1
|
Nguyen TVV, Crumpacker RH, Calderon KE, Garcia FG, Zbesko JC, Frye JB, Gonzalez S, Becktel DA, Yang T, Tavera-Garcia MA, Morrison HW, Schnellmann RG, Longo FM, Doyle KP. Post-Stroke Administration of the p75 Neurotrophin Receptor Modulator, LM11A-31, Attenuates Chronic Changes in Brain Metabolism, Increases Neurotransmitter Levels, and Improves Recovery. J Pharmacol Exp Ther 2022; 380:126-141. [PMID: 34893553 PMCID: PMC11048261 DOI: 10.1124/jpet.121.000711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 11/29/2021] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to test whether poststroke oral administration of a small molecule p75 neurotrophin receptor (p75NTR) modulator (LM11A-31) can augment neuronal survival and improve recovery in a mouse model of stroke. Mice were administered LM11A-31 for up to 12 weeks, beginning 1 week after stroke. Metabolomic analysis revealed that after 2 weeks of daily treatment, mice that received LM11A-31 were distinct from vehicle-treated mice by principal component analysis and had higher levels of serotonin, acetylcholine, and dopamine in their ipsilateral hemisphere. LM11A-31 treatment also improved redox homeostasis by restoring reduced glutathione. It also offset a stroke-induced reduction in glycolysis by increasing acetyl-CoA. There was no effect on cytokine levels in the infarct. At 13 weeks after stroke, adaptive immune cell infiltration in the infarct was unchanged in LM11A-31-treated mice, indicating that LM11A-31 does not alter the chronic inflammatory response to stroke at the site of the infarct. However, LM11A-31-treated mice had less brain atrophy, neurodegeneration, tau pathology, and microglial activation in other regions of the ipsilateral hemisphere. These findings correlated with improved recovery of motor function on a ladder test, improved sensorimotor and cognitive abilities on a nest construction test, and less impulsivity in an open field test. These data support small molecule modulation of the p75NTR for preserving neuronal health and function during stroke recovery. SIGNIFICANCE STATEMENT: The findings from this study introduce the p75 neurotrophin receptor as a novel small molecule target for promotion of stroke recovery. Given that LM11A-31 is in clinical trials as a potential therapy for Alzheimer's disease, it could be considered as a candidate for assessment in stroke or vascular dementia studies.
Collapse
Affiliation(s)
- Thuy-Vi V Nguyen
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Rachel H Crumpacker
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Kylie E Calderon
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Frankie G Garcia
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Jacob C Zbesko
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Jennifer B Frye
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Selena Gonzalez
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Danielle A Becktel
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Tao Yang
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Marco A Tavera-Garcia
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Helena W Morrison
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Rick G Schnellmann
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Frank M Longo
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| | - Kristian P Doyle
- Department of Immunobiology (T.-V.V.N., K.P.D., R.H.C., K.E.C., F.G.G., J.C.Z., J.B.F., D.A.B., M.A.T.-G.), Department of Neurology (T.-V.V.N., K.P.D., S.G.), College of Nursing (H.W.M.), Department of Pharmacology and Toxicology (R.G.S.), and Arizona Center on Aging (K.P.D.), University of Arizona, Tucson, Arizona; Department of Neurology and Neurologic Sciences, Stanford University, Stanford, California (T.Y., F.M.L.); and Southern Arizona Department of Veterans Affairs Health Care System, Tucson, Arizona (R.G.S.)
| |
Collapse
|
2
|
Selvam R, Yeh ML, Levine ES. Endogenous cannabinoids mediate the effect of BDNF at CA1 inhibitory synapses in the hippocampus. Synapse 2018; 73:e22075. [PMID: 30334291 PMCID: PMC6470051 DOI: 10.1002/syn.22075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/20/2018] [Accepted: 10/12/2018] [Indexed: 12/14/2022]
Abstract
Brain-derived neurotrophic factor (BDNF), traditionally known for promoting neuronal growth and development, is also a modulator of synaptic transmission. In addition to the well-characterized effects at excitatory synapses, BDNF has been shown to acutely suppress inhibitory neurotransmission; however, the underlying mechanisms are unclear. We have previously shown that at inhibitory synapses in layer 2/3 of the somatosensory cortex, BDNF induces the mobilization of endogenous cannabinoids (eCBs) that act retrogradely to suppress GABA release. Here, we hypothesized that in the hippocampus, BDNF acts similarly via eCB signaling to suppress GABAergic transmission. We found that the acute application of BDNF reduced the spontaneous inhibitory postsynaptic currents (sIPSCs) via postsynaptic TrkB receptor activation. The suppressive effects of BDNF required eCB signaling, as this effect on sIPSCs was prevented by a CB1 receptor antagonist. Further, blocking the postsynaptic eCB release prevented the effect of BDNF, whereas eCB reuptake inhibition enhanced the effect of BDNF. These results suggest that BDNF triggers the postsynaptic release of eCBs. To identify the specific eCB release by BDNF, we tested the effects of disrupting the synthesis or degradation of 2-arachidonoylcglycerol (2-AG). Blocking 2-AG synthesis prevented the effect of BDNF and blocking 2-AG degradation enhanced the effect of BDNF. However, there was no change in the effect of BDNF when anandamide degradation was blocked. Collectively, these results suggest that in the hippocampus, BDNF-TrkB signaling induces the postsynaptic release of the endogenous cannabinoid 2-AG, which acts retrogradely on the presynaptic CB1 receptors to suppress GABA release.
Collapse
Affiliation(s)
- Rajamani Selvam
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Mason L Yeh
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Eric S Levine
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut
| |
Collapse
|
3
|
Simmons DA. Modulating Neurotrophin Receptor Signaling as a Therapeutic Strategy for Huntington's Disease. J Huntingtons Dis 2018; 6:303-325. [PMID: 29254102 PMCID: PMC5757655 DOI: 10.3233/jhd-170275] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Huntington’s disease (HD) is an autosomal dominant neurodegenerative disorder caused by CAG repeat expansions in the IT15 gene which encodes the huntingtin (HTT) protein. Currently, no treatments capable of preventing or slowing disease progression exist. Disease modifying therapeutics for HD would be expected to target a comprehensive set of degenerative processes given the diverse mechanisms contributing to HD pathogenesis including neuroinflammation, excitotoxicity, and transcription dysregulation. A major contributor to HD-related degeneration is mutant HTT-induced loss of neurotrophic support. Thus, neurotrophin (NT) receptors have emerged as therapeutic targets in HD. The considerable overlap between NT signaling networks and those dysregulated by mutant HTT provides strong theoretical support for this approach. This review will focus on the contributions of disrupted NT signaling in HD-related neurodegeneration and how targeting NT receptors to augment pro-survival signaling and/or to inhibit degenerative signaling may combat HD pathologies. Therapeutic strategies involving NT delivery, peptidomimetics, and the targeting of specific NT receptors (e.g., Trks or p75NTR), particularly with small molecule ligands, are discussed.
Collapse
Affiliation(s)
- Danielle A Simmons
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
4
|
Perez-Rando M, Castillo-Gomez E, Bueno-Fernandez C, Nacher J. The TrkB agonist 7,8-dihydroxyflavone changes the structural dynamics of neocortical pyramidal neurons and improves object recognition in mice. Brain Struct Funct 2018; 223:2393-2408. [PMID: 29500536 DOI: 10.1007/s00429-018-1637-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/20/2018] [Indexed: 01/17/2023]
Abstract
BDNF and its receptor TrkB have important roles in neurodevelopment, neural plasticity, learning, and memory. Alterations in TrkB expression have been described in different CNS disorders. Therefore, drugs interacting with TrkB, specially agonists, are promising therapeutic tools. Among them, the recently described 7,8-dihydroxyflavone (DHF), an orally bioactive compound, has been successfully tested in animal models of these diseases. Recent studies have shown the influence of this drug on the structure of pyramidal neurons, specifically on dendritic spine density. However, there is no information yet on how DHF may alter the structural dynamics of these neurons (i.e., real-time study of the addition/elimination of dendritic spines and axonal boutons). To gain knowledge on these effects of DHF, we have performed a real-time analysis of spine and axonal dynamics in pyramidal neurons of barrel cortex, using cranial windows and 2-photon microscopy during a chronic oral treatment with this drug. After confirming TrkB expression in these neurons, we found that DHF increased the gain rates of spines and axonal boutons, as well as improved object recognition memory. These results help to understand how the activation of the BDNF-TrkB system can improve basic behavioral tasks through changes in the structural dynamics of pyramidal neurons. Moreover, they highlight DHF as a promising therapeutic vector for certain brain disorders in which this system is altered.
Collapse
Affiliation(s)
- Marta Perez-Rando
- Neurobiology Unit, Cell Biology Department, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Dr. Moliner, 50, Burjassot, 46100, Spain
| | - Esther Castillo-Gomez
- Neurobiology Unit, Cell Biology Department, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Dr. Moliner, 50, Burjassot, 46100, Spain. .,CIBERSAM: Spanish National Network for Research in Mental Health, Valencia, Spain. .,Department of Medicine, School of Medical Sciences, Universitat Jaume I, Vicente Sos Banyat s/n, 12071, Castellón de la Plana, Spain.
| | - Clara Bueno-Fernandez
- Neurobiology Unit, Cell Biology Department, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Dr. Moliner, 50, Burjassot, 46100, Spain
| | - Juan Nacher
- Neurobiology Unit, Cell Biology Department, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Dr. Moliner, 50, Burjassot, 46100, Spain. .,CIBERSAM: Spanish National Network for Research in Mental Health, Valencia, Spain. .,Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain.
| |
Collapse
|
5
|
Begum MR, Sng JCG. Molecular mechanisms of experience-dependent maturation in cortical GABAergic inhibition. J Neurochem 2017; 142:649-661. [PMID: 28628196 PMCID: PMC5599941 DOI: 10.1111/jnc.14103] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/06/2017] [Accepted: 06/09/2017] [Indexed: 12/31/2022]
Abstract
Critical periods (CP) in early post-natal life are periods of plasticity during which the neuronal circuitry is most receptive to environmental stimuli. These early experiences translate to a more permanent and sophisticated neuronal connection in the adult brain systems. Multiple studies have pointed to the development of inhibitory circuitry as one of the central factors for the onset of critical periods. We discuss several molecular mechanisms regulating inhibitory circuit maturation and CP, from gene transcription level to protein signaling level. Also, beyond the level of gene sequences, we briefly consider recent information on dynamic epigenetic regulation of gene expression through histone methylation and acetylation and their implication on timed development of the inhibitory circuitry for the onset of CP.
Collapse
Affiliation(s)
- M. Ridzwana Begum
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Judy C. G. Sng
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| |
Collapse
|
6
|
Yeh ML, Selvam R, Levine ES. BDNF-induced endocannabinoid release modulates neocortical glutamatergic neurotransmission. Synapse 2017; 71. [PMID: 28164368 DOI: 10.1002/syn.21962] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/23/2017] [Accepted: 02/01/2017] [Indexed: 01/07/2023]
Abstract
Endocannabinoids (eCBs) and neurotrophins, particularly brain-derived neurotrophic factor (BDNF), are potent neuromodulators found throughout the mammalian neocortex. Both eCBs and BDNF play critical roles in many behavioral and neurophysiological processes and are targets for the development of novel therapeutics. The effects of eCBs and BDNF are primarily mediated by the type 1 cannabinoid (CB1) receptor and the trkB tyrosine kinase receptor, respectively. Our laboratory and others have previously established that BDNF potentiates excitatory transmission by enhancing presynaptic glutamate release and modulating NMDA receptors. In contrast, we have shown that BDNF attenuates inhibitory transmission by inducing postsynaptic release of eCBs that act retrogradely to suppress GABA release in layer 2/3 of somatosensory cortex. Here, we hypothesized that BDNF also induces release of eCBs at excitatory synapses, which could have a mitigating or opposing effect on the direct presynaptic effects of BDNF. We found the highest levels of expression of CB1 and trkB and receptors in layers 2/3 and 5. Surprisingly, BDNF did not increase the frequency of spontaneous miniature excitatory postsynaptic currents (mEPSCs) onto layer 5 pyramidal neurons in somatosensory cortex, in contrast to its effects in the hippocampus and visual cortex. However, the effect of BDNF on mEPSC frequency in somatosensory cortex was unmasked by blocking CB1 receptors or disrupting eCB release. Thus, BDNF-trKB signaling regulates glutamate release in the somatosensory cortex via opposing effects, a direct presynaptic enhancement of release probability, and simultaneous postsynaptically-induced eCB release that decreases release probability via presynaptic CB1 receptors.
Collapse
Affiliation(s)
- Mason L Yeh
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT, 06030
| | - Rajamani Selvam
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT, 06030
| | - Eric S Levine
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Ave, Farmington, CT, 06030
| |
Collapse
|
7
|
Pereira PA, Millner T, Vilela M, Sousa S, Cardoso A, Madeira MD. Nerve growth factor-induced plasticity in medial prefrontal cortex interneurons of aged Wistar rats. Exp Gerontol 2016; 85:59-70. [DOI: 10.1016/j.exger.2016.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/04/2016] [Accepted: 09/20/2016] [Indexed: 01/03/2023]
|
8
|
Janssens J, Lu D, Ni B, Chadwick W, Siddiqui S, Azmi A, Etienne H, Jushaj A, van Gastel J, Martin B, Maudsley S. Development of Precision Small-Molecule Proneurotrophic Therapies for Neurodegenerative Diseases. VITAMINS AND HORMONES 2016; 104:263-311. [PMID: 28215298 DOI: 10.1016/bs.vh.2016.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Age-related neurodegenerative diseases, such as Alzheimer's disease, will represent one of the largest future burdens on worldwide healthcare systems due to the increasing proportion of elderly in our society. As deficiencies in neurotrophins are implicated in the pathogenesis of many age-related neurodegenerative disorders, it is reasonable to consider that global neurotrophin resistance may also become a major healthcare threat. Central nervous system networks are effectively maintained through aging by neuroprotective and neuroplasticity signaling mechanisms which are predominantly controlled by neurotrophin receptor signaling. Neurotrophin receptors are single pass receptor tyrosine kinases that form dimeric structures upon ligand binding to initiate cellular signaling events that control many protective and plasticity-related pathways. Declining functionality of the neurotrophin ligand-receptor system is considered one of the hallmarks of neuropathological aging. Therefore, it is imperative to develop effective therapeutic strategies to contend with this significant issue. While the therapeutic applications of cognate ligands for neurotrophin receptors are limited, the development of nonpeptidergic, small-molecule ligands can overcome these limitations, and productively regulate this important receptor system with beneficial effects. Using our advanced knowledge of the high-dimensionality complexity of receptor systems, the future generation of precision medicines targeting these systems will be an attainable goal.
Collapse
Affiliation(s)
- J Janssens
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - D Lu
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - B Ni
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - W Chadwick
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - S Siddiqui
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - A Azmi
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - H Etienne
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - A Jushaj
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - J van Gastel
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium
| | - B Martin
- Metabolism Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States
| | - S Maudsley
- Translational Neurobiology Group, University of Antwerp, Antwerpen, Belgium; Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore MD United States.
| |
Collapse
|
9
|
Nguyen TVV, Shen L, Vander Griend L, Quach LN, Belichenko NP, Saw N, Yang T, Shamloo M, Wyss-Coray T, Massa SM, Longo FM. Small molecule p75NTR ligands reduce pathological phosphorylation and misfolding of tau, inflammatory changes, cholinergic degeneration, and cognitive deficits in AβPP(L/S) transgenic mice. J Alzheimers Dis 2015; 42:459-83. [PMID: 24898660 DOI: 10.3233/jad-140036] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The p75 neurotrophin receptor (p75NTR) is involved in degenerative mechanisms related to Alzheimer's disease (AD). In addition, p75NTR levels are increased in AD and the receptor is expressed by neurons that are particularly vulnerable in the disease. Therefore, modulating p75NTR function may be a significant disease-modifying treatment approach. Prior studies indicated that the non-peptide, small molecule p75NTR ligands LM11A-31, and chemically unrelated LM11A-24, could block amyloid-β-induced deleterious signaling and neurodegeneration in vitro, and LM11A-31 was found to mitigate neuritic degeneration and behavioral deficits in a mouse model of AD. In this study, we determined whether these in vivo findings represent class effects of p75NTR ligands by examining LM11A-24 effects. In addition, the range of compound effects was further examined by evaluating tau pathology and neuroinflammation. Following oral administration, both ligands reached brain concentrations known to provide neuroprotection in vitro. Compound induction of p75NTR cleavage provided evidence for CNS target engagement. LM11A-31 and LM11A-24 reduced excessive phosphorylation of tau, and LM11A-31 also inhibited its aberrant folding. Both ligands decreased activation of microglia, while LM11A-31 attenuated reactive astrocytes. Along with decreased inflammatory responses, both ligands reduced cholinergic neurite degeneration. In addition to the amelioration of neuropathology in AD model mice, LM11A-31, but not LM11A-24, prevented impairments in water maze performance, while both ligands prevented deficits in fear conditioning. These findings support a role for p75NTR ligands in preventing fundamental tau-related pathologic mechanisms in AD, and further validate the development of these small molecules as a new class of therapeutic compounds.
Collapse
Affiliation(s)
- Thuy-Vi V Nguyen
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Lin Shen
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Lilith Vander Griend
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Lisa N Quach
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Nadia P Belichenko
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Nay Saw
- Department of Neurosurgery, Stanford University, Palo Alto, CA, USA
| | - Tao Yang
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Mehrdad Shamloo
- Department of Neurosurgery, Stanford University, Palo Alto, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA Palo Alto Veterans Affairs Health Care System, Palo Alto, CA, USA
| | - Stephen M Massa
- Department of Veterans Affairs Medical Center, San Francisco, CA, USA Department of Neurology, University of California, San Francisco, CA, USA
| | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA
| |
Collapse
|
10
|
Abstract
The endogenous cannabinoid (endocannabinoid) system is an important regulator of synaptic function. Endocannabinoids acutely modulate inhibitory and excitatory transmission, and also mediate long-term depression at GABAergic and glutamatergic synapses. Typically, endocannabinoid synthesis and release is stimulated by depolarization-induced calcium influx and/or activation of phospholipase-C (PLC) signaling triggered by mGluR activation. Recently it has been shown that brain-derived neurotrophic factor (BDNF) can also induce endocannabinoid release. Although there is growing evidence for cross-talk between BDNF and endocannabinoid signaling, little is known about the functional relevance of these interactions. In the present studies, we examined BDNF - endocannabinoid interactions in regulating activity-dependent long-term depression at inhibitory synapses (iLTD). We found that theta burst stimulation (TBS) in layer 2/3 of mouse somatosensory cortical slices can induce a form of endocannabinoid-mediated iLTD that is independent of metabotropic glutamate receptor (mGluR) activation. This endocannabinoid-dependent iLTD, however, requires endogenous BDNF-trkB signaling, as it is blocked by a trk tyrosine kinase inhibitor and by a trkB receptor antagonist, and also requires activation of diacylglycerol lipase (DAG-lipase, DGL). In addition, endocannabinoid-mediated iLTD can be induced by combining a subthreshold concentration of exogenous BDNF with weak TBS stimulation that by itself is insufficient to induce iLTD. Taken together, our results suggest that TBS can induce the release of endogenous BDNF, which triggers DGL-dependent endocannabinoid mobilization and cannabinoid receptor-dependent iLTD at layer 2/3 cortical synapses.
Collapse
|
11
|
Li M, Dai FR, Du XP, Yang QD, Zhang X, Chen Y. Infusion of BDNF into the nucleus accumbens of aged rats improves cognition and structural synaptic plasticity through PI3K-ILK-Akt signaling. Behav Brain Res 2012; 231:146-53. [PMID: 22446058 DOI: 10.1016/j.bbr.2012.03.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 03/04/2012] [Accepted: 03/07/2012] [Indexed: 01/24/2023]
Abstract
To investigate the involvement of the nucleus accumbens (NAc) in cognitive impairment and the therapeutic effects of brain-derived neurotrophic factor (BDNF) in an animal model of cognitive deficit, we infused BDNF into the NAc of cognitively impaired aged rats. Cognition was evaluated by Morris water maze test. Structural synaptic plasticity was measured by Golgi staining. Brain tissue homogenization was used to measure the changes in signal molecules. Cultured PC-12 cells expressing tyrosine kinase receptor (Trk) B/p75 neurotrophin receptor (p75(NTR)), p75(NTR) or TrkA/p75(NTR) receptors were used for BDNF stimulation assays. Significant decreases in the levels of BDNF, phosphatidylinositol-3-kinase (PI3K) and integrin-linked kinase (ILK) activity, protein kinase B (Akt) Ser⁴⁷³ phosphorylation, dendritic branching, and density of dendritic spines on medium spiny neurons were observed in the NAc. Importantly, infusion of BDNF restored cognition, synaptic plasticity, and cell signaling. In cultured PC-12 cells, BDNF activated PI3K/Akt signaling through the TrkB receptor, whereas stimulation of ILK/Akt occurred through TrkA/p75(NTR) heteroreceptor. Our study suggested that the decreased BDNF level and its downstream signaling as well as loss of synaptic plasticity in the NAc are associated with cognitive impairments in aged rats. The BDNF-activated PI3K-Akt and ILK-Akt signaling play a key role in structural synaptic plasticity. Our study also suggested that BDNF could be a mechanism-based treatment for dementia.
Collapse
Affiliation(s)
- Min Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410078, PR China
| | | | | | | | | | | |
Collapse
|
12
|
Zhou L, Too HP. Mitochondrial localized STAT3 is involved in NGF induced neurite outgrowth. PLoS One 2011; 6:e21680. [PMID: 21738764 PMCID: PMC3124549 DOI: 10.1371/journal.pone.0021680] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 06/05/2011] [Indexed: 12/28/2022] Open
Abstract
Background Signal transducer and activator of transcription 3 (STAT3) plays critical roles in neural development and is increasingly recognized as a major mediator of injury response in the nervous system. Cytokines and growth factors are known to phosphorylate STAT3 at tyrosine705 with or without the concomitant phosphorylation at serine727, resulting in the nuclear localization of STAT3 and subsequent transcriptional activation of genes. Recent evidence suggests that STAT3 may control cell function via alternative mechanisms independent of its transcriptional activity. Currently, the involvement of STAT3 mono-phosphorylated at residue serine727 (P-Ser-STAT3) in neurite outgrowth and the underlying mechanism is largely unknown. Principal Findings In this study, we investigated the role of nerve growth factor (NGF) induced P-Ser-STAT3 in mediating neurite outgrowth. NGF induced the phosphorylation of residue serine727 but not tyrosine705 of STAT3 in PC12 and primary cortical neuronal cells. In PC12 cells, serine but not tyrosine dominant negative mutant of STAT3 was found to impair NGF induced neurite outgrowth. Unexpectedly, NGF induced P-Ser-STAT3 was localized to the mitochondria but not in the nucleus. Mitochondrial STAT3 was further found to be intimately involved in NGF induced neurite outgrowth and the production of reactive oxygen species (ROS). Conclusion Taken together, the findings herein demonstrated a hitherto unrecognized novel transcription independent mechanism whereby the mitochondria localized P-Ser-STAT3 is involved in NGF induced neurite outgrowth.
Collapse
Affiliation(s)
- Lihan Zhou
- Department of Biochemistry, National University of Singapore, Singapore, Singapore
- Chemical Pharmaceutical Engineering, Singapore–Massachusetts Institute of Technology Alliance, Singapore, Singapore
| | - Heng-Phon Too
- Department of Biochemistry, National University of Singapore, Singapore, Singapore
- Chemical Pharmaceutical Engineering, Singapore–Massachusetts Institute of Technology Alliance, Singapore, Singapore
- * E-mail:
| |
Collapse
|
13
|
Lemtiri-Chlieh F, Levine ES. BDNF evokes release of endogenous cannabinoids at layer 2/3 inhibitory synapses in the neocortex. J Neurophysiol 2010; 104:1923-32. [PMID: 20719932 DOI: 10.1152/jn.00472.2010] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The neurotrophin brain-derived neurotrophic factor (BDNF) is a potent regulator of inhibitory synaptic transmission, although the locus of this effect and the underlying mechanisms are controversial. We explored a potential interaction between BDNF and endogenous cannabinoid (endocannabinoid) signaling because activation of type 1 cannabinoid (CB1) receptors potently regulates γ-aminobutyric acid (GABA) release and both trkB tyrosine kinase receptors and CB1 receptors are highly expressed at synapses in neocortical layer 2/3. Here, we found that the effects of BDNF at inhibitory cortical synapses are mediated by the release of endocannabinoids acting retrogradely at presynaptic CB1 receptors. Specifically, acute application of BDNF rapidly reduced the amplitude of inhibitory postsynaptic currents (IPSCs) via postsynaptic trkB receptor activation because intracellular delivery of the tyrosine kinase inhibitor K252a completely blocked the BDNF effect. Although triggered by postsynaptic trkB activation, BDNF exposure decreased presynaptic release probability, as evidenced by increases in the paired-pulse ratio and coefficient of variation of evoked responses. In addition, BDNF decreased the frequency but not the amplitude of action potential-independent miniature IPSCs and BDNF did not alter the postsynaptic response to locally applied GABA. These results suggest that BDNF induces the release of a retrograde messenger from the postsynaptic cell that regulates presynaptic neurotransmitter release. Consistent with a role for endocannabinoids as the retrograde signal, the effect of BDNF on IPSCs was blocked by CB1 receptor antagonists and was occluded by a cannabinoid receptor agonist. Furthermore, inhibiting endocannabinoid synthesis or transport also disrupted the BDNF effect, implicating postsynaptic endocannabinoid release triggered by BDNF.
Collapse
Affiliation(s)
- Fouad Lemtiri-Chlieh
- University of Connecticut Health Center, Department of Neuroscience, MC-3401, 263 Farmington Ave., Farmington, CT 06030, USA
| | | |
Collapse
|
14
|
Sharif A, Prevot V. ErbB receptor signaling in astrocytes: a mediator of neuron-glia communication in the mature central nervous system. Neurochem Int 2010; 57:344-58. [PMID: 20685225 DOI: 10.1016/j.neuint.2010.05.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 03/29/2010] [Accepted: 05/18/2010] [Indexed: 10/19/2022]
Abstract
Astrocytes are now recognized as active players in the developing and mature central nervous system. Each astrocyte contacts vascular structures and thousands of synapses within discrete territories. These cells receive a myriad of inputs and generate appropriate responses to regulate the function of brain microdomains. Emerging evidence has implicated receptors of the ErbB tyrosine kinase family in the integration and processing of neuronal inputs by astrocytes: ErbB receptors can be activated by a wide range of neuronal stimuli; they control critical steps of glutamate-glutamine metabolism; and they regulate the biosynthesis and release of various glial-derived neurotrophic factors, gliomediators and gliotransmitters. These key properties of astrocytic ErbB signaling in neuron-glia interactions have significance for the physiology of the mature central nervous system, as exemplified by the central control of reproduction within the hypothalamus, and are also likely to contribute to pathological situations, since both dysregulation of ErbB signaling and glial dysfunction occur in many neurological disorders.
Collapse
Affiliation(s)
- Ariane Sharif
- Inserm, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the postnatal Brain, Lille, France.
| | | |
Collapse
|
15
|
Protection of crayfish glial cells but not neurons from photodynamic injury by nerve growth factor. J Mol Neurosci 2009; 39:308-19. [PMID: 19381880 DOI: 10.1007/s12031-009-9199-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Accepted: 03/29/2009] [Indexed: 10/20/2022]
Abstract
Photodynamic treatment that causes intense oxidative stress and cell death is currently used in neurooncology. However, along with tumor cells, it may damage healthy neurons and glia. In order to study photodynamic effect on normal nerve and glial cells, we used crayfish stretch receptor, a simple system consisting of only two identified sensory neurons surrounded by glial cells. Photodynamic treatment induced firing abolition and necrosis of neurons as well as necrosis and apoptosis of glial cells. Nerve growth factor but not brain-derived neurotrophic factor or epidermal growth factor protected glial cells but not neurons from photoinduced necrosis and apoptosis. Inhibitors of tyrosine kinases or protein kinase JNK eliminated anti-apoptotic effect of nerve growth factor in photosensitized glial cells but not neurons. Therefore, these signaling proteins were involved in the anti-apoptotic activity of nerve growth factor. These data indicate the possible presence of receptors capable of recognizing murine nerve growth factor in crayfish glial cells. Thus, intercellular signaling mediated by nerve-growth-factor-like neurotrophin, receptor tyrosine kinase, and JNK may be involved in crayfish glia protection from apoptosis induced by photodynamic treatment.
Collapse
|
16
|
Medina-Aguirre I, Gutiérrez-Ospina G, Hernández-Rodríguez J, Boyzo A, Manjarrez-Gutiérrez G. Development of 5-HT(1B), SERT and thalamo-cortical afferents in early nutrionally restricted rats: an emerging explanation for delayed barrel formation. Int J Dev Neurosci 2007; 26:225-31. [PMID: 18207350 DOI: 10.1016/j.ijdevneu.2007.12.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2007] [Revised: 12/05/2007] [Accepted: 12/06/2007] [Indexed: 11/25/2022] Open
Abstract
Barrel formation is delayed in nutritionally restricted rats. The underlying cause of such delay is yet unclear. Because barrels appear upon the arrival of somatosensory thalamo-cortical afferents and the reorientation of the dendritic arborizations of cortical spiny stellate neurons, it is likely that at least one of these processes is altered by nutritional restriction. Also, the serotoninergic afferent system has been implicated in regulating barrel segregation and growth during early postnatal life. We then evaluated the pattern of immunostaining of the serotonin transporter (SERT) and of the serotonin receptor 1B (5-HT(1B)), as well as the growth and arrival time of somatosensory thalamo-cortical afferents, to infer the contribution of these elements in the delayed formation of barrels observed in nutritionally restricted rats. It was found that the rates of development and the segregation of thalamo-cortical fibers were normal in nutritionally restricted rats. SERT, but not 5-HT(1B) immunoreactivity, was decreased in the primary somatosensory cortex during barrel specification. The availability of both proteins in nutritionally restricted rats was lower than that observed in their well fed counterparts at later developmental times. It is concluded that the delayed formation of barrels observed in nutritionally restricted rats is due to a retarded reorientation of dendritic arbors of cortical neurons. This might happen as a secondary effect of decreasing the availability of SERT and/or increasing the availability of 5-HT(1B) receptor early in postnatal life.
Collapse
Affiliation(s)
- Ivett Medina-Aguirre
- Laboratorio de Patología Molecular, Unidad de Investigación Biomolecular en Cardiología, Hospital de Cardiología, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, México 06725, DF, Mexico
| | | | | | | | | |
Collapse
|
17
|
Soiampornkul R, Tong L, Thangnipon W, Balazs R, Cotman CW. Interleukin-1beta interferes with signal transduction induced by neurotrophin-3 in cortical neurons. Brain Res 2007; 1188:189-97. [PMID: 18036576 DOI: 10.1016/j.brainres.2007.10.051] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2007] [Revised: 10/11/2007] [Accepted: 10/13/2007] [Indexed: 01/19/2023]
Abstract
It was previously observed that IL-1beta interferes with BDNF-induced TrkB-mediated signal transduction and protection of cortical neurons from apoptosis evoked by deprivation from trophic support [Tong L., Balazs R., Soiampornkul R., Thangnipon W., Cotman C.W., 2007. Interleukin-1beta impairs brain derived neurotrophic factor-induced signal transduction. Neurobiol. Aging]. Here we investigated whether the effect of the cytokine on neurotrophin signaling is more general. The influence of IL-1beta on NT-3 signaling was therefore studied under conditions when NT-3 primarily activated the TrkC receptor. The cytokine reduced NT-3-induced activation of MAPK/ERK and Akt, but did not interfere with Trk receptor autophosphorylation. IL-1beta reduced tyrosine phosphorylation of the docking proteins, IRS-1 and Shc, which convey receptor activation to the downstream protein kinase cascades. These are the steps that are also inhibited by IL-1beta in BDNF-induced signal transduction. The functional consequences of the effect of IL-1beta on NT-3 signaling were severe, as NT-3 protection of the trophic support-deprived cortical neurons was abrogated. In view of the role in the maintenance and plasticity of neurons of ERK, Akt and CREB, which are activated by neurotrophins, elevated IL-1beta levels in the brain in Alzheimer's disease and other neurodegenerative diseases might contribute to the decline in cognitive functions before the pathological signs of the disease develop.
Collapse
Affiliation(s)
- Rungtip Soiampornkul
- Neuro-Behavioral Biology Center, Institute of Science and Technology for Development, Mahidol University, Salaya Campus Nakorn Pathom, Thailand
| | | | | | | | | |
Collapse
|
18
|
Mooney SM, Miller MW. Nerve growth factor neuroprotection of ethanol-induced neuronal death in rat cerebral cortex is age dependent. Neuroscience 2007; 149:372-81. [PMID: 17869443 PMCID: PMC2128252 DOI: 10.1016/j.neuroscience.2007.08.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2007] [Revised: 07/30/2007] [Accepted: 08/08/2007] [Indexed: 01/07/2023]
Abstract
Organotypic cultures of rat cortex were used to test the hypotheses that nerve growth factor (NGF) is neuroprotective for immature cortical neurons and that ethanol abolishes this neuroprotection in a developmental stage-dependent manner. Samples were obtained on gestational day (G) 16 or postnatal day (P) 3 and cultured with ethanol (0 or 400 mg/dl) and NGF (0 or 30 ng/ml) for 72 h. Dying neurons were identified as exhibiting terminal nick-end labeling, immunoreactivity for activated caspase 3, or condensed nuclear chromatin. Two cortical compartments were examined in fetal tissue: a superficial, cell-sparse marginal zone (MZ) and a cell-dense cortical plate (CP). At P3, the CP was subdivided into a cell-dense upper cortical plate (UCP) and a less densely packed lower cortical plate (LCP). Neuronal death in the MZ was affected by neither NGF nor ethanol at both ages. In the fetal CP, NGF did not affect the incidence of cell death, but ethanol increased it. Treatment with NGF caused an upregulation of the expression of Neg, a gene known to be affected by NGF and ethanol. NGF did not ameliorate the ethanol-induced death. In pups, ethanol increased the amount of death in the LCP. NGF did protect against this death. Neither ethanol nor NGF altered the incidence of cell death in the UCP. The laminar-dependent neuroprotection did not correlate with expression of NGF receptors or Neg. Thus, NGF can be protective against the neurotoxic effect of ethanol in the neonatal brain. This effect is site selective and time dependent and it targets postmigratory, differentiating neurons.
Collapse
Affiliation(s)
- S M Mooney
- Department of Neuroscience and Physiology, State University of New York-Upstate Medical University, Syracuse, NY 13210, USA.
| | | |
Collapse
|
19
|
Lovekamp-Swan T, Glendenning ML, Schreihofer DA. A high soy diet enhances neurotropin receptor and Bcl-XL gene expression in the brains of ovariectomized female rats. Brain Res 2007; 1159:54-66. [PMID: 17582385 PMCID: PMC1995131 DOI: 10.1016/j.brainres.2007.05.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2006] [Revised: 04/25/2007] [Accepted: 05/20/2007] [Indexed: 10/23/2022]
Abstract
Estrogen is a powerful neuroprotective agent with the ability to induce trophic and antiapoptotic genes. However, concerns about negative overall health consequences of estrogen replacement after menopause have led to the adoption of other strategies to obtain estrogen's benefits in the brain, including the use of selective estrogen receptor modulators, high soy diets, or isoflavone supplements. This study sought to determine the ability of a high soy diet to induce neuroprotective gene expression in the female rat brain and compare the actions of soy with estrogen. Adult ovariectomized female rats were treated with 3 days of high dose estrogen or 2 weeks of a soy-free diet, a high soy diet, or chronic low dose estrogen. Different brain regions were microdissected and subjected to real time RT-PCR for neuroprotective genes previously shown to be estrogen-regulated. The principle findings are that a high soy diet led to the widespread increase in the mRNA for neurotropin receptors TrkA and p75-NTR, and the antiapoptotic Bcl-2 family member Bcl-X(L). Immunohistochemistry confirmed increases in both TrkA and Bcl-X(L). Chronic low dose estrogen mimicked some of these effects, but acute high dose estrogen did not. The effects of a high soy diet were particularly evident in the parietal cortex and hippocampus, two regions protected by estrogen in animal models of neurological disease and injury. These results suggest that a high soy diet may provide beneficial effects to the brain similar to low dose chronic estrogen treatment such as that used for postmenopausal hormone replacement.
Collapse
Affiliation(s)
- Tara Lovekamp-Swan
- Department of Physiology, Medical College of Georgia, 1120 15th Street, Augusta, GA 30912-3000
| | - Michele L. Glendenning
- Department of Physiology, Medical College of Georgia, 1120 15th Street, Augusta, GA 30912-3000
| | - Derek A. Schreihofer
- Department of Physiology, Medical College of Georgia, 1120 15th Street, Augusta, GA 30912-3000
| |
Collapse
|
20
|
Bruns MB, Miller MW. Neurotrophin ligand-receptor systems in somatosensory cortex of adult rat are affected by repeated episodes of ethanol. Exp Neurol 2007; 204:680-92. [PMID: 17320080 PMCID: PMC1995597 DOI: 10.1016/j.expneurol.2006.12.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Revised: 12/19/2006] [Accepted: 12/28/2006] [Indexed: 12/24/2022]
Abstract
Ethanol exposure profoundly affects learning and memory and neural plasticity. Key players underlying these functions are neurotrophins. The present study explored the effects of ethanol on the distribution of neurotrophins in the cerebral cortex of the adult rat. Age- and weight-matched pairs of adult male, Long-Evans rats were fed a liquid, ethanol-containing (6.7% v/v) diet or pair-fed an isocaloric control diet three consecutive days per week for 6, 12, 18, or 24 weeks. Brains were processed immunohistochemically for nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) expression and for the expression of three neurotrophin receptors, p75, trkA, and trkB. Total numbers of immunolabeled neurons in specific layers of somatosensory cortex of ethanol- and control-fed animals were determined stereologically. Ethanol exposure induced an increase in the numbers of NGF- or BDNF-expressing neurons and in neurotrophin content per somata. These changes were (a) time and (b) laminar dependent. In contrast, the number of receptor-expressing neurons did not change due to ethanol exposure or to length of time on the ethanol diet. Thus, ethanol induces the recruitment of cortical neurons to express neurotrophins and an increase in the amount of neurotrophin expression per neuron.
Collapse
Affiliation(s)
- Marla B. Bruns
- Department of Neuroscience and Physiology, State University of New York- Upstate Medical University, Syracuse NY 13210 USA
- Developmental Exposure Alcohol Research Center, State University of New York, Binghamton NY 13902 USA and Syracuse NY 13210 USA
| | - Michael W. Miller
- Department of Neuroscience and Physiology, State University of New York- Upstate Medical University, Syracuse NY 13210 USA
- Developmental Exposure Alcohol Research Center, State University of New York, Binghamton NY 13902 USA and Syracuse NY 13210 USA
- Research Service, Veterans Affairs Medical Center, Syracuse NY 13210 USA
| |
Collapse
|
21
|
Bruns MB, Miller MW. THIS ARTICLE HAS BEEN RETRACTED: Functional nerve growth factor and trkA autocrine/paracrine circuits in adult rat cortex are revealed by episodic ethanol exposure and withdrawal. J Neurochem 2006; 100:1155-68. [PMID: 17316397 DOI: 10.1111/j.1471-4159.2006.04301.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The hypothesis tested is that cortical neurotrophins communicate through an inducible autocrine/paracrine mechanism. As ethanol (Et) can induce cortical nerve growth factor (NGF) expression, adult rats were challenged with Et on three consecutive days per week for 6 weeks. The focus of the study was layer V, the chief repository of receptor-expressing neuronal cell bodies. Brains were collected immediately after the sixth Et exposure or 72 h later [i.e., following withdrawal (WD)]. Double-label in situ hybridization-immunohistochemistry studies showed that many neuronal somata co-expressed NGF mRNA with NGF, trkA, or phosphorylated trk (p-trk), essential components of an inducible autocrine system. The frequencies of co-labeling were affected by neither Et nor WD. On the contrary, Et increased the number of NGF mRNA-expressing neurons and the amount of NGF mRNA expressed per cell. Et also increased total cortical concentration of NGF protein, the number of layer V neurons expressing trkA transcript, the amount of trkA mRNA expressed per neuron, and trkA phosphorylation. Following WD, the frequency of NGF-mRNA-expressing cells increased, although transcript and protein content fell. WD induced an increase in trkA mRNA and protein expression, however, p-trk expression was unaffected. Thus, Et treatment reveals that layer V has inducible autocrine/paracrine and anterograde neurotrophin systems. WD unveils the dynamism and recruitability of these systems.
Collapse
Affiliation(s)
- Marla B Bruns
- Department of Neuroscience and Physiology, State University of New York-Upstate Medical University, Syracuse, New York 13210, USA
| | | |
Collapse
|
22
|
Deumens R, Koopmans GC, Jaken RJP, Morren K, Comhair T, Kosar S, Steinbusch HWM, Den Bakker CGJ, Joosten EAJ. Stimulation of neurite outgrowth on neonatal cerebral astrocytes is enhanced in the presence of BDNF. Neurosci Lett 2006; 407:268-73. [PMID: 16978777 DOI: 10.1016/j.neulet.2006.08.059] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2006] [Revised: 08/19/2006] [Accepted: 08/24/2006] [Indexed: 11/22/2022]
Abstract
An area of increasing interest in spinal cord injury (SCI) research is the development of multi-factorial strategies to promote repair. In this respect, a prominent role is played by cell transplantation, the reparative effect of which can be enhanced by additional use of neurotrophic factors. Immature astrocytes have shown their merit in stimulating axon regeneration upon transplantation into the injured spinal cord. Brain-derived neurotrophic factor (BDNF) influences a wide range of descending axon tracts in the injured spinal cord. In the present study, we hypothesized that the neurite outgrowth of neonatal cortical neurons on immature astrocytes is enhanced in the presence of BDNF. To test this hypothesis, neonatal cortical neurons were cultured on neonatal astrocytes for 2 days in absence or presence of BDNF. The length of the longest neurite and the number of primary neurites per neuron were taken as measures to study neurite outgrowth. We show that BDNF dose-dependently enhanced neurite outgrowth of neonatal cerebral cortical neurons grown on immature astrocytes. Compared to conditions without BDNF, the length of the longest neurite increased by 25.5 and 28.8% in presence of 10 and 100 pg/ml BDNF, respectively. BDNF did not alter the density of the immature astrocytes. We conclude that the presence of BDNF enhances the neurite outgrowth on immature astrocytes. A multi-factorial strategy based on transplantation of neonatal astrocytes in the presence of additional BDNF is recommended and may stimulate axon regrowth after experimental injury to the central nervous system.
Collapse
Affiliation(s)
- Ronald Deumens
- Department of Psychiatry and Neuropsychology, Division Cellular Neuroscience, University of Maastricht, Maastricht, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Miller MW. Effect of prenatal exposure to ethanol on glutamate and GABA immunoreactivity in macaque somatosensory and motor cortices: Critical timing of exposure. Neuroscience 2006; 138:97-107. [PMID: 16427209 DOI: 10.1016/j.neuroscience.2005.10.060] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2005] [Revised: 10/24/2005] [Accepted: 10/26/2005] [Indexed: 11/25/2022]
Abstract
The present study explored the effects of gestational ethanol exposure on enduring changes in the distribution of projection neurons and local circuit neurons in somatosensory/motor cortex. Critical events in corticogenesis occur during macaque gestation: the first six weeks of gestation include the period of primary stem cell production and the next 18 weeks are marked by the birth, migration, early differentiation, and death of cortical neurons. Monkeys were exposed to ethanol (or saline) one day per week during the first six or during the entire 24 weeks of gestation. Offspring were killed as adolescents. Projection neurons and local circuit neurons were identified immunohistochemically with antibodies directed against glutamate and anti-GABA, respectively. In all animals, both projection neurons and local circuit neurons were distributed in all laminae of both somatosensory and motor cortices. Ethanol did not affect the size of Cresyl Violet-stained, glutamate-positive, or GABA-immunolabeled somata, however, it did decrease neuronal density. The total density of Cresyl Violet-stained neurons was reduced in monkeys treated with ethanol (or saline) one day per week during the first six weeks of gestation and during the entire 24 weeks of gestation. Similar reductions were detected for glutamate- and GABA-positive neurons. The densities of Cresyl Violet-stained and of glutamate- and GABA-expressing neurons were reduced in all cortical layers. The only exception was layer V which was unaffected in monkeys treated with ethanol (or saline) one day per week during the first six weeks of gestation and during the entire 24 weeks of gestation. Thus, the parallel effects on both neuronal subpopulations suggest that ethanol targets a population of undetermined neuronal precursors.
Collapse
Affiliation(s)
- M W Miller
- Department of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA.
| |
Collapse
|
24
|
Zhang FX, Lai CH, Tse YC, Shum DKY, Chan YS. Expression of Trk receptors in otolith-related neurons in the vestibular nucleus of rats. Brain Res 2005; 1062:92-100. [PMID: 16256078 DOI: 10.1016/j.brainres.2005.09.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2005] [Revised: 09/18/2005] [Accepted: 09/25/2005] [Indexed: 01/19/2023]
Abstract
The expression of the three Trk receptors (TrkA, TrkB, and TrkC) in otolith-related neurons within the vestibular nuclei of adult Sprague-Dawley rats was examined immunohistochemically. Conscious animals were subjected to sinusoidal linear acceleration along either the anterior-posterior (AP) or interaural (IA) axis on the horizontal plane. Neuronal activation was defined by Fos expression in cell nuclei. Control animals, viz labyrinthectomized rats subjected to stimulation and normal rats that remained stationary, showed only a few sporadically scattered Fos-labeled neurons. Among experimental rats, the number of Fos-labeled neurons and their distribution pattern in each vestibular subnucleus in animals stimulated along the antero-posterior axis were similar to those along the interaural axis. No apparent topography was observed among neurons activated along these two directions. Only about one-third of the Trk-immunoreactive neurons in the vestibular nucleus expressed Fos. Double-labeled Fos/TrkA, Fos/TrkB and Fos/TrkC neurons constituted 85-98% of the total number of Fos-labeled neurons in vestibular nuclear complex and its subgroups x and y. Our findings suggest that Trk receptors and their cognate neurotrophins in central otolith neurons may contribute to the modulation of gravity-related spatial information during horizontal head movements.
Collapse
Affiliation(s)
- F X Zhang
- Department of Physiology, Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | | | | | | | | |
Collapse
|
25
|
Sasaki S, Mori D, Toyo-oka K, Chen A, Garrett-Beal L, Muramatsu M, Miyagawa S, Hiraiwa N, Yoshiki A, Wynshaw-Boris A, Hirotsune S. Complete loss of Ndel1 results in neuronal migration defects and early embryonic lethality. Mol Cell Biol 2005; 25:7812-27. [PMID: 16107726 PMCID: PMC1190282 DOI: 10.1128/mcb.25.17.7812-7827.2005] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Regulation of cytoplasmic dynein and microtubule dynamics is crucial for both mitotic cell division and neuronal migration. NDEL1 was identified as a protein interacting with LIS1, the protein product of a gene mutated in the lissencephaly. To elucidate NDEL1 function in vivo, we generated null and hypomorphic alleles of Ndel1 in mice by targeted gene disruption. Ndel1(-/-) mice were embryonic lethal at the peri-implantation stage like null mutants of Lis1 and cytoplasmic dynein heavy chain. In addition, Ndel1(-/-) blastocysts failed to grow in culture and exhibited a cell proliferation defect in inner cell mass. Although Ndel1(+/-) mice displayed no obvious phenotypes, further reduction of NDEL1 by making null/hypomorph compound heterozygotes (Ndel1(cko/-)) resulted in histological defects consistent with mild neuronal migration defects. Double Lis1(cko/+)-Ndel1(+/-) mice or Lis1(+/-)-Ndel1(+/-) mice displayed more severe neuronal migration defects than Lis1(cko/+)-Ndel1(+/)(+) mice or Lis1(+/-)-Ndel1(+/+) mice, respectively. We demonstrated distinct abnormalities in microtubule organization and similar defects in the distribution of beta-COP-positive vesicles (to assess dynein function) between Ndel1 or Lis1-null MEFs, as well as similar neuronal migration defects in Ndel1- or Lis1-null granule cells. Rescue of these defects in mouse embryonic fibroblasts and granule cells by overexpressing LIS1, NDEL1, or NDE1 suggest that NDEL1, LIS1, and NDE1 act in a common pathway to regulate dynein but each has distinct roles in the regulation of microtubule organization and neuronal migration.
Collapse
Affiliation(s)
- Shinji Sasaki
- Department of Neuro-Science, Research Center for Genomic Medicine, Saitama Medical School, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Barrett GL, Greferath U, Barker PA, Trieu J, Bennie A. Co-expression of the P75 neurotrophin receptor and neurotrophin receptor-interacting melanoma antigen homolog in the mature rat brain. Neuroscience 2005; 133:381-92. [PMID: 15878242 DOI: 10.1016/j.neuroscience.2005.01.067] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2003] [Revised: 11/02/2004] [Accepted: 01/19/2005] [Indexed: 12/26/2022]
Abstract
The p75 neurotrophin receptor (p75(NTR)) is involved in the regulation of neuronal survival and phenotype, but its signal transduction mechanisms are poorly understood. Recent evidence has implicated the cytoplasmic protein NRAGE (neurotrophin receptor-interacting MAGE (from Melanoma AntiGEn) homolog) in p75(NTR) signaling. To gain further insight into the role of NRAGE, we investigated the co-expression of NRAGE and p75(NTR) in mature rat brain. In all areas examined, NRAGE appeared to be confined to neurons. In the basal forebrain cholinergic complex, NRAGE immunoreactivity was evident in all p75(NTR)-positive neurons. There were many more NRAGE-positive than p75(NTR)-positive neurons in these regions, however. NRAGE was also expressed in areas of the basal forebrain that did not express p75(NTR), including the lateral septal nucleus and the nucleus accumbens. A finding in marked contrast to previous studies was the presence of p75(NTR) immunoreactivity in neuronal cell bodies in the hippocampus. Hippocampal p75(NTR) immunoreactivity was apparent in rats 6 months and older, and was localized to the dentate gyrus and stratum oriens. All p75(NTR)-positive neurons in the dentate gyrus and hippocampal formation were positive for NRAGE. The majority of granular cells of the dentate gyrus and pyramidal cells in the hippocampal formation were positive for NRAGE and negative for p75(NTR). NRAGE was also present in some neuronal populations that express p75(NTR) after injury, including striatal cholinergic interneurons, and motor neurons. A region of marked disparity was the cerebral cortex, in which NRAGE immunoreactivity was widespread whereas p75(NTR) was absent. The results are consistent with an important role for NRAGE in p75(NTR) signaling, as all cells that expressed p75(NTR) also expressed NRAGE. The wider distribution of NRAGE expression suggests that NRAGE may also participate in other signaling processes.
Collapse
Affiliation(s)
- G L Barrett
- Department of Physiology, University of Melbourne, Parkville 3010, Australia.
| | | | | | | | | |
Collapse
|
27
|
Miller MW. Repeated episodic exposure to ethanol affects neurotrophin content in the forebrain of the mature rat. Exp Neurol 2004; 189:173-81. [PMID: 15296847 DOI: 10.1016/j.expneurol.2004.05.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2004] [Revised: 04/16/2004] [Accepted: 05/20/2004] [Indexed: 11/26/2022]
Abstract
Chronic exposure to ethanol can cause deficits in learning and memory. It has been suggested that withdrawal is potentially more damaging than the ethanol exposure per se. Therefore, we explored the effect of repeated episodic exposure to ethanol on key regulators of cortical activity, the neurotrophins. Rats were exposed to ethanol via a liquid diet for 3 days per week for 6-24 weeks. Control rats were pair-fed an isocaloric liquid diet or ad libitum fed chow and water. The concentrations of nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin-3 (NT-3) were determined using enzyme-linked immunosorbant assays (ELISAs). Five telencephalic structures were examined: parietal cortex, entorhinal cortex, hippocampus, the basal nucleus, and the septal nuclei. All five areas expressed each of the three neurotrophins; BDNF was most abundant and NGF the least. The parietal cortex was susceptible to ethanol exposure, NGF and BDNF content increased, and NT-3 content fell, whereas no changes were detectable in the entorhinal cortex. In the hippocampus, the amount all three neurotrophins increased following episodic ethanol exposure. Neurotrophin content in the two segments of the basal forebrain was affected; NGF and NT-3 content in the basal forebrain was reduced and NGF and BDNF content in the septal nuclei was increased by ethanol exposure. In many cases where ethanol had an effect, the change was transient so that by 24 weeks of episodic exposure, no significant changes were evident. Thus, the effects of ethanol are site- and time-dependent. This pattern differs from changes caused by chronic ethanol exposure, hence, neurotrophins must be vulnerable to the effects of withdrawal. Furthermore, the ethanol-induced changes do not appear to fit a model consistent with retrograde regulation, rather they suggest that neurotrophins act through autocrine/paracrine systems.
Collapse
Affiliation(s)
- Michael W Miller
- Department of Neuroscience and Physiology, State University of New York-Upstate Medical University, Syracuse, NY 13210, USA.
| |
Collapse
|
28
|
Liot G, Gabriel C, Cacquevel M, Ali C, MacKenzie ET, Buisson A, Vivien D. Neurotrophin-3-induced PI-3 kinase/Akt signaling rescues cortical neurons from apoptosis. Exp Neurol 2004; 187:38-46. [PMID: 15081586 DOI: 10.1016/j.expneurol.2004.01.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2003] [Revised: 12/30/2003] [Accepted: 01/08/2004] [Indexed: 10/26/2022]
Abstract
A number of cytokines including neurotrophins have been tested for their neuroprotective activity against different paradigms of neuronal death. However, as for neurotrophin-3 (NT-3), their mechanisms of action have not been fully identified. By using cultures of mouse cortical neurons, we have investigated the molecular mechanisms by which neurotrophin-3 could protect cortical neurons against apoptosis. In a model of caspase-dependent apoptosis leading to the recruitment of active initiators caspase-8 and -9 and of executioner caspase-3, we have evidenced that NT-3 displayed an anti-apoptotic effect in a dose-dependent manner. First, we showed that, in cultured cortical neurons, NT-3 could promote extracellular signal-regulated protein kinase/mitogen-activated protein kinase (ERK/MAPK) and phosphatidylinositol-3' (PI-3) kinase/Akt phosphorylation. Second, we showed that although the blockade of the Akt pathway prevented the anti-apoptotic effect of NT-3, blockade of the ERK pathway did not. Altogether, our data demonstrate that NT-3 displayed an anti-apoptotic effect on cultured cortical neurons through a mechanism involving the recruitment of the PI-3 kinase/Akt signaling pathway.
Collapse
Affiliation(s)
- Géraldine Liot
- Université de Caen, CNRS UMR 6551, Centre CYCERON, IFR 47, 14074 Caen Cedex, France
| | | | | | | | | | | | | |
Collapse
|
29
|
Miller MW, Mooney SM. Chronic exposure to ethanol alters neurotrophin content in the basal forebrain-cortex system in the mature rat: Effects on autocrine-paracrine mechanisms. ACTA ACUST UNITED AC 2004; 60:490-8. [PMID: 15307153 DOI: 10.1002/neu.20059] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neurotrophins are broadly expressed in the mammalian forebrain: notably in cerebral cortex and the basal forebrain (e.g., the septal and basal nuclei). These factors promote neuronal survival and plasticity, and have been implicated as key players in learning and memory. Chronic exposure to ethanol causes learning and memory deficits. We tested the hypothesis that ethanol affects neurotrophin expression and predicted that these changes would be consistent with alterations in retrograde or autocrine/paracrine systems. Mature rats were fed a liquid diet containing ethanol daily for 8 or 24 weeks. Weight-matched controls were pair-fed an isocaloric, isonutritive diet. Proteins from five structures (parietal and entorhinal cortices, hippocampus, and the basal and septal nuclei) were studied. ELISAs were used to determine the concentration of nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin-3 (NT-3). All three neurotrophins were detected in each structure examined. Ethanol treatment significantly (p < 0.05) affected neurotrophin expression in time- and space-dependent manners. NGF content was generally depressed by ethanol exposure, whereas NT-3 content increased. BDNF concentration was differentially affected by ethanol: it increased in the parietal cortex and the basal forebrain and decreased in the hippocampus. With the exception of NGF in the septohippocampal system, the ethanol-induced changes in connected structures were inconsistent with changes that would be predicted from a retrograde model. Thus, the present data (a) support the concept that neurotrophins act through a nonretrograde system (i.e., a local autocrine/paracrine system), and (b) that chronic exposure to ethanol disrupts these regulatory mechanisms.
Collapse
Affiliation(s)
- Michael W Miller
- Department of Neuroscience and Physiology, S.U.N.Y.-Upstate Medical University, 750 East Adams Street, Syracuse, New York 13210, USA.
| | | |
Collapse
|
30
|
Assadi AH, Zhang G, Beffert U, McNeil RS, Renfro AL, Niu S, Quattrocchi CC, Antalffy BA, Sheldon M, Armstrong DD, Wynshaw-Boris A, Herz J, D'Arcangelo G, Clark GD. Interaction of reelin signaling and Lis1 in brain development. Nat Genet 2003; 35:270-6. [PMID: 14578885 DOI: 10.1038/ng1257] [Citation(s) in RCA: 161] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2003] [Accepted: 09/29/2003] [Indexed: 11/08/2022]
Abstract
Loss-of-function mutations in RELN (encoding reelin) or PAFAH1B1 (encoding LIS1) cause lissencephaly, a human neuronal migration disorder. In the mouse, homozygous mutations in Reln result in the reeler phenotype, characterized by ataxia and disrupted cortical layers. Pafah1b1(+/-) mice have hippocampal layering defects, whereas homozygous mutants are embryonic lethal. Reln encodes an extracellular protein that regulates layer formation by interacting with VLDLR and ApoER2 (Lrp8) receptors, thereby phosphorylating the Dab1 signaling molecule. Lis1 associates with microtubules and modulates neuronal migration. We investigated interactions between the reelin signaling pathway and Lis1 in brain development. Compound mutant mice with disruptions in the Reln pathway and heterozygous Pafah1b1 mutations had a higher incidence of hydrocephalus and enhanced cortical and hippocampal layering defects. Dab1 and Lis1 bound in a reelin-induced phosphorylation-dependent manner. These data indicate genetic and biochemical interaction between the reelin signaling pathway and Lis1.
Collapse
Affiliation(s)
- Amir H Assadi
- Cain Foundation Laboratories and Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Toesca A, Giannetti S, Granato A. Overexpression of the p75 neurotrophin receptor in the sensori-motor cortex of rats exposed to ethanol during early postnatal life. Neurosci Lett 2003; 342:89-92. [PMID: 12727325 DOI: 10.1016/s0304-3940(03)00258-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Foetal alcohol syndrome is a known cause of mental retardation. It has been suggested that the anatomical and functional alterations observed in the cerebral cortex could be mediated by an interference of ethanol with developmental processes modulated by neurotrophins and/or their receptors. We have studied by immunohistochemistry the expression of the p75 neurotrophin receptor (p75 NTR) in the sensori-motor cortex of P10 and P20 rats exposed to the inhalation of ethanol during the first week of postnatal life. At both the studied ages, the number of p75 NTR immunoreactive neurons was higher in ethanol treated animals compared to controls. The increase of immunoreactive elements was relatively more marked in the motor than in the somatosensory cortex. The involvement of p75 NTR in ethanol-induced apoptosis and neural plasticity is discussed.
Collapse
Affiliation(s)
- Amelia Toesca
- Institute of Anatomy, Catholic University, L.go F. Vito 1, 00168, Rome, Italy
| | | | | |
Collapse
|
32
|
Zhang FX, Lai CH, Lai SK, Yung KKL, Shum DKY, Chan YS. Neurotrophin receptor immunostaining in the vestibular nuclei of rats. Neuroreport 2003; 14:851-5. [PMID: 12858046 DOI: 10.1097/00001756-200305060-00015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The distribution of high-affinity neurotrophin receptors in cells of the vestibular nuclear complex and its subnuclei of adult rats was examined. We noted a high density of tyrosine kinase (Trk) A- and B- and a lower density of TrkC-immunostained cells. In particular, long, intensely labelled immunostained-TrkB fibres formed networks in the neuropil. Both TrkA- and TrkB-immunostained cells were widely distributed in the lateral, medial and spinal vestibular nuclei, and were less frequently seen in the superior vestibular nucleus, x and y subnuclei. However, immunostaining for TrkC was weak in many cells within the vestibular nuclei. The widespread and abundant neuronal distribution of Trk receptors predicts that their associated neurotrophins exert significant effects on individual cells within the vestibular nuclei.
Collapse
Affiliation(s)
- F X Zhang
- Department of Physiology, The University of Hong Kong, Hong Kong, P.R. China
| | | | | | | | | | | |
Collapse
|
33
|
Kuboyama T, Tohda C, Zhao J, Nakamura N, Hattori M, Komatsu K. Axon- or dendrite-predominant outgrowth induced by constituents from Ashwagandha. Neuroreport 2002; 13:1715-20. [PMID: 12395110 DOI: 10.1097/00001756-200210070-00005] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We previously reported that the methanol extract of Ashwagandha (roots of Dunal) induced dendrite extension in a human neuroblastoma cell line. In this study, we found that six of the 18 compounds isolated from the methanol extract enhanced neurite outgrowth in human neuroblastoma SH-SY5Y cells. Double immunostaining was performed in rat cortical neurons using antibodies to phosphorylated NF-H as an axonal marker, and to MAP2 as a dendritic marker. In withanolide A-treated cells, the length of NF-H-positive processes was significantly increased compared with vehicle-treated cells, whereas, the length of MAP2-positive processes was increased by withanosides IV and VI. These results suggest that axons are predominantly extended by withanolide A, and dendrites by withanosides IV and VI.
Collapse
Affiliation(s)
- Tomoharu Kuboyama
- Research Center for Ethnomedicines, Institute of Natural Medicine, Toyama Medical and Pharmaceutical University, Toyama, Japan
| | | | | | | | | | | |
Collapse
|
34
|
Anderson RA, Robinson LLL, Brooks J, Spears N. Neurotropins and their receptors are expressed in the human fetal ovary. J Clin Endocrinol Metab 2002; 87:890-7. [PMID: 11836338 DOI: 10.1210/jcem.87.2.8221] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Mammalian ovarian development is characterized by a sequential pattern of mitotic proliferation of oogonia, initiation then arrest of meiosis, and primordial follicle formation. The factors regulating these processes are poorly understood. The neurotropins are survival and differentiation factors in the nervous system, acting via high affinity receptors of the trk protooncogene family and the low affinity p75 nerve growth factor receptor, and have also been described in the rodent ovary, where changes in NT4/TrkB gene expression have been detected at the time of primordial follicle formation. There are no data on neurotropin expression in the normal human ovary. We have investigated the expression and localization of neurotropins and their receptors in the midtrimester human fetal ovary (13-21 wk gestation). Expression of mRNA for neurotropins and their receptors was detected by RT-PCR. Clusters of oogonia were found to be the predominant site of NT4 mRNA expression using in situ hybridization. However, at later gestations granulosa cells of primordial follicles showed increased expression, with lesser expression in the enclosed oocytes. NT4 protein was also localized to the granulosa cells by immunohistochemistry and at earlier developmental stages to epithelioid cells, which were mingled with clusters of oogonia not expressing NT4. TrkB receptor protein was localized by immunohistochemistry to germ cells at all gestations examined. The p75 nerve growth factor receptor protein was exclusively expressed in the ovarian stroma. These data demonstrate the expression of neurotropins and their receptors within the human fetal ovary. Developmental changes in the pattern of expression of NT4 around the time of primordial follicle formation suggest that neurotropins may be involved in signaling between somatic cells and germ cells at this crucial stage of ovarian development.
Collapse
Affiliation(s)
- Richard A Anderson
- Medical Research Council Human Reproductive Sciences Unit, Center for Reproductive Biology, University of Edinburgh, Edinburgh, United Kingdom EH3 9ET.
| | | | | | | |
Collapse
|
35
|
Frost DO. BDNF/trkB signaling in the developmental sculpting of visual connections. PROGRESS IN BRAIN RESEARCH 2002; 134:35-49. [PMID: 11702553 DOI: 10.1016/s0079-6123(01)34004-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Neurotrophins are a family of secreted molecules that have multiple, profound actions on the structure and function of both developing and mature neurons. Neurotrophins exert their influences by signaling through the trk family of receptor tyrosine kinases and the p75 low affinity neurotrophin receptor. Here we review the contributions of neurotrophins to the development of neural circuitry in the mammalian visual system. We emphasize: (1) the role of neurotrophins as components of the cellular mechanisms by which neuroelectric activity sculpts pattern of brain connectivity; and (2) the results of recent experiments suggesting that the trafficking of neurotrophin proteins may be activity dependent.
Collapse
Affiliation(s)
- D O Frost
- Department of Pharmacology and Experimental Therapeutics, Department of Anesthesiology and Neuroscience Program, University of Maryland School of Medicine, 655 West Baltimore St., Baltimore, MD 21201, USA.
| |
Collapse
|
36
|
Endogenous brain-derived neurotrophic factor and neurotrophin-3 antagonistically regulate survival of axotomized corticospinal neurons in vivo. J Neurosci 2001. [PMID: 11331378 DOI: 10.1523/jneurosci.21-10-03492.2001] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neuronal growth factors regulate the survival of neurons by their survival and death-promoting activity on distinct populations of neurons. The neurotrophins nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin-3 (NT-3) promote neuronal survival via tyrosine kinase (Trk) receptors, whereas NGF and BDNF can also induce apoptosis in developing neurons through p75(NTR) receptors in the absence of their respective Trk receptors. Using mutant mice and inactivation of neurotrophins and their receptors with antibodies in rats, we show that endogenous NT-3 induces death of adult BDNF-dependent, axotomized corticospinal neurons (CSNs). When NT-3 is neutralized, the neurons survive even without BDNF, suggesting complete antagonism. Whereas virtually all unlesioned and axotomized CSNs express both trkB and trkC mRNA, p75 is barely detectable in unlesioned CSNs but strongly upregulated in axotomized CSNs by day 3 after lesion, the time point when cell death occurs. Blocking either cortical TrkC or p75(NTR) receptors alone prevents death, indicating that the opposing actions of NT-3 and BDNF require their respective Trk receptors, but induction of death depends on p75(NTR) cosignaling. The results show that neuronal survival can be regulated antagonistically by neurotrophins and that neurotrophins can induce neuronal death in the adult mammalian CNS. We further present evidence that signaling of tyrosine kinase receptors of the trk family can be crucially involved in the promotion of neuronal death in vivo.
Collapse
|
37
|
Abstract
In this report, we investigated whether reactive astrocytes produce neuregulins (glial growth factor 2/heregulin/acetylcholine receptor-inducing activity or neu differentiation factor) and its putative receptors, ErbB2 and ErbB3 tyrosine kinases, in the injured CNS in vivo. Significant immunoreactivities with anti-neuregulin, anti-ErbB2, and anti-ErbB3 antibodies were detected on astrocytes at the injured site 4 d after injury to the adult rat cerebral cortex. To elucidate the mechanisms for the upregulation of neuregulin expression in astrocytes, primary cultured astrocytes were treated with certain reagents, including forskolin, that are known to elevate the intracellular level of cAMP and induce marked morphological changes in astrocytes. Western blot analysis showed that the expression of a 52 kDa membrane-spanning form of a neuregulin protein was enhanced in cultured astrocytes after administration of forskolin. The upregulation of glial fibrillary acidic protein was also observed in astrocytes treated with forskolin. In contrast, inactivation of protein kinase C because of chronic treatment with phorbol ester 12-O-tetradecanoyl phorbol 13-acetate downregulated the expression of the 52 kDa isoform, although other splice variants with apparent molecular sizes of 65 and 60 kDa were upregulated. These results suggest that the enhancement of neuregulin expression at injured sites is induced, at least in part, by elevation in intracellular cAMP levels and/or a protein kinase C signaling pathway. The neuregulin expressed on reactive astrocytes may stimulate their proliferation and support the survival of neurons surrounding cortical brain wounds in vivo.
Collapse
|
38
|
Brandt JA, Churchill L, Guan Z, Fang J, Chen L, Krueger JM. Sleep deprivation but not a whisker trim increases nerve growth factor within barrel cortical neurons. Brain Res 2001; 898:105-12. [PMID: 11292453 DOI: 10.1016/s0006-8993(01)02149-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Sleep is hypothesized to influence activity-driven changes in the brain microcircuitry. A change in the barrel cortex following the removal of the mystacial whiskers in rats is a model for synaptic plasticity. This model was combined with sleep deprivation and immunoreactivity for nerve growth factor (NGF) was determined. Sleep deprivation for 6 h after light onset significantly increased the number of NGF-immunoreactive pyramidal neurons in layer V of the barrel cortex. However, unilateral trimming of mystacial whiskers did not affect NGF immunoreactivity in the contralateral or ipsilateral barrel cortices when rats were allowed to sleep. If the rats received a unilateral whisker cut at light onset, and subsequently were deprived of sleep, increases in the NGF-immunoreactive neurons were only observed in the barrel cortex on the side that received input from the remaining intact whiskers. In contrast, NGF immunoreactivity on the side contralateral to the cut whiskers decreased in sleep-deprived animals to levels below those observed in the control animals that were allowed to sleep. These results suggest that NGF expression is influenced by the interaction of sleep, afferent input and the nature of ongoing synaptic reorganization. Further, results are consistent with the hypothesis that growth factors, such as NGF, form part of the mechanism responsible for sleep regulation and that they also form one facet of sleep-related synaptic plasticity.
Collapse
Affiliation(s)
- J A Brandt
- Washington State University, College of Veterinary Medicine, Department of Veterinary and Comparative Anatomy, Pharmacology and Physiology, P.O. Box 646520, Pullman, WA 99164-6520, USA
| | | | | | | | | | | |
Collapse
|
39
|
Pitts AF, Miller MW. Expression of nerve growth factor, brain-derived neurotrophic factor, and neurotrophin-3 in the somatosensory cortex of the mature rat: coexpression with high-affinity neurotrophin receptors. J Comp Neurol 2000; 418:241-54. [PMID: 10701824 DOI: 10.1002/(sici)1096-9861(20000313)418:3<241::aid-cne1>3.0.co;2-m] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Neurotrophins, including nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin-3 (NT-3), are critical for the maintenance and plasticity of central nervous system (CNS) neurons. We tested the hypothesis that cortical neurons participate in redundant autocrine/paracrine systems. Three sets of studies determined the distribution of NGF-, BDNF-, and NT-3-expressing neurons, the frequency of neurons coexpressing NGF and BDNF, and the frequency of neurons expressing a neurotrophin and its associated high-affinity receptor. The distribution of NGF-, BDNF, and NT-3-immunoreactive neurons was identical. Neurotrophin-positive cells were parceled throughout the cortex, although the labeling frequency was not the same in all layers. More than 30% of the neurons in layers II/III, V, and VI were labeled, whereas only 5-10% of the neurons in layer IV was immunopositive for a neurotrophin. Some glia were also neurotrophin positive, particularly BDNF-positive glia. About 70% of the neurons in layers II/III and V coexpressed NGF and BDNF or coexpressed NGF and NT-3. Ligand-receptor colabeling was also common among cortical neurons. For example, nearly 70% of the NGF-, BDNF-, and NT-3-positive neurons in layer V colabeled with their respective high-affinity receptors, i.e., trkA, trkB, and trkC, respectively. Thus, (a) neurons express multiple neurotrophins and (b) cortical neurons (e.g., layer V neurons) contain the components required for autocrine/paracrine and/or anterograde communication (e.g., neurons in layer II/III support layer V neurons). These systems mean that the cortex is capable of regulating itself autonomously.
Collapse
Affiliation(s)
- A F Pitts
- Veterans Affairs Medical Center, and Department of Psychiatry, University of Iowa College of Medicine, Iowa City 52246-2208, USA
| | | |
Collapse
|