1
|
Lisek M, Tomczak J, Boczek T, Zylinska L. Calcium-Associated Proteins in Neuroregeneration. Biomolecules 2024; 14:183. [PMID: 38397420 PMCID: PMC10887043 DOI: 10.3390/biom14020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
The dysregulation of intracellular calcium levels is a critical factor in neurodegeneration, leading to the aberrant activation of calcium-dependent processes and, ultimately, cell death. Ca2+ signals vary in magnitude, duration, and the type of neuron affected. A moderate Ca2+ concentration can initiate certain cellular repair pathways and promote neuroregeneration. While the peripheral nervous system exhibits an intrinsic regenerative capability, the central nervous system has limited self-repair potential. There is evidence that significant variations exist in evoked calcium responses and axonal regeneration among neurons, and individual differences in regenerative capacity are apparent even within the same type of neurons. Furthermore, some studies have shown that neuronal activity could serve as a potent regulator of this process. The spatio-temporal patterns of calcium dynamics are intricately controlled by a variety of proteins, including channels, ion pumps, enzymes, and various calcium-binding proteins, each of which can exert either positive or negative effects on neural repair, depending on the cellular context. In this concise review, we focus on several calcium-associated proteins such as CaM kinase II, GAP-43, oncomodulin, caldendrin, calneuron, and NCS-1 in order to elaborate on their roles in the intrinsic mechanisms governing neuronal regeneration following traumatic damage processes.
Collapse
Affiliation(s)
| | | | | | - Ludmila Zylinska
- Department of Molecular Neurochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (M.L.); (J.T.); (T.B.)
| |
Collapse
|
2
|
Štepánková K, Chudíčková M, Šimková Z, Martinez-Varea N, Kubinová Š, Urdzíková LM, Jendelová P, Kwok JCF. Low oral dose of 4-methylumbelliferone reduces glial scar but is insufficient to induce functional recovery after spinal cord injury. Sci Rep 2023; 13:19183. [PMID: 37932336 PMCID: PMC10628150 DOI: 10.1038/s41598-023-46539-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 11/02/2023] [Indexed: 11/08/2023] Open
Abstract
Spinal cord injury (SCI) induces the upregulation of chondroitin sulfate proteoglycans (CSPGs) at the glial scar and inhibits neuroregeneration. Under normal physiological condition, CSPGs interact with hyaluronan (HA) and other extracellular matrix on the neuronal surface forming a macromolecular structure called perineuronal nets (PNNs) which regulate neuroplasticity. 4-methylumbelliferone (4-MU) is a known inhibitor for HA synthesis but has not been tested in SCI. We first tested the effect of 4-MU in HA reduction in uninjured rats. After 8 weeks of 4-MU administration at a dose of 1.2 g/kg/day, we have not only observed a reduction of HA in the uninjured spinal cords but also a down-regulation of CS glycosaminoglycans (CS-GAGs). In order to assess the effect of 4-MU in chronic SCI, six weeks after Th8 spinal contusion injury, rats were fed with 4-MU or placebo for 8 weeks in combination with daily treadmill rehabilitation for 16 weeks to promote neuroplasticity. 4-MU treatment reduced the HA synthesis by astrocytes around the lesion site and increased sprouting of 5-hydroxytryptamine fibres into ventral horns. However, the current dose was not sufficient to suppress CS-GAG up-regulation induced by SCI. Further adjustment on the dosage will be required to benefit functional recovery after SCI.
Collapse
Affiliation(s)
- Kateřina Štepánková
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic.
- Department of Neuroscience, Charles University, Second Faculty of Medicine, 15006, Prague, Czech Republic.
| | - Milada Chudíčková
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
| | - Zuzana Šimková
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
| | - Noelia Martinez-Varea
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
- Department of Neuroscience, Charles University, Second Faculty of Medicine, 15006, Prague, Czech Republic
| | - Šárka Kubinová
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic
- Institute of Physics, Czech Academy of Sciences, 182 21, Prague, Czech Republic
| | - Lucia Machová Urdzíková
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic.
- Department of Neuroscience, Charles University, Second Faculty of Medicine, 15006, Prague, Czech Republic.
| | - Pavla Jendelová
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic.
- Department of Neuroscience, Charles University, Second Faculty of Medicine, 15006, Prague, Czech Republic.
| | - Jessica C F Kwok
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská, 1083, Prague, Czech Republic.
- Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
3
|
Janušonis S, Haiman JH, Metzler R, Vojta T. Predicting the distribution of serotonergic axons: a supercomputing simulation of reflected fractional Brownian motion in a 3D-mouse brain model. Front Comput Neurosci 2023; 17:1189853. [PMID: 37265780 PMCID: PMC10231035 DOI: 10.3389/fncom.2023.1189853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/24/2023] [Indexed: 06/03/2023] Open
Abstract
The self-organization of the brain matrix of serotonergic axons (fibers) remains an unsolved problem in neuroscience. The regional densities of this matrix have major implications for neuroplasticity, tissue regeneration, and the understanding of mental disorders, but the trajectories of its fibers are strongly stochastic and require novel conceptual and analytical approaches. In a major extension to our previous studies, we used a supercomputing simulation to model around one thousand serotonergic fibers as paths of superdiffusive fractional Brownian motion (FBM), a continuous-time stochastic process. The fibers produced long walks in a complex, three-dimensional shape based on the mouse brain and reflected at the outer (pial) and inner (ventricular) boundaries. The resultant regional densities were compared to the actual fiber densities in the corresponding neuroanatomically-defined regions. The relative densities showed strong qualitative similarities in the forebrain and midbrain, demonstrating the predictive potential of stochastic modeling in this system. The current simulation does not respect tissue heterogeneities but can be further improved with novel models of multifractional FBM. The study demonstrates that serotonergic fiber densities can be strongly influenced by the geometry of the brain, with implications for brain development, plasticity, and evolution.
Collapse
Affiliation(s)
- Skirmantas Janušonis
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Justin H. Haiman
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Ralf Metzler
- Institute of Physics and Astronomy, University of Potsdam, Potsdam, Germany
- Asia Pacific Center for Theoretical Physics, Pohang, South Korea
| | - Thomas Vojta
- Department of Physics, Missouri University of Science and Technology, Rolla, MO, United States
| |
Collapse
|
4
|
Ghaffari-Nasab A, Badalzadeh R, Mohaddes G, Javani G, Ebrahimi-Kalan A, Alipour MR. Young Plasma Induces Antidepressant-Like Effects in Aged Rats Subjected to Chronic Mild Stress by Suppressing Indoleamine 2,3-Dioxygenase Enzyme and Kynurenine Pathway in the Prefrontal Cortex. Neurochem Res 2021; 47:358-371. [PMID: 34626305 DOI: 10.1007/s11064-021-03440-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 08/06/2021] [Accepted: 08/27/2021] [Indexed: 12/11/2022]
Abstract
Pathophysiology of depression in elderlies is linked to aging-associated increase in indoleamine 2,3-dioxygenase (IDO) levels and activity and kynurenine (Kyn) metabolites. Moreover, these aging-induced changes may alter the brain's responses to stress. Growing evidence suggested that young plasma can positively affect brain dysfunctions in old age. The present study aimed to investigate whether the antidepressant effects of young plasma administration in aged rats subjected to chronic unpredictable mild stress (CUMS) and underlying mechanisms, focusing on the prefrontal cortex (PFC). Young (3 months old) and aged (22 months old) male rats were divided into five groups; young control, aged control, aged rats subjected to CUMS (A + CUMS), aged rats subjected to CUMS and treated with young plasma (A + CUMS + YP), and aged rats subjected to CUMS and treated with old plasma (A + CUMS + OP). Plasma was injected (1 ml, intravenously) three times per week for four weeks. Young plasma significantly improved CUMS-induced depressive-like behaviors, evidenced by the increased sucrose consumption ratio in the sucrose preference test and the reduced immobility time in the forced swimming test. Furthermore, young plasma markedly reduced the levels of interferon-gamma (IFN-γ), IDO, Kyn, and Kyn to tryptophan (Kyn/Trp) ratio in PFC tissue. Expression levels of the serotonin transporter and growth-associated protein (GAP)-43 were also significantly increased after chronic administration of young plasma. These findings provide evidence for the antidepressant effect of young plasma in old age; however, whether it improves depressive behaviors or faster recovery from stress-induced deficits is required to be elucidated.
Collapse
Affiliation(s)
| | - Reza Badalzadeh
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gisou Mohaddes
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gonja Javani
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Ebrahimi-Kalan
- Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Alipour
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
5
|
Vahid-Ansari F, Albert PR. Rewiring of the Serotonin System in Major Depression. Front Psychiatry 2021; 12:802581. [PMID: 34975594 PMCID: PMC8716791 DOI: 10.3389/fpsyt.2021.802581] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/17/2021] [Indexed: 12/14/2022] Open
Abstract
Serotonin is a key neurotransmitter that is implicated in a wide variety of behavioral and cognitive phenotypes. Originating in the raphe nuclei, 5-HT neurons project widely to innervate many brain regions implicated in the functions. During the development of the brain, as serotonin axons project and innervate brain regions, there is evidence that 5-HT plays key roles in wiring the developing brain, both by modulating 5-HT innervation and by influencing synaptic organization within corticolimbic structures. These actions are mediated by 14 different 5-HT receptors, with region- and cell-specific patterns of expression. More recently, the role of the 5-HT system in synaptic re-organization during adulthood has been suggested. The 5-HT neurons have the unusual capacity to regrow and reinnervate brain regions following insults such as brain injury, chronic stress, or altered development that result in disconnection of the 5-HT system and often cause depression, anxiety, and cognitive impairment. Chronic treatment with antidepressants that amplify 5-HT action, such as selective serotonin reuptake inhibitors (SSRIs), appears to accelerate the rewiring of the 5-HT system by mechanisms that may be critical to the behavioral and cognitive improvements induced in these models. In this review, we survey the possible 5-HT receptor mechanisms that could mediate 5-HT rewiring and assess the evidence that 5-HT-mediated brain rewiring is impacting recovery from mental illness. By amplifying 5-HT-induced rewiring processes using SSRIs and selective 5-HT agonists, more rapid and effective treatments for injury-induced mental illness or cognitive impairment may be achieved.
Collapse
Affiliation(s)
- Faranak Vahid-Ansari
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Paul R Albert
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| |
Collapse
|
6
|
Rodríguez-Barrera R, Flores-Romero A, Buzoianu-Anguiano V, Garcia E, Soria-Zavala K, Incontri-Abraham D, Garibay-López M, Juárez-Vignon Whaley JJ, Ibarra A. Use of a Combination Strategy to Improve Morphological and Functional Recovery in Rats With Chronic Spinal Cord Injury. Front Neurol 2020; 11:189. [PMID: 32300328 PMCID: PMC7142263 DOI: 10.3389/fneur.2020.00189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/28/2020] [Indexed: 01/10/2023] Open
Abstract
Immunization with neural derived peptides (INDP), as well as scar removal (SR) and the use of matrices with bone marrow-mesenchymal stem cells (MSCs), have been studied separately and proven to induce a functional and morphological improvement after spinal cord injury (SCI). Herein, we evaluated the therapeutic effects of INDP combined with SR and a fibrin glue matrix (FGM) with MSCs (FGM-MSCs), on motor recovery, axonal regeneration-associated molecules and cytokine expression, axonal regeneration (catecholaminergic and serotonergic fibers), and the induction of neurogenesis after a chronic SCI. For this purpose, female adult Sprague-Dawley rats were subjected to SCI, 60 days after lesion, rats were randomly distributed in four groups: (1) Rats immunized with complete Freund's adjuvant + PBS (vehicle; PBS-I); (2) Rats with SR+ FGM-MSCs; (3) Rats with SR+ INDP + FGM-MSCs; (4) Rats only with INDP. Afterwards, we evaluated motor recovery using the BBB locomotor test. Sixty days after the therapy, protein expression of TNFα, IL-4, IL-10, BDNF, and GAP-43 were evaluated using ELISA assay. The number of catecholaminergic and serotonergic fibers were also determined. Neurogenesis was evaluated through immunofluorescence. The results show that treatment with INDP alone significantly increased motor recovery, anti-inflammatory cytokines, regeneration-associated molecules, axonal regeneration, and neurogenesis when compared to the rest of the groups. Our findings suggest that the combination therapy (SR + INDP + FGM-MSCs) modifies the non-permissive microenvironment post SCI, but it is not capable of inducing an appropriate axonal regeneration or neurogenesis when compared to the treatment with INDP alone.
Collapse
Affiliation(s)
- Roxana Rodríguez-Barrera
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico.,Proyecto CAMINA A.C., Mexico City, Mexico
| | - Adrián Flores-Romero
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico.,Proyecto CAMINA A.C., Mexico City, Mexico
| | | | - Elisa Garcia
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico.,Proyecto CAMINA A.C., Mexico City, Mexico
| | - Karla Soria-Zavala
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico.,Proyecto CAMINA A.C., Mexico City, Mexico
| | - Diego Incontri-Abraham
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico
| | - Marcela Garibay-López
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico
| | - Juan José Juárez-Vignon Whaley
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, Mexico.,Proyecto CAMINA A.C., Mexico City, Mexico
| |
Collapse
|
7
|
Affiliation(s)
- Zulvikar Syambani Ulhaq
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Maulana Malik Ibrahim Islamic State University of Malang, Batu, Indonesia
| |
Collapse
|
8
|
Donovan LJ, Spencer WC, Kitt MM, Eastman BA, Lobur KJ, Jiao K, Silver J, Deneris ES. Lmx1b is required at multiple stages to build expansive serotonergic axon architectures. eLife 2019; 8:e48788. [PMID: 31355748 PMCID: PMC6685705 DOI: 10.7554/elife.48788] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/27/2019] [Indexed: 01/18/2023] Open
Abstract
Formation of long-range axons occurs over multiple stages of morphological maturation. However, the intrinsic transcriptional mechanisms that temporally control different stages of axon projection development are unknown. Here, we addressed this question by studying the formation of mouse serotonin (5-HT) axons, the exemplar of long-range profusely arborized axon architectures. We report that LIM homeodomain factor 1b (Lmx1b)-deficient 5-HT neurons fail to generate axonal projections to the forebrain and spinal cord. Stage-specific targeting demonstrates that Lmx1b is required at successive stages to control 5-HT axon primary outgrowth, selective routing, and terminal arborization. We show a Lmx1b→Pet1 regulatory cascade is temporally required for 5-HT arborization and upregulation of the 5-HT axon arborization gene, Protocadherin-alphac2, during postnatal development of forebrain 5-HT axons. Our findings identify a temporal regulatory mechanism in which a single continuously expressed transcription factor functions at successive stages to orchestrate the progressive development of long-range axon architectures enabling expansive neuromodulation.
Collapse
Affiliation(s)
- Lauren J Donovan
- Department of NeurosciencesSchool of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - William C Spencer
- Department of NeurosciencesSchool of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Meagan M Kitt
- Department of NeurosciencesSchool of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Brent A Eastman
- Department of NeurosciencesSchool of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Katherine J Lobur
- Department of NeurosciencesSchool of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Kexin Jiao
- Department of NeurosciencesSchool of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Jerry Silver
- Department of NeurosciencesSchool of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Evan S Deneris
- Department of NeurosciencesSchool of Medicine, Case Western Reserve UniversityClevelandUnited States
| |
Collapse
|
9
|
Wang B, Wu Q, Lei L, Sun H, Michael N, Zhang X, Wang Y, Zhang Y, Ge B, Wu X, Wang Y, Xin Y, Zhao J, Li S. Long-term social isolation inhibits autophagy activation, induces postsynaptic dysfunctions and impairs spatial memory. Exp Neurol 2018; 311:213-224. [PMID: 30219732 DOI: 10.1016/j.expneurol.2018.09.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/14/2018] [Accepted: 09/12/2018] [Indexed: 12/23/2022]
Abstract
. Moreover, we found that L-PWSI increased the protein expression of p-AKT/AKT, p-mTOR/mTOR and p62, whereas the protein levels of LC3B and Beclin1 were decreased indicating an inhibition in autophagy activity. Intraperitoneal injection of rapamycin significantly potentiated fEPSP slope and cognition-related proteins expression in the L-PWSI mice. These results therefore suggest that L-PWSI induces postsynaptic dysfunction by disrupting the interaction between AMPAR, NMDAR and PSD-95, and inhibit the autophagy activity which led to impaired spatial memory and cognitive function.
Collapse
Affiliation(s)
- Bin Wang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, Dalian Medical University, Dalian, Liaoning, China.
| | - Qiong Wu
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, Dalian Medical University, Dalian, Liaoning, China
| | - Lei Lei
- Technology Centre of Target-based Nature Products for Prevention and Treatment of Ageing-related Neurodegeneration, Dalian Medical University, Dalian, Liaoning, China
| | - Hailun Sun
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, Dalian Medical University, Dalian, Liaoning, China
| | - Ntim Michael
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, Dalian Medical University, Dalian, Liaoning, China
| | - Xuan Zhang
- Technology Centre of Target-based Nature Products for Prevention and Treatment of Ageing-related Neurodegeneration, Dalian Medical University, Dalian, Liaoning, China
| | - Ying Wang
- Department of Cardiology, Institute of Heart and Vessel Diseases of Dalian Medical University, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China; Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning, China
| | - Yue Zhang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, Dalian Medical University, Dalian, Liaoning, China
| | - Biying Ge
- Technology Centre of Target-based Nature Products for Prevention and Treatment of Ageing-related Neurodegeneration, Dalian Medical University, Dalian, Liaoning, China
| | - Xuefei Wu
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, Dalian Medical University, Dalian, Liaoning, China
| | - Yue Wang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning, China
| | - Yi Xin
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, Liaoning, China.
| | - Jie Zhao
- Technology Centre of Target-based Nature Products for Prevention and Treatment of Ageing-related Neurodegeneration, Dalian Medical University, Dalian, Liaoning, China.
| | - Shao Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
10
|
Garcia LP, Witteveen JS, Middelman A, van Hulten JA, Martens GJM, Homberg JR, Kolk SM. Perturbed Developmental Serotonin Signaling Affects Prefrontal Catecholaminergic Innervation and Cortical Integrity. Mol Neurobiol 2018; 56:1405-1420. [PMID: 29948943 PMCID: PMC6400880 DOI: 10.1007/s12035-018-1105-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 05/03/2018] [Indexed: 11/26/2022]
Abstract
Proper development of the medial prefrontal cortex (mPFC), crucial for correct cognitive functioning, requires projections from, among others, the serotonergic (5-HT) and catecholaminergic systems, but it is unclear how these systems influence each other during development. Here, we describe the parallel development of the 5-HT and catecholaminergic prefrontal projection systems in rat and demonstrate a close engagement of both systems in the proximity of Cajal-Retzius cells. We further show that in the absence of the 5-HT transporter (5-HTT), not only the developing 5-HT but also the catecholaminergic system, including their projections towards the mPFC, are affected. In addition, the layer identity of the mPFC neurons and reelin-positive interneuron number and integration are altered in the absence of the 5-HTT. Together, our data demonstrate a functional interplay between the developing mPFC 5-HT and catecholaminergic systems, and call for a holistic approach in studying neurotransmitter systems-specific developmental consequences for adult behavior, to eventually allow the design of better treatment strategies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Lidiane P Garcia
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Molecular Animal Physiology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Josefine S Witteveen
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Molecular Animal Physiology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Anthonieke Middelman
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Cognitive Neuroscience, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Josephus A van Hulten
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Molecular Animal Physiology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Gerard J M Martens
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Molecular Animal Physiology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Judith R Homberg
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Cognitive Neuroscience, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Sharon M Kolk
- Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Molecular Animal Physiology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.
| |
Collapse
|
11
|
Deneris E, Gaspar P. Serotonin neuron development: shaping molecular and structural identities. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2018; 7:10.1002/wdev.301. [PMID: 29072810 PMCID: PMC5746461 DOI: 10.1002/wdev.301] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/03/2017] [Accepted: 08/24/2017] [Indexed: 02/03/2023]
Abstract
The continuing fascination with serotonin (5-hydroxytryptamine, 5-HT) as a nervous system chemical messenger began with its discovery in the brains of mammals in 1953. Among the many reasons for this decades-long interest is that the small numbers of neurons that make 5-HT influence the excitability of neural circuits in nearly every region of the brain and spinal cord. A further reason is that 5-HT dysfunction has been linked to a range of psychiatric and neurological disorders many of which have a neurodevelopmental component. This has led to intense interest in understanding 5-HT neuron development with the aim of determining whether early alterations in their generation lead to brain disease susceptibility. Here, we present an overview of the neuroanatomical organization of vertebrate 5-HT neurons, their neurogenesis, and prodigious axonal architectures, which enables the expansive reach of 5-HT neuromodulation in the central nervous system. We review recent findings that have revealed the molecular basis for the tremendous diversity of 5-HT neuron subtypes, the impact of environmental factors on 5-HT neuron development, and how 5-HT axons are topographically organized through disparate signaling pathways. We summarize studies of the gene regulatory networks that control the differentiation, maturation, and maintenance of 5-HT neurons. These studies show that the regulatory factors controlling acquisition of 5-HT-type transmitter identity continue to play critical roles in the functional maturation and the maintenance of 5-HT neurons. New insights are presented into how continuously expressed 5-HT regulatory factors control 5-HT neurons at different stages of life and how the regulatory networks themselves are maintained. WIREs Dev Biol 2018, 7:e301. doi: 10.1002/wdev.301 This article is categorized under: Nervous System Development > Vertebrates: General Principles Gene Expression and Transcriptional Hierarchies > Gene Networks and Genomics Gene Expression and Transcriptional Hierarchies > Cellular Differentiation Nervous System Development > Secondary: Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Evan Deneris
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Patricia Gaspar
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S839, Paris, France
- Sorbonne Université, Paris, France
- Institut du Fer à Moulin, Campus Jussieu, Paris, France
| |
Collapse
|
12
|
Protocadherin-αC2 is required for diffuse projections of serotonergic axons. Sci Rep 2017; 7:15908. [PMID: 29162883 PMCID: PMC5698425 DOI: 10.1038/s41598-017-16120-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 11/08/2017] [Indexed: 12/04/2022] Open
Abstract
Serotonergic axons extend diffuse projections throughout various brain areas, and serotonergic system disruption causes neuropsychiatric diseases. Loss of the cytoplasmic region of protocadherin-α (Pcdh-α) family proteins, products of the diverse clustered Pcdh genes, causes unbalanced distributions (densification and sparsification) of serotonergic axons in various target regions. However, which Pcdh-α member(s) are responsible for the phenotype is unknown. Here we demonstrated that Pcdh-αC2 (αC2), a Pcdh-α isoform, was highly expressed in serotonergic neurons, and was required for normal diffusion in single-axon-level analyses of serotonergic axons. The loss of αC2 from serotonergic neurons, but not from their target brain regions, led to unbalanced distributions of serotonergic axons. Our results suggest that αC2 expressed in serotonergic neurons is required for serotonergic axon diffusion in various brain areas. The αC2 extracellular domain displays homophilic binding activity, suggesting that its homophilic interaction between serotonergic axons regulates axonal density via αC2′s cytoplasmic domain.
Collapse
|
13
|
Spencer WC, Deneris ES. Regulatory Mechanisms Controlling Maturation of Serotonin Neuron Identity and Function. Front Cell Neurosci 2017; 11:215. [PMID: 28769770 PMCID: PMC5515867 DOI: 10.3389/fncel.2017.00215] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 07/05/2017] [Indexed: 11/29/2022] Open
Abstract
The brain serotonin (5-hydroxytryptamine; 5-HT) system has been extensively studied for its role in normal physiology and behavior, as well as, neuropsychiatric disorders. The broad influence of 5-HT on brain function, is in part due to the vast connectivity pattern of 5-HT-producing neurons throughout the CNS. 5-HT neurons are born and terminally specified midway through embryogenesis, then enter a protracted period of maturation, where they functionally integrate into CNS circuitry and then are maintained throughout life. The transcriptional regulatory networks controlling progenitor cell generation and terminal specification of 5-HT neurons are relatively well-understood, yet the factors controlling 5-HT neuron maturation are only recently coming to light. In this review, we first provide an update on the regulatory network controlling 5-HT neuron development, then delve deeper into the properties and regulatory strategies governing 5-HT neuron maturation. In particular, we discuss the role of the 5-HT neuron terminal selector transcription factor (TF) Pet-1 as a key regulator of 5-HT neuron maturation. Pet-1 was originally shown to positively regulate genes needed for 5-HT synthesis, reuptake and vesicular transport, hence 5-HT neuron-type transmitter identity. It has now been shown to regulate, both positively and negatively, many other categories of genes in 5-HT neurons including ion channels, GPCRs, transporters, neuropeptides, and other transcription factors. Its function as a terminal selector results in the maturation of 5-HT neuron excitability, firing characteristics, and synaptic modulation by several neurotransmitters. Furthermore, there is a temporal requirement for Pet-1 in the control of postmitotic gene expression trajectories thus indicating a direct role in 5-HT neuron maturation. Proper regulation of the maturation of cellular identity is critical for normal neuronal functioning and perturbations in the gene regulatory networks controlling these processes may result in long-lasting changes in brain function in adulthood. Further study of 5-HT neuron gene regulatory networks is likely to provide additional insight into how neurons acquire their mature identities and how terminal selector-type TFs function in postmitotic vertebrate neurons.
Collapse
Affiliation(s)
- William C Spencer
- Department of Neurosciences, Case Western Reserve UniversityCleveland, OH, United States
| | - Evan S Deneris
- Department of Neurosciences, Case Western Reserve UniversityCleveland, OH, United States
| |
Collapse
|
14
|
Holland SM, Thomas GM. Roles of palmitoylation in axon growth, degeneration and regeneration. J Neurosci Res 2017; 95:1528-1539. [PMID: 28150429 DOI: 10.1002/jnr.24003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 11/09/2016] [Accepted: 11/28/2016] [Indexed: 12/11/2022]
Abstract
The protein-lipid modification palmitoylation plays important roles in neurons, but most attention has focused on roles of this modification in the regulation of mature pre- and post-synapses. However, exciting recent findings suggest that palmitoylation is also critical for both the growth and integrity of neuronal axons and plays previously unappreciated roles in conveying axonal anterograde and retrograde signals. Here we review these emerging roles for palmitoylation in the regulation of axons in health and disease. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sabrina M Holland
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair)
| | - Gareth M Thomas
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair).,Department of Anatomy and Cell Biology, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA, 19140
| |
Collapse
|
15
|
Rodríguez-Barrera R, Flores-Romero A, Fernández-Presas AM, García-Vences E, Silva-García R, Konigsberg M, Blancas-Espinoza L, Buzoianu-Anguiano V, Soria-Zavala K, Suárez-Meade P, Ibarra A. Immunization with neural derived peptides plus scar removal induces a permissive microenvironment, and improves locomotor recovery after chronic spinal cord injury. BMC Neurosci 2017; 18:7. [PMID: 28056790 PMCID: PMC5217189 DOI: 10.1186/s12868-016-0331-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 12/27/2016] [Indexed: 11/17/2022] Open
Abstract
Background Immunization with neural derived peptides (INDP) as well as scar removal—separately—have shown to induce morphological and functional improvement after spinal cord injury (SCI). In the present study, we compared the effect of INDP alone versus INDP with scar removal on motor recovery, regeneration-associated and cytokine gene expression, and axonal regeneration after chronic SCI. Scar removal was conducted through a single incision with a double-bladed scalpel along the stump, and scar renewal was halted by adding α,α′-dipyridyl. Results During the chronic injury stage, two experiments were undertaken. The first experiment was aimed at testing the therapeutic effect of INDP combined with scar removal. Sixty days after therapeutic intervention, the expression of genes encoding for TNFα, IFNγ, IL4, TGFβ, BDNF, IGF1, and GAP43 was evaluated at the site of injury. Tyrosine hydroxylase and 5-hydroxytryptamine positive fibers were also studied. Locomotor evaluations showed a significant recovery in the group treated with scar removal + INDP. Moreover; this group presented a significant increase in IL4, TGFβ, BDNF, IGF1, and GAP43 expression, but a decrease of TNFα and IFNγ. Also, the spinal cord of animals receiving both treatments presented a significant increase of serotonergic and catecholaminergic fibers as compared to other the groups. The second experiment compared the results of the combined approach versus INDP alone. Rats receiving INDP likewise showed improved motor recovery, although on a lesser scale than those who received the combined treatment. An increase in inflammation and regeneration-associated gene expression, as well as in the percentage of serotonergic and catecholaminergic fibers was observed in INDP-treated rats to a lesser degree than those in the combined therapy group. Conclusions These findings suggest that INDP, both alone and in combination with scar removal, could modify the non-permissive microenvironment prevailing at the chronic phase of SCI, providing the opportunity of improving motor recovery.
Collapse
Affiliation(s)
- Roxana Rodríguez-Barrera
- Centro de Investigación en Ciencias de la Salud (CICSA), Universidad Anáhuac México Campus Norte, Huixquilucan, Estado de México, Mexico.,Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, Estado de México, Mexico.,Centro de Investigación del Proyecto CAMINA A.C., Ciudad de México, Mexico.,Posgrado en Biología Experimental, UAMI, Ciudad de México, Mexico
| | - Adrián Flores-Romero
- Centro de Investigación en Ciencias de la Salud (CICSA), Universidad Anáhuac México Campus Norte, Huixquilucan, Estado de México, Mexico.,Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, Estado de México, Mexico.,Centro de Investigación del Proyecto CAMINA A.C., Ciudad de México, Mexico
| | | | - Elisa García-Vences
- Centro de Investigación en Ciencias de la Salud (CICSA), Universidad Anáhuac México Campus Norte, Huixquilucan, Estado de México, Mexico.,Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, Estado de México, Mexico.,Centro de Investigación del Proyecto CAMINA A.C., Ciudad de México, Mexico
| | | | - Mina Konigsberg
- Lab. Bioenergética y Envejecimiento Celular, UAMI, Ciudad de México, Mexico
| | - Liliana Blancas-Espinoza
- Centro de Investigación del Proyecto CAMINA A.C., Ciudad de México, Mexico.,Hospital de Pediatría CMN Siglo XXI, Ciudad de México, Mexico
| | | | - Karla Soria-Zavala
- Centro de Investigación en Ciencias de la Salud (CICSA), Universidad Anáhuac México Campus Norte, Huixquilucan, Estado de México, Mexico.,Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, Estado de México, Mexico.,Centro de Investigación del Proyecto CAMINA A.C., Ciudad de México, Mexico
| | - Paola Suárez-Meade
- Centro de Investigación en Ciencias de la Salud (CICSA), Universidad Anáhuac México Campus Norte, Huixquilucan, Estado de México, Mexico.,Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, Estado de México, Mexico
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud (CICSA), Universidad Anáhuac México Campus Norte, Huixquilucan, Estado de México, Mexico. .,Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan, Estado de México, Mexico. .,Centro de Investigación del Proyecto CAMINA A.C., Ciudad de México, Mexico.
| |
Collapse
|
16
|
Williams KR, McAninch DS, Stefanovic S, Xing L, Allen M, Li W, Feng Y, Mihailescu MR, Bassell GJ. hnRNP-Q1 represses nascent axon growth in cortical neurons by inhibiting Gap-43 mRNA translation. Mol Biol Cell 2015; 27:518-34. [PMID: 26658614 PMCID: PMC4751602 DOI: 10.1091/mbc.e15-07-0504] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 12/01/2015] [Indexed: 11/11/2022] Open
Abstract
A novel posttranscriptional mechanism for regulating the neuronal protein GAP-43 is reported. The mRNA-binding protein hnRNP-Q1 represses Gap-43 mRNA translation by a mechanism involving a 5′ untranslated region G-quadruplex structure, which affects GAP-43 function, as demonstrated by a GAP-43–dependent increase in neurite length and number with hnRNP-Q1 knockdown. Posttranscriptional regulation of gene expression by mRNA-binding proteins is critical for neuronal development and function. hnRNP-Q1 is an mRNA-binding protein that regulates mRNA processing events, including translational repression. hnRNP-Q1 is highly expressed in brain tissue, suggesting a function in regulating genes critical for neuronal development. In this study, we have identified Growth-associated protein 43 (Gap-43) mRNA as a novel target of hnRNP-Q1 and have demonstrated that hnRNP-Q1 represses Gap-43 mRNA translation and consequently GAP-43 function. GAP-43 is a neuronal protein that regulates actin dynamics in growth cones and facilitates axonal growth. Previous studies have identified factors that regulate Gap-43 mRNA stability and localization, but it remains unclear whether Gap-43 mRNA translation is also regulated. Our results reveal that hnRNP-Q1 knockdown increased nascent axon length, total neurite length, and neurite number in mouse embryonic cortical neurons and enhanced Neuro2a cell process extension; these phenotypes were rescued by GAP-43 knockdown. Additionally, we have identified a G-quadruplex structure in the 5′ untranslated region of Gap-43 mRNA that directly interacts with hnRNP-Q1 as a means to inhibit Gap-43 mRNA translation. Therefore hnRNP-Q1–mediated repression of Gap-43 mRNA translation provides an additional mechanism for regulating GAP-43 expression and function and may be critical for neuronal development.
Collapse
Affiliation(s)
- Kathryn R Williams
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322
| | - Damian S McAninch
- Department of Chemistry and Biochemistry, Duquesne University, Pittsburgh, PA 15282
| | - Snezana Stefanovic
- Department of Chemistry and Biochemistry, Duquesne University, Pittsburgh, PA 15282
| | - Lei Xing
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322
| | - Megan Allen
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322
| | - Wenqi Li
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322
| | - Yue Feng
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322
| | | | - Gary J Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
17
|
Zhu Q, Liu Z, Wang C, Nie L, He Y, Zhang Y, Liu X, Su G. Lentiviral-mediated growth-associated protein-43 modification of bone marrow mesenchymal stem cells improves traumatic optic neuropathy in rats. Mol Med Rep 2015; 12:5691-700. [PMID: 26238991 PMCID: PMC4581804 DOI: 10.3892/mmr.2015.4132] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 06/26/2015] [Indexed: 01/03/2023] Open
Abstract
The aim of the present study was to examine the effect of growth-associated protein-43 (GAP-43) on bone marrow mesenchymal stem cell (BMSC) differentiation in a rat model of traumatic optic neuropathy (TON). GAP-43 and short hairpin (sh)RNA-GAP-43 were inserted into pGLV5 and pGLV3 lentiviral vectors, respectively. The stable control, GAP-43-overexpression and GAP-43-knockdown cell lines (GFP/BMSCs, GAP-43/BMSCs and shGAP-43/BMSCs, respectively) were established. The expression of GAP-43, neuron-specific enolase (NSE), nestin, neurofilament (NF), neuron-specific nuclear-binding protein (NeuN) and βIII-tubulin were detected in the GAP-43/BMSCs and shGAP-43/BMSCs with retinal cell-conditioned differentiation medium using semi-quantitative polymerase chain reaction (PCR), western blotting and cell immunofluorescence. In addition, the BMSCs were observed under fluorescence microscopy. The Sprague-Dawley rat models of TON were established and identified by retrograde labeling of retinal ganglion cells (RGCs) with fluoroGold (FG). The lentiviral-mediated GAP-43-modified BMSCs were then transplanted into the rat model of TON. The expression of GAP-43 was detected in the retinal tissues using qPCR and western blotting. The histopathology of the retinal tissues was observed using hematoxylin and eosin (H&E) staining. The GAP-43/BMSCs exhibited positive expression of NSE, NF, nestin and βIII-tubulin, and exhibited a neuronal phenotype. The shGAP-43/BMSCs markedly inhibited expression of NeuN, NSE, NF, nestin and βIII-tubulin induced by retinal cell-conditioned differentiation medium. The FG staining revealed that the number of labeled RGCs were significantly decreased in the TON model rats, compared with normal rats (P<0.05). The H&E staining revealed that the degree of pathological changes was improved in the GAP-43/BMSC group, compared with the GFP/BMSC and shGAP-43/BMSC groups. In conclusion, GAP-43 promoted BMSC differentiation into neuron-like cells, and intravitreally injected GAP-43/BMSCs promoted the process of nerve repair in a rat model of TON.
Collapse
Affiliation(s)
- Qi Zhu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Zaoxia Liu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Chenguang Wang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Lili Nie
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yuxi He
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yan Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Xin Liu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Guanfang Su
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
18
|
Zhang ZC, Luan F, Xie CY, Geng DD, Wang YY, Ma J. Low-frequency transcranial magnetic stimulation is beneficial for enhancing synaptic plasticity in the aging brain. Neural Regen Res 2015. [PMID: 26199608 PMCID: PMC4498353 DOI: 10.4103/1673-5374.158356] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
In the aging brain, cognitive function gradually declines and causes a progressive reduction in the structural and functional plasticity of the hippocampus. Transcranial magnetic stimulation is an emerging and novel neurological and psychiatric tool used to investigate the neurobiology of cognitive function. Recent studies have demonstrated that low-frequency transcranial magnetic stimulation (≤1 Hz) ameliorates synaptic plasticity and spatial cognitive deficits in learning-impaired mice. However, the mechanisms by which this treatment improves these deficits during normal aging are still unknown. Therefore, the current study investigated the effects of transcranial magnetic stimulation on the brain-derived neurotrophic factor signal pathway, synaptic protein markers, and spatial memory behavior in the hippocampus of normal aged mice. The study also investigated the downstream regulator, Fyn kinase, and the downstream effectors, synaptophysin and growth-associated protein 43 (both synaptic markers), to determine the possible mechanisms by which transcranial magnetic stimulation regulates cognitive capacity. Transcranial magnetic stimulation with low intensity (110% average resting motor threshold intensity, 1 Hz) increased mRNA and protein levels of brain-derived neurotrophic factor, tropomyosin receptor kinase B, and Fyn in the hippocampus of aged mice. The treatment also upregulated the mRNA and protein expression of synaptophysin and growth-associated protein 43 in the hippocampus of these mice. In conclusion, brain-derived neurotrophic factor signaling may play an important role in sustaining and regulating structural synaptic plasticity induced by transcranial magnetic stimulation in the hippocampus of aging mice, and Fyn may be critical during this regulation. These responses may change the structural plasticity of the aging hippocampus, thereby improving cognitive function.
Collapse
Affiliation(s)
- Zhan-Chi Zhang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Feng Luan
- Department of Otorhinolaryngology, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Chun-Yan Xie
- Second Surgical Department, Qinghe Public Hospital of Hebei Province, Xingtai, Hebei Province, China
| | - Dan-Dan Geng
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Yan-Yong Wang
- Department of Neurology, First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China ; Hebei Key Laboratory for Brain Aging and Cognitive Neuroscience, Shijiazhuang, Hebei Province, China
| | - Jun Ma
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei Province, China ; Hebei Key Laboratory for Brain Aging and Cognitive Neuroscience, Shijiazhuang, Hebei Province, China
| |
Collapse
|
19
|
Conditional anterograde tracing reveals distinct targeting of individual serotonin cell groups (B5-B9) to the forebrain and brainstem. Brain Struct Funct 2014; 221:535-61. [PMID: 25403254 PMCID: PMC4750555 DOI: 10.1007/s00429-014-0924-4] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 10/16/2014] [Indexed: 01/10/2023]
Abstract
Serotoninergic innervation of the central nervous system is provided by hindbrain raphe nuclei (B1–B9). The extent to which each raphe subdivision has distinct topographic organization of their projections is still unclear. We provide a comprehensive description of the main targets of the rostral serotonin (5-HT) raphe subgroups (B5–B9) in the mouse brain. Adeno-associated viruses that conditionally express GFP under the control of the 5-HT transporter promoter were used to label small groups of 5-HT neurons in the dorsal (B7d), ventral (B7v), lateral (B7l), and caudal (B6) subcomponents of the dorsal raphe (DR) nucleus as well as in the rostral and caudal parts of the median raphe (MR) nucleus (B8 and B5, respectively), and in the supralemniscal (B9) cell group. We illustrate the distinctive and largely non-overlapping projection areas of these cell groups: for instance, DR (B7) projects to basal parts of the forebrain, such as the amygdala, whereas MR (B8) is the main 5-HT source to the hippocampus, septum, and mesopontine tegmental nuclei. Distinct subsets of B7 have preferential brain targets: B7v is the main source of 5-HT for the cortex and amygdala while B7d innervates the hypothalamus. We reveal for the first time the target areas of the B9 cell group, demonstrating projections to the caudate, prefrontal cortex, substantia nigra, locus coeruleus and to the raphe cell groups. The broad topographic organization of the different raphe subnuclei is likely to underlie the different functional roles in which 5-HT has been implicated in the brain. The present mapping study could serve as the basis for genetically driven specific targeting of the different subcomponents of the mouse raphe system.
Collapse
|
20
|
Glover ME, Pugh PC, Jackson NL, Cohen JL, Fant AD, Akil H, Clinton SM. Early-life exposure to the SSRI paroxetine exacerbates depression-like behavior in anxiety/depression-prone rats. Neuroscience 2014; 284:775-797. [PMID: 25451292 DOI: 10.1016/j.neuroscience.2014.10.044] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 10/27/2014] [Accepted: 10/28/2014] [Indexed: 12/26/2022]
Abstract
Selective serotonin reuptake inhibitor (SSRI) antidepressants are the mainstay treatment for the 10-20% of pregnant and postpartum women who suffer major depression, but the effects of SSRIs on their children's developing brain and later emotional health are poorly understood. SSRI use during pregnancy can elicit antidepressant withdrawal in newborns and increase toddlers' anxiety and social avoidance. In rodents, perinatal SSRI exposure increases adult depression- and anxiety-like behavior, although certain individuals are more vulnerable to these effects than others. Our study establishes a rodent model of individual differences in susceptibility to perinatal SSRI exposure, utilizing selectively bred Low Responder (bLR) and High Responder (bHR) rats that were previously bred for high versus low behavioral response to novelty. Pregnant bHR/bLR females were chronically treated with the SSRI paroxetine (10 mg/kg/day p.o.) to examine its effects on offspring's emotional behavior and gene expression in the developing brain. Paroxetine treatment had minimal effect on bHR/bLR dams' pregnancy outcomes or maternal behavior. We found that bLR offspring, naturally prone to an inhibited/anxious temperament, were susceptible to behavioral abnormalities associated with perinatal SSRI exposure (which exacerbated their Forced Swim Test immobility), while high risk-taking bHR offspring were resistant. Microarray studies revealed robust perinatal SSRI-induced gene expression changes in the developing bLR hippocampus and amygdala (postnatal days 7-21), including transcripts involved in neurogenesis, synaptic vesicle components, and energy metabolism. These results highlight the bLR/bHR model as a useful tool to explore the neurobiology of individual differences in susceptibility to perinatal SSRI exposure.
Collapse
Affiliation(s)
- M E Glover
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama-Birmingham, USA
| | - P C Pugh
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama-Birmingham, USA
| | - N L Jackson
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama-Birmingham, USA
| | - J L Cohen
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama-Birmingham, USA
| | - A D Fant
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - H Akil
- Molecular and Behavioral Neuroscience Institute, University of Michigan, USA
| | - S M Clinton
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama-Birmingham, USA.
| |
Collapse
|
21
|
Repetitive transcranial magnetic stimulation (rTMS) influences spatial cognition and modulates hippocampal structural synaptic plasticity in aging mice. Exp Gerontol 2014; 58:256-68. [DOI: 10.1016/j.exger.2014.08.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Revised: 07/27/2014] [Accepted: 08/26/2014] [Indexed: 01/09/2023]
|
22
|
Vuillaume ML, Delrue MA, Naudion S, Toutain J, Fergelot P, Arveiler B, Lacombe D, Rooryck C. Expanding the clinical phenotype at the 3q13.31 locus with a new case of microdeletion and first characterization of the reciprocal duplication. Mol Genet Metab 2013; 110:90-7. [PMID: 23920044 DOI: 10.1016/j.ymgme.2013.07.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 07/11/2013] [Accepted: 07/11/2013] [Indexed: 11/30/2022]
Abstract
Congenital deletions at the 3q13.31 locus have been recently described as a novel microdeletion syndrome characterized by developmental delay, postnatal overgrowth, hypoplastic male genitalia and characteristic facial features. A common critical region of overlapping of 580kb was delineated including two strong candidate genes for developmental delay: DRD3 and ZBTB20. In this report, we describe a new case of 3q13.31 microdeletion identified by array-CGH in a 16year-old girl sharing clinical features commonly observed in the 3q13.31 microdeletion syndrome. This girl had a microdeletion of 7.39Mb spanning the common critical region of overlapping. More interestingly, we report for the first time the existence of a microduplication reciprocal to the microdeletion syndrome. This familial 2.76Mb microduplication identified by array-CGH was carried by two brothers and their father. The phenotype shared by the brothers resembled the phenotype related to the 3q13.31 microdeletion syndrome including especially severe intellectual disability, developmental delay, behavioral abnormalities and obesity. This microduplication involves three strong candidate genes for the developmental delay ZBTB20, LSAMP and GAP43. Further molecular characterization showed that DRD3, another strong candidate gene for developmental delay, was not included in the duplicated region. However, a dosage alteration of this gene cannot be completely excluded as the duplication was inverted at proximity of this gene, as revealed by FISH analysis. Finally, we hypothesized that the phenotype shared by the two brothers could be related to a gene dosage imbalance even if gene expression could not be measured in relevant tissues such as brain or adipocytes.
Collapse
Affiliation(s)
- Marie-Laure Vuillaume
- CHU Bordeaux, Service de Génétique Médicale, Place Amélie Raba Léon, Bordeaux, France.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Shuvarikov A, Campbell IM, Dittwald P, Neill NJ, Bialer MG, Moore C, Wheeler PG, Wallace SE, Hannibal MC, Murray MF, Giovanni MA, Terespolsky D, Sodhi S, Cassina M, Viskochil D, Moghaddam B, Herman K, Brown CW, Beck CR, Gambin A, Cheung SW, Patel A, Lamb AN, Shaffer LG, Ellison JW, Ravnan JB, Stankiewicz P, Rosenfeld JA. Recurrent HERV-H-mediated 3q13.2-q13.31 deletions cause a syndrome of hypotonia and motor, language, and cognitive delays. Hum Mutat 2013; 34:1415-23. [PMID: 23878096 DOI: 10.1002/humu.22384] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 07/11/2013] [Indexed: 11/09/2022]
Abstract
We describe the molecular and clinical characterization of nine individuals with recurrent, 3.4-Mb, de novo deletions of 3q13.2-q13.31 detected by chromosomal microarray analysis. All individuals have hypotonia and language and motor delays; they variably express mild to moderate cognitive delays (8/9), abnormal behavior (7/9), and autism spectrum disorders (3/9). Common facial features include downslanting palpebral fissures with epicanthal folds, a slightly bulbous nose, and relative macrocephaly. Twenty-eight genes map to the deleted region, including four strong candidate genes, DRD3, ZBTB20, GAP43, and BOC, with important roles in neural and/or muscular development. Analysis of the breakpoint regions based on array data revealed directly oriented human endogenous retrovirus (HERV-H) elements of ~5 kb in size and of >95% DNA sequence identity flanking the deletion. Subsequent DNA sequencing revealed different deletion breakpoints and suggested nonallelic homologous recombination (NAHR) between HERV-H elements as a mechanism of deletion formation, analogous to HERV-I-flanked and NAHR-mediated AZFa deletions. We propose that similar HERV elements may also mediate other recurrent deletion and duplication events on a genome-wide scale. Observation of rare recurrent chromosomal events such as these deletions helps to further the understanding of mechanisms behind naturally occurring variation in the human genome and its contribution to genetic disease.
Collapse
Affiliation(s)
- Andrey Shuvarikov
- Signature Genomic Laboratories, PerkinElmer, Inc, Spokane, Washington
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Magnetic stimulation modulates structural synaptic plasticity and regulates BDNF–TrkB signal pathway in cultured hippocampal neurons. Neurochem Int 2013. [DOI: 10.1016/j.neuint.2012.11.010] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
25
|
Muneoka K, Funahashi H, Ogawa T, Whitaker-Azmitia PM, Shioda S. Shared features of S100B immunohistochemistry and cytochrome oxidase histochemistry in the ventroposterior thalamus and lateral habenula in neonatal rats. Int J Dev Neurosci 2012; 30:499-505. [PMID: 22627026 DOI: 10.1016/j.ijdevneu.2012.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 05/10/2012] [Accepted: 05/10/2012] [Indexed: 10/28/2022] Open
Abstract
The ventroposterior thalamus and the habenular nuclei of the epithalamus are relevant to the monoaminergic system functionally and anatomically. The glia-derived S100B protein plays a critical role in the development of the nervous system including the monoaminergic systems. In this study, we performed an immunohistochemical study of glia-related proteins including S100B, serotonin transporter, and microtubule-associated protein 2, as well as cytochrome oxidase histochemistry in neonatal rats. Results showed the same findings for S100B immunohistochemistry between the ventroposterior thalamus and the lateral habenula at postnatal day 7: intense staining in cell bodies of astrocytes, diffusely spread immunoproduct in the intercellular space, and S100B-free areas as well as a strong reaction to cytochrome oxidase histochemistry. Further common features were the scarcity of glial fibrillary acidic protein-positive astrocytes and the few apoptotic cells observed. The results of the cytochrome oxidase reaction suggested that S100B is released actively into intercellular areas in restricted brain regions showing high neuronal activity at postnatal day 7. Pathology of the ventroposterior thalamus and the habenula is suggested in mental disorders, and S100B might be a key factor for investigations in these areas.
Collapse
Affiliation(s)
- Katsumasa Muneoka
- Department of Anatomy I, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo 142-8555, Japan.
| | | | | | | | | |
Collapse
|
26
|
Patodia S, Raivich G. Downstream effector molecules in successful peripheral nerve regeneration. Cell Tissue Res 2012; 349:15-26. [PMID: 22580509 DOI: 10.1007/s00441-012-1416-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 03/19/2012] [Indexed: 12/16/2022]
Abstract
The robust axon regeneration that occurs following peripheral nerve injury is driven by transcriptional activation of the regeneration program and by the expression of a wide range of downstream effector molecules from neuropeptides and neurotrophic factors to adhesion molecules and cytoskeletal adaptor proteins. These regeneration-associated effector molecules regulate the actin-tubulin machinery of growth-cones, integrate intracellular signalling and stimulatory and inhibitory signals from the local environment and translate them into axon elongation. In addition to the neuronally derived molecules, an important transcriptional component is found in locally activated Schwann cells and macrophages, which release a number of cytokines, growth factors and neurotrophins that support neuronal survival and axonal regeneration and that might provide directional guidance cues towards appropriate peripheral targets. This review aims to provide a comprehensive up-to-date account of the transcriptional regulation and functional role of these effector molecules and of the information that they can give us with regard to the organisation of the regeneration program.
Collapse
Affiliation(s)
- Smriti Patodia
- Centre for Perinatal Brain Protection and Repair, University College London, Chenies Mews 86-96, London, WC1E 6HX, UK
| | | |
Collapse
|
27
|
Zhao JC, Zhang LX, Zhang Y, Shen YF. The differential regulation of Gap43 gene in the neuronal differentiation of P19 cells. J Cell Physiol 2012; 227:2645-53. [PMID: 21938722 DOI: 10.1002/jcp.23006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Growth associated protein 43 (Gap43) is a neuron-specific phosphoprotein, which plays critical role in axon growth and synapses functions during neurogenesis. Here we identified two transcription start sites (TSSs) of the mouse Gap43 gene designated as a proximal site at +1, and a distal TSS at -414. RT-qPCR data reveal that the transcripts from +1 increase 10-fold on day-1 post-all-trans retinoic acid (RA) treatment, reached a peak value at day-4 and gradually reduced. By contrast, the distal TSS directs a late, remarkably sharp increase of the transcripts from the day-5 on. An intense signal of Gap43 at the neurites and neural network is determined by the efficient transcription of the distal promoter as shown in Northern blot and RT-qPCR assay. In addition, the targeting of p300 in combination with a differential enrichment of Brm to Brg1 change at the distal promoter region of the gene is induced under RA treatment. The over hundreds of GA rich stretches and the GAGAG elements located between the two TSSs may take parts in the differential transcription of the two TSSs of the Gap43. Our findings provide the first evidence on the identification and differential transcription of the two TSSs of the mouse Gap43 gene, and the preferential distribution of their protein products in the specific stages of RA induced P19 differentiation. These data suggest the efficient transcription of the distal promoter of Gap43 is an important mark for the transition of P19 cells from the progenitor stage into neuronal differentiation.
Collapse
Affiliation(s)
- Ji-cheng Zhao
- National Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | | | | | | |
Collapse
|
28
|
Kim YG, Lee YI. Differential Expressions of Synaptogenic Markers between Primary Cultured Cortical and Hippocampal Neurons. Exp Neurobiol 2012; 21:61-7. [PMID: 22792026 PMCID: PMC3381213 DOI: 10.5607/en.2012.21.2.61] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Accepted: 02/13/2012] [Indexed: 11/19/2022] Open
Abstract
Primary dissociated neuronal cultures are widely used research tools to investigate of pathological mechanisms and to treat various central and peripheral nervous system problems including trauma and degenerative neuronal diseases. We introduced a protocol that utilizes hippocampal and cortical neurons from embryonic day 17 or 18 mice. We applied appropriate markers (GAP-43 and synaptophysin) to investigate whether neurite outgrowth and synaptogenesis can be distinguished at a particular period of time. GAP-43 was found along the neural processes in a typical granular pattern, and its expression increased proportionally as neurites lengthened during the early in vitro period. Unlike GAP-43, granular immunoreactive patterns of synaptophysin along the neurites were clearly found from day 2 in vitro with relatively high immunoreactive levels. Expression of synaptic markers from cortical neurons reached peak level earlier than that of hippocampal neurons, although neurite outgrowths of hippocampal neurons were faster than those of cortical neurons. The amount of peak synaptic markers expressed was also higher in cortical neurons than that in hippocampal neurons. These results strongly suggest the usefulness of primary cultured neurons from mice embryos for synaptic function and plasticity studies, because of their clear and typical patterns of morphology that establish synapses. Results from this study also suggest the proper amount of time in vitro according to neuronal types (cortical or hippocampal) when utilized in experiments related with synaptogenesis or synaptic activities.
Collapse
Affiliation(s)
- Yun-Gi Kim
- Department of NanoBio Medical Science, College of Medicine, Dankook University, Cheonan 330-714, Korea
| | | |
Collapse
|
29
|
Dudok JJ, Groffen AJA, Toonen RFT, Verhage M. Deletion of Munc18-1 in 5-HT neurons results in rapid degeneration of the 5-HT system and early postnatal lethality. PLoS One 2011; 6:e28137. [PMID: 22140524 PMCID: PMC3226659 DOI: 10.1371/journal.pone.0028137] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2011] [Accepted: 11/02/2011] [Indexed: 12/30/2022] Open
Abstract
The serotonin (5-HT) system densely innervates many brain areas and is important for proper brain development. To specifically ablate the 5-HT system we generated mutant mice carrying a floxed Munc18-1 gene and Cre recombinase driven by the 5-HT-specific serotonin reuptake transporter (SERT) promoter. The majority of mutant mice died within a few days after birth. Immunohistochemical analysis of brains of these mice showed that initially 5-HT neurons are formed and the cortex is innervated with 5-HT projections. From embryonic day 16 onwards, however, 5-HT neurons started to degenerate and at postnatal day 2 hardly any 5-HT projections were present in the cortex. The 5-HT system of mice heterozygous for the floxed Munc18-1 allele was indistinguishable from control mice. These data show that deletion of Munc18-1 in 5-HT neurons results in rapid degeneration of the 5-HT system and suggests that the 5-HT system is important for postnatal survival.
Collapse
Affiliation(s)
- Jacobus J. Dudok
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Alexander J. A. Groffen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Ruud F. T. Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Kiyasova V, Gaspar P. Development of raphe serotonin neurons from specification to guidance. Eur J Neurosci 2011; 34:1553-62. [DOI: 10.1111/j.1460-9568.2011.07910.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
31
|
Han L, Itoh K, Yaoi T, Moriwaki S, Kato S, Nakamura K, Fushiki S. Prenatal and Lactational Exposure to Bisphenol A in Mice Alters Expression of Genes Involved in Cortical Barrel Development without Morphological Changes. Acta Histochem Cytochem 2011; 44:25-33. [PMID: 21448315 PMCID: PMC3061449 DOI: 10.1267/ahc.10042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 01/05/2011] [Indexed: 01/01/2023] Open
Abstract
It has been reported that premature infants in neonatal intensive care units are exposed to a high rate of bisphenol A (BPA), an endocrine disrupting chemical. Our previous studies demonstrated that corticothalamic projection was disrupted by prenatal exposure to BPA, which persisted even in adult mice. We therefore analyzed whether prenatal and lactational exposure to low doses of BPA affected the formation of the cortical barrel, the barreloid of the thalamus, and the barrelette of the brainstem in terms of the histology and the expression of genes involved in the barrel development. Pregnant mice were injected subcutaneously with 20 µg/kg of BPA daily from embryonic day 0 (E0) to postnatal 3 weeks (P3W), while the control mice received a vehicle alone. The barrel, barreloid and barrelette of the adult mice were examined by cytochrome C oxidase (COX) staining. There were no significant differences in the total and septal areas and the patterning of the posterior medial barrel subfield (PMBSF), barreloid and barrelette, between the BPA-exposure and control groups in the adult mice. The developmental study at postnatal day 1 (PD1), PD4 and PD8 revealed that the cortical barrel vaguely appeared at PD4 and completely formed at PD8 in both groups. The expression pattern of some genes was spatiotemporally altered depending on the sex and the treatment. These results suggest that the trigeminal projection and the thalamic relay to the cortical barrel were spared after prenatal and lactational exposure to low doses of BPA, although prenatal exposure to BPA was previously shown to disrupt the corticothalamic projection.
Collapse
Affiliation(s)
- Longzhe Han
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| | - Kyoko Itoh
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| | - Takeshi Yaoi
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| | - Sanzo Moriwaki
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| | - Shingo Kato
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| | - Keiko Nakamura
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| | - Shinji Fushiki
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| |
Collapse
|
32
|
Richerson GB, Buchanan GF. The serotonin axis: Shared mechanisms in seizures, depression, and SUDEP. Epilepsia 2011; 52 Suppl 1:28-38. [PMID: 21214537 DOI: 10.1111/j.1528-1167.2010.02908.x] [Citation(s) in RCA: 150] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
There is a growing appreciation that patients with seizures are also affected by a number of comorbid conditions, including an increase in prevalence of depression (Kanner, 2009), sleep apnea (Chihorek et al., 2007), and sudden death (Ryvlin et al., 2006; Tomson et al., 2008). The mechanisms responsible for these associations are unclear. Herein we discuss the possibility that underlying pathology in the serotonin (5-HT) system of patients with epilepsy lowers the threshold for seizures, while also increasing the risk of depression and sudden death. We propose that postictal dysfunction of 5-HT neurons causes depression of breathing and arousal in some epilepsy patients, and this can lead to sudden unexpected death in epilepsy (SUDEP). We further draw parallels between SUDEP and sudden infant death syndrome (SIDS), which may share pathophysiologic mechanisms, and which have both been linked to defects in the 5-HT system.
Collapse
|
33
|
Chronic administration of 13-cis-retinoic acid does not alter the number of serotoninergic neurons in the mouse raphe nuclei. Neuroscience 2011; 172:66-73. [PMID: 20977931 DOI: 10.1016/j.neuroscience.2010.10.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Revised: 10/18/2010] [Accepted: 10/19/2010] [Indexed: 12/24/2022]
Abstract
The synthetic retinoid 13-cis-retinoic acid (13-cis-RA), prescribed for the treatment of severe nodular acne, has been linked to an increased incidence of depression. Chronic treatment studies in rodents have shown that 13-cis-RA induces an increase in depression-related behaviours and a functional uncoupling of the hippocampus and dorsal raphe nucleus (DRN). Changes in the number of serotoninergic neurons in the DRN have been reported in depressed human patients. Given that retinoids have apoptotic effects, we hypothesized that a decrease in the number of serotoninergic neurons in the DRN or median raphe nucleus (MRN) would lead to decreased serotoninergic tone and in turn to the behavioural changes seen with 13-cis-RA administration. Here, we used immunolabelling and unbiased stereological methods to estimate the number of serotonin (5-hydroxytryptamine, 5-HT) neurons in the MRN and DRN of vehicle control and 13-cis-RA-treated adult mice. In the MRN, the number of 5-HT immunolabelled cells was 1815±194 in control, compared with 1954±111 in 13-cis-RA treated tissues. The number of 5-HT immunolabelled cells was much higher in the DRN, with 7148±377 cells in the control, compared with 7578±424 in the 13-cis-RA treated group. Further analysis of the DRN revealed that there were no changes in the number of 5-HT neurons within distinct subregions of the DRN. Similarly, changes in the density of serotoninergic neurons or in the volume of the MRN or DRN were not observed in 13-cis-RA treated animals. These data show that apoptotic actions of 13-cis-RA do not occur in vivo at drug concentrations that induce changes in depression-related behaviour and functional uncoupling of the DRN and hippocampus. The potential pro-depressant behavioural and molecular effects associated with chronic administration of 13-cis-RA may result from changes in serotoninergic activity rather than changes in the number of serotoninergic neurons.
Collapse
|
34
|
Zaccaria KJ, Lagace DC, Eisch AJ, McCasland JS. Resistance to change and vulnerability to stress: autistic-like features of GAP43-deficient mice. GENES, BRAIN, AND BEHAVIOR 2010; 9:985-96. [PMID: 20707874 PMCID: PMC2975747 DOI: 10.1111/j.1601-183x.2010.00638.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
There is an urgent need for animal models of autism spectrum disorder (ASD) to understand the underlying pathology and facilitate development and testing of new treatments. The synaptic growth-associated protein-43 (GAP43) has recently been identified as an autism candidate gene of interest. Our previous studies show many brain abnormalities in mice lacking one allele for GAP43 [GAP43 (+/-)] that are consistent with the disordered connectivity theory of ASD. Thus, we hypothesized that GAP43 (+/-) mice would show at least some autistic-like behaviors. We found that GAP43 (+/-) mice, relative to wild-type (+/+) littermates, displayed resistance to change, consistent with one of the diagnostic criteria for ASD. GAP43 (+/-) mice also displayed stress-induced behavioral withdrawal and anxiety, as seen in many autistic individuals. In addition, both GAP43 (+/-) mice and (+/+) littermates showed low social approach and lack of preference for social novelty, consistent with another diagnostic criterion for ASD. This low sociability is likely because of the mixed C57BL/6J 129S3/SvImJ background. We conclude that GAP43 deficiency leads to the development of a subset of autistic-like behaviors. As these behaviors occur in a mouse that displays disordered connectivity, we propose that future anatomical and functional studies in this mouse may help uncover underlying mechanisms for these specific behaviors. Strain-specific low sociability may be advantageous in these studies, creating a more autistic-like environment for study of the GAP43-mediated deficits of resistance to change and vulnerability to stress.
Collapse
Affiliation(s)
- K J Zaccaria
- Department of Cell & Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA.
| | | | | | | |
Collapse
|
35
|
Lovastatin induces neuronal differentiation and apoptosis of embryonal carcinoma and neuroblastoma cells: enhanced differentiation and apoptosis in combination with dbcAMP. Mol Cell Biochem 2010; 345:1-11. [PMID: 20694854 DOI: 10.1007/s11010-010-0553-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Accepted: 07/23/2010] [Indexed: 10/19/2022]
Abstract
Differentiation-based therapeutics are an underutilized but a potentially significant treatment option for cancer patients. We show that lovastatin, a competitive inhibitor of the rate-limiting enzyme of mevalonate synthesis HMG-CoA reductase, is able to induce tumour cell differentiation and apoptosis in vitro. We used embryonal carcinoma (EC) and neuroblastoma (NB) cell lines and found that lovastatin promoted apoptosis and induced expression of the neuronal differentiation markers, tyrosine hydroxylase (TH), and growth-associated protein 43. The apoptotic and differentiation responses were time and dose-dependant and rescued by the co-administration of mevalonate. The expression of TH is regulated primarily by a cyclic AMP (cAMP) response element (CRE) in its promoter. Lovastatin enhanced the expression of a CRE-driven luciferase construct in P19 cells. Furthermore, combining lovastatin with 1 mM dibutyryladenosine 3',5'-cyclic monophosphate treatments induced higher expression from the CRE construct, enhanced differentiation and cytotoxicity. This study suggests the potential of combining these therapeutic approaches in EC and NB patients.
Collapse
|
36
|
Serotonin: a regulator of neuronal morphology and circuitry. Trends Neurosci 2010; 33:424-34. [PMID: 20561690 DOI: 10.1016/j.tins.2010.05.005] [Citation(s) in RCA: 213] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Revised: 05/17/2010] [Accepted: 05/18/2010] [Indexed: 11/22/2022]
Abstract
Serotonin is an important neuromodulator associated with a wide range of physiological effects in the central nervous system. The exact mechanisms whereby serotonin influences brain development are not well understood, although studies in invertebrate and vertebrate model organisms are beginning to unravel a regulatory role for serotonin in neuronal morphology and circuit formation. Recent data suggest a developmental window during which altered serotonin levels permanently influence neuronal circuitry, however, the temporal constraints and molecular mechanisms responsible are still under investigation. Growing evidence suggests that alterations in early serotonin signaling contribute to a number of neurodevelopmental and neuropsychiatric disorders. Thus, understanding how altered serotonin signaling affects neuronal morphology and plasticity, and ultimately animal physiology and pathophysiology, will be of great significance.
Collapse
|
37
|
Rooney GE, Howard L, O'Brien T, Windebank AJ, Barry FP. Elevation of cAMP in mesenchymal stem cells transiently upregulates neural markers rather than inducing neural differentiation. Stem Cells Dev 2009; 18:387-98. [PMID: 18554089 DOI: 10.1089/scd.2008.0080] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The aims of this research were to examine the neural expression profile of undifferentiated mesenchymal stem cells (MSCs), to define a serum-free environment that would support the survival of MSCs, and to assess the effects of elevated cyclic adenosine monophosphate (cAMP) levels on MSC morphology and expression of neural markers. The neural profile of MSCs was assessed using immunocytochemistry and real-time polymerase chain reaction (PCR) techniques. These MSCs were then cultured in varying serum-free environments, with flow cytometry analysis of cell viability and apoptosis. The effects of forskolin and 8 bromo-cAMP treatment on MSC morphology and expression of neural markers were assessed using light microscopy, immunocytochemistry and real-time PCR analysis. Expression of the neural markers nestin, vimentin, beta III tubulin, glial fibrillary acidic protein, and the tropomyosin-related kinases was demonstrated in normal undifferentiated MSCs. Furthermore, MSCs cultured in serum-free conditions containing ascorbic acid 2-phosphate demonstrated greater long-term survival and reduced apoptosis. In contrast, forskolin increased the percentage of cells undergoing apoptosis. Culture in the presence of forskolin induced a 6-fold increase in beta III tubulin expression after a 6-h exposure time, which was accompanied by dramatic changes in morphology. However, this effect was transient, with the cells reverting to their normal morphology by 24 h. Treatment with 8 bromo-cAMP induced similar increases in beta III tubulin expression without such dramatic morphological changes. Factors which increase the concentration of cAMP induce transient changes in expression of neural markers that appear to be the result of cellular adaptation to changes in culture conditions rather than a real process of neural differentiation.
Collapse
Affiliation(s)
- Gemma E Rooney
- Regenerative Medicine Institute, National Centre for Biomedical and Engineering Science, National University of Ireland, Galway, Ireland
| | | | | | | | | |
Collapse
|
38
|
Zhao HS, Chen SJ, Wu N, Wang XQ, Yin ZQ, Wang Y. LEDGFp52 controls rat retinal ganglion cell neurite growth in culture and regulates specific neuronal growth-associated genes and protein production. J Int Med Res 2008; 36:815-29. [PMID: 18652779 DOI: 10.1177/147323000803600425] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
We investigated the regulation of primary neurite growth and expression of specific growth-associated genes by lens epithelium-derived growth factor (LEDGF) in rat retinal ganglion cells (RGCs). A pAd-LEDGFp52 adenovirus vector and a siRNA-LEDGFp52 eucaryotic expression vector were transfected into cultured RGCs. Transfection with pAd-LEDGFp52 significantly increased the number of neurites and their lengths compared with untransfected control RGCs. The expression of growth associated protein 43 (GAP43), microtubule-associated protein 2 (MAP2), and low-molecular-weight neurofilament (NF-L) genes and proteins were also significantly up-regulated. In contrast, the introduction of siRNA-LEDGFp52 significantly decreased the number and length of neurites, and significantly down-regulated the expression GAP43, NF-L and MAP2 genes and proteins compared with controls. Our findings suggest that LEDGFp52 might act as a dendritic arborization gene as well as an axonal elongation gene in RGCs and that it might be beneficial to the functional recovery of regenerating RGCs.
Collapse
Affiliation(s)
- H S Zhao
- Southwest Eye Hospital/Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
39
|
A critical importance of polyamine site in NMDA receptors for neurite outgrowth and fasciculation at early stages of P19 neuronal differentiation. Exp Cell Res 2008; 314:2603-17. [PMID: 18586028 DOI: 10.1016/j.yexcr.2008.06.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Revised: 05/27/2008] [Accepted: 06/05/2008] [Indexed: 01/16/2023]
Abstract
We have investigated the role of N-methyl-d-aspartate receptors (NMDARs) and gamma-aminobutyric acid receptors type A (GABA(A)Rs) at an early stage of P19 neuronal differentiation. The subunit expression was profiled in 24-hour intervals with RT-PCR and functionality of the receptors was verified via fluo-3 imaging of Ca(2+) dynamics in the immature P19 neurons showing that both NMDA and GABA excite neuronal bodies, but only polyamine-site sensitive NMDAR stimulation leads to enhanced Ca(2+) signaling in the growth cones. Inhibition of NR1/NR2B NMDARs by 1 muM ifenprodil severely impaired P19 neurite extension and fasciculation, and this negative effect was fully reversible by polyamine addition. In contrast, GABA(A)R antagonism by a high dose of 200 microM bicuculline had no observable effect on P19 neuronal differentiation and fasciculation. Except for the differential NMDAR and GABA(A)R profiles of Ca(2+) signaling within the immature P19 neurons, we have also shown that inhibition of NR1/NR2B NMDARs strongly decreased mRNA level of NCAM-180, which has been previously implicated as a regulator of neuronal growth cone protrusion and neurite extension. Our data thus suggest a critical role of NR1/NR2B NMDARs during the process of neuritogenesis and fasciculation of P19 neurons via differential control of local growth cone Ca(2+) surges and NCAM-180 signaling.
Collapse
|
40
|
Donovan SL, McCasland JS. GAP-43 is critical for normal targeting of thalamocortical and corticothalamic, but not trigeminothalamic axons in the whisker barrel system. Somatosens Mot Res 2008; 25:33-47. [PMID: 18344146 DOI: 10.1080/08990220701830696] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mice lacking the growth-associated protein GAP-43 (KO) show disrupted cortical topography and no barrels. Whisker-related patterns of cells are normal in the KO brainstem trigeminal complex (BSTC), while the pattern in KO ventrobasal thalamus (VB) is somewhat compromised. To better understand the basis for VB and cortical abnormalities, we used small placements of DiI to trace axonal projections between BSTC, VB, and barrel cortex in wildtype (WT) and GAP-43 KO mice. The trigeminothalamic (TT) pathway consists of axons from cells in the Nucleus Prinicipalis that project to the contralateral VB thalamus. DiI-labeled KO TT axons crossed the midline from BSTC and projected to contralateral VB normally, consistent with normal BSTC cytoarchitecture. By contrast, the KO thalamocortical axons (TCA) projection was highly abnormal. KO TCAs showed delays of 1-2 days in initial ingrowth to cortex. Postnatally, KO TCAs showed multiple pathfinding errors near intermediate targets, and were abnormally fasciculated within the internal capsule (IC). Interestingly, most individually labeled KO TCAs terminated in deep layers instead of in layer IV as in WT. This misprojection is consistent with birthdating analysis in KO mice, which revealed that neurons normally destined for layer IV remain in deep cortical layers. Early outgrowth of KO corticofugal (CF) axons was similar for both genotypes. However, at P7 KO CF fibers remained bundled as they entered the IC, and exhibited few terminal branches in VB. Thus, the establishment of axonal projections between thalamus and cortex are disrupted in GAP-43 KO mice.
Collapse
Affiliation(s)
- Stacy L Donovan
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | | |
Collapse
|
41
|
Estivill-Torrús G, Llebrez-Zayas P, Matas-Rico E, Santín L, Pedraza C, De Diego I, Del Arco I, Fernández-Llebrez P, Chun J, De Fonseca FR. Absence of LPA1 signaling results in defective cortical development. ACTA ACUST UNITED AC 2007; 18:938-50. [PMID: 17656621 DOI: 10.1093/cercor/bhm132] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Lysophosphatidic acid (LPA) is a simple phospholipid with extracellular signaling properties mediated by specific G protein-coupled receptors. At least 2 LPA receptors, LPA(1) and LPA(2), are expressed in the developing brain, the former enriched in the neurogenic ventricular zone (VZ), suggesting a normal role in neurogenesis. Despite numerous studies reporting the effects of exogenous LPA using in vitro neural models, the first LPA(1) loss-of-function mutants reported did not show gross cerebral cortical defects in the 50% that survived perinatal demise. Here, we report a role for LPA(1) in cortical neural precursors resulting from analysis of a variant of a previously characterized LPA(1)-null mutant that arose spontaneously during colony expansion. These LPA(1)-null mice, termed maLPA(1), exhibit almost complete perinatal viability and show a reduced VZ, altered neuronal markers, and increased cortical cell death that results in a loss of cortical layer cellularity in adults. These data support LPA(1) function in normal cortical development and suggest that the presence of genetic modifiers of LPA(1) influences cerebral cortical development.
Collapse
|
42
|
Luellen BA, Bianco LE, Schneider LM, Andrews AM. Reduced brain-derived neurotrophic factor is associated with a loss of serotonergic innervation in the hippocampus of aging mice. GENES BRAIN AND BEHAVIOR 2007; 6:482-90. [PMID: 17156118 DOI: 10.1111/j.1601-183x.2006.00279.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) regulates monoamine neuronal growth, survival and function in development and throughout adulthood. At 18 months of age, mice with constitutive reductions in BDNF expression show decreased serotonin innervation in the hippocampus compared with age-matched wildtype mice. It is not known, however, whether age-accelerated loss of serotonergic innervation in BDNF(+/-) mice occurs in other brain regions, advances beyond 18 months or is associated with alterations in other neurotransmitter systems. In this study, immunocytochemistry was used to assess serotonergic and catecholaminergic innervation in 26-month-old BDNF(+/-) mice. Age-related loss of serotonin axons in the hippocampus was potentiated in BDNF(+/-) mice compared with wildtype mice at this late age, particularly in the CA1 subregion. By contrast, aging BDNF(+/-) mice showed increased serotonin innervation of the basomedial nucleus of the amygdala. In the noradrenergic system, BDNF(+/-) mice showed reduced numbers of cell bodies and fibers in the locus coeruleus compared with age-matched wildtype mice, whereas no changes were observed in dopaminergic innervation with respect to genotype. In vivo zero net flux microdialysis in awake mice showed a significant decrease in extracellular serotonin levels in the hippocampus in BDNF(+/-) mice at 20 months of age. Thus, reduced BDNF is associated with altered serotonergic and noradrenergic innervation in aging mice and, in particular, with accelerated loss of serotonergic innervation to the hippocampus that is manifest as a decrease in basal neurotransmission.
Collapse
Affiliation(s)
- B A Luellen
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802-4615, USA
| | | | | | | |
Collapse
|
43
|
Albright MJ, Weston MC, Inan M, Rosenmund C, Crair MC. Increased thalamocortical synaptic response and decreased layer IV innervation in GAP-43 knockout mice. J Neurophysiol 2007; 98:1610-25. [PMID: 17581849 DOI: 10.1152/jn.00219.2007] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The growth-associated protein, GAP-43, is an axonally localized neuronal protein with high expression in the developing brain and in regenerating neurites. Mice that lack GAP-43 (GAP-43 -/-) fail to form a whisker-related barrel map. In this study, we use GAP-43 -/- mice to examine GAP-43 synaptic function in the context of thalamocortical synapse development and cortical barrel map formation. Examination of thalamocortical synaptic currents in an acute brain slice preparation and in autaptic thalamic neurons reveals that GAP-43 -/- synapses have larger alpha-amino-3-hydroxyl-5-methyl-4-isoxazolepropionate receptor (AMPAR)-mediated currents than controls despite similar AMPAR function and normal probability of vesicular release. Interestingly, GAP-43 -/- synapses are less sensitive to blockade by a competitive glutamate receptor antagonist, suggesting higher levels of neurotransmitter in the cleft during synaptic transmission. Field excitatory postsynaptic potentials (EPSPs) from GAP-43 -/- thalamocortical synapses reveal a reduced fiber response, and anatomical analysis shows reduced thalamic innervation of barrel cortex in GAP-43 -/- mice. Despite this fact synaptic responses in the field EPSPs are similar in GAP-43 -/- mice and wild-type littermate controls, and the ratio of AMPAR-mediated to N-methyl-d-aspartate receptor (NMDAR)-mediated currents (AMPAR:NMDAR ratio) is larger than normal. This suggests that GAP-43 -/- mice form fewer thalamocortical synapses in layer IV because of decreased anatomical innervation of the cortex, but the remaining contacts are individually stronger possibly due to increased neurotransmitter concentration in the synaptic cleft. Together, these results indicate that in addition to its well known role in axonal pathfinding GAP-43 plays a functional role in regulating neurotransmitter release.
Collapse
Affiliation(s)
- Michael J Albright
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | |
Collapse
|
44
|
Hohmann CF, Walker EM, Boylan CB, Blue ME. Neonatal serotonin depletion alters behavioral responses to spatial change and novelty. Brain Res 2007; 1139:163-77. [PMID: 17296168 PMCID: PMC1974858 DOI: 10.1016/j.brainres.2006.12.095] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2006] [Revised: 10/19/2006] [Accepted: 12/29/2006] [Indexed: 10/23/2022]
Abstract
Multiple brain disorders that show serotonergic imbalances have a developmental onset. Experimental models indicate a role for serotonin as a morphogen in brain development. To selectively study the effects of serotonin depletions on cortical structural development and subsequent behavior, we developed a mouse model in which a serotonin neurotoxin, 5,7-dihydroxytryptamine (5,7-DHT), is injected into the medial forebrain bundle (mfb) on the day of birth. Littermates with saline injections into the mfb and age matched mice served as controls. This study characterized the extent and duration of serotonergic denervation after the selective neonatal lesion and investigated effects on exploratory behavior, spatial learning and anxiety in mice of both sexes. We report significant decreases in the serotonergic (5-HT) innervation to cortex and hippocampus, but not to subcortical forebrain structures in 5,7-DHT-lesioned mice. The depletion of 5-HT fibers in cortical areas was long lasting in lesioned mice but autoradiographic binding to high affinity 5-HT transporters was only transiently reduced. Male but not female lesioned mice reduced their exploration significantly in response to spatial rearrangement and object novelty, suggesting increased anxiety in response to change but normal spatial cognition. Our data show that developmental disruptions in the serotonergic innervation of cortex and hippocampus are sufficient to induce permanent, sex specific, behavioral alterations. These results may have significant implications for understanding brain disorders presenting with cortical morphogenetic abnormalities and altered serotonin neurotransmission, such as autism, schizophrenia and affective disorders.
Collapse
|
45
|
Kwaśnicka-Crawford DA, Carson AR, Scherer SW. IQCJ-SCHIP1, a novel fusion transcript encoding a calmodulin-binding IQ motif protein. Biochem Biophys Res Commun 2006; 350:890-9. [PMID: 17045569 DOI: 10.1016/j.bbrc.2006.09.136] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2006] [Accepted: 09/21/2006] [Indexed: 10/24/2022]
Abstract
The existence of transcripts that span two adjacent, independent genes is considered rare in the human genome. This study characterizes a novel human fusion gene named IQCJ-SCHIP1. IQCJ-SCHIP1 is the longest isoform of a complex transcriptional unit that bridges two separate genes that encode distinct proteins, IQCJ, a novel IQ motif containing protein and SCHIP1, a schwannomin interacting protein that has been previously shown to interact with the Neurofibromatosis type 2 (NF2) protein. IQCJ-SCHIP1 is located on the chromosome 3q25 and comprises a 1692-bp transcript encompassing 11 exons spanning 828kb of the genomic DNA. We show that IQCJ-SCHIP1 mRNA is highly expressed in the brain. Protein encoded by the IQCJ-SCHIP1 gene was localized to cytoplasm and actin-rich regions and in differentiated PC12 cells was also seen in neurite extensions.
Collapse
|
46
|
Vohra BP, Tsuji K, Nagashimada M, Uesaka T, Wind D, Armon J, Enomoto H, Heuckeroth RO. Differential gene expression and functional analysis implicate novel mechanisms in enteric nervous system precursor migration and neuritogenesis. Dev Biol 2006; 298:259-71. [PMID: 16904662 PMCID: PMC1952185 DOI: 10.1016/j.ydbio.2006.06.033] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2006] [Revised: 05/17/2006] [Accepted: 06/22/2006] [Indexed: 10/24/2022]
Abstract
Enteric nervous system (ENS) development requires complex interactions between migrating neural-crest-derived cells and the intestinal microenvironment. Although some molecules influencing ENS development are known, many aspects remain poorly understood. To identify additional molecules critical for ENS development, we used DNA microarray, quantitative real-time PCR and in situ hybridization to compare gene expression in E14 and P0 aganglionic or wild type mouse intestine. Eighty-three genes were identified with at least two-fold higher expression in wild type than aganglionic bowel. ENS expression was verified for 39 of 42 selected genes by in situ hybridization. Additionally, nine identified genes had higher levels in aganglionic bowel than in WT animals suggesting that intestinal innervation may influence gene expression in adjacent cells. Strikingly, many synaptic function genes were expressed at E14, a time when the ENS is not needed for survival. To test for developmental roles for these genes, we used pharmacologic inhibitors of Snap25 or vesicle-associated membrane protein (VAMP)/synaptobrevin and found reduced neural-crest-derived cell migration and decreased neurite extension from ENS precursors. These results provide an extensive set of ENS biomarkers, demonstrate a role for SNARE proteins in ENS development and highlight additional candidate genes that could modify Hirschsprung's disease penetrance.
Collapse
Affiliation(s)
- Bhupinder P.S. Vohra
- Department of Pediatrics and Department of Molecular Biology and Pharmacology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8208, St. Louis, MO 63110 U.S.A
| | - Keiji Tsuji
- Laboratory for Neuronal Differentiation and Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Mayumi Nagashimada
- Laboratory for Neuronal Differentiation and Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
- Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshihiro Uesaka
- Laboratory for Neuronal Differentiation and Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Daniel Wind
- Department of Pediatrics and Department of Molecular Biology and Pharmacology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8208, St. Louis, MO 63110 U.S.A
| | - Jennifer Armon
- Department of Pediatrics and Department of Molecular Biology and Pharmacology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8208, St. Louis, MO 63110 U.S.A
| | - Hideki Enomoto
- Laboratory for Neuronal Differentiation and Regeneration, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Robert O. Heuckeroth
- Department of Pediatrics and Department of Molecular Biology and Pharmacology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8208, St. Louis, MO 63110 U.S.A
| |
Collapse
|
47
|
Janusonis S, Anderson GM, Shifrovich I, Rakic P. Ontogeny of brain and blood serotonin levels in 5-HT receptor knockout mice: potential relevance to the neurobiology of autism. J Neurochem 2006; 99:1019-31. [PMID: 16981893 DOI: 10.1111/j.1471-4159.2006.04150.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The most consistent neurochemical finding in autism has been elevated group mean levels of blood platelet 5-hydroxytryptamine (5-HT, serotonin). The origin and significance of this platelet hyperserotonemia remain poorly understood. The 5-HT(1A) receptor plays important roles in the developing brain and is also expressed in the gut, the main source of platelet 5-HT. Post-natal tissue levels of 5-HT, 5-hydroxyindoleacetic acid (5-HIAA) and tryptophan were examined in the brain, duodenum and blood of 5-HT(1A) receptor-knockout and wild-type mice. At 3 days after birth, the knockout mice had lower mean brain 5-HT levels and normal mean platelet 5-HT levels. Also, at 3 days after birth, the mean tryptophan levels in the brain, duodenum and blood of the knockout mice were around 30% lower than those of the wild-type mice. By 2 weeks after birth, the mean brain 5-HT levels of the knockout mice normalized, but their mean platelet 5-HT levels became 24% higher than normal. The possible causes of these dynamic shifts were explored by examining correlations between central and peripheral levels of 5-HT, 5-HIAA and tryptophan. The results are discussed in relation to the possible role of 5-HT in the ontogeny of autism.
Collapse
Affiliation(s)
- Skirmantas Janusonis
- Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut, USA.
| | | | | | | |
Collapse
|
48
|
McIlvain V, McCasland JS. GAP-43 heterozygous mice show delayed barrel patterning, differentiation of radial glia, and downregulation of GAP-43. ACTA ACUST UNITED AC 2006; 288:143-57. [PMID: 16435363 DOI: 10.1002/ar.a.20291] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
GAP-43 heterozygous (HZ) mice exhibit abnormal thalamocortical pathfinding, fasciculation, and terminal arborization at postnatal day 7 (P7). Here we tested whether these defects are correlated with delayed development of HZ cortical patterns. We assessed the rate of barrel segregation and radial glia differentiation in wild-type (WT) and HZ cortices. Since GAP-43 is involved in some forms of neural plasticity, we also compared the duration of the critical period for lesion-induced plasticity in both genotypes. Cytochrome oxidase histochemistry revealed a delay of approximately 1 day in barrel pattern formation in GAP-43 HZ mice. GAP-43 WT barrels showed complete segregation between P2-P3, while HZ barrels did not reach the same level of segregation until P3-P4. We found a similar delay in the transformation of radial glia from monopolar to multipolar phenotypes, from P5 in WT to P7 in HZ cortex. Radial glial cells represent many of the neuronal progenitors in developing cortex and aid in cell migration. Thus, the delay in radial glial differentiation may contribute to the delay in HZ barrel segregation. Interestingly, we found no change in the extent of the critical period for HZ cortical responsiveness to early peripheral damage or in the time course of the cortical response. As expected, GAP-43 expression in HZ cortex is significantly reduced early in development. However, HZ GAP-43 expression remains at maximum levels after P9, when it is normally downregulated. As a result, HZ GAP-43 expression is near-normal by P26, by which time near-normal barrel dimensions have been restored. Our findings indicate that GAP-43 deficiency leads to early delays in barrel development and suggest that these failures are followed by homeostatic responses, including prolonged GAP-43 expression. These compensatory mechanisms may rescue normal cortical reorganization in neonates and near-normal barrel morphology and GAP-43 expression in adulthood.
Collapse
Affiliation(s)
- Vera McIlvain
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York 13210, USA
| | | |
Collapse
|
49
|
Luellen BA, Szapacs ME, Materese CK, Andrews AM. The neurotoxin 2′-NH2-MPTP degenerates serotonin axons and evokes increases in hippocampal BDNF. Neuropharmacology 2006; 50:297-308. [PMID: 16288930 DOI: 10.1016/j.neuropharm.2005.09.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2005] [Revised: 09/03/2005] [Accepted: 09/05/2005] [Indexed: 11/15/2022]
Abstract
1-Methyl-4-(2'-aminophenyl)-1,2,3,6-tetrahydropyridine (2'-NH2-MPTP) causes long-term depletions in cortical and hippocampal serotonin (5-HT) and norepinephrine (NE) that are accompanied by acute elevations in glial fibrillary acidic protein (GFAP) and argyrophilia. To further investigate the hypothesis that these changes are reflective of serotonergic and noradrenergic axonal degeneration, 2'-NH2-MPTP was administered to mice and innervation densities were determined immunocytochemically. Regional responses of the neurotrophin, brain-derived neurotrophic factor (BDNF), to putative damage were also assessed. Three days after 2'-NH2-MPTP, 5-HT axons exhibited a beaded, tortuous appearance indicative of ongoing degeneration. At 21 days, numbers of serotonin axons were significantly decreased, with the greatest axonal losses occurring in cortex and hippocampus. Serotonin axons in the amygdala were contrastingly spared long-term damage, as were 5-HT and NE cell bodies in the brain stem. BDNF protein levels were selectively increased in the hippocampus 3 days post-dose and returned to normal 21 days later. These results, in conjunction with previous findings, demonstrate that 2'-NH2-MPTP causes degeneration of serotonergic axons innervating the cortex and hippocampus on par with depletions in neurotransmitter levels. Moreover, damage to the hippocampus, a brain region important for learning and memory, and the modulation of anxiety and stress responsiveness, results in a transitory increase in BDNF.
Collapse
Affiliation(s)
- Beth A Luellen
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | |
Collapse
|
50
|
Yang Y, Xie Y, Chai H, Fan M, Liu S, Liu H, Bruce I, Wu W. Microarray analysis of gene expression patterns in adult spinal motoneurons after different types of axonal injuries. Brain Res 2006; 1075:1-12. [PMID: 16460709 DOI: 10.1016/j.brainres.2005.12.060] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Revised: 10/05/2005] [Accepted: 12/04/2005] [Indexed: 10/25/2022]
Abstract
Three experimental models of axonal injuries in adult rat spinal motoneurons were established to investigate changes of gene expression in response to such injuries. We took advantage of cDNA microarray analysis to determine the differential expression of genes in injured motoneurons following distal axotomy or root avulsion in the absence or presence of BDNF. The major finding was that, in response to proximal axonal injury (avulsion), expression of genes that are known to facilitate neuronal survival and axonal regeneration (e.g., IGFRII, PI3K, IGFBP-6, GSTs, GalR2) were down-regulated; but following treatment with BDNF they were up-regulated. In addition, the expression of genes known to be involved in apoptosis and DNA damage (e.g., ANX5, TS, ALR) were down-regulated in BDNF-treated animals with avulsion. Furthermore, many functional families of genes previously shown to play roles in the pathophysiology of axonal injury, including SNAP-25A, SV2B, Ras-related ras3a/4b, ERK1/2, 14-3-3 proteins, proteasome proteins, oncogenes, GAP-43, and NMDAR1, were altered after either distal axotomy or avulsion injury. Some of the changes in gene expression, including Lim-2, FRAG1, GlaR2, GSTs, ALR, TS, ANX3/5, and nhe1/2, are first reported here in injured motoneurons. The differential expression of genes identified by the expression arrays was confirmed by gene-specific RT-PCR for eight genes (GAP-43, IGFR II, Lim-2, MIF, NDAP1, TS, PCC3, and FRAG1) and by in situ hybridization for Lim-2. These results suggest that abnormal regulation of particular biochemical pathways may induce motoneuron death after ventral root avulsion in adult animals. This study presents an approach for selecting specific genes and their products that may be involved in motoneuron degeneration following axonal injuries.
Collapse
Affiliation(s)
- Yi Yang
- Department of Anatomy, Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | | | | | | | | | | | | | | |
Collapse
|