1
|
Zhang F, Chen J, Li Y, Ye J, Wang C. Neuronal Scaffold Protein ARMS Interacts with Synaptotagmin-4 C2AB through the Ankyrin Repeat Domain with an Unexpected Mode. Int J Mol Sci 2023; 24:16993. [PMID: 38069318 PMCID: PMC10707181 DOI: 10.3390/ijms242316993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/21/2023] [Accepted: 11/26/2023] [Indexed: 12/18/2023] Open
Abstract
The ankyrin repeat-rich membrane spanning (ARMS), a transmembrane neuronal scaffold protein, plays a fundamental role in neuronal physiology, including neuronal development, polarity, differentiation, survival and angiogenesis, through interactions with diverse partners. Previous studies have shown that the ARMS negatively regulates brain-derived neurotrophic factor (BDNF) secretion by interacting with Synaptotagmin-4 (Syt4), thereby affecting neurogenesis and the development and function of the nervous system. However, the molecular mechanisms of the ARMS/Syt4 complex assembly remain unclear. Here, we confirmed that the ARMS directly interacts with Syt4 through its N-terminal ankyrin repeats 1-8. Unexpectedly, both the C2A and C2B domains of Syt4 are necessary for binding with the ARMS. We then combined the predicted complex structural models from AlphaFold2 with systematic biochemical analyses using point mutagenesis to underline the molecular basis of ARMS/Syt4 complex formation and to identify two conserved residues, E15 and W72, of the ARMS, as essential residues mediating the assembly of the complex. Furthermore, we showed that ARMS proteins are unable to interact with Syt1 or Syt3, indicating that the interaction between ARMS and Syt4 is specific. Taken together, the findings from this study provide biochemical details on the interaction between the ARMS and Syt4, thereby offering a biochemical basis for the further understanding of the potential mechanisms and functional implications of the ARMS/Syt4 complex formation, especially with regard to the modulation of BDNF secretion and associated neuropathies.
Collapse
Affiliation(s)
- Fa Zhang
- Hefei National Research Center for Physical Sciences at the Microscale, MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Jiasheng Chen
- Hefei National Research Center for Physical Sciences at the Microscale, MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Yahong Li
- Hefei National Research Center for Physical Sciences at the Microscale, MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Jin Ye
- Hefei National Research Center for Physical Sciences at the Microscale, MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Chao Wang
- Hefei National Research Center for Physical Sciences at the Microscale, MOE Key Laboratory for Membraneless Organelles & Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Biomedical Sciences and Health Laboratory of Anhui Province, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
2
|
Orciani C, Hall H, Pentz R, Foret MK, Do Carmo S, Cuello AC. Long-term nucleus basalis cholinergic depletion induces attentional deficits and impacts cortical neurons and BDNF levels without affecting the NGF synthesis. J Neurochem 2022; 163:149-167. [PMID: 35921478 DOI: 10.1111/jnc.15683] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 11/26/2022]
Abstract
Basal forebrain cholinergic neurons (BFCNs) represent the main source of cholinergic innervation to the cortex and hippocampus and degenerate early in Alzheimer's disease (AD) progression. Phenotypic maintenance of BFCNs depends on levels of mature nerve growth factor (mNGF) and mature brain-derived neurotrophic factor (mBDNF), produced by target neurons and retrogradely transported to the cell body. Whether a reciprocal interaction where BFCN inputs impact neurotrophin availability and affect cortical neuronal markers is unknown. To address our hypothesis, we immunolesioned the nucleus basalis (nb), a basal forebrain cholinergic nuclei projecting mainly to the cortex, by bilateral stereotaxic injection of 192-IgG-Saporin (the cytotoxin Saporin binds p75ntr receptors expressed exclusively by BFCNs) in 2.5-month-old Wistar rats. At six months post-lesion, Saporin-injected rats (SAP) showed an impairment in a modified version of the 5-Choice Serial Reaction Time Task (5-choice task). Post-mortem analyses of the brain revealed a reduction of Choline Acetyltransferase-immunoreactive neurons compared to wild-type controls. A diminished number of cortical vesicular acetylcholine transporter-immunoreactive boutons was accompanied by a reduction in BDNF mRNA, mBDNF protein levels, markers of glutamatergic (vGluT1) and GABAergic (GAD65) neurons in the SAP-group compared to the controls. NGF mRNA, NGF precursor and mNGF protein levels were not affected. Additionally, cholinergic markers correlated with the attentional deficit and BDNF levels. Our findings demonstrate that while cholinergic nb loss impairs cognition and reduces cortical neuron markers, it produces differential effects on neurotrophin availability, affecting BDNF but not NGF levels.
Collapse
Affiliation(s)
- Chiara Orciani
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Helene Hall
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Rowan Pentz
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Morgan K Foret
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Sonia Do Carmo
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - A Claudio Cuello
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada.,Department of Pharmacology, Oxford University, US (Visiting Professor)
| |
Collapse
|
3
|
The role of serotonin neurotransmission in rapid antidepressant actions. Psychopharmacology (Berl) 2022; 239:1823-1838. [PMID: 35333951 DOI: 10.1007/s00213-022-06098-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 02/16/2022] [Indexed: 10/18/2022]
Abstract
RATIONALE Ketamine has rapid antidepressant effects that represent a significant advance in treating depression, but its poor safety and tolerability limit its clinical utility. Accreting evidence suggests that serotonergic neurotransmission participates in the rapid antidepressant effects of ketamine and hallucinogens. Thus, understanding how serotonin contributes to these effects may allow identification of novel rapid antidepressant mechanisms with improved tolerability. OBJECTIVE The goal of this paper is to understand how serotonergic mechanisms participate in rapid antidepressant mechanisms. METHODS We review the relevance of serotonergic neurotransmission for rapid antidepressant effects and evaluate the role of 5-HT1A, 5-HT1B, 5-HT2A, and 5-HT4 receptors in synaptic plasticity, BDNF signaling, and GSK-3β activity. Subsequently, we develop hypotheses on the relationship of these receptor systems to rapid antidepressant effects. RESULTS We found that 5-HT1A and 5-HT1B receptors may participate in ketamine's rapid antidepressant mechanisms, while agonists at 5-HT2A and 5-HT4 receptors may independently behave as rapid antidepressants. 5-HT1A, 5-HT2A, and 5-HT4 receptors increase synaptic plasticity in the cortex or hippocampus but do not consistently increase BDNF signaling. We found that 5-HT1A and 5-HT1B receptors may participate in rapid antidepressant mechanisms as a consequence of increased BDNF signaling, rather than a cause. 5-HT2A and 5-HT4 receptor agonists may increase BDNF signaling, but these relationships are tenuous and need more study. Finally, we found that ketamine and several serotonergic receptor systems may mechanistically converge on reduced GSK-3β activity. CONCLUSIONS We find it plausible that serotonergic neurotransmission participates in rapid antidepressant mechanisms by increasing synaptic plasticity, perhaps through GSK-3β inhibition.
Collapse
|
4
|
Influenza A Virus (H1N1) Infection Induces Microglial Activation and Temporal Dysbalance in Glutamatergic Synaptic Transmission. mBio 2021; 12:e0177621. [PMID: 34700379 PMCID: PMC8546584 DOI: 10.1128/mbio.01776-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Influenza A virus (IAV) causes respiratory tract disease and is responsible for seasonal and reoccurring epidemics affecting all age groups. Next to typical disease symptoms, such as fever and fatigue, IAV infection has been associated with behavioral alterations presumably contributing to the development of major depression. Previous experiments using IAV/H1N1 infection models have shown impaired hippocampal neuronal morphology and cognitive abilities, but the underlying pathways have not been fully described. In this study, we demonstrate that infection with a low-dose non-neurotrophic H1N1 strain of IAV causes ample peripheral immune response followed by a temporary blood-brain barrier disturbance. Although histological examination did not reveal obvious pathological processes in the brains of IAV-infected mice, detailed multidimensional flow cytometric characterization of immune cells uncovered subtle alterations in the activation status of microglial cells. More specifically, we detected an altered expression pattern of major histocompatibility complex classes I and II, CD80, and F4/80 accompanied by elevated mRNA levels of CD36, CD68, C1QA, and C3, suggesting evolved synaptic pruning. To closer evaluate how these profound changes affect synaptic balance, we established a highly sensitive multiplex flow cytometry-based approach called flow synaptometry. The introduction of this novel technique enabled us to simultaneously quantify the abundance of pre- and postsynapses from distinct brain regions. Our data reveal a significant reduction of VGLUT1 in excitatory presynaptic terminals in the cortex and hippocampus, identifying a subtle dysbalance in glutamatergic synapse transmission upon H1N1 infection in mice. In conclusion, our results highlight the consequences of systemic IAV-triggered inflammation on the central nervous system and the induction and progression of neuronal alterations. IMPORTANCE Influenza A virus (IAV) causes mainly respiratory tract disease with fever and fatigue but is also associated with behavioral alterations in humans. Here, we demonstrate that infection with a low-dose non-neurotrophic H1N1 strain of IAV causes peripheral immune response followed by a temporary blood-brain barrier disturbance. Characterization of immune cells uncovered subtle alterations in the activation status of microglia cells that might reshape neuronal synapses. We established a highly sensitive multiplex flow cytometry-based approach called flow synaptometry to more closely study the synapses. Thus, we detected a specific dysbalance in glutamatergic synapse transmission upon H1N1 infection in mice. In conclusion, our results highlight the consequences of systemic IAV-triggered inflammation on the central nervous system and the induction and progression of neuronal alterations.
Collapse
|
5
|
Fyk-Kolodziej BE, Mueller PJ. Sedentary Conditions Promote Subregionally Specific Changes in Brain-Derived Neurotrophic Factor in the Rostral Ventrolateral Medulla. Front Physiol 2021; 12:756542. [PMID: 34721079 PMCID: PMC8548431 DOI: 10.3389/fphys.2021.756542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/13/2021] [Indexed: 11/30/2022] Open
Abstract
A sedentary lifestyle is the top preventable cause of death and accounts for substantial socioeconomic costs to society. The rostral ventrolateral medulla regulates blood pressure under normal and pathophysiological states, and demonstrates inactivity-related structural and functional neuroplasticity, which is subregionally specific. The purpose of this study was to examine pro- and mature forms of brain-derived neurotrophic factor (BDNF) and their respective receptors in the male rat rostral ventrolateral medulla (RVLM) and its rostral extension following sedentary vs. active (running wheels) conditions (10-12weeks). We used subregionally specific Western blotting to determine that the mature form of BDNF and its ratio to its pro-form were lower in more caudal subregions of the rostral ventrolateral medulla of sedentary rats but higher in the rostral extension when both were compared to active rats. The full-length form of the tropomyosin receptor kinase B receptor and the non-glycosylated form of the 75 kilodalton neurotrophin receptor were lower in sedentary compared to active rats. The rostrocaudal patterns of expression of the mature form of BDNF and the full-length form of the tropomyosin receptor kinase B receptor were remarkably similar to the subregionally specific patterns of enhanced dendritic branching, neuronal activity, and glutamate-mediated increases in sympathetic nerve activity observed in previous studies performed in sedentary rats. Our studies suggest signaling pathways related to BDNF within subregions of both the rostral ventrolateral medulla and its rostral extension contribute to cardiovascular disease and premature death related to a sedentary lifestyle.
Collapse
Affiliation(s)
| | - Patrick J. Mueller
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
6
|
Abstract
Neurotrophin-3 (NT-3) belongs to a family of growth factors called neurotrophins whose actions are centered in the nervous system. NT-3 is structurally related to other neurotrophins like brain-derived neurotrophic factor. The expression of NT-3 starts with the onset of neurogenesis and continues throughout life. A wealth of information links NT-3 to the growth, differentiation, and survival of hippocampal cells as well as sympathetic and sensory neurons. These studies have described the distribution of NT-3 and its receptors throughout development and in the mature nervous system. Prior works has begun to cell-type specific impact of NT-3 as well as identify the signaling pathways involved. However, much less is known about how NT-3 regulates synaptic transmission. This chapter focuses role of NT-3 in the modulation of synaptic transmission.
Collapse
|
7
|
Qiu F, Zhou Y, Deng Y, Yi J, Gong M, Liu N, Wei C, Xiang S. Knockdown of TNFAIP1 prevents di-(2-ethylhexyl) phthalate-induced neurotoxicity by activating CREB pathway. CHEMOSPHERE 2020; 241:125114. [PMID: 31683445 DOI: 10.1016/j.chemosphere.2019.125114] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/10/2019] [Accepted: 10/12/2019] [Indexed: 06/10/2023]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is a widely used plasticizer. It has neurotoxicity and exposure to it causes impairment of neurodevelopment, behavior and cognition. However, the molecular mechanisms responsible for the DEHP-induced neurotoxicity are not yet clearly defined. Tumor necrosis factor-induced protein 1 (TNFAIP1) was first discovered in umbilical vein endothelial cells and was further found to be important in the progress of Alzheimer's disease. Herein we explore the mechanism of TNFAIP1 in DEHP-induced neurotoxicity with the involvement of cyclic AMP response elements binding protein (CREB) signaling pathway in a mouse neuroblastoma cell line (N2a cells). We found that exposure to DEHP induced apoptosis and downregulated the expression of brain-derived neurotrophic factor (BDNF), synaptic proteins PSD 95 and synapsin-1 while upregulated the expression of TNFAIP1 and decreased the levels of phosphorylated Akt, CaMK Ⅳ, catalytic subunits of PKA and CREB in CREB signaling pathway. Knockdown of TNFAIP1 using TNFAIP1 small interfering RNA (siRNA) expression vector prevented DEHP from inhibiting CREB pathway, thus reduced apoptosis and restored expression of BDNF, PSD 95 and synapsin-1. Our data indicate that downregulation of TNFAIP1 prevents DEHP-induced neurotoxicity via activating CREB pathway. Therefore, TNFAIP1 is a potential target for relieving the DEHP-induced neurotoxicity and related neurological disorders.
Collapse
Affiliation(s)
- Feng Qiu
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Yubo Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Yeke Deng
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Junzhi Yi
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Mengting Gong
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Ning Liu
- School of Medicine, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Chenxi Wei
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China.
| | - Shuanglin Xiang
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China.
| |
Collapse
|
8
|
Charsar BA, Brinton MA, Locke K, Chen AY, Ghosh B, Urban MW, Komaravolu S, Krishnamurthy K, Smit R, Pasinelli P, Wright MC, Smith GM, Lepore AC. AAV2-BDNF promotes respiratory axon plasticity and recovery of diaphragm function following spinal cord injury. FASEB J 2019; 33:13775-13793. [PMID: 31577916 DOI: 10.1096/fj.201901730r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
More than half of spinal cord injury (SCI) cases occur in the cervical region, leading to respiratory dysfunction due to damaged neural circuitry that controls critically important muscles such as the diaphragm. The C3-C5 spinal cord is the location of phrenic motor neurons (PhMNs) that are responsible for diaphragm activation; PhMNs receive bulbospinal excitatory drive predominately from supraspinal neurons of the rostral ventral respiratory group (rVRG). Cervical SCI results in rVRG axon damage, PhMN denervation, and consequent partial-to-complete paralysis of hemidiaphragm. In a rat model of C2 hemisection SCI, we expressed the axon guidance molecule, brain-derived neurotrophic factor (BDNF), selectively at the location of PhMNs (ipsilateral to lesion) to promote directed growth of rVRG axons toward PhMN targets by performing intraspinal injections of adeno-associated virus serotype 2 (AAV2)-BDNF vector. AAV2-BDNF promoted significant functional diaphragm recovery, as assessed by in vivo electromyography. Within the PhMN pool ipsilateral to injury, AAV2-BDNF robustly increased sprouting of both spared contralateral-originating rVRG axons and serotonergic fibers. Furthermore, AAV2-BDNF significantly increased numbers of putative monosynaptic connections between PhMNs and these sprouting rVRG and serotonergic axons. These findings show that targeting circuit plasticity mechanisms involving the enhancement of synaptic inputs from spared axon populations is a powerful strategy for restoring respiratory function post-SCI.-Charsar, B. A., Brinton, M. A., Locke, K., Chen, A. Y., Ghosh, B., Urban, M. W., Komaravolu, S., Krishnamurthy, K., Smit, R., Pasinelli, P., Wright, M. C., Smith, G. M., Lepore, A. C. AAV2-BDNF promotes respiratory axon plasticity and recovery of diaphragm function following spinal cord injury.
Collapse
Affiliation(s)
- Brittany A Charsar
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Michael A Brinton
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Katherine Locke
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Anna Y Chen
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Biswarup Ghosh
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Mark W Urban
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Sreeya Komaravolu
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Karthik Krishnamurthy
- Department of Neuroscience, Jefferson Weinberg Amyotrophic Lateral Sclerosis (ALS) Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Rupert Smit
- Department of Anatomy and Cell Biology, Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Piera Pasinelli
- Department of Neuroscience, Jefferson Weinberg Amyotrophic Lateral Sclerosis (ALS) Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Megan C Wright
- Department of Biology, Arcadia University, Philadelphia, Pennsylvania, USA
| | - George M Smith
- Department of Anatomy and Cell Biology, Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Angelo C Lepore
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
9
|
Altshuler DB, Wang L, Zhao L, Miklja Z, Linzey J, Brezzell A, Kakaizada S, Krishna S, Orringer DA, Briceño EM, Gabel N, Hervey-Jumper SL. BDNF, COMT, and DRD2 polymorphisms and ability to return to work in adult patients with low- and high-grade glioma. Neurooncol Pract 2019; 6:375-385. [PMID: 31555452 PMCID: PMC6753359 DOI: 10.1093/nop/npy059] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Cognitive and language dysfunction is common among patients with glioma and has a significant impact on survival and health-related quality of life (HRQOL). Little is known about the factors that make individual patients more or less susceptible to the cognitive sequelae of the disease. A better understanding of the individual and population characteristics related to cognitive function in glioma patients is required to appropriately stratify patients, prognosticate, and develop more efficacious treatment regimens. There is evidence that allelic variation among genes involved in neurotransmission and synaptic plasticity are related to neurocognitive performance in states of health and neurologic disease. METHODS We studied the association of single-nucleotide polymorphism variations in brain-derived neurotrophic factor (BDNF, rs6265), dopamine receptor 2 (DRD2, rs1076560), and catechol-O-methyltransferase (COMT, rs4680) with neurocognitive function and ability to return to work in glioma patients at diagnosis and at 3 months. We developed a functional score based on the number of high-performance alleles that correlates with the capacity for patients to return to work. RESULTS Patients with higher-performing alleles have better scores on neurocognitive testing with the Repeatable Battery for the Assessment of Neuropsychological Status and Stroop test, but not the Trail Making Test. CONCLUSIONS A better understanding of the genetic contributors to neurocognitive performance in glioma patients and capacity for functional recovery is necessary to develop improved treatment strategies based on patient-specific factors.
Collapse
Affiliation(s)
| | - Lin Wang
- Department of Neurosurgery, University of Michigan, Ann Arbor, USA
| | - Lili Zhao
- Department of Biostatistics, University of Michigan, Ann Arbor, USA
| | - Zachary Miklja
- Department of Neurosurgery, University of Michigan, Ann Arbor, USA
| | - Joey Linzey
- Department of Neurosurgery, University of Michigan, Ann Arbor, USA
| | - Amanda Brezzell
- Department of Neurosurgery, University of Michigan, Ann Arbor, USA
| | - Sofia Kakaizada
- Department of Neurosurgery, University of California San Francisco, USA
| | - Saritha Krishna
- Department of Neurosurgery, University of California San Francisco, USA
| | - Daniel A Orringer
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, USA
| | - Emily M Briceño
- Department of Neurosurgery, Physical Medicine and Rehabilitation, University of Michigan, Ann Arbor, USA
| | - Nicolette Gabel
- Department of Neurosurgery, Physical Medicine and Rehabilitation, University of Michigan, Ann Arbor, USA
| | - Shawn L Hervey-Jumper
- Department of Neurosurgery, University of Michigan, Ann Arbor, USA
- Department of Neurosurgery, University of California San Francisco, USA
| |
Collapse
|
10
|
Pradhan J, Noakes PG, Bellingham MC. The Role of Altered BDNF/TrkB Signaling in Amyotrophic Lateral Sclerosis. Front Cell Neurosci 2019; 13:368. [PMID: 31456666 PMCID: PMC6700252 DOI: 10.3389/fncel.2019.00368] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022] Open
Abstract
Brain derived neurotrophic factor (BDNF) is well recognized for its neuroprotective functions, via activation of its high affinity receptor, tropomysin related kinase B (TrkB). In addition, BDNF/TrkB neuroprotective functions can also be elicited indirectly via activation of adenosine 2A receptors (A2aRs), which in turn transactivates TrkB. Evidence suggests that alterations in BDNF/TrkB, including TrkB transactivation by A2aRs, can occur in several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Although enhancing BDNF has been a major goal for protection of dying motor neurons (MNs), this has not been successful. Indeed, there is emerging in vitro and in vivo evidence suggesting that an upregulation of BDNF/TrkB can cause detrimental effects on MNs, making them more vulnerable to pathophysiological insults. For example, in ALS, early synaptic hyper-excitability of MNs is thought to enhance BDNF-mediated signaling, thereby causing glutamate excitotoxicity, and ultimately MN death. Moreover, direct inhibition of TrkB and A2aRs has been shown to protect MNs from these pathophysiological insults, suggesting that modulation of BDNF/TrkB and/or A2aRs receptors may be important in early disease pathogenesis in ALS. This review highlights the relevance of pathophysiological actions of BDNF/TrkB under certain circumstances, so that manipulation of BDNF/TrkB and A2aRs may give rise to alternate neuroprotective therapeutic strategies in the treatment of neural diseases such as ALS.
Collapse
Affiliation(s)
- Jonu Pradhan
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Peter G Noakes
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Mark C Bellingham
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
11
|
Theobromine Improves Working Memory by Activating the CaMKII/CREB/BDNF Pathway in Rats. Nutrients 2019; 11:nu11040888. [PMID: 31010016 PMCID: PMC6520707 DOI: 10.3390/nu11040888] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/05/2019] [Accepted: 04/17/2019] [Indexed: 01/08/2023] Open
Abstract
Theobromine (TB) is a primary methylxanthine found in cacao beans. cAMP-response element-binding protein (CREB) is a transcription factor, which is involved in different brain processes that bring about cellular changes in response to discrete sets of instructions, including the induction of brain-derived neurotropic factor (BDNF). Ca2+/calmodulin-dependent protein kinase II (CaMKII) has been strongly implicated in the memory formation of different species as a key regulator of gene expression. Here we investigated whether TB acts on the CaMKII/CREB/BDNF pathway in a way that might improve the cognitive and learning function in rats. Male Wistar rats (5 weeks old) were divided into two groups. For 73 days, the control rats (CN rats) were fed a normal diet, while the TB-fed rats (TB rats) received the same food, but with a 0.05% TB supplement. To assess the effects of TB on cognitive and learning ability in rats: The radial arm maze task, novel object recognition test, and Y-maze test were used. Then, the brain was removed and the medial prefrontal cortex (mPFC) was isolated for Western Blot, real-time PCR and enzyme-linked immunosorbent assay. Phosphorylated CaMKII (p-CaMKII), phosphorylated CREB (p-CREB), and BDNF level in the mPFC were measured. In all the behavior tests, working memory seemed to be improved by TB ingestion. In addition, p-CaMKII and p-CREB levels were significantly elevated in the mPFC of TB rats in comparison to those of CN rats. We also found that cortical BDNF protein and mRNA levels in TB rats were significantly greater than those in CN rats. These results suggest that orally supplemented TB upregulates the CaMKII/CREB/BDNF pathway in the mPFC, which may then improve working memory in rats.
Collapse
|
12
|
The primate-specific peptide Y-P30 regulates morphological maturation of neocortical dendritic spines. PLoS One 2019; 14:e0211151. [PMID: 30759095 PMCID: PMC6373909 DOI: 10.1371/journal.pone.0211151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/08/2019] [Indexed: 12/20/2022] Open
Abstract
The 30-amino acid peptide Y-P30 corresponds to the N-terminus of the primate-specific, sweat gland-derived dermcidin prepropeptide. Previous work has revealed that Y-P30 enhances the interaction of pleiotrophin and syndecans-2/3, and thus represents a natural ligand to study this signaling pathway. In immature neurons, Y-P30 activates the c-Src and p42/44 ERK kinase pathway, increases the amount of F-actin in axonal growth cones, and promotes neuronal survival, cell migration and axonal elongation. The action of Y-P30 on axonal growth requires syndecan-3 and heparan sulfate side chains. Whether Y-P30 has the potential to influence dendrites and dendritic protrusions has not been explored. The latter is suggested by the observations that syndecan-2 expression increases during postnatal development, that syndecan-2 becomes enriched in dendritic spines, and that overexpression of syndecan-2 in immature neurons results in a premature morphological maturation of dendritic spines. Here, analysing rat cortical pyramidal and non-pyramidal neurons in organotypic cultures, we show that Y-P30 does not alter the development of the dendritic arborization patterns. However, Y-P30 treatment decreases the density of apical, but not basal dendritic protrusions at the expense of the filopodia. Analysis of spine morphology revealed an unchanged mushroom/stubby-to-thin spine ratio and a shortening of the longest decile of dendritic protrusions. Whole-cell recordings from cortical principal neurons in dissociated cultures grown in the presence of Y-P30 demonstrated a decrease in the frequency of glutamatergic mEPSCs. Despite these differences in protrusion morphology and synaptic transmission, the latter likely attributable to presynaptic effects, calcium event rate and amplitude recorded in pyramidal neurons in organotypic cultures were not altered by Y-P30 treatment. Together, our data suggest that Y-P30 has the capacity to decelerate spinogenesis and to promote morphological, but not synaptic, maturation of dendritic protrusions.
Collapse
|
13
|
Novais A, Silva A, Ferreira AC, Falcão AM, Sousa N, Palha JA, Marques F, Sousa JC. Adult Hippocampal Neurogenesis Modulation by the Membrane-Associated Progesterone Receptor Family Member Neudesin. Front Cell Neurosci 2018; 12:463. [PMID: 30534059 PMCID: PMC6275434 DOI: 10.3389/fncel.2018.00463] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/13/2018] [Indexed: 11/13/2022] Open
Abstract
Neudesin (Neuron-derived neurotrophic factor, NENF), a membrane-associated progesterone receptor family (MAPR) member, is a neuron secreted protein with neurotrophic properties during embryonic stages. However, its role in the adult brain is still poorly addressed. In this study we have used neudesin-null (Nenf−/−) mice and performed a characterization of the proliferation state of the adult neurogenic niches, the adult subventricular zone (SVZ) and the hippocampus subgranular zone (SGZ). Nenf−/− males did not presented any deficits in proliferation in the SVZ neither in vivo nor in vitro. On the other hand a decrease in cell proliferation in the SGZ was observed, as well as a decrease in the number of newborn neurons in the dentate gyrus (DG) that was accompanied by impaired context discrimination in a contextual fear conditioning (CFC) task. Since NENF neurotrophic action is suggested to occur via the formation of a progesterone stability complex for the activation of non-genomic cascade, we further evaluated progesterone metabolism in the absence of NENF. Interestingly, expression of progesterone catabolic rate-determining enzyme, 5-α-reductase was upregulated in the DG of Nenf−/−, together with a significant increase in the expression of the δGABAA receptor gene, involved in DG tonic inhibition. Taken together, these findings add in vivo evidence on the neurotrophic properties of NENF in the adult brain. Furthermore, the mechanism of action of NENF in this process might implicate neurosteroids modulation, at least in the DG.
Collapse
Affiliation(s)
- Ashley Novais
- Life and Health Sciences Research Institute (ICVS), Neuroscience Domain, School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Guimarães, Portugal
| | - Alberto Silva
- Life and Health Sciences Research Institute (ICVS), Neuroscience Domain, School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Guimarães, Portugal
| | - Ana Catarina Ferreira
- Life and Health Sciences Research Institute (ICVS), Neuroscience Domain, School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Guimarães, Portugal
| | - Ana Mendanha Falcão
- Life and Health Sciences Research Institute (ICVS), Neuroscience Domain, School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), Neuroscience Domain, School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Guimarães, Portugal
| | - Joana Almeida Palha
- Life and Health Sciences Research Institute (ICVS), Neuroscience Domain, School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Guimarães, Portugal
| | - Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), Neuroscience Domain, School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Guimarães, Portugal
| | - João Carlos Sousa
- Life and Health Sciences Research Institute (ICVS), Neuroscience Domain, School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Guimarães, Portugal
| |
Collapse
|
14
|
Regulation of BDNF Release by ARMS/Kidins220 through Modulation of Synaptotagmin-IV Levels. J Neurosci 2018; 38:5415-5428. [PMID: 29769266 DOI: 10.1523/jneurosci.1653-17.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 04/07/2018] [Accepted: 05/03/2018] [Indexed: 11/21/2022] Open
Abstract
BDNF is a growth factor with important roles in the nervous system in both physiological and pathological conditions, but the mechanisms controlling its secretion are not completely understood. Here, we show that ARMS/Kidins220 negatively regulates BDNF secretion in neurons from the CNS and PNS. Downregulation of the ARMS/Kidins220 protein in the adult mouse brain increases regulated BDNF secretion, leading to its accumulation in the striatum. Interestingly, two mouse models of Huntington's disease (HD) showed increased levels of ARMS/Kidins220 in the hippocampus and regulated BDNF secretion deficits. Importantly, reduction of ARMS/Kidins220 in hippocampal slices from HD mice reversed the impaired regulated BDNF release. Moreover, there are increased levels of ARMS/Kidins220 in the hippocampus and PFC of patients with HD. ARMS/Kidins220 regulates Synaptotagmin-IV levels, which has been previously observed to modulate BDNF secretion. These data indicate that ARMS/Kidins220 controls the regulated secretion of BDNF and might play a crucial role in the pathogenesis of HD.SIGNIFICANCE STATEMENT BDNF is an important growth factor that plays a fundamental role in the correct functioning of the CNS. The secretion of BDNF must be properly controlled to exert its functions, but the proteins regulating its release are not completely known. Using neuronal cultures and a new conditional mouse to modulate ARMS/Kidins220 protein, we report that ARMS/Kidins220 negatively regulates BDNF secretion. Moreover, ARMS/Kidins220 is overexpressed in two mouse models of Huntington's disease (HD), causing an impaired regulation of BDNF secretion. Furthermore, ARMS/Kidins220 levels are increased in brain samples from HD patients. Future studies should address whether ARMS/Kidins220 has any function on the pathophysiology of HD.
Collapse
|
15
|
Therapeutic Potentials of Synapses after Traumatic Brain Injury: A Comprehensive Review. Neural Plast 2017; 2017:4296075. [PMID: 28491479 PMCID: PMC5405590 DOI: 10.1155/2017/4296075] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/09/2017] [Accepted: 03/14/2017] [Indexed: 12/26/2022] Open
Abstract
Massive studies have focused on the understanding of the pathobiology of cellular and molecular changes and injury mechanisms after traumatic brain injury (TBI), but very few studies have specially discussed the role of synapses in the context of TBI. This paper specifically highlights the role and therapeutic potentials of synapses after TBI. First, we review and conclude how synapses interact with constant structural, metabolic, neuroendocrine, and inflammatory mechanisms after TBI. Second, we briefly describe several key synaptic proteins involved in neuroplasticity, which may be novel neuronal targets for specific intervention. Third, we address therapeutic interventions in association with synapses after TBI. Finally, we concisely discuss the study gaps in the synapses after TBI, in hopes that this would provide more insights for future studies. Synapses play an important role in TBI; while the understandings on the synaptic participation in the treatments and prognosis of TBI are lacking, more studies in this area are warranted.
Collapse
|
16
|
Ramku E, Ramku R, Spanca D, Zhjeqi V. Functional Pattern of Increasing Concentrations of Brain-Derived Neurotrophic Factor in Spiral Ganglion: Implications for Research on Cochlear Implants. Open Access Maced J Med Sci 2017; 5:121-125. [PMID: 28507614 PMCID: PMC5420760 DOI: 10.3889/oamjms.2017.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 02/22/2017] [Accepted: 02/25/2017] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND: As previously various studies have suggested application of brain-derived neurotrophic factor (BDNF) may be considered as a promising future therapy for hearing deficits, in particular for the improvement of cochlear neurone loss during cochlear implantation. AIM: The present study’s aim was to establish the upper threshold of the concentration of BDNF in Naval Medical Research Institute (NMRI) mice spiral ganglion outgrowth. METHODS: Spiral ganglion explants were prepared from post-natal day 4 (p4) (NMRI) mice of both sexes under the approval and guidelines of the regional council of Hearing Research Institute Tubingen. RESULTS: Spiral ganglion explants were cultured at postnatal days 4 in the presence of different concentrations of BDNF as described under methods. We chose an age of postnatal day (P4) and concentrations of BDNF 0; 6; 12.5; 25 and 50 ƞg/ml. Averaged neurite outgrowth is measured in 4 different cultures that were treated with different concentrations. Results show that with increasing concentrations of BDNF, the neurite density increases. CONCLUSION: The present finding show evidence that BDNF has a clear incremental effect on the number of neurites of spiral ganglia in the prehearing organ, but less on the neurite length. The upper threshold of exogenous BNDF concentration on spiral ganglion explant is 25 ƞg/ml. This means that concentration beyond this level has no further incremental impact. Therefore our suggestion for hydrogel concentration in NMRA mice in future research should be 25 ƞg/ml.
Collapse
Affiliation(s)
- Emina Ramku
- University Clinical Center, Prishtina, Kosovo
| | - Refik Ramku
- Private Polyclinic OTOKIRURGJIA, Prishtina, Kosovo
| | | | | |
Collapse
|
17
|
Franklin DJ, Grossberg S. A neural model of normal and abnormal learning and memory consolidation: adaptively timed conditioning, hippocampus, amnesia, neurotrophins, and consciousness. COGNITIVE, AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2017; 17:24-76. [PMID: 27905080 PMCID: PMC5272895 DOI: 10.3758/s13415-016-0463-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
How do the hippocampus and amygdala interact with thalamocortical systems to regulate cognitive and cognitive-emotional learning? Why do lesions of thalamus, amygdala, hippocampus, and cortex have differential effects depending on the phase of learning when they occur? In particular, why is the hippocampus typically needed for trace conditioning, but not delay conditioning, and what do the exceptions reveal? Why do amygdala lesions made before or immediately after training decelerate conditioning while those made later do not? Why do thalamic or sensory cortical lesions degrade trace conditioning more than delay conditioning? Why do hippocampal lesions during trace conditioning experiments degrade recent but not temporally remote learning? Why do orbitofrontal cortical lesions degrade temporally remote but not recent or post-lesion learning? How is temporally graded amnesia caused by ablation of prefrontal cortex after memory consolidation? How are attention and consciousness linked during conditioning? How do neurotrophins, notably brain-derived neurotrophic factor (BDNF), influence memory formation and consolidation? Is there a common output path for learned performance? A neural model proposes a unified answer to these questions that overcome problems of alternative memory models.
Collapse
Affiliation(s)
- Daniel J Franklin
- Center for Adaptive Systems, Graduate Program in Cognitive and Neural Systems, and Departments of Mathematics, Psychological & Brain Sciences, and Biomedical Engineering, Boston University, 677 Beacon Street, Room 213, Boston, MA, 02215, USA
| | - Stephen Grossberg
- Center for Adaptive Systems, Graduate Program in Cognitive and Neural Systems, and Departments of Mathematics, Psychological & Brain Sciences, and Biomedical Engineering, Boston University, 677 Beacon Street, Room 213, Boston, MA, 02215, USA.
| |
Collapse
|
18
|
D'Amico D, Gener T, de Lagrán MM, Sanchez-Vives MV, Santos M, Dierssen M. Infralimbic Neurotrophin-3 Infusion Rescues Fear Extinction Impairment in a Mouse Model of Pathological Fear. Neuropsychopharmacology 2017; 42:462-472. [PMID: 27534266 PMCID: PMC5399232 DOI: 10.1038/npp.2016.154] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 08/05/2016] [Accepted: 08/07/2016] [Indexed: 11/09/2022]
Abstract
The inability to properly extinguish fear memories constitutes the foundation of several anxiety disorders, including panic disorder. Recent findings show that boosting prefrontal cortex synaptic plasticity potentiates fear extinction, suggesting that therapies that augment synaptic plasticity could prove useful in rescue of fear extinction impairments in this group of disorders. Previously, we reported that mice with selective deregulation of neurotrophic tyrosine kinase receptor, type 3 expression (TgNTRK3) exhibit increased fear memories accompanied by impaired extinction, congruent with an altered activation pattern of the amygdala-hippocampus-medial prefrontal cortex fear circuit. Here we explore the specific role of neurotrophin 3 and its cognate receptor in the medial prefrontal cortex, and its involvement in fear extinction in a pathological context. In this study we combined molecular, behavioral, in vivo pharmacology and ex vivo electrophysiological recordings in TgNTRK3 animals during contextual fear extinction processes. We show that neurotrophin 3 protein levels are increased upon contextual fear extinction in wild-type animals but not in TgNTRK3 mice, which present deficits in infralimbic long-term potentiation. Importantly, infusion of neurotrophin 3 to the medial prefrontal cortex of TgNTRK3 mice rescues contextual fear extinction and ex vivo local application improves medial prefrontal cortex synaptic plasticity. This effect is blocked by inhibition of extracellular signal-regulated kinase phosphorylation through peripheral administration of SL327, suggesting that rescue occurs via this pathway. Our results suggest that stimulating neurotrophin 3-dependent medial prefrontal cortex plasticity could restore contextual fear extinction deficit in pathological fear and could constitute an effective treatment for fear-related disorders.
Collapse
Affiliation(s)
- Davide D'Amico
- Systems Biology Programme, Centre for Genomic Regulation (CRG)/Barcelona Biomedical Research Park, Barcelona, Spain,Universitat Pompeu Fabra (UPF), Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Thomas Gener
- Systems Biology Programme, Centre for Genomic Regulation (CRG)/Barcelona Biomedical Research Park, Barcelona, Spain,Universitat Pompeu Fabra (UPF), Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Maria Martínez de Lagrán
- Systems Biology Programme, Centre for Genomic Regulation (CRG)/Barcelona Biomedical Research Park, Barcelona, Spain,Universitat Pompeu Fabra (UPF), Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Maria V Sanchez-Vives
- Systems Neuroscience, IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain,Life & Medical Sciences, ICREA (Instituciò Catalana de Recerca i Estudis Avançats), Barcelona, Spain
| | - Mónica Santos
- Systems Biology Programme, Centre for Genomic Regulation (CRG)/Barcelona Biomedical Research Park, Barcelona, Spain,Universitat Pompeu Fabra (UPF), Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Mara Dierssen
- Systems Biology Programme, Centre for Genomic Regulation (CRG)/Barcelona Biomedical Research Park, Barcelona, Spain,Universitat Pompeu Fabra (UPF), Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain,Barcelona Institute for Science and Technology (BIST), Barcelona, Spain,Systems Biology Programme, Center for Genomic Regulation, (CRG)/Barcelona Biomedical Research Park, C/ Dr Aiguader 88, Barcelona 08003, Spain, Tel: +34 933160140, Fax +34 93 316 00 99, E-mail:
| |
Collapse
|
19
|
Cobianchi S, Arbat-Plana A, López-Álvarez VM, Navarro X. Neuroprotective Effects of Exercise Treatments After Injury: The Dual Role of Neurotrophic Factors. Curr Neuropharmacol 2017; 15:495-518. [PMID: 27026050 PMCID: PMC5543672 DOI: 10.2174/1570159x14666160330105132] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 02/19/2016] [Accepted: 03/03/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Shared connections between physical activity and neuroprotection have been studied for decades, but the mechanisms underlying this effect of specific exercise were only recently brought to light. Several evidences suggest that physical activity may be a reasonable and beneficial method to improve functional recovery in both peripheral and central nerve injuries and to delay functional decay in neurodegenerative diseases. In addition to improving cardiac and immune functions, physical activity may represent a multifunctional approach not only to improve cardiocirculatory and immune functions, but potentially modulating trophic factors signaling and, in turn, neuronal function and structure at times that may be critical for neurodegeneration and regeneration. METHODS Research content related to the effects of physical activity and specific exercise programs in normal and injured nervous system have been reviewed. RESULTS Sustained exercise, particularly if applied at moderate intensity and early after injury, exerts anti-inflammatory and pro-regenerative effects, and may boost cognitive and motor functions in aging and neurological disorders. However, newest studies show that exercise modalities can differently affect the production and function of brain-derived neurotrophic factor and other neurotrophins involved in the generation of neuropathic conditions. These findings suggest the possibility that new exercise strategies can be directed to nerve injuries with therapeutical benefits. CONCLUSION Considering the growing burden of illness worldwide, understanding of how modulation of neurotrophic factors contributes to exercise-induced neuroprotection and regeneration after peripheral nerve and spinal cord injuries is a relevant topic for research, and represents the beginning of a new non-pharmacological therapeutic approach for better rehabilitation of neural disorders.
Collapse
Affiliation(s)
- Stefano Cobianchi
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autonoma de Barcelona, Bellaterra, Spain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | - Ariadna Arbat-Plana
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autonoma de Barcelona, Bellaterra, Spain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | - Víctor M. López-Álvarez
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autonoma de Barcelona, Bellaterra, Spain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | - Xavier Navarro
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autonoma de Barcelona, Bellaterra, Spain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| |
Collapse
|
20
|
Nair B, Wong-Riley MTT. Transcriptional Regulation of Brain-derived Neurotrophic Factor Coding Exon IX: ROLE OF NUCLEAR RESPIRATORY FACTOR 2. J Biol Chem 2016; 291:22583-22593. [PMID: 27624937 DOI: 10.1074/jbc.m116.742304] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/09/2016] [Indexed: 01/22/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is an active neurotrophin abundantly expressed throughout the nervous system. It plays an important role in synaptic transmission, plasticity, neuronal proliferation, differentiation, survival, and death. The Bdnf gene in rodents has eight non-coding exons and only a single coding exon (IX). Despite its recognized regulation by neuronal activity, relatively little is known about its transcriptional regulation, and even less about the transcription factor candidates that may play such a role. The goal of the present study was to probe for such a candidate that may regulate exon IX in the rat Bdnf gene. Our in silico analysis revealed tandem binding sites for nuclear respiratory factor 2 (NRF-2) on the promoter of exon IX. NRF-2 is of special significance because it co-regulates the expressions of mediators of energy metabolism (cytochrome c oxidase) and mediators of neuronal activity (glutamatergic receptors). To test our hypothesis that NRF-2 also regulates the Bdnf gene, we performed electrophoretic mobility shift assay (EMSA), chromatin immunoprecipitation (ChIP), promoter cloning, and site-directed mutagenesis, real-time quantitative PCR (RT-qPCR), and Western blotting analysis. Results indicate that NRF-2 functionally regulates exon IX of the rat Bdnf gene. The binding sites of NRF-2 are conserved between rats and mice. Overexpressing NRF-2 up-regulated the expression of Bdnf exon IX, whereas knocking down NRF-2 down-regulated such expression. These findings are consistent with our hypothesis that NRF-2, in addition to regulating the coupling between neuronal activity and energy metabolism, also regulates the expression of BDNF, which is intimately associated with energy-demanding neuronal activity.
Collapse
Affiliation(s)
- Bindu Nair
- From the Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Margaret T T Wong-Riley
- From the Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| |
Collapse
|
21
|
Amyloid-Beta Induced Changes in Vesicular Transport of BDNF in Hippocampal Neurons. Neural Plast 2016; 2016:4145708. [PMID: 26881108 PMCID: PMC4736975 DOI: 10.1155/2016/4145708] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/26/2015] [Accepted: 11/29/2015] [Indexed: 12/15/2022] Open
Abstract
The neurotrophin brain derived neurotrophic factor (BDNF) is an important growth factor in the CNS. Deficits in transport of this secretory protein could underlie neurodegenerative diseases. Investigation of disease-related changes in BDNF transport might provide insights into the cellular mechanism underlying, for example, Alzheimer's disease (AD). To analyze the role of BDNF transport in AD, live cell imaging of fluorescently labeled BDNF was performed in hippocampal neurons of different AD model systems. BDNF and APP colocalized with low incidence in vesicular structures. Anterograde as well as retrograde transport of BDNF vesicles was reduced and these effects were mediated by factors released from hippocampal neurons into the extracellular medium. Transport of BDNF was altered at a very early time point after onset of human APP expression or after acute amyloid-beta(1-42) treatment, while the activity-dependent release of BDNF remained unaffected. Taken together, extracellular cleavage products of APP induced rapid changes in anterograde and retrograde transport of BDNF-containing vesicles while release of BDNF was unaffected by transgenic expression of mutated APP. These early transport deficits might lead to permanently impaired brain functions in the adult brain.
Collapse
|
22
|
Neurotrophin-3 Enhances the Synaptic Organizing Function of TrkC-Protein Tyrosine Phosphatase σ in Rat Hippocampal Neurons. J Neurosci 2015; 35:12425-31. [PMID: 26354911 DOI: 10.1523/jneurosci.1330-15.2015] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neurotrophin-3 (NT-3) and its high-affinity receptor TrkC play crucial trophic roles in neuronal differentiation, axon outgrowth, and synapse development and plasticity in the nervous system. We demonstrated previously that postsynaptic TrkC functions as a glutamatergic synapse-inducing (synaptogenic) cell adhesion molecule trans-interacting with presynaptic protein tyrosine phosphatase σ (PTPσ). Given that NT-3 and PTPσ bind distinct domains of the TrkC extracellular region, here we tested the hypothesis that NT-3 modulates TrkC/PTPσ binding and synaptogenic activity. NT-3 enhanced PTPσ binding to cell surface-expressed TrkC and facilitated the presynapse-inducing activity of TrkC in rat hippocampal neurons. Imaging of recycling presynaptic vesicles combined with TrkC knockdown and rescue approaches demonstrated that NT-3 rapidly potentiates presynaptic function via binding endogenous postsynaptic TrkC in a tyrosine kinase-independent manner. Thus, NT-3 positively modulates the TrkC-PTPσ complex for glutamatergic presynaptic assembly and function independently from TrkC kinase activation. Our findings provide new insight into synaptic roles of neurotrophin signaling and mechanisms controlling synaptic organizing complexes. Significance statement: Although many synaptogenic adhesion complexes have been identified in recent years, little is known about modulatory mechanisms. Here, we demonstrate a novel role of neurotrophin-3 in synaptic assembly and function as a positive modulator of the TrkC-protein tyrosine phosphatase σ complex. This study provides new insight into the involvement of neurotrophin signaling in synapse development and plasticity, presenting a molecular mechanism that may underlie previous observations of short- and long-term enhancement of presynaptic function by neurotrophin. Given the links of synaptogenic adhesion molecules to autism and schizophrenia, this study might also contribute to a better understanding of the pathogenesis of these disorders and provide a new direction for ameliorating imbalances in synaptic signaling networks.
Collapse
|
23
|
NT-3 protein levels are enhanced in the hippocampus of PRG1-deficient mice but remain unchanged in PRG1/LPA2 double mutants. Neurosci Lett 2015; 612:145-148. [PMID: 26687273 DOI: 10.1016/j.neulet.2015.12.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/03/2015] [Accepted: 12/09/2015] [Indexed: 12/29/2022]
Abstract
The plasticity-related gene 1 (PRG1) modulates bioactive lipids at the postsynaptic density and is a novel player in neuronal plasticity and regulation of glutamatergic transmission at principal neurons. PRG1, a neuronal molecule, is highly expressed during development and regeneration processes at the postsynaptic density, modulates synaptic lysophosphatidic acid (LPA) levels and is related to epilepsy and brain injury. In the present study, we analyzed the interaction between the synaptic molecules PRG1 and LPA2R with other plasticity-related molecules the neurotrophins. The protein levels of NGF, BDNF and NT-3 were measured using ELISA in hippocampal tissue of homozygous (PRG(-/-)) and heterozygous (PRG(+/-)) PRG1 deficient mice and compared to their wild type (PRG(+/+)/WT) littermates. In the hippocampus, protein levels of NT-3 were significantly increased in PRG(-/-) mice (compared to WT-litters) while protein levels of NGF and BDNF were not affected. Since PRG1 deficiency leads to increased neuronal excitability and higher hippocampal network activity, which may well influence neurotrophin levels, we further assessed PRG1 deficient mice on an LPA2-receptor (LPA2R) deficient background, reported to normalize hippocampal over-excitability in PRG1(-/-) mice. However, on an LPA2R deficient background, protein levels of NT-3 in PRG1(-/-) mice (PRG1(-/-)/LPA2R(-/-)) were not significantly different when compared to WT animals. Since PRG1 deficient mice showed over-excitability in glutamatergic neurons. This was normalized by additional LPA2R deletion, and we conclude the increased NT3-levels were directly or indirectly attributable to increased hippocampal network activity, possibly exerting a protective effect against over-excitability.
Collapse
|
24
|
Ciriello J, Moreau JM, McCoy AM, Jones DL. Leptin dependent changes in the expression of tropomyosin receptor kinase B protein in nucleus of the solitary tract to acute intermittent hypoxia. Neurosci Lett 2015; 602:115-9. [PMID: 26163463 DOI: 10.1016/j.neulet.2015.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 06/30/2015] [Accepted: 07/03/2015] [Indexed: 11/25/2022]
Abstract
To investigate the possibility that leptin exerts an effect in NTS by inducing changes in the expression of pre- and/or post-synaptic proteins, experiments were done in Sprague-Dawley wild-type rats (WT) rats and leptin-deficient rats (Lep(Δ151/Δ151); KILO rat) exposed to 8h of continuous intermittent hypoxia (IH) or normoxia. Protein was extracted from the caudal medial NTS and analyzed by western blot for the expression of brain-derived neurotrophic factor (BDNF), tropomyosin receptor kinase B (TrkB), synaptophysin, synaptopodin and growth-associated protein-43 (GAP-43). In WT rats, BDNF and GAP 43 protein expression levels were not altered after IH or normoxia, although there was a trend towards an increase in BDNF expression. On the other hand, after IH, protein expression of both isoforms of the BDNF receptor TrkB (gp95 and gp145) was higher. Furthermore, synaptophysin protein expression was lower compared to normoxic WT rats. In the KILO rat, no changes were observed in the protein expression of BDNF, TrkB, or GAP 43 after IH when compared to KILO normoxic controls. However, synaptophysin was lower in the IH exposed KILO rat compared to normoxic controls, as found in the WT rat. Expression of synaptopodin was not detected in NTS in either IH or normoxic animals of all groups. These results suggest that leptin released during IH may contribute to neurotrophic changes occurring within NTS and that these changes may be associated with altered chemoreceptor reflex function.
Collapse
Affiliation(s)
- John Ciriello
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| | - Jason M Moreau
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Aaron M McCoy
- Sigma Advanced Genetic Engineering Laboratory, Sigma-ldrich Corp., St. Louis, MO 63146, USA
| | - Douglas L Jones
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
25
|
Neumann JT, Thompson JW, Raval AP, Cohan CH, Koronowski KB, Perez-Pinzon MA. Increased BDNF protein expression after ischemic or PKC epsilon preconditioning promotes electrophysiologic changes that lead to neuroprotection. J Cereb Blood Flow Metab 2015; 35:121-30. [PMID: 25370861 PMCID: PMC4294405 DOI: 10.1038/jcbfm.2014.185] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 10/02/2014] [Accepted: 10/03/2014] [Indexed: 01/06/2023]
Abstract
Ischemic preconditioning (IPC) via protein kinase C epsilon (PKCɛ) activation induces neuroprotection against lethal ischemia. Brain-derived neurotrophic factor (BDNF) is a pro-survival signaling molecule that modulates synaptic plasticity and neurogenesis. Interestingly, BDNF mRNA expression increases after IPC. In this study, we investigated whether IPC or pharmacological preconditioning (PKCɛ activation) promoted BDNF-induced neuroprotection, if neuroprotection by IPC or PKCɛ activation altered neuronal excitability, and whether these changes were BDNF-mediated. We used both in vitro (hippocampal organotypic cultures and cortical neuronal-glial cocultures) and in vivo (acute hippocampal slices 48 hours after preconditioning) models of IPC or PKCɛ activation. BDNF protein expression increased 24 to 48 hours after preconditioning, where inhibition of the BDNF Trk receptors abolished neuroprotection against oxygen and glucose deprivation (OGD) in vitro. In addition, there was a significant decrease in neuronal firing frequency and increase in threshold potential 48 hours after preconditioning in vivo, where this threshold modulation was dependent on BDNF activation of Trk receptors in excitatory cortical neurons. In addition, 48 hours after PKCɛ activation in vivo, the onset of anoxic depolarization during OGD was significantly delayed in hippocampal slices. Overall, these results suggest that after IPC or PKCɛ activation, there are BDNF-dependent electrophysiologic modifications that lead to neuroprotection.
Collapse
Affiliation(s)
- Jake T Neumann
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [3] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - John W Thompson
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Ami P Raval
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [3] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Charles H Cohan
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [3] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [4] Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Kevin B Koronowski
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [3] Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Miguel A Perez-Pinzon
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [3] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [4] Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
26
|
Abstract
Pharmaceuticals and medical devices hold the promise of enhancing brain function, not only of those suffering from neurodevelopmental, neuropsychiatric or neurodegenerative illnesses, but also of healthy individuals. However, a number of lifestyle interventions are proven cognitive enhancers, improving attention, problem solving, reasoning, learning and memory or even mood. Several of these interventions, such as physical exercise, cognitive, mental and social stimulation, may be described as environmental enrichments of varying types. Use of these non-pharmacological cognitive enhancers circumvents some of the ethical considerations associated with pharmaceutical or technological cognitive enhancement, being low in cost, available to the general population and presenting low risk to health and well-being. In this chapter, there will be particular focus on the effects of exercise and enrichment on learning and memory and the evidence supporting their efficacy in humans and in animal models will be described.
Collapse
Affiliation(s)
- Áine M Kelly
- Department of Physiology, School of Medicine, Level 2, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland,
| |
Collapse
|
27
|
Smith PA. BDNF: No gain without pain? Neuroscience 2014; 283:107-23. [DOI: 10.1016/j.neuroscience.2014.05.044] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 05/16/2014] [Accepted: 05/21/2014] [Indexed: 12/22/2022]
|
28
|
Hutson PH, Tarazi FI, Madhoo M, Slawecki C, Patkar AA. Preclinical pharmacology of amphetamine: Implications for the treatment of neuropsychiatric disorders. Pharmacol Ther 2014; 143:253-64. [DOI: 10.1016/j.pharmthera.2014.03.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 03/14/2014] [Indexed: 11/28/2022]
|
29
|
Pollak DD, Minh BQ, Cicvaric A, Monje FJ. A novel fibroblast growth factor receptor family member promotes neuronal outgrowth and synaptic plasticity in aplysia. Amino Acids 2014; 46:2477-88. [PMID: 25059541 PMCID: PMC4200351 DOI: 10.1007/s00726-014-1803-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 06/30/2014] [Indexed: 02/06/2023]
Abstract
Fibroblast Growth Factor (FGF) Receptors (FGFRs) regulate essential biological processes, including embryogenesis, angiogenesis, cellular growth and memory-related long-term synaptic plasticity. Whereas canonical FGFRs depend exclusively on extracellular Immunoglobulin (Ig)-like domains for ligand binding, other receptor types, including members of the tropomyosin-receptor-kinase (Trk) family, use either Ig-like or Leucine-Rich Repeat (LRR) motifs, or both. Little is known, however, about the evolutionary events leading to the differential incorporation of LRR domains into Ig-containing tyrosine kinase receptors. Moreover, although FGFRs have been identified in many vertebrate species, few reports describe their existence in invertebrates. Information about the biological relevance of invertebrate FGFRs and evolutionary divergences between them and their vertebrate counterparts is therefore limited. Here, we characterized ApLRRTK, a neuronal cell-surface protein recently identified in Aplysia. We unveiled ApLRRTK as the first member of the FGFRs family deprived of Ig-like domains that instead contains extracellular LRR domains. We describe that ApLRRTK exhibits properties typical of canonical vertebrate FGFRs, including promotion of FGF activity, enhancement of neuritic outgrowth and signaling via MAPK and the transcription factor CREB. ApLRRTK also enhanced the synaptic efficiency of neurons known to mediate in vivo memory-related defensive behaviors. These data reveal a novel molecular regulator of neuronal function in invertebrates, provide the first evolutionary linkage between LRR proteins and FGFRs and unveil an unprecedented mechanism of FGFR gene diversification in primeval central nervous systems.
Collapse
Affiliation(s)
- Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Center for Physiology and Pharmacology, Schwarzspanierstrasse 17, 1090, Vienna, Austria
| | | | | | | |
Collapse
|
30
|
Salter M, Beggs S. Sublime Microglia: Expanding Roles for the Guardians of the CNS. Cell 2014; 158:15-24. [DOI: 10.1016/j.cell.2014.06.008] [Citation(s) in RCA: 331] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Indexed: 12/31/2022]
|
31
|
Hullugundi SK, Ferrari MD, van den Maagdenberg AMJM, Nistri A. The mechanism of functional up-regulation of P2X3 receptors of trigeminal sensory neurons in a genetic mouse model of familial hemiplegic migraine type 1 (FHM-1). PLoS One 2013; 8:e60677. [PMID: 23577145 PMCID: PMC3618040 DOI: 10.1371/journal.pone.0060677] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 03/01/2013] [Indexed: 12/05/2022] Open
Abstract
A knock-in (KI) mouse model of FHM-1 expressing the R192Q missense mutation of the Cacna1a gene coding for the α1 subunit of CaV2.1 channels shows, at the level of the trigeminal ganglion, selective functional up-regulation of ATP -gated P2X3 receptors of sensory neurons that convey nociceptive signals to the brainstem. Why P2X3 receptors are constitutively more responsive, however, remains unclear as their membrane expression and TRPV1 nociceptor activity are the same as in wildtype (WT) neurons. Using primary cultures of WT or KI trigeminal ganglia, we investigated whether soluble compounds that may contribute to initiating (or maintaining) migraine attacks, such as TNFα, CGRP, and BDNF, might be responsible for increasing P2X3 receptor responses. Exogenous application of TNFα potentiated P2X3 receptor-mediated currents of WT but not of KI neurons, most of which expressed both the P2X3 receptor and the TNFα receptor TNFR2. However, sustained TNFα neutralization failed to change WT or KI P2X3 receptor currents. This suggests that endogenous TNFα does not regulate P2X3 receptor responses. Nonetheless, on cultures made from both genotypes, exogenous TNFα enhanced TRPV1 receptor-mediated currents expressed by a few neurons, suggesting transient amplification of TRPV1 nociceptor responses. CGRP increased P2X3 receptor currents only in WT cultures, although prolonged CGRP receptor antagonism or BDNF neutralization reduced KI currents to WT levels. Our data suggest that, in KI trigeminal ganglion cultures, constitutive up-regulation of P2X3 receptors probably is already maximal and is apparently contributed by basal CGRP and BDNF levels, thereby rendering these neurons more responsive to extracellular ATP.
Collapse
Affiliation(s)
- Swathi K. Hullugundi
- Neuroscience Department, International School for Advanced Studies (SISSA),Trieste, Italy
| | - Michel D. Ferrari
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Arn M. J. M. van den Maagdenberg
- Department of Neurology, Leiden University Medical Centre, Leiden, The Netherlands
- Department of Human Genetics, Leiden Genetics University Medical Centre, Leiden, The Netherlands
| | - Andrea Nistri
- Neuroscience Department, International School for Advanced Studies (SISSA),Trieste, Italy
- * E-mail:
| |
Collapse
|
32
|
Abstract
It has been suggested that long-term modifications of synaptic transmission constitute the foundation of the processes by which information is stored in the central nervous system. A group of proteins called neurotrophins are considered powerful molecular mediators in central synaptic plasticity. Among these, brain-derived neurotrophic factor (BDNF) as well as neurotrophin-3 (NT-3) have emerged as having key roles in the neurobiological mechanisms related to learning and memory. In this chapter, we review the studies that have represented a significant step forward in understanding the role played by BDNF and NT-3 in long-term synaptic plasticity. The effects of BDNF and NT-3 on synaptic plasticity can be of a permissive nature, establishing the conditions under which plastic changes can take place, or it may be instructive, directly modifying the communication and morphology of synapses. The actions carried out by BDNF include its capacity to contribute to the stabilization and maturation of already-existing synapses, as well as to generate new synaptic contacts. One important finding that highlights the participation of these neurotrophins in synaptic plasticity is the observation that adding BDNF or NT-3 gives rise to drastic long-term increases in synaptic transmission, similar to the long-term potentiation in the hippocampus and neocortex of mammals. Because neurotrophins modulate both the electrical properties and the structural organization of the synapse, these proteins have been considered important biological markers of learning and memory processes.
Collapse
Affiliation(s)
- Andrea Gómez-Palacio-Schjetnan
- División de Investigación y Estudios de Posgrado, Facultad de Psicologia, Universidad Nacional Autónoma de México, 04510, México, D.F., Mexico
| | | |
Collapse
|
33
|
Soligo M, Nori SL, Protto V, Florenzano F, Manni L. Acupuncture and Neurotrophin Modulation. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2013; 111:91-124. [DOI: 10.1016/b978-0-12-411545-3.00005-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
|
34
|
Takano K, Yamasaki H, Kawabe K, Moriyama M, Nakamura Y. Imipramine induces brain-derived neurotrophic factor mRNA expression in cultured astrocytes. J Pharmacol Sci 2012; 120:176-86. [PMID: 23076128 DOI: 10.1254/jphs.12039fp] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Depression is one of the most prevalent and livelihood-threatening forms of mental illnesses and the neural circuitry underlying depression remains incompletely understood. Recent studies suggest that the neuronal plasticity involved with brain-derived neurotrophic factor (BDNF) plays an important role in the recovery from depression. Some antidepressants are reported to induce BDNF expression in vivo; however, the mechanisms have been considered solely in neurons and not fully elucidated. In the present study, we evaluated the effects of imipramine, a classic tricyclic antidepressant drug, on BDNF expression in cultured rat brain astrocytes. Imipramine dose-dependently increased BDNF mRNA expression in astrocytes. The imipramine-induced BDNF increase was suppressed with inhibitors for protein kinase A (PKA) or MEK/ERK. Moreover, imipramine exposure activated transcription factor cAMP response element binding protein (CREB) in a dose-dependent manner. These results suggested that imipramine induced BDNF expression through CREB activation via PKA and/or ERK pathways. Imipramine treatment in depression might exert antidepressant action through BDNF production from astrocytes, and glial BDNF expression might be a target of developing novel antidepressants.
Collapse
Affiliation(s)
- Katsura Takano
- Laboratory of Integrative Physiology in Veterinary Sciences, Osaka Prefecture University, Izumisano, Osaka, Japan.
| | | | | | | | | |
Collapse
|
35
|
Tlili A, Hoischen A, Ripoll C, Benabou E, Badel A, Ronan A, Touraine R, Grattau Y, Stora S, van Bon B, de Vries B, Menten B, Bockaert N, Gecz J, Antonarakis SE, Campion D, Potier MC, Bléhaut H, Delabar JM, Janel N. BDNF and DYRK1A are variable and inversely correlated in lymphoblastoid cell lines from Down syndrome patients. Mol Neurobiol 2012; 46:297-303. [PMID: 22669612 DOI: 10.1007/s12035-012-8284-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 05/24/2012] [Indexed: 11/24/2022]
Abstract
Down syndrome or trisomy 21 is the most common genetic disorder leading to mental retardation. One feature is impaired short- and long-term spatial memory, which has been linked to altered brain-derived neurotrophic factor (BDNF) levels. Mouse models of Down syndrome have been used to assess neurotrophin levels, and reduced BDNF has been demonstrated in brains of adult transgenic mice overexpressing Dyrk1a, a candidate gene for Down syndrome phenotypes. Given the link between DYRK1A overexpression and BDNF reduction in mice, we sought to assess a similar association in humans with Down syndrome. To determine the effect of DYRK1A overexpression on BDNF in the genomic context of both complete trisomy 21 and partial trisomy 21, we used lymphoblastoid cell lines from patients with complete aneuploidy of human chromosome 21 (three copies of DYRK1A) and from patients with partial aneuploidy having either two or three copies of DYRK1A. Decreased BDNF levels were found in lymphoblastoid cell lines from individuals with complete aneuploidy as well as those with partial aneuploidies conferring three DYRK1A alleles. In contrast, lymphoblastoid cell lines from individuals with partial trisomy 21 having only two DYRK1A copies displayed increased BDNF levels. A negative correlation was also detected between BDNF and DYRK1A levels in lymphoblastoid cell lines with complete aneuploidy of human chromosome 21. This finding indicates an upward regulatory role of DYRK1A expression on BDNF levels in lymphoblastoid cell lines and emphasizes the role of genetic variants associated with psychiatric disorders.
Collapse
Affiliation(s)
- Asma Tlili
- Unit of Functional and Adaptive Biology, Sorbonne Paris Cité, Univ. Paris Diderot, EAC-CNRS 4413, 75013 Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Exogenous BDNF enhances the integration of chronically injured axons that regenerate through a peripheral nerve grafted into a chondroitinase-treated spinal cord injury site. Exp Neurol 2012; 239:91-100. [PMID: 23022460 DOI: 10.1016/j.expneurol.2012.09.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Revised: 08/03/2012] [Accepted: 09/20/2012] [Indexed: 12/20/2022]
Abstract
Although axons lose some of their intrinsic capacity for growth after their developmental period, some axons retain the potential for regrowth after injury. When provided with a growth-promoting substrate such as a peripheral nerve graft (PNG), severed axons regenerate into and through the graft; however, they stop when they reach the glial scar at the distal graft-host interface that is rich with inhibitory chondroitin sulfate proteoglycans. We previously showed that treatment of a spinal cord injury site with chondroitinase (ChABC) allows axons within the graft to traverse the scar and reinnervate spinal cord, where they form functional synapses. While this improvement in outgrowth was significant, it still represented only a small percentage (<20%) of axons compared to the total number of axons that regenerated into the PNG. Here we tested whether providing exogenous brain-derived neurotrophic factor (BDNF) via lentivirus in tissue distal to the PNG would augment regeneration beyond a ChABC-treated glial interface. We found that ChABC treatment alone promoted axonal regeneration but combining ChABC with BDNF-lentivirus did not increase the number of axons that regenerated back into spinal cord. Combining BDNF with ChABC did increase the number of spinal cord neurons that were trans-synaptically activated during electrical stimulation of the graft, as indicated by c-Fos expression, suggesting that BDNF overexpression improved the functional significance of axons that did reinnervate distal spinal cord tissue.
Collapse
|
37
|
Schober ME, Block B, Requena DF, Hale MA, Lane RH. Developmental traumatic brain injury decreased brain derived neurotrophic factor expression late after injury. Metab Brain Dis 2012; 27:167-73. [PMID: 22527999 PMCID: PMC3383795 DOI: 10.1007/s11011-012-9309-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Accepted: 04/15/2012] [Indexed: 01/06/2023]
Abstract
Pediatric traumatic brain injury (TBI) is a major cause of acquired cognitive dysfunction in children. Hippocampal Brain Derived Neurotrophic Factor (BDNF) is important for normal cognition. Little is known about the effects of TBI on BDNF levels in the developing hippocampus. We used controlled cortical impact (CCI) in the 17 day old rat pup to test the hypothesis that CCI would first increase rat hippocampal BDNF mRNA/protein levels relative to SHAM and Naïve rats by post injury day (PID) 2 and then decrease BDNF mRNA/protein by PID14. Relative to SHAM, CCI did not change BDNF mRNA/protein levels in the injured hippocampus in the first 2 days after injury but did decrease BDNF protein at PID14. Surprisingly, BDNF mRNA decreased at PID 1, 3, 7 and 14, and BDNF protein decreased at PID 2, in SHAM and CCI hippocampi relative to Naïve. In conclusion, TBI decreased BDNF protein in the injured rat pup hippocampus 14 days after injury. BDNF mRNA levels decreased in both CCI and SHAM hippocampi relative to Naïve, suggesting that certain aspects of the experimental paradigm (such as craniotomy, anesthesia, and/or maternal separation) may decrease the expression of BDNF in the developing hippocampus. While BDNF is important for normal cognition, no inferences can be made regarding the cognitive impact of any of these factors. Such findings, however, suggest that meticulous attention to the experimental paradigm, and possible inclusion of a Naïve group, is warranted in studies of BDNF expression in the developing brain after TBI.
Collapse
|
38
|
Chavez-Valdez R, Mason A, Nunes AR, Northington FJ, Tankersley C, Ahlawat R, Johnson SM, Gauda EB. Effect of hyperoxic exposure during early development on neurotrophin expression in the carotid body and nucleus tractus solitarii. J Appl Physiol (1985) 2012; 112:1762-72. [PMID: 22422797 DOI: 10.1152/japplphysiol.01609.2011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Synaptic activity can modify expression of neurotrophins, which influence the development of neuronal circuits. In the newborn rat, early hyperoxia silences the synaptic activity and input from the carotid body, impairing the development and function of chemoreceptors. The purpose of this study was to determine whether early hyperoxic exposure, sufficient to induce hypoplasia of the carotid body and decrease the number of chemoafferents, would also modify neurotrophin expression within the nucleus tractus solitarii (nTS). Rat pups were exposed to hyperoxia (fraction of inspired oxygen 0.60) or normoxia until 7 or 14 days of postnatal development (PND). In the carotid body, hyperoxia decreased brain-derived neurotrophic factor (BDNF) protein expression by 93% (P = 0.04) after a 7-day exposure, followed by a decrease in retrogradely labeled chemoafferents by 55% (P = 0.004) within the petrosal ganglion at 14 days. Return to normoxia for 1 wk after a 14-day hyperoxic exposure did not reverse this effect. In the nTS, hyperoxia for 7 days: 1) decreased BDNF gene expression by 67% and protein expression by 18%; 2) attenuated upregulation of BDNF mRNA levels in response to acute hypoxia; and 3) upregulated p75 neurotrophic receptor, truncated tropomyosin kinase B (inactive receptor), and cleaved caspase-3. These effects were not observed in the locus coeruleus (LC). Hyperoxia for 14 days also decreased tyrosine hydroxylase levels by 18% (P = 0.04) in nTS but not in the LC. In conclusion, hyperoxic exposure during early PND reduces neurotrophin levels in the carotid body and the nTS and shifts the balance of neurotrophic support from prosurvival to proapoptotic in the nTS, the primary brain stem site for central integration of sensory and autonomic inputs.
Collapse
Affiliation(s)
- Raul Chavez-Valdez
- Department of Pediatrics, Division of Neonatology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-3200, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Boyce VS, Park J, Gage FH, Mendell LM. Differential effects of brain-derived neurotrophic factor and neurotrophin-3 on hindlimb function in paraplegic rats. Eur J Neurosci 2011; 35:221-32. [PMID: 22211901 PMCID: PMC3509221 DOI: 10.1111/j.1460-9568.2011.07950.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We compared the effect of viral administration of brain-derived neurotrophic factor (BDNF) or neurotrophin 3 (NT-3) on locomotor recovery in adult rats with complete thoracic (T10) spinal cord transection injuries, in order to determine the effect of chronic neurotrophin expression on spinal plasticity. At the time of injury, BDNF, NT-3 or green fluorescent protein (GFP) (control) was delivered to the lesion via adeno-associated virus (AAV) constructs. AAV–BDNF was significantly more effective than AAV–NT-3 in eliciting locomotion. In fact, AAV–BDNF-treated rats displayed plantar, weight-supported hindlimb stepping on a stationary platform, that is, without the assistance of a moving treadmill and without step training. Rats receiving AAV–NT-3 or AAV–GFP were incapable of hindlimb stepping during this task, despite provision of balance support. AAV–NT-3 treatment did promote the recovery of treadmill-assisted stepping, but this required continuous perineal stimulation. In addition, AAV–BDNF-treated rats were sensitized to noxious heat, whereas AAV–NT-3-treated and AAV–GFP-treated rats were not. Notably, AAV–BDNF-treated rats also developed hindlimb spasticity, detracting from its potential clinical applicability via the current viral delivery method. Intracellular recording from triceps surae motoneurons revealed that AAV–BDNF significantly reduced motoneuron rheobase, suggesting that AAV–BDNF promoted the recovery of over-ground stepping by enhancing neuronal excitability. Elevated nuclear c-Fos expression in interneurons located in the L2 intermediate zone after AAV–BDNF treatment indicated increased activation of interneurons in the vicinity of the locomotor central pattern generator. AAV–NT-3 treatment reduced motoneuron excitability, with little change in c-Fos expression. These results support the potential for BDNF delivery at the lesion site to reorganize locomotor circuits.
Collapse
Affiliation(s)
- Vanessa S Boyce
- Department of Neurobiology and Behavior, Life Sciences Building, Room 532, State University of New York at Stony Brook, Stony Brook, NY 11794-5230, USA
| | | | | | | |
Collapse
|
40
|
Carreño FR, Walch JD, Dutta M, Nedungadi TP, Cunningham JT. Brain-derived neurotrophic factor-tyrosine kinase B pathway mediates NMDA receptor NR2B subunit phosphorylation in the supraoptic nuclei following progressive dehydration. J Neuroendocrinol 2011; 23:894-905. [PMID: 21848649 PMCID: PMC3183156 DOI: 10.1111/j.1365-2826.2011.02209.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We studied the effects of water deprivation (WD) on the phosphorylation of tyrosine kinase B (TrkB) and NMDA receptor subunits in the supraoptic nucleus (SON) of the rat. Laser capture microdissection and quantitative reverse transcriptase polymerase chain reaction was used to demonstrate brain-derived neurotrophic factor (BDNF) and TrkB gene expression in vasopressin SON neurones. Immunohistochemistry confirmed BDNF staining in vasopressin neurones, whereas staining for phosphorylated TrkB was increased following WD. Western blot analysis of brain punches containing the SON revealed that tyrosine phosphorylation of TrkB (pTrkBY(515)), serine phosphorylation of NR1 (pNR1S(866) or pNR1) and tyrosine phosphorylation of NR2B subunits (pNR2BY(1472) or pNR2B) were significantly increased in WD animals compared to controls. Access to water for 2 h reduced pTrkBY(515) content to control levels without affecting pNR1 or pNR2B. Four hours of rehydration was needed to reduce pNR1 and pNR2B to control levels. To test whether increased phosphorylation of TrkB in the present study is mediated by BDNF, a group of animals were instrumented with right SON cannula coupled to mini-osmotic pumps filled with vehicle or TrkB-Fc fusion protein, which prevents BDNF binding to TrkB. In the left SON contralateral to the cannula, TrkB phosphorylation was significantly enhanced following WD. Separate analysis of the right SON, which received TrkB-Fc, showed that the TrkB receptor phosphorylation following WD was significantly attenuated. Although increased pNR1S(866) following WD was not affected by local infusion of TrkB-Fc, pNR2BY(1472) was significantly reduced. Co-immunoprecipitation revealed an increased physical interaction between Fyn kinase and NR2B and TrkB in the SON following WD. Thus, activation of TrkB in the SON following WD may affect cellular excitability through the phosphorylation of NR2B subunits.
Collapse
Affiliation(s)
- Flávia Regina Carreño
- Department of Integrative Physiology, and Cardiovascular Research Institute, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Blvd, Fort Worth, TX 76107
| | - Joseph D Walch
- Department of Integrative Physiology, and Cardiovascular Research Institute, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Blvd, Fort Worth, TX 76107
- Department of Pharmacology and Neuroscience, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229
| | - Mayurika Dutta
- Department of Integrative Physiology, and Cardiovascular Research Institute, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Blvd, Fort Worth, TX 76107
| | - Thekkethil P. Nedungadi
- Department of Integrative Physiology, and Cardiovascular Research Institute, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Blvd, Fort Worth, TX 76107
| | - J. Thomas Cunningham
- Department of Integrative Physiology, and Cardiovascular Research Institute, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Blvd, Fort Worth, TX 76107
| |
Collapse
|
41
|
JIP3 mediates TrkB axonal anterograde transport and enhances BDNF signaling by directly bridging TrkB with kinesin-1. J Neurosci 2011; 31:10602-14. [PMID: 21775604 DOI: 10.1523/jneurosci.0436-11.2011] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF), secreted from target tissues, binds and activates TrkB receptors, located on axonal terminals of the innervating neurons, and thereby initiates retrograde signaling. Long-range anterograde transport of TrkB in axons and dendrites requires kinesin-mediated transport. However, it remains unknown whether anterograde TrkB transport mechanisms are the same in axons versus in dendrites. Here, we show that c-Jun NH(2)-terminal kinase-interacting protein 3 (JIP3) binds directly to TrkB, via a minimal 12 aa domain in the TrkB juxtamembrane region, and links TrkB to kinesin-1. The JIP3/TrkB interaction selectively drives TrkB anterograde transport in axons but not in dendrites of rat hippocampal neurons. Moreover, we find that TrkB axonal transport mediated by JIP3 could regulate BDNF-induced Erk activation and axonal filopodia formation. Our findings demonstrate a role for JIP3-mediated TrkB anterograde axonal transport in recruiting more TrkB into distal axons and facilitating BDNF-induced retrograde signaling and synapse modulation, which provides a novel mechanism of how the TrkB anterograde transport can be coupled to BDNF signaling in distal axons.
Collapse
|
42
|
McGinty JF, Bache AJ, Coleman NT, Sun WL. The Role of BDNF/TrkB Signaling in Acute Amphetamine-Induced Locomotor Activity and Opioid Peptide Gene Expression in the Rat Dorsal Striatum. Front Syst Neurosci 2011; 5:60. [PMID: 21811445 PMCID: PMC3143721 DOI: 10.3389/fnsys.2011.00060] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 06/27/2011] [Indexed: 01/15/2023] Open
Abstract
Exposure to psychostimulants increases brain-derived neurotrophic factor (BDNF) mRNA and protein levels in the cerebral cortex and subcortical structures. Because BDNF is co-localized with dopamine and glutamate in afferents to the striatum of rats, it may be co-released with those neurotransmitters upon stimulation. Further, there may be an interaction between the intracellular signaling cascades activated by dopamine, glutamate, and TrkB receptors in medium spiny striatal neurons. In the present study, the effect of acute amphetamine administration on TrkB phosphorylation, as an indirect indicator of activation, and striatal gene expression, was evaluated. In Experiment 1, 15 min or 2 h after a single saline or amphetamine (2.5 mg/kg, i.p.) injection, the caudate–putamen (CPu), nucleus accumbens (NAc), and dorsomedial prefrontal cortex (dmPFC) were extracted and processed for phospho (p)-TrkB immunoreactivity. Immunoprecipitation analyses indicated that neither the tyrosine phosphorylation (p-Tyr) or autophosphorylation sites of TrkB (706) were changed in NAc, CPu, or dmPFC 15 min after amphetamine administration. In contrast, p-Tyr and the PLCγ phosphorylation site of TrkB (816) were increased in the NAc and CPu 2 h after amphetamine. In Experiment 2, intra-striatal infusion of the tyrosine kinase inhibitor, K252a, increased amphetamine-induced vertical activity but not total distance traveled. In addition, K252a inhibited amphetamine-induced preprodynorphin, but not preproenkephalin, mRNA expression in the striatum. These data indicate that acute amphetamine administration induces p-TrkB activation and signaling in a time- and brain region-dependent manner and that TrkB/BDNF signaling plays an important role in amphetamine-induced behavior and striatal gene expression.
Collapse
Affiliation(s)
- Jacqueline F McGinty
- Department of Neurosciences, Medical University of South Carolina Charleston, SC, USA
| | | | | | | |
Collapse
|
43
|
Bi C, Yue X, Zhou R, Plummer MR. EphA activation overrides the presynaptic actions of BDNF. J Neurophysiol 2011; 105:2364-74. [PMID: 21411563 DOI: 10.1152/jn.00564.2010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The adult pattern of neural connectivity is shaped by repulsive and attractive factors, many of which are modulated by activity. Although much is known about the actions of these factors when studied in isolation, little is known about how they interact. To address this question, we examined the effects of sequential or coapplication of brain-derived neurotrophic factor (BDNF) and Fc-conjugated ephrin-A5 or EphA5 in cultured embryonic hippocampal neurons. BDNF promotes neurite outgrowth and synapse formation, and when applied acutely, it elicits an increase in ongoing synaptic activity. Members of the ephrin family of ligands and receptors can be repulsive and prevent formation of synaptic contacts. Acute exposure to either ephrin-A5-Fc or EphA5-Fc transiently enhanced synaptic activity when applied alone, but when applied prior to BDNF, they dramatically reduced the electrophysiological effects of the neurotrophin. Conversely, BDNF had no effect on subsequently applied ephrin-A5-Fc or EphA5-Fc. Consistent with this, ephrin-A5-Fc also prevented BDNF-induced activation of p42/44 MAPK. The effect of ephrin-A5-Fc appears to be presynaptic, as it prevented the BDNF-induced increase in spontaneous miniature postsynaptic current frequency, whereas EphA5-Fc did not. These results suggest that these factors can be categorized differently, with the contact-mediated activation of EphA receptors by ephrin-A5 overriding the diffusion-mediated effect of BDNF.
Collapse
Affiliation(s)
- Caixia Bi
- Rutgers University, Department of Cell Biology & Neuroscience, Nelson Laboratories, 604 Allison Rd., Piscataway, NJ 08854-8082, USA
| | | | | | | |
Collapse
|
44
|
Pearson-Fuhrhop KM, Cramer SC. Genetic influences on neural plasticity. PM R 2011; 2:S227-40. [PMID: 21172685 DOI: 10.1016/j.pmrj.2010.09.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Accepted: 09/13/2010] [Indexed: 01/07/2023]
Abstract
Neural plasticity refers to the capability of the brain to alter function or structure in response to a range of events and is a crucial component of both functional recovery after injury and skill learning in healthy individuals. A number of factors influence neural plasticity and recovery of function after brain injury. The current review considers the impact of genetic factors. Polymorphisms in the human genes coding for brain-derived neurotrophic factor and apolipoprotein E have been studied in the context of plasticity and stroke recovery and are discussed here in detail. Several processes involved in plasticity and stroke recovery, such as depression or pharmacotherapy effects, are modulated by other genetic polymorphisms and are also discussed. Finally, new genetic polymorphisms that have not been studied in the context of stroke are proposed as new directions for study. A better understanding of genetic influences on recovery and response to therapy might allow improved treatment after a number of forms of central nervous system injury.
Collapse
|
45
|
Ali I, Salzberg MR, French C, Jones NC. Electrophysiological insights into the enduring effects of early life stress on the brain. Psychopharmacology (Berl) 2011; 214:155-73. [PMID: 21165736 DOI: 10.1007/s00213-010-2125-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Accepted: 11/29/2010] [Indexed: 12/31/2022]
Abstract
Increasing evidence links exposure to stress early in life to long-term alterations in brain function, which in turn have been linked to a range of psychiatric and neurological disorders in humans. Electrophysiological approaches to studying these causal pathways have been relatively underexploited. Effects of early life stress on neuronal electrophysiological properties offer a set of potential mechanisms for these susceptibilities, notably in the case of epilepsy. Thus, we review experimental evidence for altered cellular and circuit electrophysiology resulting from exposure to early life stress. Much of this work focuses on limbic long-term potentiation, but other studies address alterations in electrophysiological properties of ion channels, neurotransmitter systems, and the autonomic nervous system. We discuss mechanisms which may mediate these effects, including influences of early life stress on key components of brain synaptic transmission, particularly glutamate, GABA and 5-HT receptors, and influences on neuroplasticity (primarily neurogenesis and synaptic density) and on neuronal network activity. The existing literature, although small, provides strong evidence that early life stress induces enduring, often robust effects on a range of electrophysiological properties, suggesting further study of enduring effects of early life stress employing electrophysiological methods and concepts will be productive in illuminating disease pathophysiology.
Collapse
Affiliation(s)
- Idrish Ali
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Melbourne, VIC, Australia
| | | | | | | |
Collapse
|
46
|
Calcium-dependent activator protein for secretion 2 (CAPS2) promotes BDNF secretion and is critical for the development of GABAergic interneuron network. Proc Natl Acad Sci U S A 2010; 108:373-8. [PMID: 21173225 DOI: 10.1073/pnas.1012220108] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Calcium-dependent activator protein for secretion 2 (CAPS2) is a dense-core vesicle-associated protein that is involved in the secretion of BDNF. BDNF has a pivotal role in neuronal survival and development, including the development of inhibitory neurons and their circuits. However, how CAPS2 affects BDNF secretion and its biological significance in inhibitory neurons are largely unknown. Here we reveal the role of CAPS2 in the regulated secretion of BDNF and show the effect of CAPS2 on the development of hippocampal GABAergic systems. We show that CAPS2 is colocalized with BDNF, both synaptically and extrasynaptically in axons of hippocampal neurons. Overexpression of exogenous CAPS2 in hippocampal neurons of CAPS2-KO mice enhanced depolarization-induced BDNF exocytosis events in terms of kinetics, frequency, and amplitude. We also show that in the CAPS2-KO hippocampus, BDNF secretion is reduced, and GABAergic systems are impaired, including a decreased number of GABAergic neurons and their synapses, a decreased number of synaptic vesicles in inhibitory synapses, and a reduced frequency and amplitude of miniature inhibitory postsynaptic currents. Conversely, excitatory neurons in the CAPS2-KO hippocampus were largely unaffected with respect to field excitatory postsynaptic potentials, miniature excitatory postsynaptic currents, and synapse number and morphology. Moreover, CAPS2-KO mice exhibited several GABA system-associated deficits, including reduced late-phase long-term potentiation at CA3-CA1 synapses, decreased hippocampal theta oscillation frequency, and increased anxiety-like behavior. Collectively, these results suggest that CAPS2 promotes activity-dependent BDNF secretion during the postnatal period that is critical for the development of hippocampal GABAergic networks.
Collapse
|
47
|
Correlation of cognitive performance and morphological changes in neocortical pyramidal neurons in aging. Neurobiol Aging 2010; 33:1466-80. [PMID: 21163553 DOI: 10.1016/j.neurobiolaging.2010.10.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 10/12/2010] [Accepted: 10/16/2010] [Indexed: 12/24/2022]
Abstract
It is well established that the cerebral cortex undergoes extensive remodeling in aging. In this study, we used behaviorally characterized rats to correlate age-related morphological changes with cognitive impairment. For this, young and aged animals were tested in the Morris water maze to evaluate their cognitive performance. Following behavioral characterization, the animals were perfused and a combination of intracellular labeling and immunohistochemistry was applied. Using this approach, we characterized the dendritic morphology of cortical pyramidal neurons as well as the pattern of glutamatergic and GABAergic appositions on their cell bodies and dendrites. We focused on the association region of the parietal cortex (LtPA) and the medial prefrontal cortex (mPFC) for their involvement in the Morris water maze task. We found an age-related atrophy of distal basal dendrites that did not differ between aged cognitively unimpaired (AU) and aged cognitively impaired animals (AI). Dendritic spines and glutamatergic appositions generally decreased from young to AU and from AU to AI rats. On the other hand, GABAergic appositions only showed a trend towards a decrease in AU rats. Collectively, the data show that the ratio of excitatory/inhibitory inputs was only altered in AI animals. When cortical cholinergic varicosities were labeled on alternate sections, we found that AI animals also had a significant reduction of cortical cholinergic boutons compared with AU or young animals. In aged animals, the density of cortical cholinergic varicosities correlated with the excitatory/inhibitory ratio. Our data suggest that both cholinergic atrophy and an imbalance towards inhibition may contribute to the observed age-associated behavioral impairment.
Collapse
|
48
|
Baig BJ, Whalley HC, Hall J, McIntosh AM, Job DE, Cunningham-Owens DG, Johnstone EC, Lawrie SM. Functional magnetic resonance imaging of BDNF val66met polymorphism in unmedicated subjects at high genetic risk of schizophrenia performing a verbal memory task. Psychiatry Res 2010; 183:195-201. [PMID: 20708907 DOI: 10.1016/j.pscychresns.2010.06.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 04/28/2010] [Accepted: 06/22/2010] [Indexed: 12/31/2022]
Abstract
Multiple strands of evidence suggest a role for Brain Derived Neurotrophic Factor (BDNF) in the pathophysiology of schizophrenia. It is not yet clear, however, how BDNF may contribute to altered brain function seen in the disorder, or in those at high genetic risk. The current study examines functional imaging correlates of the BDNF val66met polymorphism in a population at high genetic risk of schizophrenia. Subjects at high genetic risk for the disorder (n=58) provided both BDNF genotyping and fMRI data while performing a verbal memory task. During encoding, participants were presented with a word and asked to make a 'living'/'non-living' classification. During retrieval, individuals were requested to make an 'old'/'new' word classification. For encoding, we report decreased activation of the inferior occipital cortex and a trend in the cingulate cortex in Val homozygote individuals relative to Met carriers. For retrieval, we report decreases in activation in the prefrontal, cingulate cortex and bilateral posterior parietal regions in Val homozygote individuals versus Met carriers. These findings add to previous evidence suggesting that genetic variation in the BDNF gene modulates prefrontal and limbic functioning and suggests that it may contribute to differences in brain function seen in those at high risk of the disorder.
Collapse
Affiliation(s)
- Benjamin J Baig
- Division of Psychiatry, University of Edinburgh, Kennedy Tower, Royal Edinburgh Hospital, Morningside Park, Edinburgh, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Calissano P, Matrone C, Amadoro G. Nerve growth factor as a paradigm of neurotrophins related to Alzheimer's disease. Dev Neurobiol 2010; 70:372-83. [PMID: 20186703 DOI: 10.1002/dneu.20759] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Converging lines of evidence on the possible connection between NGF signaling and Alzheimer's diseases (AD) are unraveling new facets which could depict this neurotrophin (NTF) in a more central role. AD animal models have provided evidence that a shortage of NGF supply may induce an AD-like syndrome. In vitro experiments, moreover, are delineating a possible temporal and causal link between APP amiloydogenic processing and altered post-translational tau modifications. After NGF signaling interruption, the pivotal upstream players of the amyloid cascade (APP, beta-secretase, and active form of gamma-secretase) are up-regulated, leading to an increased production of amyloid beta peptide (Abeta) and to its intracellular aggregation in molecular species of different sizes. Contextually, the Abeta released pool generates an autocrine toxic loop in the same healthy neurons. At the same time tau protein undergoes anomalous, GSKbeta-mediated, phosphorylation at specific pathogenetic sites (Ser262 and Thr 231), caspase(s) and calpain- I- mediated truncation, detachment from microtubules with consequent cytoskeleton collapse and axonal transport impairment. All these events are inhibited when the amyloidogenic processing is reduced by beta and gamma secretase inhibitors or anti-Abeta antibodies and appear to be causally correlated to TrkA, p75CTF, Abeta, and PS1 molecular association in an Abeta-mediated fashion. In this scenario, the so-called trophic action exerted by NGF (and possibly also by other neurotrophins) in these targets neurons is actually the result of an anti-amyloidogenic activity.
Collapse
Affiliation(s)
- P Calissano
- Institute of Neurobiology and Molecular Medicine, C.N.R. Fondazione Santa Lucia, Italy.
| | | | | |
Collapse
|
50
|
Li B, Arime Y, Hall FS, Uhl GR, Sora I. Impaired spatial working memory and decreased frontal cortex BDNF protein level in dopamine transporter knockout mice. Eur J Pharmacol 2010; 628:104-7. [PMID: 19932884 PMCID: PMC3724416 DOI: 10.1016/j.ejphar.2009.11.036] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Revised: 10/26/2009] [Accepted: 11/10/2009] [Indexed: 10/20/2022]
Abstract
Brain-derived neurotrophic factor (BDNF), one of the key brain neurotrophins, has been implicated in neuronal plasticity and memory. Recent studies document the importance of BDNF for normal long-term memory functions. However, there are few studies of the roles of BDNF in short-term memory. Dopamine is likely to play important roles in BDNF gene expression in specific brain regions, including frontal cortical regions that are implicated in short-term working memory processes that include spontaneous alternation. We have thus tested spatial working memory in dopamine transporter knockout (DAT KO) and wild-type mice. Spontaneous alternation in the Y-maze, an index of short-term spatial working memory in mice, was significantly decreased in DAT KO mice compared to wild-type mice. BDNF protein was significantly decreased in frontal cortex, though not in striatum or hippocampus, of the DAT KO mice. The data support the hypothesis that impaired spatial working memory in DAT KO mice may be related to decreased frontal cortical BDNF in these animals, and document apparent roles for BDNF in a short-term memory process.
Collapse
Affiliation(s)
- BingJin Li
- Department of Biological Psychiatry, Tohoku University Graduate School of Medicine, Japan
- Morden Research Center for Traditional Chinese Medicine of Shanxi University, TaiYuan, China
| | - Yosefu Arime
- Department of Biological Psychiatry, Tohoku University Graduate School of Medicine, Japan
| | - F. Scott Hall
- Molecular Neurobiology Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Baltimore, MD, USA
| | - George R. Uhl
- Molecular Neurobiology Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Baltimore, MD, USA
| | - Ichiro Sora
- Department of Biological Psychiatry, Tohoku University Graduate School of Medicine, Japan
| |
Collapse
|