRF as they pertain to gene expression in the mammalian nervous system. In the first part we consider basic facts about the expression and activity of these transcription factors: the organization of the encoding genes and their promoters, the second messenger cascades converging on their regulatory promoter sites, the control of their transcription, the binding to dimeric partners and to specific DNA sequences, their trans-activation potential, and their posttranslational modifications. In the second part we describe the expression and possible roles of these transcription factors in neural tissue: in the quiescent brain, during pre- and postnatal development, following sensory stimulation, nerve transection (axotomy), neurodegeneration and apoptosis, hypoxia-ischemia, generalized and limbic seizures, long-term potentiation and learning, drug dependence and withdrawal, and following stimulation by neurotransmitters, hormones and neurotrophins. We also describe their expression and possible roles in glial cells. Finally, we discuss the relevance of their expression for nervous system functioning under normal and patho-physiological conditions.
Collapse
Affiliation(s)
- T Herdegen
- Institute of Pharmacology, University of Kiel, Hospitalstrasse 4, 24105, Kiel,
| | | |
Collapse
|
46
|
Hopkins DA, Plumier JC, Currie RW. Induction of the 27-kDa heat shock protein (Hsp27) in the rat medulla oblongata after vagus nerve injury. Exp Neurol 1998; 153:173-83. [PMID: 9784277 DOI: 10.1006/exnr.1998.6870] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The 27-kDa heat shock protein (Hsp27) is constitutively expressed in motor and sensory neurons of the brainstem. Hsp27 is also rapidly induced in the nervous system following oxidative and cellular metabolic stress. In this study, we examined the distribution of Hsp27 in the rat medulla oblongata by means of immunohistochemistry after the vagus nerve was cut or crushed. After vagal injury, rats were allowed to survive for 6, 12, 24 h, 2, 4, 7, 10, 14, 30, or 90 days. Vagus nerve lesions resulted in a time-dependent up-regulation of Hsp27 in vagal motor and nodose ganglion sensory neurons that expressed Hsp27 constitutively and de novo induction in neurons that did not express Hsp27 constitutively. In the dorsal motor nucleus of the vagus nerve (DMV) and nucleus ambiguus, the levels of Hsp27 in motor neurons were elevated within 24 h of injury and persisted for up to 90 days. Vagal afferents to the nucleus of the tractus solitarius (NTS) and area postrema showed increases in Hsp27 levels within 4 days that were still present 90 days postinjury. In addition, increases in Hsp27 staining of axons in the NTS and DMV suggest that vagus nerve injury resulted in sprouting of afferent axons and spread into areas of the dorsal vagal complex not normally innervated by the vagus. Our observations are consistent with the possibility that Hsp27 plays a role in long-term survival of distinct subpopulations of injured vagal motor and sensory neurons.
Collapse
Affiliation(s)
- D A Hopkins
- Department of Anatomy and Neurobiology, Dalhousie University, Halifax, Nova Scota, B3H 4H7, Canada
| | | | | |
Collapse
|
47
|
Grimm R, Tischmeyer W. Complex patterns of immediate early gene induction in rat brain following brightness discrimination training and pseudotraining. Behav Brain Res 1997; 84:109-16. [PMID: 9079777 DOI: 10.1016/s0166-4328(97)83330-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Following training of rats on a footshock-motivated brightness discrimination task in a Y-maze, different sets of transcription factor encoding immediate early genes (IEGs) were induced in anatomically distinct brain regions. As revealed by Northern analysis, mRNA levels of c-fos, jun-B and zif/268 increased in the hippocampus, while the expression of c-jun remained unchanged over a period of 7 h. In the cerebral cortex, c-jun was induced in addition to the other genes examined. In contrast, only c-fos, but not c-jun or zif/268 mRNAs were increased in the cerebellum. The induction of IEGs was rapid and transient, reaching maximal levels immediately after training and returning to basal levels within 2 h. Similar spatiotemporal expression patterns were observed in rats that received identical, but unpaired, stimuli in a pseudotraining procedure. Our results suggest that the initial prerequisites of learning, such as stimulus novelty, lead to an increased expression of IEG mRNAs after training and pseudotraining as an early necessary but not sufficient precondition for memory consolidation. Additional converging inputs might control at the transcriptional, translational or post-translational level the synthesis and biological effectiveness of proteins necessary to complete the formation of the memory trace in trained animals.
Collapse
Affiliation(s)
- R Grimm
- Federal Institute for Neurobiology, Magdeburg, Germany
| | | |
Collapse
|
48
|
Abstract
A large proportion of neurons die during normal development of the nervous system via an active process known as apoptosis. We counted the total number of neurons and apoptotic neurons in the superior cervical ganglion of the GH Wistar rat strain, which possesses a neurotrophic deficit leading to excessive perinatal cell death, and in its normal counterpart (N) by using the optical disector method to quantify the extent of apoptosis during postnatal development. Total neuron numbers fell between postnatal days 3 and 14 by 10 and 40% in N and GH, respectively. In GH ganglia, 1.5% of neurons were apoptotic at any given time, as determined by the presence of condensed chromatin clumps. Some types of cell death have been associated with expression of the immediate-early genes c-fos and c-jun. Therefore, we used histological and immunocytochemical techniques to characterise individual neurons and to detect the products of these immediate-early genes during developmental cell death. All apoptotic cells were immunopositive for c-jun protein, whereas no c-jun protein was detected in nonapoptotic cells. Conversely, members of the fos family of transcription factors were detected in the nucleus of 60% of nonapoptotic cells but in only a minor proportion of cells undergoing apoptosis. These results indicate that c-jun occurs in neurons that are committed to die. This is the first situation in which the presence of jun protein has been correlated with normal programmed cell death in individual apoptotic neurons.
Collapse
Affiliation(s)
- A Messina
- Department of Physiology, Melbourne University, Royal Melbourne Hospital, Royal Parade Parkville, Australia
| | | | | |
Collapse
|
49
|
Martín G, Seguí J, Díaz-Villoslada P, Montalbán X, Planas AM, Ferrer I. Jun expression is found in neurons located in the vicinity of subacute plaques in patients with multiple sclerosis. Neurosci Lett 1996; 212:95-8. [PMID: 8832647 DOI: 10.1016/0304-3940(96)12776-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The expression of members of the Fos and Jun families is examined by immunohistochemistry in the brains of two patients with multiple sclerosis (MS). Strong c-Jun immunoreactivity is observed in the cytoplasm of neurons located in the vicinity of subacute plaques, but not in neurons of brain compartments not compromised by MS and in the neighborhood of chronic plaques. Strong c-Jun immunoreactivity also contrast with weak c-Jun immunoreactivity of corresponding neurons in control brains. In addition, punctate Jun D immunoreactivity is observed in the neuropil of the same areas that express c-Jun. No immunoreaction is found to c-Fos, Fos-related antigens and Jun B in these areas. The present results suggest that selective Jun neuronal expression in the vicinity of subacute plaques is a consistent reaction to demyelination and axonal damage.
Collapse
Affiliation(s)
- G Martín
- Unitat de Neuropatología, Hospital Prínceps d'Espanya, Universitat de Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
50
|
Ferrer I, Olive M, Ribera J, Planas AM. Naturally occurring (programmed) and radiation-induced apoptosis are associated with selective c-Jun expression in the developing rat brain. Eur J Neurosci 1996; 8:1286-98. [PMID: 8752598 DOI: 10.1111/j.1460-9568.1996.tb01297.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Expression of the different members of transcription factors Fos and Jun was examined in the developing rat brain. Constitutive expression of c-Fos, Fos-related antigens, Jun B and Jun D, as revealed with immunohistochemistry, is higher and more widely distributed in the developing rat brain than in the adult. Selective strong c-Jun expression is observed in the cytoplasm and nuclei of apoptotic cells during the whole process of naturally occurring (programmed) cell death. Cells expressing strong c-Jun immunoreactivity are undetermined cells, neurons and astrocytes. Selective c-Jun expression is also observed following ionizing radiation in rats aged 3 days. Induction of c-jun mRNA, as revealed with in situ hybridization, occurs between 5 and 15 min following gamma-irradiation. Strong c-Jun protein expression appears at 2 h, peaks at 6 h and decreases thereafter to reach normal levels 48 h after gamma-ray exposure. Strong c-Jun protein expression is coincidental with endonuclease activation, as revealed with the method of in situ labelling of nuclear DNA fragmentation, and is restricted to apoptotic cells. Cycloheximide injection at the time of irradiation blocks c-Jun expression, indicating that c-Jun immunoreactivity is attributable to de novo protein synthesis. These observations demonstrate in vivo selective strong c-Jun expression associated with programmed cell death and ionizing radiation-induced apoptosis in the developing rat brain.
Collapse
Affiliation(s)
- I Ferrer
- Unitat de Neuropatalogia, Servei d'Anatomia Patològica, Hospital Princeps d'Espanya, Universitat de Barcelona, 08907 Hospitalet de Llobregat, Spain
| | | | | | | |
Collapse
|