1
|
Sakamaki Y, Shobudani M, Ojiro R, Ozawa S, Tang Q, Zou X, Ebizuka Y, Karasawa A, Woo G, Yoshida T, Shibutani M. Suppression of Hippocampal Neurogenesis and Oligodendrocyte Maturation Similar to Developmental Hypothyroidism by Maternal Exposure of Rats to Ammonium Perchlorate, a Gunpowder Raw Material and Known Environmental Contaminant. ENVIRONMENTAL TOXICOLOGY 2025; 40:30-53. [PMID: 39248596 PMCID: PMC11628657 DOI: 10.1002/tox.24413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 07/18/2024] [Accepted: 08/17/2024] [Indexed: 09/10/2024]
Abstract
The environmental contaminant perchlorate raises concern for hypothyroidism-related brain disorders in children. This study investigated the effects of developmental perchlorate exposure on hippocampal neurogenesis and oligodendrocyte (OL) development. Pregnant Sprague-Dawley rats were administered with ammonium perchlorate (AP) in drinking water at concentrations of 0 (control), 300, and 1000 ppm from gestation day 6 until weaning [postnatal day (PND) 21]. On PND 21, offspring displayed decreased serum triiodothyronine and thyroxine concentrations at 1000 ppm and thyroid follicular epithelial cell hyperplasia at ≥300 ppm (accompanying increased proliferation activity at 1000 ppm). Hippocampal neurogenesis indicated suppressed proliferation of neurogenic cells at ≥300 ppm, causing decreases in type-1 neural stem cells (NSCs) and type-2a neural progenitor cells. In addition, an increase of SST+ GABAergic interneurons and decreasing trend for ARC+ granule cells were observed at 1000 ppm. CNPase+ mature OLs were decreased in number in the dentate gyrus hilus at ≥300 ppm. At PND 77, thyroid changes had disappeared; however, the decrease of type-1 NSCs and increase of SST+ interneurons persisted, CCK+ interneurons were increased, and white matter tissue area was decreased at 1000 ppm. Obtained results suggest an induction of hypothyroidism causing suppressed hippocampal neurogenesis (targeting early neurogenic processes and decreased synaptic plasticity of granule cells involving ameliorative interneuron responses) and suppressed OL maturation during the weaning period. In adulthood, suppression of neurogenesis continued, and white matter hypoplasia was evident. Observed brain changes were similar to those caused by developmental hypothyroidism, suggesting that AP-induced developmental neurotoxicity was due to hypothyroidism.
Collapse
Affiliation(s)
- Yuri Sakamaki
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of AgricultureTokyo University of Agriculture and TechnologyTokyoJapan
| | - Momoka Shobudani
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of AgricultureTokyo University of Agriculture and TechnologyTokyoJapan
| | - Ryota Ojiro
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of AgricultureTokyo University of Agriculture and TechnologyTokyoJapan
- Cooperative Division of Veterinary Sciences, Graduate School of AgricultureTokyo University of Agriculture and TechnologyTokyoJapan
| | - Shunsuke Ozawa
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of AgricultureTokyo University of Agriculture and TechnologyTokyoJapan
- Cooperative Division of Veterinary Sciences, Graduate School of AgricultureTokyo University of Agriculture and TechnologyTokyoJapan
| | - Qian Tang
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of AgricultureTokyo University of Agriculture and TechnologyTokyoJapan
- Cooperative Division of Veterinary Sciences, Graduate School of AgricultureTokyo University of Agriculture and TechnologyTokyoJapan
| | - Xinyu Zou
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of AgricultureTokyo University of Agriculture and TechnologyTokyoJapan
- Cooperative Division of Veterinary Sciences, Graduate School of AgricultureTokyo University of Agriculture and TechnologyTokyoJapan
| | - Yuri Ebizuka
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of AgricultureTokyo University of Agriculture and TechnologyTokyoJapan
| | - Ayumi Karasawa
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of AgricultureTokyo University of Agriculture and TechnologyTokyoJapan
| | - Gye‐Hyeong Woo
- Laboratory of Histopathology, Department of Clinical Laboratory ScienceSemyung UniversityJecheonChungbukRepublic of Korea
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of AgricultureTokyo University of Agriculture and TechnologyTokyoJapan
- Cooperative Division of Veterinary Sciences, Graduate School of AgricultureTokyo University of Agriculture and TechnologyTokyoJapan
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of AgricultureTokyo University of Agriculture and TechnologyTokyoJapan
- Cooperative Division of Veterinary Sciences, Graduate School of AgricultureTokyo University of Agriculture and TechnologyTokyoJapan
| |
Collapse
|
2
|
Spero V, Scherma M, D'Amelio S, Collu R, Dedoni S, Camoglio C, Siddi C, Fratta W, Molteni R, Fadda P. Activity-based anorexia (ABA) model: Effects on brain neuroinflammation, redox balance and neuroplasticity during the acute phase. Neurochem Int 2024; 180:105842. [PMID: 39244038 DOI: 10.1016/j.neuint.2024.105842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/14/2024] [Accepted: 08/25/2024] [Indexed: 09/09/2024]
Abstract
Several evidences suggest that immuno-inflammatory responses are involved in the pathogenesis of anorexia nervosa (AN). Herein we investigate the possible alteration of key mediators of inflammation, redox balance, and neuroplasticity in the brain of rats showing an anorexic-like phenotype. We modeled AN in adolescent female rats using the activity-based anorexia (ABA) paradigm and measured gene expression levels of targets of interest in the prefrontal cortex (PFC) and dorsal hippocampus (DH). We observed reduced mRNA levels of pro-inflammatory cytokines IL-1β and TNF-α, the inflammasome NLRP3, and the microglial marker CD11b in both PFC and DH of ABA animals. Conversely, the mRNA of IL-6, which acts as both a pro-inflammatory and anti-inflammatory cytokine, was increased. Moreover, we observed an overall upregulation of different antioxidant enzymes in PFC, while their profile was not affected or opposite in the DH, with the exception of MT1α. Interestingly, ABA animals showed elevated levels of the neuroplasticity marker BDNF in both PFC and DH. Our data indicate that ABA induction is associated with anatomical-specific cerebral alteration of mediators of neuroinflammation, oxidative balance and neuroplasticity. Although more research should be conducted, these results add important information about the role of these systems in the complex AN etiopathogenesis.
Collapse
Affiliation(s)
- Vittoria Spero
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Maria Scherma
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Sabrina D'Amelio
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Roberto Collu
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, USA
| | - Simona Dedoni
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Chiara Camoglio
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Carlotta Siddi
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Walter Fratta
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Raffaella Molteni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Paola Fadda
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy; CNR Institute of Neuroscience - Cagliari, National Research Council, Cagliari, Italy.
| |
Collapse
|
3
|
Tsybko A, Eremin D, Ilchibaeva T, Khotskin N, Naumenko V. CDNF Exerts Anxiolytic, Antidepressant-like, and Procognitive Effects and Modulates Serotonin Turnover and Neuroplasticity-Related Genes. Int J Mol Sci 2024; 25:10343. [PMID: 39408672 PMCID: PMC11482483 DOI: 10.3390/ijms251910343] [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: 08/15/2024] [Revised: 09/17/2024] [Accepted: 09/21/2024] [Indexed: 10/19/2024] Open
Abstract
Cerebral dopamine neurotrophic factor (CDNF) is an unconventional neurotrophic factor because it does not bind to a known specific receptor on the plasma membrane and functions primarily as an unfolded protein response (UPR) regulator in the endoplasmic reticulum. Data on the effects of CDNF on nonmotor behavior and monoamine metabolism are limited. Here, we performed the intracerebroventricular injection of a recombinant CDNF protein at doses of 3, 10, and 30 μg in C57BL/6 mice. No adverse effects of the CDNF injection on feed and water consumption or locomotor activity were observed for 3 days afterwards. Decreases in body weight and sleep duration were transient. CDNF-treated animals demonstrated improved performance on the operant learning task and a substantial decrease in anxiety and behavioral despair. CDNF in all the doses enhanced serotonin (5-HT) turnover in the murine frontal cortex, hippocampus, and midbrain. This alteration was accompanied by changes in the mRNA levels of the 5-HT1A and 5-HT7 receptors and in monoamine oxidase A mRNA and protein levels. We found that CDNF dramatically increased c-Fos mRNA levels in all investigated brain areas but elevated the phosphorylated-c-Fos level only in the midbrain. Similarly, enhanced CREB phosphorylation was found in the midbrain in experimental animals. Additionally, the upregulation of a spliced transcript of XBP1 (UPR regulator) was detected in the midbrain and frontal cortex. Thus, we can hypothesize that exogenous CDNF modulates the UPR pathway and overall neuronal activation and enhances 5-HT turnover, thereby affecting learning and emotion-related behavior.
Collapse
Affiliation(s)
- Anton Tsybko
- The Federal Research Center, Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia; (D.E.); (T.I.); (N.K.); (V.N.)
| | | | | | | | | |
Collapse
|
4
|
Jara JS, Avci HX, Kouremenou I, Doulazmi M, Bakouche J, Dubacq C, Goyenvalle C, Mariani J, Lohof AM, Sherrard RM. Pax3 induces target-specific reinnervation through axon collateral expression of PSA-NCAM. Prog Neurobiol 2024; 232:102560. [PMID: 38097036 DOI: 10.1016/j.pneurobio.2023.102560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/07/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023]
Abstract
Damaged or dysfunctional neural circuits can be replaced after a lesion by axon sprouting and collateral growth from undamaged neurons. Unfortunately, these new connections are often disorganized and rarely produce clinical improvement. Here we investigate how to promote post-lesion axonal collateral growth, while retaining correct cellular targeting. In the mouse olivocerebellar path, brain-derived neurotrophic factor (BDNF) induces correctly-targeted post-lesion cerebellar reinnervation by remaining intact inferior olivary axons (climbing fibers). In this study we identified cellular processes through which BDNF induces this repair. BDNF injection into the denervated cerebellum upregulates the transcription factor Pax3 in inferior olivary neurons and induces rapid climbing fiber sprouting. Pax3 in turn increases polysialic acid-neural cell adhesion molecule (PSA-NCAM) in the sprouting climbing fiber path, facilitating collateral outgrowth and pathfinding to reinnervate the correct targets, cerebellar Purkinje cells. BDNF-induced reinnervation can be reproduced by olivary Pax3 overexpression, and abolished by olivary Pax3 knockdown, suggesting that Pax3 promotes axon growth and guidance through upregulating PSA-NCAM, probably on the axon's growth cone. These data indicate that restricting growth-promotion to potential reinnervating afferent neurons, as opposed to stimulating the whole circuit or the injury site, allows axon growth and appropriate guidance, thus accurately rebuilding a neural circuit.
Collapse
Affiliation(s)
- J Sebastian Jara
- Sorbonne Université & CNRS, IBPS-B2A, Biological Adaptation and Ageing, Paris France
| | - Hasan X Avci
- Sorbonne Université & CNRS, IBPS-B2A, Biological Adaptation and Ageing, Paris France
| | - Ioanna Kouremenou
- Sorbonne Université & CNRS, IBPS-B2A, Biological Adaptation and Ageing, Paris France
| | - Mohamed Doulazmi
- Sorbonne Université & CNRS, IBPS-B2A, Biological Adaptation and Ageing, Paris France
| | - Joelle Bakouche
- Sorbonne Université & CNRS, IBPS-B2A, Biological Adaptation and Ageing, Paris France
| | - Caroline Dubacq
- Sorbonne Université, CNRS & INSERM, IBPS-NPS, Neurosciences Paris Seine, Paris France
| | - Catherine Goyenvalle
- Sorbonne Université & CNRS, IBPS-B2A, Biological Adaptation and Ageing, Paris France
| | - Jean Mariani
- Sorbonne Université & CNRS, IBPS-B2A, Biological Adaptation and Ageing, Paris France
| | - Ann M Lohof
- Sorbonne Université & CNRS, IBPS-B2A, Biological Adaptation and Ageing, Paris France
| | - Rachel M Sherrard
- Sorbonne Université & CNRS, IBPS-B2A, Biological Adaptation and Ageing, Paris France.
| |
Collapse
|
5
|
Scherma M, Palmas MF, Pisanu A, Masia P, Dedoni S, Camoglio C, Fratta W, Carta AR, Fadda P. Induction of Activity-Regulated Cytoskeleton-Associated Protein and c-Fos Expression in an Animal Model of Anorexia Nervosa. Nutrients 2023; 15:3830. [PMID: 37686862 PMCID: PMC10490422 DOI: 10.3390/nu15173830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/19/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Anorexia nervosa (AN) is a complex eating disorder characterized by reduced caloric intake to achieve body-weight loss. Furthermore, over-exercise is commonly reported. In recent years, animal models of AN have provided evidence for neuroplasticity changes in specific brain areas of the mesocorticolimbic circuit, which controls a multitude of functions including reward, emotion, motivation, and cognition. The activity-regulated cytoskeleton-associated protein (Arc) is an immediate early gene that modulates several forms of synaptic plasticity and has been linked to neuropsychiatric illness. Since the role of Arc in AN has never been investigated, in this study we evaluated whether the anorexic-like phenotype reproduced by the activity-based anorexia (ABA) model may impact its expression in selected brain regions that belong to the mesocorticolimbic circuit (i.e., prefrontal cortex, nucleus accumbens, and hippocampus). The marker of neuronal activation c-Fos was also assessed. We found that the expression of both markers increased in all the analyzed brain areas of ABA rats in comparison to the control groups. Moreover, a negative correlation between the density of Arc-positive cells and body-weight loss was found. Together, our findings suggest the importance of Arc and neuroplasticity changes within the brain circuits involved in dysfunctional behaviors associated with AN.
Collapse
Affiliation(s)
- Maria Scherma
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (M.F.P.); (S.D.); (C.C.); (W.F.); (A.R.C.); (P.F.)
| | - Maria Francesca Palmas
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (M.F.P.); (S.D.); (C.C.); (W.F.); (A.R.C.); (P.F.)
| | - Augusta Pisanu
- Neuroscience Institute, Section of Cagliari, National Research Council (CNR), 09042 Cagliari, Italy;
| | - Paolo Masia
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (M.F.P.); (S.D.); (C.C.); (W.F.); (A.R.C.); (P.F.)
| | - Simona Dedoni
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (M.F.P.); (S.D.); (C.C.); (W.F.); (A.R.C.); (P.F.)
| | - Chiara Camoglio
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (M.F.P.); (S.D.); (C.C.); (W.F.); (A.R.C.); (P.F.)
| | - Walter Fratta
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (M.F.P.); (S.D.); (C.C.); (W.F.); (A.R.C.); (P.F.)
| | - Anna R. Carta
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (M.F.P.); (S.D.); (C.C.); (W.F.); (A.R.C.); (P.F.)
| | - Paola Fadda
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (M.F.P.); (S.D.); (C.C.); (W.F.); (A.R.C.); (P.F.)
- Neuroscience Institute, Section of Cagliari, National Research Council (CNR), 09042 Cagliari, Italy;
| |
Collapse
|
6
|
Kemenesi-Gedei PB, Csabafi KA, Kis G. Inflammatory Orofacial Pain Activates Peptidergic Neurons and Upregulates the Oxytocin Receptor Expression in Trigeminal Ganglion. Biomedicines 2023; 11:2419. [PMID: 37760859 PMCID: PMC10525584 DOI: 10.3390/biomedicines11092419] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
The majority of orofacial pain is caused by musculoskeletal and neuropathological diseases related to inflammatory processes that lead even to transcriptional alterations in the trigeminal ganglion (TG) neurons. The hypothalamic nonapeptide oxytocin has been reported to modulate nociception via binding and activating its receptor in primary sensory neurons. The purpose of this study was to analyze the gene expression of the oxytocin receptor (OTR), c-Fos, an indicator of neuronal activity, and α-calcitonin gene-related peptide (αCGRP), a characteristic neurotransmitter of the peptidergic trigeminal primary afferents in an animal model of inflammation-induced orofacial pain. Carrageenan was unilaterally injected into the vibrissal pads of male and female adult Wistar rats. RT-qPCR was performed to analyze the levels of mRNA expression in TGs 24 h after injection. The gene expression analysis revealed higher fold changes regarding the c-Fos (mean ± S.E: ♀: 3.9 ± 0.19; ♂: 3.55 ± 0.18) and αCGRP (♀: 2.84 ± 0.13; ♂: 3.39 ± 0.47) expression levels of mRNA, and a moderate rise in the expression of the OTR mRNA (♀: 1.52 ± 0.07; ♂: 1.49 ± 0.07) was observed in comparison to both vehicle(saline)-treated and untreated controls. Our results furnish evidence for inflammation-induced activation of peptidergic neurons, and it is suggested that oxytocin modulates inflammation-induced nociception by enhancing their signaling capacity due to its elevated expression in the sensory ganglion cells, thus providing new therapies for orofacial pain relief that target the OTRs.
Collapse
Affiliation(s)
- Péter Bátor Kemenesi-Gedei
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Krisztina Anna Csabafi
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Gyöngyi Kis
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, 6720 Szeged, Hungary
| |
Collapse
|
7
|
Arc-Mediated Synaptic Plasticity Regulates Cognitive Function in a Migraine Mouse Model. Brain Sci 2023; 13:brainsci13020331. [PMID: 36831874 PMCID: PMC9954307 DOI: 10.3390/brainsci13020331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/08/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023] Open
Abstract
Previous clinical and basic studies have shown that migraine is associated with cognitive impairment, anxiety, and depression. It severely affects the quality of life. In this study, C57BL/6 mice were randomly divided into four groups: IS group, IS+M group, and IS+S group with repeated application of dural inflammatory soup (IS) stimulation to establish a migraine model, followed by PBS, memantine, and sumatriptan interventions, respectively; the blank control group underwent the same treatment procedure but with PBS instead of IS and intervention drugs. The cognitive function of the mice was used as the main outcome indicator. After application of the IS, mice showed reduced pain threshold for mechanical stimulation, decreased learning memory capacity, attention deficit, a reduced number of dendritic spines in hippocampal neurons, and altered synaptic ultrastructure. The cognitive function indexes of mice in the IS+M group recovered with changes in Arc protein expression to a level not statistically different from that of the Control group, while the IS and IS+S groups remained at lower levels. The present results suggest that Arc-mediated synaptic plasticity may be an essential mechanism of cognitive dysfunction in migraine.
Collapse
|
8
|
Vermehren-Schmaedick A, Olah MJ, Ramunno-Johnson D, Lidke KA, Cohen MS, Vu TQ. Molecular-Scale Dynamics of Long Range Retrograde Brain-Derived Neurotrophic Factor Transport Shaped by Cellular Spatial Context. Front Neurosci 2022; 16:835815. [PMID: 35431786 PMCID: PMC9008462 DOI: 10.3389/fnins.2022.835815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/03/2022] [Indexed: 12/11/2022] Open
Abstract
Retrograde neurotrophin (NT) transport is a specialized form of signal transduction used to conduct information from axons to the cell bodies of central and peripheral nervous system neurons. It is activated upon NT-Trk receptor binding, NT-Trk internalization into signaling endosomes, and their motion along the axon toward the cell body. Brain-derived neurotrophic factor (BDNF) is an abundant NT that modulates key brain and spinal cord functions, and defects in BDNF trafficking are associated with neuronal death, neurodegenerative diseases and in nerve injury. Decades of study have yielded impressive progress in elucidating NT retrograde transport; however, much information remains unclear. For example, while it is known that NT function is dependent on tight control of NT-receptor intracellular trafficking, data describing the precise spatiotemporal molecular dynamics of their axonal to somatic transport are lacking. In past work, we showed the use of discrete, photo-bleaching-resistant quantum dot (QD)-BNDF probes to activate and track BDNF-TrkB receptor internalization; this revealed a rich diversity of molecular motions that intracellular BDNF signaling endosomes undergo within the soma of nodose ganglia sensory neurons. Here, we used combined techniques of discrete QD-BDNF tracking with compartmented microfluidic chambers to characterize retrograde BDNF-TrkB transport over long-ranging distances of primary dorsal root ganglion sensory neuronal axons. Our new findings show that axonal retrograde motion is comprised of heterogeneous mixtures of diffusive behaviors, pauses, and variations in net molecular-motor-dependent transport speeds. Notably, specific molecular dynamic features such as NT speed were dependent on spatial context that could be categorized in distance from distal axons and proximity to the soma and were not entirely dictated by active motor transport speed. The important implication is recognition that NT-receptor retrograde transport is comprised of molecular dynamics, which change over the course of long-range trafficking to shape overall transport and possibly signaling.
Collapse
Affiliation(s)
- Anke Vermehren-Schmaedick
- Department of Biomedical Engineering, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, United States
| | - Mark J. Olah
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM, United States
| | - Damien Ramunno-Johnson
- Department of Biomedical Engineering, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| | - Keith A. Lidke
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM, United States
| | - Michael S. Cohen
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, United States
| | - Tania Q. Vu
- Department of Biomedical Engineering, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
9
|
Eggert S, Kins S, Endres K, Brigadski T. Brothers in arms: proBDNF/BDNF and sAPPα/Aβ-signaling and their common interplay with ADAM10, TrkB, p75NTR, sortilin, and sorLA in the progression of Alzheimer's disease. Biol Chem 2022; 403:43-71. [PMID: 34619027 DOI: 10.1515/hsz-2021-0330] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/16/2021] [Indexed: 12/22/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is an important modulator for a variety of functions in the central nervous system (CNS). A wealth of evidence, such as reduced mRNA and protein level in the brain, cerebrospinal fluid (CSF), and blood samples of Alzheimer's disease (AD) patients implicates a crucial role of BDNF in the progression of this disease. Especially, processing and subcellular localization of BDNF and its receptors TrkB and p75 are critical determinants for survival and death in neuronal cells. Similarly, the amyloid precursor protein (APP), a key player in Alzheimer's disease, and its cleavage fragments sAPPα and Aβ are known for their respective roles in neuroprotection and neuronal death. Common features of APP- and BDNF-signaling indicate a causal relationship in their mode of action. However, the interconnections of APP- and BDNF-signaling are not well understood. Therefore, we here discuss dimerization properties, localization, processing by α- and γ-secretase, relevance of the common interaction partners TrkB, p75, sorLA, and sortilin as well as shared signaling pathways of BDNF and sAPPα.
Collapse
Affiliation(s)
- Simone Eggert
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, D-67663 Kaiserslautern, Germany
| | - Stefan Kins
- Department of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Str. 13, D-67663 Kaiserslautern, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg-University Mainz, D-55131 Mainz, Germany
| | - Tanja Brigadski
- Department of Informatics and Microsystem Technology, University of Applied Sciences Kaiserslautern, D-66482 Zweibrücken, Germany
| |
Collapse
|
10
|
Sun GC, Lee YJ, Lee YC, Yu HF, Wang DC. Exercise prevents the impairment of learning and memory in prenatally phthalate-exposed male rats by improving the expression of plasticity-related proteins. Behav Brain Res 2021; 413:113444. [PMID: 34245761 DOI: 10.1016/j.bbr.2021.113444] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/27/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022]
Abstract
Regular exercise has been identified to facilitate neuroplasticity that maximize functional outcome after brain injuries. Brain-derived neurotrophic factor (BDNF) has emerged as a key facilitator of neuroplasticity after exercise. The activity-regulated cytoskeleton associated protein (Arc) is induced by BDNF and N-methyl-d-aspartic acid receptor (NMDAR), contributing to functional modification of neuroplasticity in the hippocampus. Meanwhile, early-life exposure to neuroendocrine disruptor di-(2-ethylhexyl)-phthalate (DEHP) is a risk factor for behavioral deficits, but the mechanisms responsible for DEHP-induced neurotoxicity are not well understood. The purpose of this study is to investigate whether hippocampal Arc expression is impaired by DEHP exposure and to examine the protective role of exercise in the prenatally DEHP-exposed male rats. Sprague Dawley dams were fed with vehicle or DEHP during gestation. The male offspring were trained to treadmill running for 5 weeks followed by examination of behavioral and biochemical outcomes. The results showed that DEHP-exposed rats exhibited impairment of spatial learning and memory as well as down-regulations of BDNF, NMDAR, Arc, and synaptophysin. Importantly, aerobic exercise during childhood-adolescence prevented the impairment of learning and memory by recovering the expressions of BDNF, NMDAR, Arc, and synaptophysin. These findings suggest that exercise may provide beneficial effects on ameliorating the impairment of neuroplasticity in the prenatally DEHP-exposed male rats at late adolescence.
Collapse
Affiliation(s)
- Gwo-Ching Sun
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Yi-Ju Lee
- Department of Sports Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Yi-Ching Lee
- Department of Food and Beverage Services, Tainan University of Technology, Tainan, Taiwan.
| | - Hsien-Fu Yu
- Department of Sports Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Dean-Chuan Wang
- Department of Sports Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
11
|
Acuña-Hinrichsen F, Covarrubias-Pinto A, Ishizuka Y, Stolzenbach MF, Martin C, Salazar P, Castro MA, Bramham CR, Otth C. Herpes Simplex Virus Type 1 Neuronal Infection Triggers the Disassembly of Key Structural Components of Dendritic Spines. Front Cell Neurosci 2021; 15:580717. [PMID: 33708072 PMCID: PMC7940845 DOI: 10.3389/fncel.2021.580717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 02/01/2021] [Indexed: 11/13/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a widespread neurotropic virus. Primary infection of HSV-1 in facial epithelium leads to retrograde axonal transport to the central nervous system (CNS) where it establishes latency. Under stressful conditions, the virus reactivates, and new progeny are transported anterogradely to the primary site of infection. During the late stages of neuronal infection, axonal damage can occur, however, the impact of HSV-1 infection on the morphology and functional integrity of neuronal dendrites during the early stages of infection is unknown. We previously demonstrated that acute HSV-1 infection in neuronal cell lines selectively enhances Arc protein expression - a major regulator of long-term synaptic plasticity and memory consolidation, known for being a protein-interaction hub in the postsynaptic dendritic compartment. Thus, HSV-1 induced Arc expression may alter the functionality of infected neurons and negatively impact dendritic spine dynamics. In this study we demonstrated that HSV-1 infection induces structural disassembly and functional deregulation in cultured cortical neurons, an altered glutamate response, Arc accumulation within the somata, and decreased expression of spine scaffolding-like proteins such as PSD-95, Drebrin and CaMKIIβ. However, whether these alterations are specific to the HSV-1 infection mechanism or reflect a secondary neurodegenerative process remains to be determined.
Collapse
Affiliation(s)
- Francisca Acuña-Hinrichsen
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Post-graduate Program, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Adriana Covarrubias-Pinto
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Post-graduate Program, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
- Institute of Biochemistry II, Goethe University School of Medicine, Frankfurt am Main, Germany
- Institute of Biochemistry and Microbiology, Faculty of Science, Universidad Austral de Chile, Valdivia, Chile
| | - Yuta Ishizuka
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Carolina Martin
- School of Medical Technology, Austral University of Chile, Puerto Montt, Chile
| | - Paula Salazar
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Maite A. Castro
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Institute of Biochemistry and Microbiology, Faculty of Science, Universidad Austral de Chile, Valdivia, Chile
- Janelia Research Campus, HHMI, VA, United States
| | | | - Carola Otth
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
12
|
Cortical influences of serotonin and glutamate on layer V pyramidal neurons. PROGRESS IN BRAIN RESEARCH 2021; 261:341-378. [PMID: 33785135 DOI: 10.1016/bs.pbr.2020.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Layer V pyramidal neurons constitute principle output neurons of the medial prefrontal cortex (mPFC)/neocortex to subcortical regions including the intralaminar/midline thalamic nuclei, amygdala, basal ganglia, brainstem nuclei and the spinal cord. The effects of 5-hydroxytryptamine (5-HT) on layer V pyramidal cells primarily reflect a range of excitatory influences through 5-HT2A receptors and inhibitory influences through non-5-HT2A receptors, including 5-HT1A receptors. While the 5-HT2A receptor is primarily a postsynaptic receptor on throughout the apical dendritic field of 5-HT2A receptors, activation of a minority of 5-HT2A receptors also appears to increase spontaneous excitatory postsynaptic currents/potentials (EPSCs/EPSPs) via a presynaptic effect on thalamocortical terminals arising from the midline and intralaminar thalamic nuclei. Activation of 5-HT2A receptors by the phenethylamine hallucinogen also appears to increase asynchronous release of glutamate upon the layer V pyramidal dendritic field, an effect that is suppressed by 5-HT itself through non-5-HT2A receptors. Serotonergic hallucinogens acting on 5-HT2A receptors also appears to increase gene expression of immediate early genes (iEG) and other receptors appearing to induce an iEG-like response like BDNF. Psychedelic hallucinogens acting on 5-HT2A receptors also induce head twitches in rodents that appear related to induction of glutamate release. These electrophysiological, biochemical and behavioral effects of serotonergic hallucinogens appear to be related to modulating glutamatergic thalamocortical neurotransmission and/or shifting the balance toward 5-HT2A receptor activation and away from non-5-HT2A receptor activation. These 5-HT2A receptor induced responses are modulated by feedback homeostatic mechanisms through mGlu2, mGlu4, and mGlu8 presynaptic receptors on thalamocortical terminals. These 5-HT2A receptor and glutamatergic interactions also appear to play a role on higher cortical functions of the mPFC such as motoric impulsivity and antidepressant-like behavioral responses on the differential-reinforcement-of low rate 72-s (DRL 72-s schedule). These mutually opposing effects between 5-HT2A receptor and mGlu autoreceptor activation (e.g., blocking 5-HT2A receptors and enhancing activity at mGlu2 receptors) may play a clinical role with respect to currently prescribed or novel antidepressant drugs. Thus, there is an important balance between 5-HT2A receptor activation and activation of mGlu autoreceptors on prefrontal cortical layer V pyramidal cells with respect to the electrophysiological, biochemical and behavioral effects serotonergic hallucinogenic drugs.
Collapse
|
13
|
Livingstone RW, Elder MK, Barrett MC, Westlake CM, Peppercorn K, Tate WP, Abraham WC, Williams JM. Secreted Amyloid Precursor Protein-Alpha Promotes Arc Protein Synthesis in Hippocampal Neurons. Front Mol Neurosci 2019; 12:198. [PMID: 31474829 PMCID: PMC6702288 DOI: 10.3389/fnmol.2019.00198] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/30/2019] [Indexed: 12/22/2022] Open
Abstract
Secreted amyloid precursor protein-α (sAPPα) is a neuroprotective and memory-enhancing molecule, however, the mechanisms through which sAPPα promotes these effects are not well understood. Recently, we have shown that sAPPα enhances cell-surface expression of glutamate receptors. Activity-related cytoskeletal-associated protein Arc (Arg3.1) is an immediate early gene capable of modulating long-term potentiation, long-term depression and homeostatic plasticity through regulation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor localization. Accordingly, we hypothesized that sAPPα may enhance synaptic plasticity, in part, by the de novo synthesis of Arc. Using primary cortical and hippocampal neuronal cultures we found that sAPPα (1 nM, 2 h) enhances levels of Arc mRNA and protein. Arc protein levels were increased in both the neuronal somata and dendrites in a Ca2+/calmodulin-dependent protein kinase II-dependent manner. Additionally, dendritic Arc expression was dependent upon activation of mitogen-activated protein kinase and protein kinase G. The enhancement of dendritic Arc protein was significantly reduced by antagonism of N-methyl-D-aspartate (NMDA) and nicotinic acetylcholine (α7nACh) receptors, and fully eliminated by dual application of these antagonists. This effect was further corroborated in area CA1 of acute hippocampal slices. These data suggest sAPPα-regulated plasticity within hippocampal neurons is mediated by cooperation of NMDA and α7nACh receptors to engage a cascade of signal transduction molecules to enhance the transcription and translation of Arc.
Collapse
Affiliation(s)
- Rhys W Livingstone
- Department of Anatomy, Brain Health Research Centre, Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Megan K Elder
- Department of Anatomy, Brain Health Research Centre, Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Maya C Barrett
- Department of Anatomy, Brain Health Research Centre, Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Courteney M Westlake
- Department of Anatomy, Brain Health Research Centre, Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Katie Peppercorn
- Department of Biochemistry, Brain Health Research Centre, Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Warren P Tate
- Department of Biochemistry, Brain Health Research Centre, Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Wickliffe C Abraham
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Joanna M Williams
- Department of Anatomy, Brain Health Research Centre, Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| |
Collapse
|
14
|
Abd-Elrahman KS, Ferguson SSG. Modulation of mTOR and CREB pathways following mGluR5 blockade contribute to improved Huntington's pathology in zQ175 mice. Mol Brain 2019; 12:35. [PMID: 30961637 PMCID: PMC6454676 DOI: 10.1186/s13041-019-0456-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 03/29/2019] [Indexed: 01/07/2023] Open
Abstract
Huntington’s disease (HD) is a neurodegenerative disorder caused by a genetic abnormality in the huntingtin gene that leads to a polyglutamine repeat expansion of the huntingtin protein. The cleaved polyglutamine expansion of mutant huntingtin (mHTT) protein can form aggregates strongly correlated with HD progression. We have previously shown that the inhibition of mGluR5 using CTEP, a selective negative allosteric mGluR5 modulator, can delay disease progression and reduce in mHTT aggregates in the zQ175 mouse model of HD. This was paralleled by enhanced catalytic activity of Unc-51-like kinase 1 (ULK1), a kinase modulated by mammalian target of rapamycin (mTOR) and key regulator of autophagy initiation. In the present study, we show that CTEP can correct aberrant phosphoinositide 3-kinase (PI3K)/Akt/mTOR signaling detected in zQ175 mice that may underlie the enhanced ULK1 activity and activation of autophagy. We also show that CTEP can facilitate cAMP response element-binding protein (CREB)-mediated expression of brain-derived neurotrophic factor (BDNF) to foster neuronal survival and reduce apoptosis. Taken together, our findings provide the molecular evidence for how targeting mGluR5 using a well-tolerated selective NAM can mitigate two critical mechanisms of neurodegeneration, autophagy and apoptosis.
Collapse
Affiliation(s)
- Khaled S Abd-Elrahman
- University of Ottawa Brain and Mind Institute, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, K1H 8M5, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, K1H 8M5, Canada.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Stephen S G Ferguson
- University of Ottawa Brain and Mind Institute, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, K1H 8M5, Canada. .,Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, K1H 8M5, Canada.
| |
Collapse
|
15
|
Thomas TC, Stockhausen EM, Law LM, Khodadad A, Lifshitz J. Rehabilitation modality and onset differentially influence whisker sensory hypersensitivity after diffuse traumatic brain injury in the rat. Restor Neurol Neurosci 2018; 35:611-629. [PMID: 29036852 DOI: 10.3233/rnn-170753] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND As rehabilitation strategies advance as therapeutic interventions, the modality and onset of rehabilitation after traumatic brain injury (TBI) are critical to optimize treatment. Our laboratory has detected and characterized a late-onset, long-lasting sensory hypersensitivity to whisker stimulation in diffuse brain-injured rats; a deficit that is comparable to visual or auditory sensory hypersensitivity in humans with an acquired brain injury. OBJECTIVE We hypothesize that the modality and onset of rehabilitation therapies will differentially influence sensory hypersensitivity in response to the Whisker Nuisance Task (WNT) as well as WNT-induced corticosterone (CORT) stress response in diffuse brain-injured rats and shams. METHODS After midline fluid percussion brain injury (FPI) or sham surgery, rats were assigned to one of four rehabilitative interventions: (1) whisker sensory deprivation during week one or (2) week two or (3) whisker stimulation during week one or (4) week two. At 28 days following FPI and sham procedures, sensory hypersensitivity was assessed using the WNT. Plasma CORT was evaluated immediately following the WNT (aggravated levels) and prior to the pre-determined endpoint 24 hours later (non-aggravated levels). RESULTS Deprivation therapy during week two elicited significantly greater sensory hypersensitivity to the WNT compared to week one (p < 0.05), and aggravated CORT levels in FPI rats were significantly lower than sham levels. Stimulation therapy during week one resulted in low levels of sensory hypersensitivity to the WNT, similar to deprivation therapy and naïve controls, however, non-aggravated CORT levels in FPI rats were significantly higher than sham. CONCLUSION These data indicate that modality and onset of sensory rehabilitation can differentially influence FPI and sham rats, having a lasting impact on behavioral and stress responses to the WNT, emphasizing the necessity for continued evaluation of modality and onset of rehabilitation after TBI.
Collapse
Affiliation(s)
- Theresa Currier Thomas
- Barrow Neurological Institute at Phoenix Children's Hospital - Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, AZ, USA.,Phoenix VA Healthcare System - Phoenix, AZ, USA.,Spinal Cord and Brain Injury Research Center, University of Kentucky Chandler Medical Center - Lexington, KY, USA
| | - Ellen Magee Stockhausen
- Core Medical Group, Manchester, NH, USA.,Spinal Cord and Brain Injury Research Center, University of Kentucky Chandler Medical Center - Lexington, KY, USA
| | - L Matthew Law
- Barrow Neurological Institute at Phoenix Children's Hospital - Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, AZ, USA
| | - Aida Khodadad
- Barrow Neurological Institute at Phoenix Children's Hospital - Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, AZ, USA
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital - Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, AZ, USA.,Phoenix VA Healthcare System - Phoenix, AZ, USA.,Neuroscience Program, Arizona State University - Tempe, AZ, USA.,Spinal Cord and Brain Injury Research Center, University of Kentucky Chandler Medical Center - Lexington, KY, USA
| |
Collapse
|
16
|
Paolantoni C, Ricciardi S, De Paolis V, Okenwa C, Catalanotto C, Ciotti MT, Cattaneo A, Cogoni C, Giorgi C. Arc 3' UTR Splicing Leads to Dual and Antagonistic Effects in Fine-Tuning Arc Expression Upon BDNF Signaling. Front Mol Neurosci 2018; 11:145. [PMID: 29755318 PMCID: PMC5934489 DOI: 10.3389/fnmol.2018.00145] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/10/2018] [Indexed: 01/02/2023] Open
Abstract
Activity-regulated cytoskeletal associated protein (Arc) is an immediate-early gene critically involved in synaptic plasticity and memory consolidation. Arc mRNA is rapidly induced by synaptic activation and a portion is locally translated in dendrites where it modulates synaptic strength. Being an activity-dependent effector of homeostatic balance, regulation of Arc is uniquely tuned to result in short-lived bursts of expression. Cis-Acting elements that control its transitory expression post-transcriptionally reside primarily in Arc mRNA 3′ UTR. These include two conserved introns which distinctively modulate Arc mRNA stability by targeting it for destruction via the nonsense mediated decay pathway. Here, we further investigated how splicing of the Arc mRNA 3′ UTR region contributes to modulate Arc expression in cultured neurons. Unexpectedly, upon induction with brain derived neurotrophic factor, translational efficiency of a luciferase reporter construct harboring Arc 3′ UTR is significantly upregulated and this effect is dependent on splicing of Arc introns. We find that, eIF2α dephosphorylation, mTOR, ERK, PKC, and PKA activity are key to this process. Additionally, CREB-dependent transcription is required to couple Arc 3′ UTR-splicing to its translational upregulation, suggesting the involvement of de novo transcribed trans-acting factors. Overall, splicing of Arc 3′ UTR exerts a dual and unique effect in fine-tuning Arc expression upon synaptic signaling: while inducing mRNA decay to limit the time window of Arc expression, it also elicits translation of the decaying mRNA. This antagonistic effect likely contributes to the achievement of a confined yet efficient burst of Arc protein expression, facilitating its role as an effector of synapse-specific plasticity.
Collapse
Affiliation(s)
- Chiara Paolantoni
- European Brain Research Institute Rita Levi-Montalcini Rome, Rome, Italy.,Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Simona Ricciardi
- European Brain Research Institute Rita Levi-Montalcini Rome, Rome, Italy.,Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
| | - Veronica De Paolis
- European Brain Research Institute Rita Levi-Montalcini Rome, Rome, Italy.,Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Chinenye Okenwa
- European Brain Research Institute Rita Levi-Montalcini Rome, Rome, Italy
| | - Caterina Catalanotto
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Maria T Ciotti
- Institute of Cell Biology and Neurobiology, National Research Council, Rome, Italy
| | - Antonino Cattaneo
- European Brain Research Institute Rita Levi-Montalcini Rome, Rome, Italy.,Bio@SNS Laboratory, Scuola Normale Superiore, Pisa, Italy
| | - Carlo Cogoni
- Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy
| | - Corinna Giorgi
- European Brain Research Institute Rita Levi-Montalcini Rome, Rome, Italy
| |
Collapse
|
17
|
Hunter CJ, Remenyi J, Correa SA, Privitera L, Reyskens KMSE, Martin KJ, Toth R, Frenguelli BG, Arthur JSC. MSK1 regulates transcriptional induction of Arc/Arg3.1 in response to neurotrophins. FEBS Open Bio 2017; 7:821-834. [PMID: 28593137 PMCID: PMC5458472 DOI: 10.1002/2211-5463.12232] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/23/2017] [Accepted: 03/31/2017] [Indexed: 12/29/2022] Open
Abstract
The immediate early gene activity‐regulated cytoskeletal protein (Arc)/Arg3.1 and the neurotrophin brain‐derived neurotrophic factor (BDNF) play important roles in synaptic plasticity and learning and memory in the mammalian brain. However, the mechanisms by which BDNF regulates the expression of Arc/Arg3.1 are unclear. In this study, we show that BDNF acts via the ERK1/2 pathway to activate the nuclear kinase mitogen‐ and stress‐activated protein kinase 1 (MSK1). MSK1 then induces Arc/Arg3.1 expression via the phosphorylation of histone H3 at the Arc/Arg3.1 promoter. MSK1 can also phosphorylate the transcription factor cyclic‐AMP response element‐binding protein (CREB) on Ser133. However, this is not required for BDNF‐induced Arc.Arg3.1 transcription as a Ser133Ala knockin mutation had no effect on Arc/Arg3.1 induction. In parallel, ERK1/2 directly activates Arc/Arg3.1 mRNA transcription via at least one serum response element on the promoter, which bind a complex of the Serum Response Factor (SRF) and a Ternary Complex Factor (TCF).
Collapse
Affiliation(s)
- Chris J Hunter
- MRC Protein Phosphorylation Unit College of Life Sciences Sir James Black Centre University of Dundee UK
| | - Judit Remenyi
- Wellcome Trust Centre for Gene Regulation and Expression Wellcome Trust Building College of Life Sciences University of Dundee UK
| | - Sonia A Correa
- Bradford School of Pharmacy Faculty of Life Sciences University of Bradford UK
| | | | - Kathleen M S E Reyskens
- Division of Cell Signalling and Immunology Wellcome Trust Building College of Life Sciences University of Dundee UK
| | - Kirsty J Martin
- MRC Protein Phosphorylation Unit College of Life Sciences Sir James Black Centre University of Dundee UK
| | - Rachel Toth
- MRC Protein Phosphorylation Unit College of Life Sciences Sir James Black Centre University of Dundee UK
| | | | - J Simon C Arthur
- Division of Cell Signalling and Immunology Wellcome Trust Building College of Life Sciences University of Dundee UK
| |
Collapse
|
18
|
Bacopa monniera (CDRI-08) Upregulates the Expression of Neuronal and Glial Plasticity Markers in the Brain of Scopolamine Induced Amnesic Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:837012. [PMID: 26413129 PMCID: PMC4564643 DOI: 10.1155/2015/837012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/20/2015] [Indexed: 12/18/2022]
Abstract
Preclinical studies on animal models have discerned the antiamnesic and memory-enhancing potential of Bacopa monniera (Brahmi) crude extract and standardized extracts. These studies primarily focus on behavioral consequences. However, lack of information on molecular underpinnings has limited the clinical trials of the potent herb in human subjects. In recent years, researchers highlight plasticity markers as molecular correlates of amnesia and being crucial to design therapeutic targets. In the present report, we have investigated the effect of a special extract of B. monniera (CDRI-08) on the expression of key neuronal (BDNF and Arc) and glial (GFAP) plasticity markers in the cerebrum of scopolamine induced amnesic mice. Pre- and postadministration of CDRI-08 ameliorated amnesic effect of scopolamine by decreasing acetyl cholinesterase activity and drastically upregulating the mRNA and protein expression of BDNF, Arc, and GFAP in mouse cerebrum. Interestingly, the plant extract per se elevated BDNF and Arc expression as compared to control but GFAP was unaltered. In conclusion, our findings provide the first molecular evidence for antiamnesic potential of CDRI-08 via enhancement of both neuronal and glial plasticity markers. Further investigations on detailed molecular pathways would encourage therapeutic application of the extract in memory disorders.
Collapse
|
19
|
Lie MEK, Overgaard A, Mikkelsen JD. Effect of a postnatal high-fat diet exposure on puberty onset, estrous cycle regularity, and kisspeptin expression in female rats. Reprod Biol 2013; 13:298-308. [PMID: 24287038 DOI: 10.1016/j.repbio.2013.08.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 08/19/2013] [Accepted: 08/31/2013] [Indexed: 10/26/2022]
Abstract
Kisspeptin, encoded by Kiss1, plays a key role in pubertal maturation and reproduction as a positive upstream regulator of the hypothalamic-pituitary-gonadal (HPG) axis. To examine the role of high-fat diet (HFD) on puberty onset, estrous cycle regularity, and kisspeptin expression, female rats were exposed to HFD in distinct postnatal periods. Three groups of rats were exposed to HFD containing 60% energy from fat during the pre-weaning period (postnatal day (PND) 1-16, HFD PND 1-16), post-weaning period (HFD PND 21-34), or during both periods (HFD PND 1-34). Puberty onset, evaluated by vaginal opening, was monitored on days 30-34. Leptin, estradiol (E2), Kiss1 mRNA levels, and number of kisspeptin-immunoreactive cells in the anteroventral periventricular nucleus (AVPV) and arcuate nucleus (ARC) were measured at day 34. Body weight increased only in rats exposed to HFD during post-weaning period, whereas the timing of vaginal opening was unaffected in all three groups. Leptin, Kiss1 mRNA levels, and number of kisspeptin-immunoreactive cells at day 34 were not affected by HFD. Additionally, the estrous cycle regularity was monitored in rats exposed to HFD for 40 days from weaning. Leptin, E2, and Kiss1 mRNA levels in the AVPV and ARC were measured after the HFD exposure. Thirty-three percent of rats exposed to HFD exhibited irregular estrous cycles and a two-fold increase in leptin. By contrast, E2 level and Kiss1 mRNA levels were not affected by the treatment. These data show that postnatal HFD exposure induced irregular estrous cycles, but had no effect on puberty onset or kisspeptin.
Collapse
Affiliation(s)
- Maria E K Lie
- Neurobiology Research Unit, University Hospital Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Ø, Denmark
| | | | | |
Collapse
|
20
|
Jørgensen CV, Klein AB, EL-Sayed M, Knudsen GM, Mikkelsen JD. Metabotropic glutamate receptor 2 and corticotrophin-releasing factor receptor-1 gene expression is differently regulated by BDNF in rat primary cortical neurons. Synapse 2013; 67:794-800. [DOI: 10.1002/syn.21689] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 06/03/2013] [Indexed: 01/19/2023]
Affiliation(s)
| | - Anders B. Klein
- Neurobiology Research Unit, University Hospital Rigshospitalet; Copenhagen Denmark
| | - Mona EL-Sayed
- Neurobiology Research Unit, University Hospital Rigshospitalet; Copenhagen Denmark
| | - Gitte M. Knudsen
- Neurobiology Research Unit, University Hospital Rigshospitalet; Copenhagen Denmark
| | - Jens D. Mikkelsen
- Neurobiology Research Unit, University Hospital Rigshospitalet; Copenhagen Denmark
| |
Collapse
|
21
|
Induction of the plasticity-associated immediate early gene Arc by stress and hallucinogens: role of brain-derived neurotrophic factor. Int J Neuropsychopharmacol 2013; 16:405-15. [PMID: 22404904 DOI: 10.1017/s1461145712000168] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Exposure to stress and hallucinogens in adulthood evokes persistent alterations in neurocircuitry and emotional behaviour. The structural and functional changes induced by stress and hallucinogen exposure are thought to involve transcriptional alterations in specific effector immediate early genes. The immediate early gene, activity regulated cytoskeletal-associated protein (Arc), is important for both activity and experience dependent plasticity. We sought to examine whether trophic factor signalling through brain-derived neurotrophic factor (BDNF) contributes to the neocortical regulation of Arc mRNA in response to distinct stimuli such as immobilization stress and the hallucinogen 2,5-dimethoxy-4-iodoamphetamine (DOI). Acute exposure to either immobilization stress or DOI induced Arc mRNA levels within the neocortex. BDNF infusion into the neocortex led to a robust up-regulation of local Arc transcript expression. Further, baseline Arc mRNA expression in the neocortex was significantly decreased in inducible BDNF knockout mice with an adult-onset, forebrain specific BDNF loss. The induction of Arc mRNA levels in response to both acute immobilization stress or a single administration of DOI was significantly attenuated in the inducible BDNF knockout mice. Taken together, our results implicate trophic factor signalling through BDNF in the regulation of cortical Arc mRNA expression, both under baseline conditions and following stress and hallucinogen exposure. These findings suggest the possibility that the regulation of Arc expression via BDNF provides a molecular substrate for the structural and synaptic plasticity observed following stimuli such as stress and hallucinogens.
Collapse
|
22
|
Ramirez-Amaya V, Angulo-Perkins A, Chawla MK, Barnes CA, Rosi S. Sustained transcription of the immediate early gene Arc in the dentate gyrus after spatial exploration. J Neurosci 2013; 33:1631-9. [PMID: 23345235 PMCID: PMC6618719 DOI: 10.1523/jneurosci.2916-12.2013] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 11/12/2012] [Accepted: 12/01/2012] [Indexed: 11/21/2022] Open
Abstract
After spatial exploration in rats, Arc mRNA is expressed in ∼2% of dentate gyrus (DG) granule cells, and this proportion of Arc-positive neurons remains stable for ∼8 h. This long-term presence of Arc mRNA following behavior is not observed in hippocampal CA1 pyramidal cells. We report here that in rats ∼50% of granule cells with cytoplasmic Arc mRNA, induced some hours previously during exploration, also show Arc expression in the nucleus. This suggests that recent transcription can occur long after the exploration behavior that elicited it. To confirm that the delayed nuclear Arc expression was indeed recent transcription, Actinomycin D was administered immediately after exploration. This treatment resulted in inhibition of recent Arc expression both when evaluated shortly after exploratory behavior as well as after longer time intervals. Together, these data demonstrate a unique kinetic profile for Arc transcription in hippocampal granule neurons following behavior that is not observed in other cell types. Among a number of possibilities, this sustained transcription may provide a mechanism that ensures that the synaptic connection weights in the sparse population of granule cells recruited during a given behavioral event are able to be modified.
Collapse
Affiliation(s)
- Victor Ramirez-Amaya
- Redes Neuronales Plásticas Laboratory, Department of Neurobiología Conductual y Cognitiva Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México.
| | | | | | | | | |
Collapse
|
23
|
Chu YF, Yen CT, Lee LJ. Neonatal whisker clipping alters behavior, neuronal structure and neural activity in adult rats. Behav Brain Res 2012; 238:124-33. [PMID: 23098795 DOI: 10.1016/j.bbr.2012.10.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 10/09/2012] [Accepted: 10/13/2012] [Indexed: 12/31/2022]
Abstract
Early experience plays critical roles during the development of sensory systems. For example, neonatal surgical manipulations of the whiskers in rodents lead to altered neural activity and behaviors later in life. However, while surgical procedures damage the sensory pathway; it is hard to examine the impact of whisker deprivation on adult animals. To address this issue, we performed a neonatal whisker clipping (WC0-3) paradigm, a non-invasive procedure, from the day of birth (P0) to postnatal day (P) 3, and examined behavioral performances in their adult age. With fully regrown whiskers, the WC0-3 rats exhibited shorter crossable distance than controls in a gap-crossing task, suggesting a defect in their whisker-specific tactile function. In their somatosensory cortex, the layer IV spiny stellate neurons had reduced dendritic complexity and spine density. After exploration in a novel environment, the expression of an activity-dependent immediate early gene, c-fos, increased dramatically in the somatosensory cortex. However, in WC0-3 rats, the number of c-Fos positive cells was less than those in control rats, indicating a fault in transducing sensory-related neural activity between cortical layers in WC0-3 rats. Together, our results demonstrate the roles of early tactile experience on the establishment of layer-specific excitatory connection in the barrel cortex. Early sensory insufficiency would leave long-lasting functional deficits in the sensory system.
Collapse
Affiliation(s)
- Yu-Fei Chu
- Graduate Institute of Zoology, National Taiwan University, Taipei, Taiwan
| | | | | |
Collapse
|
24
|
Jenks BG, Kuribara M, Kidane AH, Kramer BMR, Roubos EW, Scheenen WJJM. The role of brain-derived neurotrophic factor in the regulation of cell growth and gene expression in melanotrope cells of Xenopus laevis. Gen Comp Endocrinol 2012; 177:315-21. [PMID: 22248443 DOI: 10.1016/j.ygcen.2012.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Revised: 01/01/2012] [Accepted: 01/02/2012] [Indexed: 12/13/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is, despite its name, also found outside the central nervous system (CNS), but the functional significance of this observation is largely unknown. This review concerns the expression of BDNF in the pituitary gland. While the presence of the neurotrophin in the mammalian pituitary gland is well documented its functional significance remains obscure. Studies on the pars intermedia of the pituitary of the amphibian Xenopus laevis have shown that BDNF is produced by the neuroendocrine melanotrope cells, its expression is physiologically regulated, and the melanotrope cells themselves express receptors for the neurotrophin. The neurotrophin has been shown to act as an autocrine factor on the melanotrope to promote cell growth and regulate gene expression. In doing so BDNF supports the physiological function of the cell to produce and release α-melanophore-stimulating hormone for the purpose of adjusting the animal's skin color to that of its background.
Collapse
Affiliation(s)
- Bruce G Jenks
- Department of Cellular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
25
|
Effect of brain-derived neurotrophic factor on behavior and key members of the brain serotonin system in genetically predisposed to behavioral disorders mouse strains. Neuroscience 2012; 214:59-67. [DOI: 10.1016/j.neuroscience.2012.04.031] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 04/04/2012] [Accepted: 04/07/2012] [Indexed: 11/20/2022]
|