1
|
Deng C, Reinhard S, Hennlein L, Eilts J, Sachs S, Doose S, Jablonka S, Sauer M, Moradi M, Sendtner M. Impaired dynamic interaction of axonal endoplasmic reticulum and ribosomes contributes to defective stimulus-response in spinal muscular atrophy. Transl Neurodegener 2022; 11:31. [PMID: 35650592 PMCID: PMC9161492 DOI: 10.1186/s40035-022-00304-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 04/28/2022] [Indexed: 11/19/2022] Open
Abstract
Background Axonal degeneration and defects in neuromuscular neurotransmission represent a pathological hallmark in spinal muscular atrophy (SMA) and other forms of motoneuron disease. These pathological changes do not only base on altered axonal and presynaptic architecture, but also on alterations in dynamic movements of organelles and subcellular structures that are not necessarily reflected by static histopathological changes. The dynamic interplay between the axonal endoplasmic reticulum (ER) and ribosomes is essential for stimulus-induced local translation in motor axons and presynaptic terminals. However, it remains enigmatic whether the ER and ribosome crosstalk is impaired in the presynaptic compartment of motoneurons with Smn (survival of motor neuron) deficiency that could contribute to axonopathy and presynaptic dysfunction in SMA. Methods Using super-resolution microscopy, proximity ligation assay (PLA) and live imaging of cultured motoneurons from a mouse model of SMA, we investigated the dynamics of the axonal ER and ribosome distribution and activation. Results We observed that the dynamic remodeling of ER was impaired in axon terminals of Smn-deficient motoneurons. In addition, in axon terminals of Smn-deficient motoneurons, ribosomes failed to respond to the brain-derived neurotrophic factor stimulation, and did not undergo rapid association with the axonal ER in response to extracellular stimuli. Conclusions These findings implicate impaired dynamic interplay between the ribosomes and ER in axon terminals of motoneurons as a contributor to the pathophysiology of SMA and possibly also other motoneuron diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s40035-022-00304-2.
Collapse
Affiliation(s)
- Chunchu Deng
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078, Würzburg, Germany
| | - Sebastian Reinhard
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-University Wuerzburg, 97074, Würzburg, Germany
| | - Luisa Hennlein
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078, Würzburg, Germany
| | - Janna Eilts
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-University Wuerzburg, 97074, Würzburg, Germany
| | - Stefan Sachs
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-University Wuerzburg, 97074, Würzburg, Germany
| | - Sören Doose
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-University Wuerzburg, 97074, Würzburg, Germany
| | - Sibylle Jablonka
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078, Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, Julius-Maximilians-University Wuerzburg, 97074, Würzburg, Germany
| | - Mehri Moradi
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078, Würzburg, Germany.
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, 97078, Würzburg, Germany.
| |
Collapse
|
2
|
Priya R, Paredes MF, Karayannis T, Yusuf N, Liu X, Jaglin X, Graef I, Alvarez-Buylla A, Fishell G. Activity Regulates Cell Death within Cortical Interneurons through a Calcineurin-Dependent Mechanism. Cell Rep 2019; 22:1695-1709. [PMID: 29444424 DOI: 10.1016/j.celrep.2018.01.007] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 09/26/2017] [Accepted: 12/30/2017] [Indexed: 02/03/2023] Open
Abstract
We demonstrate that cortical interneurons derived from ventral eminences, including the caudal ganglionic eminence, undergo programmed cell death. Moreover, with the exception of VIP interneurons, this occurs in a manner that is activity-dependent. In addition, we demonstrate that, within interneurons, Calcineurin, a calcium-dependent protein phosphatase, plays a critical role in sequentially linking activity to maturation (E15-P5) and survival (P5-P20). Specifically, embryonic inactivation of Calcineurin results in a failure of interneurons to morphologically mature and prevents them from undergoing apoptosis. By contrast, early postnatal inactivation of Calcineurin increases apoptosis. We conclude that Calcineurin serves a dual role of promoting first the differentiation of interneurons and, subsequently, their survival.
Collapse
Affiliation(s)
- Rashi Priya
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, Smilow Research Center, New York University School of Medicine, 522 First Avenue, New York, NY 10016, USA
| | - Mercedes Francisca Paredes
- Department of Neurological Surgery, The Eli and Edythe Broad Center of Regeneration Medicine, Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Theofanis Karayannis
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, Smilow Research Center, New York University School of Medicine, 522 First Avenue, New York, NY 10016, USA
| | - Nusrath Yusuf
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, Smilow Research Center, New York University School of Medicine, 522 First Avenue, New York, NY 10016, USA; Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Xingchen Liu
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, Smilow Research Center, New York University School of Medicine, 522 First Avenue, New York, NY 10016, USA
| | - Xavier Jaglin
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, Smilow Research Center, New York University School of Medicine, 522 First Avenue, New York, NY 10016, USA; Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Isabella Graef
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Arturo Alvarez-Buylla
- Department of Neurological Surgery, The Eli and Edythe Broad Center of Regeneration Medicine, Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gord Fishell
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, Smilow Research Center, New York University School of Medicine, 522 First Avenue, New York, NY 10016, USA; Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE; Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Stanley Center at the Broad Institute, 75 Ames Street, Cambridge, MA 02142, USA.
| |
Collapse
|
3
|
Brandenburg JE, Gransee HM, Fogarty MJ, Sieck GC. Differences in lumbar motor neuron pruning in an animal model of early onset spasticity. J Neurophysiol 2018; 120:601-609. [PMID: 29718808 DOI: 10.1152/jn.00186.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Motor neuron (MN) development in early onset spasticity is poorly understood. For example, spastic cerebral palsy (sCP), the most common motor disability of childhood, is poorly predicted by brain imaging, yet research remains focused on the brain. By contrast, MNs, via the motor unit and neurotransmitter signaling, are the target of most therapeutic spasticity treatments and are the final common output of motor control. MN development in sCP is a critical knowledge gap, because the late embryonic and postnatal periods are not only when the supposed brain injury occurs but also are critical times for spinal cord neuromotor development. Using an animal model of early onset spasticity [ spa mouse (B6.Cg- Glrbspa/J) with a glycine (Gly) receptor mutation], we hypothesized that removal of effective glycinergic neurotransmitter inputs to MNs during development will influence MN pruning (including primary dendrites) and MN size. Spa (Glrb-/-) and wild-type (Glrb+/+) mice, ages 4-9 wk, underwent unilateral retrograde labeling of the tibialis anterior muscle MNs via peroneal nerve dip in tetramethylrhodamine. After 3 days, mice were euthanized and perfused with 4% paraformaldehyde, and the spinal cord was excised and processed for confocal imaging. Spa mice had ~61% fewer lumbar tibialis anterior MNs ( P < 0.01), disproportionately affecting larger MNs. Additionally, a ~23% reduction in tibialis anterior MN somal surface area ( P < 0.01) and a 12% increase in primary dendrites ( P = 0.046) were observed. Thus MN pruning and MN somal surface area are abnormal in early onset spasticity. Fewer and smaller MNs may contribute to the spastic phenotype. NEW & NOTEWORTHY Motor neuron (MN) development in early onset spasticity is poorly understood. In an animal model of early onset spasticity, spa mice, we found ~61% fewer lumbar tibialis anterior MNs compared with controls. This MN loss disproportionately affected larger MNs. Thus number and heterogeneity of the MN pool are decreased in spa mice, likely contributing to the spastic phenotype.
Collapse
Affiliation(s)
- Joline E Brandenburg
- Department of Physical Medicine and Rehabilitation, Mayo Clinic College of Medicine , Rochester, Minnesota.,Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine , Rochester, Minnesota
| | - Heather M Gransee
- Department of Anesthesiology, Mayo Clinic College of Medicine , Rochester, Minnesota
| | - Matthew J Fogarty
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine , Rochester, Minnesota.,School of Biomedical Sciences, The University of Queensland , Brisbane , Australia
| | - Gary C Sieck
- Department of Physical Medicine and Rehabilitation, Mayo Clinic College of Medicine , Rochester, Minnesota.,Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine , Rochester, Minnesota.,Department of Anesthesiology, Mayo Clinic College of Medicine , Rochester, Minnesota
| |
Collapse
|
4
|
Changes in neurotrophic factors of adult rat laryngeal muscles during nerve regeneration. Neuroscience 2016; 333:44-53. [DOI: 10.1016/j.neuroscience.2016.07.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 07/01/2016] [Accepted: 07/02/2016] [Indexed: 02/06/2023]
|
5
|
Aarse J, Herlitze S, Manahan-Vaughan D. The requirement of BDNF for hippocampal synaptic plasticity is experience-dependent. Hippocampus 2016; 26:739-51. [PMID: 26662461 PMCID: PMC5066736 DOI: 10.1002/hipo.22555] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2015] [Indexed: 12/05/2022]
Abstract
Brain‐derived neurotrophic factor (BDNF) supports neuronal survival, growth, and differentiation and has been implicated in forms of hippocampus‐dependent learning. In vitro, a specific role in hippocampal synaptic plasticity has been described, although not all experience‐dependent forms of synaptic plasticity critically depend on BDNF. Synaptic plasticity is likely to enable long‐term synaptic information storage and memory, and the induction of persistent (>24 h) forms, such as long‐term potentiation (LTP) and long‐term depression (LTD) is tightly associated with learning specific aspects of a spatial representation. Whether BDNF is required for persistent (>24 h) forms of LTP and LTD, and how it contributes to synaptic plasticity in the freely behaving rodent has never been explored. We examined LTP, LTD, and related forms of learning in the CA1 region of freely dependent mice that have a partial knockdown of BDNF (BDNF+/−). We show that whereas early‐LTD (<90min) requires BDNF, short‐term depression (<45 min) does not. Furthermore, BDNF is required for LTP that is induced by mild, but not strong short afferent stimulation protocols. Object‐place learning triggers LTD in the CA1 region of mice. We observed that object‐place memory was impaired and the object‐place exploration failed to induce LTD in BDNF+/− mice. Furthermore, spatial reference memory, that is believed to be enabled by LTP, was also impaired. Taken together, these data indicate that BDNF is required for specific, but not all, forms of hippocampal‐dependent information storage and memory. Thus, very robust forms of synaptic plasticity may circumvent the need for BDNF, rather it may play a specific role in the optimization of weaker forms of plasticity. The finding that both learning‐facilitated LTD and spatial reference memory are both impaired in BDNF+/− mice, suggests moreover, that it is critically required for the physiological encoding of hippocampus‐dependent memory. © 2015 The Authors Hippocampus Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Janna Aarse
- Department of Neurophysiology, Medical Faculty.,International Graduate School of Neuroscience
| | - Stefan Herlitze
- Faculty of Biology and Biotechnology, Department of Zoology and Neurobiology Ruhr University, Bochum, 44780 Bochum, Germany
| | - Denise Manahan-Vaughan
- Department of Neurophysiology, Medical Faculty.,International Graduate School of Neuroscience
| |
Collapse
|
6
|
Alder J, Fujioka W, Giarratana A, Wissocki J, Thakkar K, Vuong P, Patel B, Chakraborty T, Elsabeh R, Parikh A, Girn HS, Crockett D, Thakker-Varia S. Genetic and pharmacological intervention of the p75NTR pathway alters morphological and behavioural recovery following traumatic brain injury in mice. Brain Inj 2015; 30:48-65. [PMID: 26579945 DOI: 10.3109/02699052.2015.1088963] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PRIMARY OBJECTIVE Neurotrophin levels are elevated after TBI, yet there is minimal regeneration. It was hypothesized that the pro-neurotrophin/p75NTR pathway is induced more than the mature neurotrophin/Trk pathway and that interfering with p75 signalling improves recovery following TBI. RESEARCH DESIGN Lateral Fluid Percussion (LFP) injury was performed on wildtype and p75 mutant mice. In addition, TrkB agonist 7,8 Dihydroxyflavone or p75 antagonist TAT-Pep5 were tested. Western blot and immunohistochemistry revealed biochemical and cellular changes. Morris Water Maze and Rotarod tests demonstrated cognitive and vestibulomotor function. MAIN OUTCOMES AND RESULTS p75 was up-regulated and TrkB was down-regulated 1 day post-LFP. p75 mutant mice as well as mice treated with the p75 antagonist or the TrkB agonist exhibited reduced neuronal death and degeneration and less astrocytosis. The cells undergoing apoptosis appear to be neurons rather than glia. There was improved motor function and spatial learning in p75 mutant mice and mice treated with the p75 antagonist. CONCLUSIONS Many of the pathological and behavioural consequences of TBI might be due to activation of the pro-neurotrophin/p75 toxic pathway overriding the protective mechanisms of the mature neurotrophin/Trk pathway. Targeting p75 can be a novel strategy to counteract the damaging effects of TBI.
Collapse
MESH Headings
- Animals
- Apoptosis/physiology
- Astrocytes/metabolism
- Astrocytes/pathology
- Brain Injuries, Traumatic/drug therapy
- Brain Injuries, Traumatic/genetics
- Brain Injuries, Traumatic/metabolism
- Brain-Derived Neurotrophic Factor/metabolism
- Cognition/physiology
- Flavones/pharmacology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Mutant Strains
- Nerve Growth Factors/metabolism
- Receptor, trkB/agonists
- Receptor, trkB/genetics
- Receptor, trkB/metabolism
- Receptors, Nerve Growth Factor/antagonists & inhibitors
- Receptors, Nerve Growth Factor/genetics
- Receptors, Nerve Growth Factor/metabolism
- Sensory Receptor Cells/metabolism
- Sensory Receptor Cells/pathology
Collapse
Affiliation(s)
- Janet Alder
- a Rutgers Robert Wood Johnson Medical School , Piscataway , NJ , USA
| | - Wendy Fujioka
- a Rutgers Robert Wood Johnson Medical School , Piscataway , NJ , USA
| | - Anna Giarratana
- a Rutgers Robert Wood Johnson Medical School , Piscataway , NJ , USA
| | - Jenna Wissocki
- a Rutgers Robert Wood Johnson Medical School , Piscataway , NJ , USA
| | - Keya Thakkar
- a Rutgers Robert Wood Johnson Medical School , Piscataway , NJ , USA
| | - Phung Vuong
- a Rutgers Robert Wood Johnson Medical School , Piscataway , NJ , USA
| | - Bijal Patel
- a Rutgers Robert Wood Johnson Medical School , Piscataway , NJ , USA
| | | | - Rami Elsabeh
- a Rutgers Robert Wood Johnson Medical School , Piscataway , NJ , USA
| | - Ankit Parikh
- a Rutgers Robert Wood Johnson Medical School , Piscataway , NJ , USA
| | - Hartaj S Girn
- a Rutgers Robert Wood Johnson Medical School , Piscataway , NJ , USA
| | - David Crockett
- a Rutgers Robert Wood Johnson Medical School , Piscataway , NJ , USA
| | | |
Collapse
|
7
|
Klausmeyer A, Stern D, Wiese S. Isolation and culture of spinal cord motor neurons. ACTA ACUST UNITED AC 2015; 66:1.9.1-1.9.10. [PMID: 25727328 DOI: 10.1002/0471143030.cb0109s66] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Isolated spinal motoneurons are a powerful tool for studying basic mechanisms of neurite growth and survival. Since motoneurons are a minor population of developing spinal cord cells, they need to be purified and enriched to separate them from non-neuronal cells. Therefore, the particular feature of embryonic motoneurons to express the low affinity neurotrophin receptor p75(NTR) is used to separate the motoneurons from other contaminating cells. Two ways are described to isolate embryonic motoneurons: the basic protocol taking advantage of the ability of p75(NTR) to bind lectin, and an alternative method using an antibody against p75(NTR) for a panning procedure. These protocols comprise suggestions for the cultivation of the isolated motoneurons for experiments regarding neural outgrowth and survival as well as instruction for the preparation of proteins of the cells.
Collapse
Affiliation(s)
- Alice Klausmeyer
- Institute for Cell Morphology and Molecular Neurobiology, Group for Cell Biology, Ruhr-University, Bochum, Germany
| | - D Stern
- Institute for Cell Morphology and Molecular Neurobiology, Group for Cell Biology, Ruhr-University, Bochum, Germany
| | - S Wiese
- Institute for Cell Morphology and Molecular Neurobiology, Group for Cell Biology, Ruhr-University, Bochum, Germany
| |
Collapse
|
8
|
Tomàs J, Santafé MM, Garcia N, Lanuza MA, Tomàs M, Besalduch N, Obis T, Priego M, Hurtado E. Presynaptic membrane receptors in acetylcholine release modulation in the neuromuscular synapse. J Neurosci Res 2014; 92:543-54. [PMID: 24464361 DOI: 10.1002/jnr.23346] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 11/11/2013] [Accepted: 11/19/2013] [Indexed: 01/06/2023]
Abstract
Over the past few years, we have studied, in the mammalian neuromuscular junction (NMJ), the local involvement in transmitter release of the presynaptic muscarinic ACh autoreceptors (mAChRs), purinergic adenosine autoreceptors (P1Rs), and trophic factor receptors (TFRs; for neurotrophins and trophic cytokines) during development and in the adult. At any given moment, the way in which a synapse works is largely the logical outcome of the confluence of these (and other) metabotropic signalling pathways on intracellular kinases, which phosphorylate protein targets and materialize adaptive changes. We propose an integrated interpretation of the complementary function of these receptors in the adult NMJ. The activity of a given receptor group can modulate a given combination of spontaneous, evoked, and activity-dependent release characteristics. For instance, P1Rs can conserve resources by limiting spontaneous quantal leak of ACh (an A1 R action) and protect synapse function, because stimulation with adenosine reduces the magnitude of depression during repetitive activity. The overall outcome of the mAChRs seems to contribute to upkeep of spontaneous quantal output of ACh, save synapse function by decreasing the extent of evoked release (mainly an M2 action), and reduce depression. We have also identified several links among P1Rs, mAChRs, and TFRs. We found a close dependence between mAChR and some TFRs and observed that the muscarinic group has to operate correctly if the tropomyosin-related kinase B receptor (trkB) is also to operate correctly, and vice versa. Likewise, the functional integrity of mAChRs depends on P1Rs operating normally.
Collapse
Affiliation(s)
- Josep Tomàs
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Taylor AR, Gifondorwa DJ, Robinson MB, Strupe JL, Prevette D, Johnson JE, Hempstead BL, Oppenheim RW, Milligan CE. Motoneuron programmed cell death in response to proBDNF. Dev Neurobiol 2012; 72:699-712. [PMID: 21834083 PMCID: PMC3233653 DOI: 10.1002/dneu.20964] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Motoneurons (MN) as well as most neuronal populations undergo a temporally and spatially specific period of programmed cell death (PCD). Several factors have been considered to regulate the survival of MNs during this period, including availability of muscle-derived trophic support and activity. The possibility that target-derived factors may also negatively regulate MN survival has been considered, but not pursued. Neurotrophin precursors, through their interaction with p75(NTR) and sortilin receptors have been shown to induce cell death during development and following injury in the CNS. In this study, we find that muscle cells produce and secrete proBDNF. ProBDNF through its interaction with p75(NTR) and sortilin, promotes a caspase-dependent death of MNs in culture. We also provide data to suggest that proBDNF regulates MN PCD during development in vivo.
Collapse
Affiliation(s)
- AR Taylor
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine Winston-Salem, NC
| | - DJ Gifondorwa
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine Winston-Salem, NC
| | - MB Robinson
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine Winston-Salem, NC
| | - JL Strupe
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine Winston-Salem, NC
| | - D Prevette
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine Winston-Salem, NC
| | - JE Johnson
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine Winston-Salem, NC
| | - BL Hempstead
- Department of Medicine Cornell University Medical Center, NY
| | - RW Oppenheim
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine Winston-Salem, NC
- Interdisciplinary Neuroscience Program, Wake Forest University School of Medicine Winston-Salem, NC
- ALS Center, Wake Forest University School of Medicine Winston-Salem, NC
| | - CE Milligan
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine Winston-Salem, NC
- Interdisciplinary Neuroscience Program, Wake Forest University School of Medicine Winston-Salem, NC
- ALS Center, Wake Forest University School of Medicine Winston-Salem, NC
| |
Collapse
|
10
|
|
11
|
Gould TW, Oppenheim RW. Motor neuron trophic factors: therapeutic use in ALS? BRAIN RESEARCH REVIEWS 2011; 67:1-39. [PMID: 20971133 PMCID: PMC3109102 DOI: 10.1016/j.brainresrev.2010.10.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2010] [Revised: 10/12/2010] [Accepted: 10/18/2010] [Indexed: 12/12/2022]
Abstract
The modest effects of neurotrophic factor (NTF) treatment on lifespan in both animal models and clinical studies of Amyotropic Lateral Sclerosis (ALS) may result from any one or combination of the four following explanations: 1.) NTFs block cell death in some physiological contexts but not in ALS; 2.) NTFs do not rescue motoneurons (MNs) from death in any physiological context; 3.) NTFs block cell death in ALS but to no avail; and 4.) NTFs are physiologically effective but limited by pharmacokinetic constraints. The object of this review is to critically evaluate the role of both NTFs and the intracellular cell death pathway itself in regulating the survival of spinal and cranial (lower) MNs during development, after injury and in response to disease. Because the role of molecules mediating MN survival has been most clearly resolved by the in vivo analysis of genetically engineered mice, this review will focus on studies of such mice expressing reporter, null or other mutant alleles of NTFs, NTF receptors, cell death or ALS-associated genes.
Collapse
Affiliation(s)
- Thomas W Gould
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1010, USA.
| | | |
Collapse
|
12
|
Garcia N, Tomàs M, Santafe MM, Lanuza MA, Besalduch N, Tomàs J. Blocking p75NTR receptors alters polyinnervationz of neuromuscular synapses during development. J Neurosci Res 2011; 89:1331-41. [DOI: 10.1002/jnr.22620] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 12/20/2010] [Accepted: 01/13/2011] [Indexed: 01/17/2023]
|
13
|
Garcia N, Santafe MM, Tomàs M, Lanuza MA, Besalduch N, Tomàs J. Involvement of brain-derived neurotrophic factor (BDNF) in the functional elimination of synaptic contacts at polyinnervated neuromuscular synapses during development. J Neurosci Res 2010; 88:1406-19. [PMID: 20029969 DOI: 10.1002/jnr.22320] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We use immunohistochemistry to describe the localization of brain-derived neurotrophic factor (BDNF) and its receptors trkB and p75(NTR) in the neuromuscular synapses of postnatal rats (P6-P7) during the synapse elimination period. The receptor protein p75(NTR) is present in the nerve terminal, muscle cell and glial Schwann cell whereas BDNF and trkB proteins can be detected mainly in the pre- and postsynaptic elements. Exogenously applied BDNF (10 nM for 3 hr or 50 nM for 1 hr) increases ACh release from singly and dually innervated synapses. This effect may be specific for BDNF because the neurotrophin NT-4 (2-8 nM) does not modulate release at P6-P7. Blocking the receptors trkB and p75(NTR) (with K-252a and anti-p75-192-IgG, respectively) completely abolishes the potentiating effect of exogenous BDNF. In addition, exogenous BDNF transiently recruits functionally depressed silent terminals, and this effect seems to be mediated by trkB. Calcium ions, the L-type voltage-dependent calcium channels and protein kinase C are involved in BDNF-mediated nerve ending recruitment. Blocking experiments suggest that endogenous BDNF could operate through p75(NTR) receptors coupled to potentiate ACh release in all nerve terminals because the anti-p75-192-IgG reduces release. However, blocking the trkB receptor (K-252a) or neutralizing endogenous BDNF with the trkB-IgG fusion protein reveals a trkB-mediated release inhibition on almost mature strong endings in dual junctions. Taken together these results suggest that a BDNF-induced p75(NTR)-mediated ACh release potentiating mechanism and a BDNF-induced trkB-mediated release inhibitory mechanism may contribute to developmental synapse disconnection.
Collapse
Affiliation(s)
- N Garcia
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Carrer Sant Llorenç, Spain
| | | | | | | | | | | |
Collapse
|
14
|
Garcia N, Tomàs M, Santafe MM, Lanuza MA, Besalduch N, Tomàs J. Localization of brain-derived neurotrophic factor, neurotrophin-4, tropomyosin-related kinase b receptor, and p75 NTR receptor by high-resolution immunohistochemistry on the adult mouse neuromuscular junction. J Peripher Nerv Syst 2010; 15:40-9. [PMID: 20433604 DOI: 10.1111/j.1529-8027.2010.00250.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Neurotrophins and their receptors, the trk receptor tyrosine kinases (trks) and p75(NTR), are differentially expressed among the cell types that make up synapses. It is important to determine the precise location of these molecules involved in neurotransmission. Here we use immunostaining and Western blotting to study the localization and expression of neurotrophin brain-derived neurotrophic factor (BDNF) and neurotrophin-4 (NT-4) and the receptors tropomyosin-related kinase b (trkB) and p75(NTR) at the adult neuromuscular junction. Our confocal immunofluorescence results on the whole mounts of the mouse Levator auris longus muscle and on semithin cross-sections showed that BDNF, NT-4, trkB, and p75(NTR) were localized on the three cells in the neuromuscular synapse (motor axons, post-synaptic muscle and Schwann cells).
Collapse
Affiliation(s)
- Neus Garcia
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
| | | | | | | | | | | |
Collapse
|
15
|
Abstract
Neurotrophic factors (NTFs) are a pleiotropic group of secreted growth factors that regulate multiple aspects of neuronal development, including the regressive event of cell death. Skeletal muscleinnervating lower motoneurons (MNs) of the brain stem and spinal cord comprise one population of central neurons in which programmed cell death (PCD) during embryogenesis has been actively investigated, as much for reasons of technical facility as clinical relevance. The precise identity of NTF-dependent MNs has remained unclear, with most studies simply reporting losses or gains across the entire spinal cord or individual brain-stem nuclei. However, MNs are grouped into highly heterogenous populations based on transcriptional identity, target innervation, and physiological function. Therefore, recent work has focused on the effects of NTF overexpression or deletion on the survival of these MN subpopulations. Together with the recent progress attained in the generation of conditional mutant mice, in which the function of an NTF or its receptor can be eliminated specifically in MNs, these recent studies have begun to define the differential trophic requirements for MN subpopulations during PCD. The intent of this review is to summarize these recent findings and to discuss their significance with respect to neurotrophic theory.
Collapse
|
16
|
Bhargava S. Role of nerve growth factor and its receptor in the morphogenesis of neural tube in early chick embryo. Gen Comp Endocrinol 2007; 153:141-6. [PMID: 17418844 DOI: 10.1016/j.ygcen.2007.02.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2006] [Revised: 02/22/2007] [Accepted: 02/24/2007] [Indexed: 12/11/2022]
Abstract
Expression of p75 nerve growth factor receptor (p75(NTR)) in the early neurogenesis of chick embryo showed that nerve growth factor receptor (NGFR) is localized in presumptive neuroectoblast and endoblast in the chick gastrula but not in the mesoblast. By stages 9, 10, and 11, NGFR positive cells were located distinctly in the region where the neural folds converge, meet and fuse. NGFR expression was also seen in developing notochord and somites, wherein the reaction was localized on the cell surfaces. Strong p75(NTR) reaction was seen on the roof of the neural tube where it detaches from the head ectoderm by stage 12. The study revealed that p75(NTR) is co-expressed with NGF in the same developmental stage(s) and in areas, where cell death occurs during neuronal development. Further, when the endogenous levels of NGF signaling were blocked by anti-NGF antibody, abnormalities were observed at the anterior end of the neural tube formation. As a result, embryos showed open neural tubes and a few were bent on one side of the body axis. In a small proportion of embryos, diffused somites were observed. The findings supports and confirms our previous study that NGF signaling plays a significant role in the shaping of neural tube in chick embryos through p75(NTR)-NGF receptor.
Collapse
Affiliation(s)
- Shobha Bhargava
- Molecular Embryology Laboratory, Department of Zoology, University of Pune, Pune 411 007, India.
| |
Collapse
|
17
|
Brunet N, Tarabal O, Portero-Otín M, Oppenheim RW, Esquerda JE, Calderó J. Survival and death of mature avian motoneurons in organotypic slice culture: trophic requirements for survival and different types of degeneration. J Comp Neurol 2007; 501:669-90. [PMID: 17299760 DOI: 10.1002/cne.21157] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We have developed an organotypic culture technique that uses slices of chick embryo spinal cord, in which trophic requirements for long-term survival of mature motoneurons (MNs) were studied. Slices were obtained from E16 chick embryos and maintained for up to 28 days in vitro (DIV) in a basal medium. Under these conditions, most MNs died. To promote MN survival, 14 different trophic factors were assayed. Among these 14, glial cell line-derived neurotrophic factor (GDNF) and vascular endothelial growth factor were the most effective. GDNF was able to promote MN survival for at least 28 DIV. K(+) depolarization or caspase inhibition prevented MN death but also induced degenerative-like changes in rescued MNs. Agents that elevate cAMP levels promoted the survival of a proportion of MNs for at least 7 DIV. Examination of dying MNs revealed that, in addition to cells exhibiting a caspase-3-dependent apoptotic pattern, some MNs died by a caspase-3-independent mechanism and displayed autophagic vacuoles, an extremely convoluted nucleus, and a close association with microglia. This organotypic spinal cord slice culture may provide a convenient model for testing conditions that promote survival of mature-like MNs that are affected in late-onset MN disease such as amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Núria Brunet
- Unitat de Neurobiologia Cel.lular, Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina, Universitat de Lleida and IRB Lleida, 25008 Lleida, Catalonia, Spain
| | | | | | | | | | | |
Collapse
|
18
|
Frebel K, Wiese S. Signalling molecules essential for neuronal survival and differentiation. Biochem Soc Trans 2006; 34:1287-90. [PMID: 17073803 DOI: 10.1042/bst0341287] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Motoneurons are made in excess throughout development. Initial analysis of the mechanisms that lead to apoptotic cell death during later stages of development and the early postnatal period led to the discovery of neurotrophic factors. These factors comprise different families acting through different tyrosine kinase receptors. Intracellular signalling cascades that lead to the survival of neurons are, on the one hand, the Ras/Raf (Ras-activated factor)/MAPK (mitogen-activated protein kinase) pathway and, on the other, the PI3K (phosphoinositide 3-kinase)/Akt (protein kinase B) pathway. The initial thought of these factors acting as single molecules in separate cascades has been converted into a model in which the dynamics of interaction of these pathways and the subcellular diverse functions of the key regulators have been taken into account. Bag1 (Bcl-2-associated athanogene 1), a molecule that was originally found to act as a co-chaperone of Hsp70 (heat-shock protein 70), also interacts with B-Raf, C-Raf and Akt to phosphorylate Bad (Bcl-2/Bcl-XL-antagonist, causing cell death), a pro-apoptotic member of the Bcl-2 family, and leads to specific subcellular distribution of phosphorylated Akt and B-Raf. These functions lead to survival of embryonic neural stem cells and therefore serve as a key event to regulate the viability of these cells.
Collapse
Affiliation(s)
- K Frebel
- Institute for Clinical Neurobiology, Julius-Maximilians University of Würzburg, Josef Schneider Strasse 11, D97080 Würzburg, Germany
| | | |
Collapse
|
19
|
Pitts EV, Potluri S, Hess DM, Balice-Gordon RJ. Neurotrophin and Trk-mediated signaling in the neuromuscular system. Int Anesthesiol Clin 2006; 44:21-76. [PMID: 16849956 DOI: 10.1097/00004311-200604420-00004] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
20
|
Ko IK, Kato K, Iwata H. Parallel analysis of multiple surface markers expressed on rat neural stem cells using antibody microarrays. Biomaterials 2005; 26:4882-91. [PMID: 15763268 DOI: 10.1016/j.biomaterials.2004.11.049] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2004] [Accepted: 11/24/2004] [Indexed: 01/28/2023]
Abstract
Neural stem cells are the attractive cell source for functional regeneration of damaged central nervous tissues by means of cell transplantation or in situ induction of differentiated neural cells. Such stem cell therapies require the prospective identification and isolation of neural stem cells. However they are difficult due to limited information on surface markers. This study aimed at developing an antibody microarray that permits parallel analysis of multiple surface antigens expressed on neural stem cells present in a neurosphere-forming cell population. A microarray was prepared by micro-spotting antibodies directed to surface antigens and ligands for membrane-associated receptors onto the patterned monolayer of alkanethiols self-assembled on a gold-evaporated glass plate. Neurosphere-forming cells were subjected to a cell-binding assay on the microarray followed by immunofluorescent staining of nestin, an intracellular marker of neural stem cells. It was demonstrated that such a cell based assay facilitated to examine the specificity of surface antigens for nestin-positive neural stem cells. Furthermore, the microarray could also be used to assess proliferation capability of cells bound to individual spots. These results suggest that the microarray-based strategy will provide a useful tool for the parallel analysis of surface markers expressed on a specific cell type in a heterogeneous population.
Collapse
Affiliation(s)
- In Kap Ko
- Institute for Frontier Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | |
Collapse
|
21
|
Reddypalli S, Roll K, Lee HK, Lundell M, Barea-Rodriguez E, Wheeler EF. p75NTR-mediated signaling promotes the survival of myoblasts and influences muscle strength. J Cell Physiol 2005; 204:819-29. [PMID: 15754321 DOI: 10.1002/jcp.20330] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
During muscle development, the p75(NTR) is expressed transiently on myoblasts. The temporal expression pattern of the receptor raises the possibility that the receptor is influencing muscle development. To test this hypothesis, p75(NTR)-deficient mutant mice were tested for muscle strength by using a standard wire gripe strength test and were found to have significantly decreased strength relative to that of normal mice. When normal mybolasts were examined in vivo for expression of NGF receptors, p75(NTR) was detected on myoblasts but the high affinity NGF receptor, trk A, was not co-expressed with p75(NTR). In vitro, proliferating C2C12 and primary myoblasts co-expressed the p75(NTR) and MyoD, but immunofluorescent analysis of primary myoblasts and RT-PCR analysis of C2C12 mRNA revealed that myoblasts were devoid of trk A. In contrast to the cell death functions that characterize the p75(NTR) in neurons, p75(NTR)-positive primary and C2C12 myoblasts did not differentiate or undergo apoptosis in response to neurotrophins. Rather, myoblasts survived and even proliferated when grown at subconfluent densities in the presence of the neurotrophins. Furthermore, when myoblasts treated with NGF were lysed and immunoprecipitated with antibodies against phosphorylated I-kappaB and AKT, the cells contained increased levels of both phospho-proteins, both of which promote cell survival. By contrast, neurotrophin-treated myoblasts did not induce phosphorylation of Map Kinase p42/44 or p38, indicating the survival was not mediated by the trk A receptor. Taken together, the data indicate that the p75(NTR) mediates survival of myoblasts prior to differentiation and that the activity of this receptor during myogenesis is important for developing muscle.
Collapse
|
22
|
Mousavi K, Parry DJ, Jasmin BJ. BDNF rescues myosin heavy chain IIB muscle fibers after neonatal nerve injury. Am J Physiol Cell Physiol 2004; 287:C22-9. [PMID: 14973145 DOI: 10.1152/ajpcell.00583.2003] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neonatal sciatic nerve injury is known to result in an extensive loss of lumbar motor neurons as well as the disappearance of their respective muscle fibers in the hindlimb musculature. The loss of motor neurons and muscle fibers can be prevented by immediate administration of target-derived neurotrophic factors to the site of injury. In the present study, we investigated the role of ciliary neurotrophic factor (CNTF) and brain-derived neurotrophic factor (BDNF) in the survival and maturation of a subset of motor neurons innervating the extensor digitorum longus (EDL) and tibialis anterior (TA) muscles. We have shown that combined administration of CNTF and BDNF prevented the loss of motor units after neonatal nerve injury and contributed to the maintenance of muscle mass. Importantly, this combined neurotrophin regimen also prevented the disappearance of muscle fibers that express myosin heavy chain IIB (MyHC IIB) in both EDL and TA muscles 3 mo after neonatal sciatic nerve crush. In parallel studies, we observed a higher level of BDNF in EDL muscle during the critical period of development when motor neurons are highly susceptible to target removal. Given our previous findings that combined administration of CNTF with neurotrophin-3 (NT-3) or neurotrophin-4/5 (NT-4/5) did not result in the rescue of MyHC IIB fibers in EDL, the present results show the importance of muscle-derived BDNF in the survival and maturation of a subpopulation of motor neurons and of MyHC IIB muscle fibers during neonatal development of the neuromuscular system.
Collapse
Affiliation(s)
- Kambiz Mousavi
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | | | | |
Collapse
|
23
|
Vernon EM, Oppenheim RW, Johnson JE. Distinct muscle targets do not vary in the developmental regulation of brain-derived neurotrophic factor. J Comp Neurol 470:317-329,2004. J Comp Neurol 2004; 470:330-7. [PMID: 14755520 DOI: 10.1002/cne.20018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Developing neurons depend on many target-derived signals. One of these signals is the neurotrophin brain-derived neurotrophic factor (BDNF). Exogenous application of BDNF in vitro and in vivo rescues a population of lumbar motoneurons from programmed cell death. Given that BDNF does not rescue all motoneurons and that motoneurons differ in trophic factor receptor expression, subpopulations of motoneurons may have different sensitivities to the factor. These differences may be reflected in distinct target muscles specialized to produce different protein concentrations, or muscles may contain equal amounts of the factor and receptor expression determines motoneuron responsiveness. By using a sensitive electrochemiluminescent immunoassay (ECLIA), we measured normal developmental changes in BDNF protein concentration in anatomically and functionally distinct chick embryonic thigh muscles from E6 to E18. We found that there were no significant differences in BDNF protein concentration between muscles classified according to function (fast vs. slow) or anatomical position (flexor vs. extensor) and that the quantity of BDNF in the target did not appear to be activity dependent. These results suggest that, during development, the differences in the response of motoneurons to BDNF are not due to the anatomical or functional diversity of muscle targets. J. Comp. Neurol. 470:330-337, 2004.
Collapse
Affiliation(s)
- Elizabeth Marie Vernon
- Neuroscience Program, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | | | |
Collapse
|
24
|
Rabizadeh S, Bredesen DE. Ten years on: mediation of cell death by the common neurotrophin receptor p75(NTR). Cytokine Growth Factor Rev 2003; 14:225-39. [PMID: 12787561 DOI: 10.1016/s1359-6101(03)00018-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The common neurotrophin receptor p75(NTR) remains one of the most enigmatic of the tumor necrosis factor receptor (TNFR) superfamily: on the one hand, it displays a death domain and has been shown to be capable of mediating programmed cell death (PCD) upon ligand binding; on the other hand, its death domain is of type II (unlike that of Fas or TNFR I), and it has also been shown to be capable of mediating cell death in response to the withdrawal of ligand. Thus, p75(NTR) may function as a death receptor-similar to Fas or TNFR I-or a dependence receptor-similar to deleted in colorectal cancer (DCC) or uncoordinated gene-5 homologues 1-3 (UNC5H1-3). Here, we review the data relating to the mediation of PCD by p75(NTR), and suggest that one reasonable model for the apparently paradoxical effects of p75(NTR) is that this receptor functions as a "quality control" in that it is capable of mediating PCD in at least four situations: (1). withdrawal of neurotrophins; (2). exposure to mismatched neurotrophins; (3). exposure to unprocessed neurotrophins; and (4). exposure of inappropriately immature cells to neurotrophins. Results to date suggest that these functions are mediated through different underlying mechanisms, and that their respective signaling pathways are cell type and co-receptor dependent.
Collapse
Affiliation(s)
- Shahrooz Rabizadeh
- The Buck Institute for Age Research, 8001 Redwood Blvd, Novato, CA 94945-1400, USA.
| | | |
Collapse
|
25
|
Homma S, Yaginuma H, Vinsant S, Seino M, Kawata M, Gould T, Shimada T, Kobayashi N, Oppenheim RW. Differential expression of the GDNF family receptors RET and GFRalpha1, 2, and 4 in subsets of motoneurons: a relationship between motoneuron birthdate and receptor expression. J Comp Neurol 2003; 456:245-59. [PMID: 12528189 DOI: 10.1002/cne.10529] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Previous studies have demonstrated the expression of specific members of the glial cell line-derived neurotrophic factor (GDNF) receptor family alpha (GFRalpha) in subsets of motoneurons (MNs) in the developing mouse spinal cord. We examined the expression pattern of GFRalpha and RET in the avian lumbar spinal cord during the period of programmed cell death (PCD) of MNs by using double labeling in situ hybridization and immunohistochemistry. In the lateral motor column (LMC) of the lumbar spinal cord, a laminar organization of GFRalpha expression was observed: GFRalpha1-positive MNs were located in the medial LMC; GFRalpha1-, 2-, and 4-positive MNs were situated in the lateral LMC; and GFRalpha4-positive MNs were located in the intermediate LMC. The species of GFRalpha receptor that was expressed in MNs was found to be related to their birthdates. The expression of subpopulation-specific transcriptional factors was also used to define MNs that express a specific pattern of GFRalpha. This analysis suggests that motor pools as defined by these transcriptional factors have unique expression patterns of GFRalpha receptor. Early limb bud ablation did not affect the expression of GFRalpha in the spinal cord, indicating that regulation of receptor expression is independent of target-derived signals. Finally, GDNF mRNA expression was found in the limb during the PCD period of MNs. In conclusion, these results indicate that time of withdrawal from the mitotic cycle may specify the expression pattern of GFRalpha in subsets of MNs and that GDNF may function as a target-derived neurotrophic factor for specific subpopulations of MNs.
Collapse
Affiliation(s)
- Shunsaku Homma
- Department of Anatomy, School of Medicine, Fukushima Medical University, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Fraley GS, Ulibarri C. Development of androgen receptor and p75(NTR) mRNAs and peptides in the lumbar spinal cord of the gerbil. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2002; 137:101-14. [PMID: 12220702 DOI: 10.1016/s0165-3806(02)00412-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Development of sex differences in the spinal cord appears to be largely under the control of androgen and although neurotrophins may also have a role. Spinal cords of male and female neonatal gerbils (postnatal days 1, 5, 7, 10, 23) and adult gerbils (postnatal day 150) were examined to determine the relative temporal expression of androgen receptor (AR) and the low-affinity neurotrophin receptor (p75) mRNAs within the spinal nucleus of the bulbocavernosus (SNB) and dorsolateral nucleus (DLN). Furthermore, prepubertal male gerbils were placed into one of six gonadal hormone treatment groups at weaning: Either sham castrate, castrated with gonadal hormone replacement, or castrated without gonadal hormone replacement. Ten weeks later gerbils were aldehyde-perfused, spinal cords removed and processed for presence of AR and p75 immunoreactivity (ir) in motoneurons of the SNB and DLN. During neonatal development, there were significant increases in androgen receptor mRNA within the SNB and DLN. In the SNB, the increase in androgen receptor mRNA preceded the increase in p75 mRNA. Peripubertally, significantly more SNB than DLN motoneurons contained AR- and p75-ir. These data demonstrate that AR expression occurs along the same developmental time frame as the development of the SNB and DLN and the organizational effects of androgens on their development continues through puberty in the male gerbil.
Collapse
MESH Headings
- Aging/metabolism
- Animals
- Animals, Newborn
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Estradiol/metabolism
- Female
- Gerbillinae
- Immunohistochemistry
- Male
- Motor Neurons/cytology
- Motor Neurons/drug effects
- Motor Neurons/metabolism
- Peptides/genetics
- Peptides/metabolism
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Receptor, Nerve Growth Factor
- Receptors, Androgen/drug effects
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Receptors, Nerve Growth Factor/drug effects
- Receptors, Nerve Growth Factor/genetics
- Receptors, Nerve Growth Factor/metabolism
- Sex Characteristics
- Sex Differentiation/drug effects
- Sex Differentiation/genetics
- Spinal Cord/cytology
- Spinal Cord/growth & development
- Spinal Cord/metabolism
- Testosterone/metabolism
Collapse
Affiliation(s)
- Gregory Scott Fraley
- Program in Neuroscience and Department of Veterinary and Comparative Anatomy, Pharmacology, and Physiology, College of Veterinary Medicine, Washington State University, Pullman, WA 99161-6520 USA.
| | | |
Collapse
|
27
|
Bhargava S, Modak SP. Expression of nerve growth factor during the development of nervous system in early chick embryo. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2002; 136:43-9. [PMID: 12036516 DOI: 10.1016/s0165-3806(02)00346-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the chick gastrula, nerve growth factor (NGF) is localized to the endoblast mesoblast presumptive head ectoderm but not in the presumptive neuroectoblast. During early morphogenesis the dorsal body ectoderm presumptive neural crest cells exhibit strong NGF positive cell surface reaction. NGF appears to be a marker of cells participating in morphogenetic movements but not early neural differentiation. NGF is localized where neural folds fuse and cells die allowing detachment of the neural tube from head ectoderm as well as in dead cells in the neurocoele. NGF reactivity in cells lining the evaginated extremities of the optic vesicle the floor of the neural tube the splanchnopleure heart primordia the inner outer surfaces of somites is suggestive of the role of NGF in primitive organ shaping.
Collapse
Affiliation(s)
- Shobha Bhargava
- Molecular Embryology Laboratory, Department of Zoology, University of Pune, Pune 411 007, India.
| | | |
Collapse
|
28
|
Naeem A, Abbas L, Guthrie S. Comparison of the effects of HGF, BDNF, CT-1, CNTF, and the branchial arches on the growth of embryonic cranial motor neurons. JOURNAL OF NEUROBIOLOGY 2002; 51:101-14. [PMID: 11932952 DOI: 10.1002/neu.10048] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In the developing embryo, axon growth and guidance depend on cues that include diffusible molecules. We have shown previously that the branchial arches and hepatocyte growth factor (HGF) are growth-promoting and chemoattractant for young embryonic cranial motor axons. HGF is produced in the branchial arches of the embryo, but a number of lines of evidence suggest that HGF is unlikely to be the only factor involved in the growth and guidance of these axons. Here we investigate whether other neurotrophic factors could be involved in the growth of young cranial motor neurons in explant cultures. We find that brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF) and cardiotrophin-1 (CT-1) all promote the outgrowth of embryonic cranial motor neurons, while glial cell line-derived neurotrophic factor (GDNF) and neurotrophin-3 (NT-3) fail to affect outgrowth. We next examined whether HGF and the branchial arches had similar effects on motor neuron subpopulations at different axial levels. Our results show that HGF acts as a generalized rather than a specific neurotrophic factor and guidance cue for cranial motor neurons. Although the branchial arches also had general growth-promoting effects on all motor neuron subpopulations, they chemoattracted different axial levels differentially, with motor neurons from the caudal hindbrain showing the most striking response.
Collapse
Affiliation(s)
- Arifa Naeem
- MRC Centre for Developmental Neurobiology, 4th Floor New Hunt's House, King's College, Guy's Campus, London SE1 1 UL, UK
| | | | | |
Collapse
|
29
|
Nishimune H, Vasseur S, Wiese S, Birling MC, Holtmann B, Sendtner M, Iovanna JL, Henderson CE. Reg-2 is a motoneuron neurotrophic factor and a signalling intermediate in the CNTF survival pathway. Nat Cell Biol 2000; 2:906-14. [PMID: 11146655 DOI: 10.1038/35046558] [Citation(s) in RCA: 128] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cytokines that are related to ciliary neurotrophic factor (CNTF) are physiologically important survival factors for motoneurons, but the mechanisms by which they prevent neuronal cell death remain unknown. Reg-2/PAP I (pancreatitis-associated protein I), referred to here as Reg-2, is a secreted protein whose expression in motoneurons during development is dependent on cytokines. Here we show that CNTF-related cytokines induce Reg-2 expression in cultured motoneurons. Purified Reg-2 can itself act as an autocrine/paracrine neurotrophic factor for a subpopulation of motoneurons, by stimulating a survival pathway involving phosphatidylinositol-3-kinase, Akt kinase and NF-kappaB. Blocking Reg-2 expression in motoneurons using Reg-2 antisense adenovirus specifically abrogates the survival effect of CNTF on cultured motoneurons, indicating that Reg-2 expression is a necessary step in the CNTF survival pathway. Reg-2 shows a unique pattern of expression in late embryonic spinal cord: it is progressively upregulated in individual motoneurons on a cell-by-cell basis, indicating that only a fraction of motoneurons in a given motor pool may be exposed to cytokines. Thus, Reg-2 is a neurotrophic factor for motoneurons, and is itself an obligatory intermediate in the survival signalling pathway of CNTF-related cytokines.
Collapse
Affiliation(s)
- H Nishimune
- INSERM U.382, IBDM (CNRS - INSERM - Université de la Méditerranée), Campus de Luminy - Case 907, 13288 Marseille Cedex 9, France
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Gould TW, Burek MJ, Sosnowski JM, Prevette D, Oppenheim RW. The spatial-temporal gradient of naturally occurring motoneuron death reflects the time of prior exit from the cell cycle and position within the lateral motor column. Dev Biol 1999; 216:611-21. [PMID: 10642796 DOI: 10.1006/dbio.1999.9490] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Embryonic lumbar spinal motoneurons (MNs) are characterized by a period of programmed cell death (PCD) that spans several days and occurs in a rostrocaudal gradient. The generation of these MNs also takes place in a temporal-spatial gradient, such that MNs within rostral lumbar segments exit the cell cycle earlier and MNs within progressively caudal regions are born later. In vitro studies have shown that the latest born spinal MNs, presumably through the possession of endogenous "survival properties," are also the last to acquire their trophic dependence. If the birth date and therefore spinal cord location of lumbar spinal MNs influence the spatial-temporal pattern of PCD, then earlier born MNs should die sooner and be located more rostrally than those generated later. Alternatively, if the time at which MNs die during development is unrelated to their prior exit from the cell cycle, those born at various phases should die throughout the period of PCD. We report here that lumbar MNs generated during the earliest part (embryonic day 2-3) of the proliferative period in the developing chick spinal cord tend to die during the earliest stages of the PCD period and that MNs born in successive 12-h intervals die at correspondingly later periods during PCD. Furthermore, the spatial progression of PCD of these subpopulations of MNs occurs in a rostrocaudal gradient. Finally, while MNs do appear to die in a mediolateral gradient during the period of MN PCD, this pattern is only partly accounted for by MNs born in consecutive intervals. These data support the notion that the timing and rostrocaudal location of MNs undergoing PCD reflect their time of exit from the cell cycle.
Collapse
Affiliation(s)
- T W Gould
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | | | | | | | |
Collapse
|
31
|
Gould TW, Burek MJ, Ishihara R, Lo AC, Prevette D, Oppenheim RW. Androgens rescue avian embryonic lumbar spinal motoneurons from injury-induced but not naturally occurring cell death. JOURNAL OF NEUROBIOLOGY 1999; 41:585-95. [PMID: 10590181 DOI: 10.1002/(sici)1097-4695(199912)41:4<585::aid-neu13>3.0.co;2-#] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The regulation of survival of spinal motoneurons (MNs) has been shown to depend during development and after injury on a variety of neurotrophic molecules produced by skeletal muscle target tissue. Increasing evidence also suggests that other sources of trophic support prevent MNs from undergoing naturally occurring or injury-induced death. We have examined the role of endogenous and exogenous androgens on the survival of developing avian lumbar spinal MNs during their period of programmed cell death (PCD) between embryonic day (E)6 and E11 or after axotomy on E12. We found that although treatment with testosterone, dihydrotestosterone (DHT), or the androgen receptor antagonist flutamide (FL) failed to affect the number of these MNs during PCD, administration of DHT from E12 to E15 following axotomy on E12 significantly attenuated injury-induced MN death. This effect was inhibited by cotreatment with FL, whereas treatment with FL alone did not affect MN survival. Finally, we examined the spinal cord at various times during development and following axotomy on E12 for the expression of androgen receptor using the polyclonal PG-21 antibody. Our results suggest that exogenously applied androgens are capable of rescuing MNs from injury-induced cell death and that they act directly on these cells via an androgen receptor-mediated mechanism. By contrast, endogenous androgens do not appear to be involved in the regulation of normal PCD of developing avian MNs.
Collapse
Affiliation(s)
- T W Gould
- Department of Neurobiology and Anatomy and the Neuroscience Program, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, North Carolina 27157-1010, USA
| | | | | | | | | | | |
Collapse
|
32
|
Steljes TPV, Kinoshita Y, Wheeler EF, Oppenheim RW, von Bartheld CS. Neurotrophic factor regulation of developing avian oculomotor neurons: Differential effects of BDNF and GDNF. ACTA ACUST UNITED AC 1999. [DOI: 10.1002/(sici)1097-4695(19991105)41:2<295::aid-neu11>3.0.co;2-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
33
|
Dolcet X, Egea J, Soler RM, Martin-Zanca D, Comella JX. Activation of phosphatidylinositol 3-kinase, but not extracellular-regulated kinases, is necessary to mediate brain-derived neurotrophic factor-induced motoneuron survival. J Neurochem 1999; 73:521-31. [PMID: 10428047 DOI: 10.1046/j.1471-4159.1999.0730521.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Chick embryo spinal cord motoneurons develop a trophic response to some neurotrophins when they are maintained in culture in the presence of muscle extract. Thus, after 2 days in culture, brain-derived neurotrophic factor (BDNF) promotes motoneuron survival. In the present study we have analyzed the intracellular pathways that may be involved in the BDNF-induced motoneuron survival. We have observed that BDNF activated the extracellular-regulated kinase (ERK) mitogen-activated protein (MAP) kinase and the phosphatidylinositol (PI) 3-kinase pathways. To examine the contribution of these pathways to the survival effect triggered by BDNF, we used PD 98059, a specific inhibitor of MAP kinase kinase, and LY 294002, a selective inhibitor of PI 3-kinase. PD 98059, at doses that significantly reduced the phosphorylation of ERKs, did not show any prominent effect on neuronal survival. However, LY 294002 at doses that inhibited the phosphorylation of Akt, a down-stream element of the PI 3-kinase, completely abolished the motoneuron survival effects of BDNF. Moreover, cell death triggered by LY 294002 treatment exhibited features similar to those observed after muscle extract deprivation. Our results suggest that the PI 3-kinase pathway plays an important role in the survival effect triggered by BDNF on motoneurons, whereas activation of the ERK MAP kinase pathway is not relevant.
Collapse
Affiliation(s)
- X Dolcet
- Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina, Universitat de Lleida, Catalonia, Spain
| | | | | | | | | |
Collapse
|
34
|
Crockett DP, Wang L, Zhang RX, Egger MD. Distribution of the low-affinity neurotrophin receptor (p75) in the developing trigeminal brainstem complex in the rat. Anat Rec (Hoboken) 1999; 254:549-65. [PMID: 10203262 DOI: 10.1002/(sici)1097-0185(19990401)254:4<549::aid-ar10>3.0.co;2-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The low-affinity neurotrophin receptor (p75) binds all members of the neurotrophin family. In the rat, during the first week postpartum, dense p75-immunoreactivity (IR) is present throughout all components of the trigeminal brainstem complex (TBC), largely associated with primary sensory afferents. Within subnucleus caudalis (SpC) of the TBC, intense p75-IR is present in all laminae at birth. During the second and third postnatal weeks, p75-IR in SpC gradually fades within the deeper laminae, becoming generally restricted in the adult to laminae I and II. Similar declines in p75-IR intensity occur in the subnucleus oralis (SpO); in the SpO in the adult, p75-IR is confined to the dorsalmost portion of SpO. In subnucleus interpolaris, an emerging, vibrissa-related pattern of p75-IR is detectable on PD0 (first 24 hr postpartum), which becomes fully differentiated during PD4-PD7. However, this pattern gradually disappears during the third postnatal week. Ventrally in the nucleus principalis (PrV), a pattern of p75-IR that mirrors the topographical arrangement of the vibrissae is detectable by PD0-PD1, is fully differentiated by the end of the first postnatal week, and persists into adulthood. Perinatal unilateral sectioning of the infraorbital nerve on PD0-PD1, but not as late as PD4, disrupts p75-IR patterning in the adult PrV. Although p75 appears to be associated with primary afferent pattern formation, to determine whether it is essential, we examined mutant mice unable to form functional p75. In the TBC of these knockout mice, examined as adults, patterns of cytochrome oxidase staining (which parallel those of p75-IR) appeared to be normal. In summary, during early development, p75 is widely expressed in the TBC during periods of active synaptogenesis and pattern formation, whereas in the adult, its expression is restricted to association with populations of primary sensory afferents. However, the absence of functional p75 in genetically altered mice does not appear to prevent primary afferent pattern formation.
Collapse
Affiliation(s)
- D P Crockett
- Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, Piscataway 08854-5635, USA.
| | | | | | | |
Collapse
|
35
|
Bradley DM, Beaman FD, Moore DB, Kidd K, Heaton MB. Neurotrophic factors BDNF and GDNF protect embryonic chick spinal cord motoneurons from ethanol neurotoxicity in vivo. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 1999; 112:99-106. [PMID: 9974163 DOI: 10.1016/s0165-3806(98)00155-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Maternal consumption of ethanol is widely recognized as a leading cause of mental and physical deficits. Many populations of the central nervous system are affected by the teratogenic effects of ethanol. Neurotrophic factors (NTFs) have been shown to protect against ethanol neurotoxicity in culture, although there have been no demonstrations of such protection in vivo, in specific neuronal populations. Previous studies have demonstrated that ethanol is toxic to developing chick embryo motoneurons when administered from embryonic day 10 (E10) to E15. NTFs such as brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) have been shown to support developing spinal cord motoneurons, and when exogenously applied, decrease naturally occurring cell death, and protect against axotomy. The concurrent delivery of BDNF or GDNF with ethanol to the embryonic chick from E10 to E15 was designed to examine the capacity of these NTFs to provide in vivo neuroprotection for this ethanol-sensitive motoneuron population. Analysis of motoneuron numbers indicated that both BDNF and GDNF provided protection to developing spinal cord motoneurons from ethanol toxicity, restoring motoneuron numbers to control levels. This study represents the first demonstration of in vivo neuroprotection from ethanol toxicity with respect to specific neuronal populations.
Collapse
Affiliation(s)
- D M Bradley
- University of Florida Brain Institute, Gainesville 32610-0244, USA
| | | | | | | | | |
Collapse
|
36
|
Jungbluth S, Koentges G, Lumsden A. Coordination of early neural tube development by BDNF/trkB. Development 1997; 124:1877-85. [PMID: 9169835 DOI: 10.1242/dev.124.10.1877] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neurotrophins signal through members of the trk family of tyrosine kinase receptors and are known to regulate several neuronal properties. Although initially characterized by their ability to prevent naturally occurring cell death of subsets of neurons during development, neurotrophins can also regulate the proliferation and differentiation of precursor cells. Here we report a novel involvement of neurotrophins in early development of the neural tube. We demonstrate that a functional trkB receptor is expressed by motor neuron progenitors in the ventral neural tube and that treatment of ventral neural tube explants with the trkB ligand Brain-Derived Neurotrophic Factor (BDNF) leads to a significant increase in the number of motor neurons. The only BDNF expression detectable at this stage is by a subset of ventrally projecting interneurons in the dorsal neural tube; ablating this region in vivo leads to a reduction of motor neuron numbers. This loss can be prevented by simultaneous treatment with BDNF. We propose that BDNF produced by dorsal interneurons stimulates proliferation and/or differentiation of motor neuron progenitors after anterograde axonal transport and release in proximity to the trkB-expressing motor neuron precursors, thereby coordinating development between dorsal and ventral regions of the neural tube.
Collapse
Affiliation(s)
- S Jungbluth
- Department of Developmental Neurobiology, United Medical and Dental School, Guy's Hospital, London, UK
| | | | | |
Collapse
|