1
|
Conroy JN, Coulson EJ. High-affinity TrkA and p75 neurotrophin receptor complexes: A twisted affair. J Biol Chem 2022; 298:101568. [PMID: 35051416 PMCID: PMC8889134 DOI: 10.1016/j.jbc.2022.101568] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 10/27/2022] Open
Abstract
Neurotrophin signaling is essential for normal nervous system development and adult function. Neurotrophins are secreted proteins that signal via interacting with two neurotrophin receptor types: the multifaceted p75 neurotrophin receptor and the tropomyosin receptor kinase receptors. In vivo, neurons compete for the limited quantities of neurotrophins, a process that underpins neural plasticity, axonal targeting, and ultimately survival of the neuron. Thirty years ago, it was discovered that p75 neurotrophin receptor and tropomyosin receptor kinase A form a complex and mediate high-affinity ligand binding and survival signaling; however, despite decades of functional and structural research, the mechanism of modulation that yields this high-affinity complex remains unclear. Understanding the structure and mechanism of high-affinity receptor generation will allow development of pharmaceuticals to modulate this function for treatment of the many nervous system disorders in which altered neurotrophin expression or signaling plays a causative or contributory role. Here we re-examine the key older literature and integrate it with more recent studies on the topic of how these two receptors interact. We also identify key outstanding questions and propose a model of inside-out allosteric modulation to assist in resolving the elusive high-affinity mechanism and complex.
Collapse
Affiliation(s)
- Jacinta N Conroy
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Elizabeth J Coulson
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia; Clem Jones Centre for Ageing and Disease Research, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
2
|
Franco ML, Nadezhdin KD, Light TP, Goncharuk SA, Soler-Lopez A, Ahmed F, Mineev KS, Hristova K, Arseniev AS, Vilar M. Interaction between the transmembrane domains of neurotrophin receptors p75 and TrkA mediates their reciprocal activation. J Biol Chem 2021; 297:100926. [PMID: 34216618 PMCID: PMC8327350 DOI: 10.1016/j.jbc.2021.100926] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 11/23/2022] Open
Abstract
The neurotrophin receptors p75 and tyrosine protein kinase receptor A (TrkA) play important roles in the development and survival of the nervous system. Biochemical data suggest that p75 and TrkA reciprocally regulate the activities of each other. For instance, p75 is able to regulate the response of TrkA to lower concentrations of nerve growth factor (NGF), and TrkA promotes shedding of the extracellular domain of p75 by α-secretases in a ligand-dependent manner. The current model suggests that p75 and TrkA are regulated by means of a direct physical interaction; however, the nature of such interaction has been elusive thus far. Here, using NMR in micelles, multiscale molecular dynamics, FRET, and functional studies, we identified and characterized the direct interaction between TrkA and p75 through their respective transmembrane domains (TMDs). Molecular dynamics of p75-TMD mutants suggests that although the interaction between TrkA and p75 TMDs is maintained upon mutation, a specific protein interface is required to facilitate TrkA active homodimerization in the presence of NGF. The same mutations in the TMD protein interface of p75 reduced the activation of TrkA by NGF as well as reducing cell differentiation. In summary, we provide a structural model of the p75-TrkA receptor complex necessary for neuronal development stabilized by TMD interactions.
Collapse
Affiliation(s)
- María L Franco
- Unit of Molecular Basis of Neurodegeneration, Institute of Biomedicine CSIC, València, Spain
| | - Kirill D Nadezhdin
- Department of Structural Biology, Laboratory of NMR-Spectroscopy, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russian Federation
| | - Taylor P Light
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sergey A Goncharuk
- Department of Structural Biology, Laboratory of NMR-Spectroscopy, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russian Federation; Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Moscow, Russian Federation
| | - Andrea Soler-Lopez
- Unit of Molecular Basis of Neurodegeneration, Institute of Biomedicine CSIC, València, Spain
| | - Fozia Ahmed
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Konstantin S Mineev
- Department of Structural Biology, Laboratory of NMR-Spectroscopy, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russian Federation; Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Moscow, Russian Federation
| | - Kalina Hristova
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Alexander S Arseniev
- Department of Structural Biology, Laboratory of NMR-Spectroscopy, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russian Federation.
| | - Marçal Vilar
- Unit of Molecular Basis of Neurodegeneration, Institute of Biomedicine CSIC, València, Spain.
| |
Collapse
|
3
|
Asuni GP, Speidell A, Mocchetti I. Neuronal apoptosis induced by morphine withdrawal is mediated by the p75 neurotrophin receptor. J Neurochem 2021; 158:169-181. [PMID: 33742683 PMCID: PMC10176599 DOI: 10.1111/jnc.15355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/22/2021] [Accepted: 03/15/2021] [Indexed: 01/01/2023]
Abstract
Morphine withdrawal evokes neuronal apoptosis through mechanisms that are still under investigation. We have previously shown that morphine withdrawal increases the levels of pro-brain-derived neurotrophic factor (BDNF), a proneurotrophin that promotes neuronal apoptosis through the binding and activation of the pan-neurotrophin receptor p75 (p75NTR). In this work, we sought to examine whether morphine withdrawal increases p75NTR-driven signaling events. We employed a repeated morphine treatment-withdrawal paradigm in order to investigate biochemical and histological indicators of p75NTR-mediated neuronal apoptosis in mice. We found that repeated cycles of spontaneous morphine withdrawal promote an accumulation of p75NTR in hippocampal synapses. At the same time, TrkB, the receptor that is crucial for BDNF-mediated synaptic plasticity in the hippocampus, was decreased, suggesting that withdrawal alters the neurotrophin receptor environment to favor synaptic remodeling and apoptosis. Indeed, we observed evidence of neuronal apoptosis in the hippocampus, including activation of c-Jun N-terminal kinase (JNK) and increased active caspase-3. These effects were not seen in saline or morphine-treated mice which had not undergone withdrawal. To determine whether p75NTR was necessary in promoting these outcomes, we repeated these experiments in p75NTR heterozygous mice. The lack of one p75NTR allele was sufficient to prevent the increases in phosphorylated JNK and active caspase-3. Our results suggest that p75NTR participates in the neurotoxic and proinflammatory state evoked by morphine withdrawal. Because p75NTR activation negatively influences synaptic repair and promotes cell death, preventing opioid withdrawal is crucial for reducing neurotoxic mechanisms accompanying opioid use disorders.
Collapse
Affiliation(s)
- Gino P. Asuni
- Laboratory of Preclinical Neurobiology, Georgetown University Medical Center, Washington DC, USA
| | - Andrew Speidell
- Laboratory of Preclinical Neurobiology, Georgetown University Medical Center, Washington DC, USA
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington DC, USA
| | - Italo Mocchetti
- Laboratory of Preclinical Neurobiology, Georgetown University Medical Center, Washington DC, USA
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington DC, USA
| |
Collapse
|
4
|
Howard L, Wyatt S, Davies AM. Neuregulin-4 contributes to the establishment of cutaneous sensory innervation. Dev Neurobiol 2021; 81:139-148. [PMID: 33369884 PMCID: PMC8365430 DOI: 10.1002/dneu.22803] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 11/30/2022]
Abstract
Recent work has shown that neuregulin‐4 (NRG4) is a physiological regulator of the growth of sympathetic axons and CNS dendrites in the developing nervous system. Here, we have investigated whether NRG4 plays a role in sensory axon growth and the establishment of cutaneous sensory innervation. Imaging early nerve fibers in the well‐characterized cutaneous trigeminal territory, the brachial plexus, and thorax revealed very marked and highly significant decreases in nerve fiber length and branching density in Nrg4−/− embryos compared with Nrg4+/+ littermates. NRG4 promoted neurotrophin‐independent sensory axon growth from correspondingly early trigeminal ganglion and DRG neurons in culture but not from enteroceptive nodose ganglion neurons. High levels of Nrg4 mRNA were detected in cutaneous tissues but not in sensory ganglia. Our findings suggest that NRG4 is an important target‐derived factor that participates in the establishment of early cutaneous sensory innervation.
Collapse
Affiliation(s)
- Laura Howard
- School of Biosciences, Cardiff University, Cardiff, UK
| | - Sean Wyatt
- School of Biosciences, Cardiff University, Cardiff, UK
| | - Alun M Davies
- School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
5
|
Qin Z, Gonsalvez DG, Wood RJ, Daemi F, Yoo S, Ivanusic JJ, Coulson EJ, Murray SS, Xiao J. Partial deletion of p75 NTR in large-diameter DRG neurons exerts no influence upon the survival of peripheral sensory neurons in vivo. J Neurosci Res 2020; 98:1987-1998. [PMID: 32585763 DOI: 10.1002/jnr.24665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 04/20/2020] [Accepted: 05/14/2020] [Indexed: 01/19/2023]
Abstract
The p75 neurotrophin receptor (p75NTR ) is required for maintaining peripheral sensory neuron survival and function; however, the underlying cellular mechanism remains unclear. The general view is that expression of p75NTR by the neuron itself is required for maintaining sensory neuron survival and myelination in the peripheral nervous system (PNS). Adopting a neuronal-specific conditional knockout strategy, we demonstrate the partial depletion of p75NTR in neurons exerts little influence upon maintaining sensory neuron survival and peripheral nerve myelination in health and after demyelinating neuropathy. Our data show that the density and total number of dorsal root ganglion (DRG) neurons in 2-month-old mice is not affected following the deletion of p75NTR in large-diameter myelinating neurons, as assessed by stereology. Adopting experimental autoimmune neuritis induced in adult male mice, an animal model of demyelinating peripheral neuropathy, we identify that deleting p75NTR in myelinating neurons exerts no influence upon the disease progression, the total number of DRG neurons, and the extent of myelin damage in the sciatic nerve, indicating that the expression of neuronal p75NTR is not essential for maintaining peripheral neuron survival and myelination after a demyelinating insult in vivo. Together, results of this study suggest that the survival and myelination of peripheral sensory neurons is independent of p75NTR expressed by a subtype of neurons in vivo. Thus, our findings provide new insights into the mechanism underpinning p75NTR -mediated neuronal survival in the PNS.
Collapse
Affiliation(s)
- Zuoheng Qin
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - David G Gonsalvez
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Rhiannon J Wood
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Fatemeh Daemi
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Sangwon Yoo
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Jason J Ivanusic
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Elizabeth J Coulson
- School of Biomedical Sciences, Queensland University, Brisbane, QLD, Australia
| | - Simon S Murray
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Junhua Xiao
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
6
|
Association of distinct type 1 bone morphogenetic protein receptors with different molecular pathways and survival outcomes in neuroblastoma. Neuronal Signal 2020; 4:NS20200006. [PMID: 32714600 PMCID: PMC7366490 DOI: 10.1042/ns20200006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 12/21/2022] Open
Abstract
Neuroblastoma (NB) is a paediatric cancer that arises in the sympathetic nervous system. Patients with stage 4 tumours have poor outcomes and 20% of high-risk cases have MYCN amplification. The bone morphogenetic proteins (BMPs) play roles in sympathetic neuritogenesis, by signalling through bone morphogenetic protein receptor (BMPR)2 and either BMPR1A or BMPR1B. Alterations in BMPR2 expression have been reported in NB; it is unknown if the expression of BMPR1A or BMPR1B is altered. We report lower BMPR2 and BMPR1B, and higher BMPR1A, expression in stage 4 and in MYCN-amplified NB. Kaplan–Meier plots showed that high BMPR2 or BMPR1B expression was linked to better survival, while high BMPR1A was linked to worse survival. Gene ontology enrichment and pathway analyses revealed that BMPR2 and BMPR1B co-expressed genes were enriched in those associated with NB differentiation. BMPR1A co-expressed genes were enriched in those associated with cell proliferation. Moreover, the correlation between BMPR2 and BMPR1A was strengthened, while the correlation between BMPR2 and BMPR1B was lost, in MYCN-amplified NB. This suggested that differentiation should decrease BMPR1A and increase BMPR1B expression. In agreement, nerve growth factor treatment of cultured sympathetic neurons decreased Bmpr1a expression and increased Bmpr1b expression. Overexpression of dominant negative BMPR1B, treatment with a BMPR1B inhibitor and treatment with GDF5, which signals via BMPR1B, showed that BMPR1B signalling is required for optimal neuritogenesis in NB cells, suggesting that loss of BMPR1B may alter neuritogenesis. The present study shows that expression of distinct BMPRs is associated with different survival outcomes in NB.
Collapse
|
7
|
Calhan OY, Wyatt S, Davies AM. CD40L reverse signaling suppresses prevertebral sympathetic axon growth and tissue innervation. Dev Neurobiol 2020; 79:949-962. [PMID: 32077240 PMCID: PMC7187455 DOI: 10.1002/dneu.22735] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/13/2020] [Accepted: 02/17/2020] [Indexed: 12/30/2022]
Abstract
CD40‐activated CD40L reverse signaling is a major physiological regulator of the growth of neural processes in the developing nervous system. Previous work on superior cervical ganglion (SCG) neurons of the paravertebral sympathetic chain has shown that CD40L reverse signaling enhances NGF‐promoted axon growth and tissue innervation. Here we show that CD40L reverse signaling has the opposite function in prevertebral ganglion (PVG) sympathetic neurons. During a circumscribed perinatal window of development, PVG neurons cultured from Cd40–/– mice had substantially larger, more exuberant axon arbors in the presence of NGF than PVG neurons cultured from wild‐type mice. Tissues that receive their sympathetic innervation from PVG neurons were markedly hyperinnervated in Cd40–/– mice compared with wild‐type mice. The exuberant axonal growth phenotype of cultured CD40‐deficient perinatal PVG neurons was pared back to wild‐type levels by activating CD40L reverse signaling with a CD40‐Fc chimeric protein, but not by activating CD40 forward signaling with CD40L. The co‐expression of CD40 and CD40L in PVG neurons suggests that these proteins engage in an autocrine signaling loop in these neurons. Our work shows that CD40L reverse signaling is a physiological regulator of NGF‐promoted sympathetic axon growth and tissue innervation with opposite effects in paravertebral and prevertebral neurons.
Collapse
Affiliation(s)
| | - Sean Wyatt
- School of Biosciences, Cardiff University, Cardiff, UK
| | | |
Collapse
|
8
|
Howard L, McWilliams TG, Wyatt S, Davies AM. CD40 forward signalling is a physiological regulator of early sensory axon growth. Development 2019; 146:dev.176495. [PMID: 31488565 PMCID: PMC6765180 DOI: 10.1242/dev.176495] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 08/14/2019] [Indexed: 01/19/2023]
Abstract
Multiple members of the tumour necrosis factor superfamily (TNFSF) regulate the growth and branching of neural processes late in development, when neurons are establishing and refining connections. Here, we present the first evidence that a TNFSF member acts much earlier in development, when axons are growing to their targets. CD40L transiently enhanced axon growth from embryonic mouse DRG neurons cultured at this early stage. Early spinal nerves of embryos lacking the CD40L receptor (Cd40−/− mice) were significantly shorter in vivo than those of Cd40+/+ littermates. CD40L was synthesized in early DRG targets and was co-expressed with CD40 in early DRG neurons. Whereas CD40L enhanced early axon growth independently of neurotrophins, disruption of a CD40L/CD40 autocrine loop impaired early neurotrophin-promoted axon growth. In marked contrast to the widespread regulation of axon and dendrite growth by CD40L reverse signalling later in development, CD40-Fc, which activates reverse signalling, had no effect on early sensory axon growth. These results suggest that CD40 forward signalling is a novel physiological regulator of early axon growth that acts by target-derived and autocrine mechanisms. Summary: CD40L, a novel physiological regulator of early sensory axon growth at the stage when sensory axons are growing to their targets, activates CD40 forward signalling by target-derived and autocrine mechanisms.
Collapse
Affiliation(s)
- Laura Howard
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AT, UK
| | - Thomas G McWilliams
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AT, UK
| | - Sean Wyatt
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AT, UK
| | - Alun M Davies
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AT, UK
| |
Collapse
|
9
|
Chen XQ, Mobley WC. Exploring the Pathogenesis of Alzheimer Disease in Basal Forebrain Cholinergic Neurons: Converging Insights From Alternative Hypotheses. Front Neurosci 2019; 13:446. [PMID: 31133787 PMCID: PMC6514132 DOI: 10.3389/fnins.2019.00446] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/18/2019] [Indexed: 01/01/2023] Open
Abstract
Alzheimer disease (AD) represents an oncoming epidemic that without an effective treatment promises to exact extraordinary financial and emotional burdens (Apostolova, 2016). Studies of pathogenesis are essential for defining critical molecular and cellular events and for discovering therapies to prevent or mitigate their effects. Through studies of neuropathology, genetic and cellular, and molecular biology recent decades have provided many important insights. Several hypotheses have been suggested. Documentation in the 1980s of selective loss of cholinergic neurons of the basal forebrain, followed by clinical improvement in those treated with inhibitors of acetylycholinesterase, supported the "cholinergic hypothesis of age-related cognitive dysfunction" (Bartus et al., 1982). A second hypothesis, prompted by the selective loss of cholinergic neurons and the discovery of central nervous system (CNS) neurotrophic factors, including nerve growth factor (NGF), prompted the "deficient neurotrophic hypothesis" (Chen et al., 2018). The most persuasive hypothesis, the amyloid cascade hypothesis first proposed more than 25 years ago (Selkoe and Hardy, 2016), is supported by a wealth of observations. Genetic studies were exceptionally important, pointing to increased dose of the gene for the amyloid precursor protein (APP) in Down syndrome (DS) and a familial AD (FAD) due to duplication of APP and to mutations in APP and in the genes for Presenilin 1 and 2 (PSEN1, 2), which encode the γ-secretase enzyme that processes APP (Dorszewska et al., 2016). The "tau hypothesis" noted the prominence of tau-related pathology and its correlation with dementia (Kametani and Hasegawa, 2018). Recent interest in induction of microglial activation in the AD brain, as well as other manifestations of inflammation, supports the "inflammatory hypothesis" (Mcgeer et al., 2016). We place these findings in the context of the selective, but by no means unique, involvement of BFCNs and their trophic dependence on NGF signaling and speculate as to how pathogenesis in these neurons is initiated, amplified and ultimately results in their dysfunction and death. In so doing we attempt to show how the different hypotheses for AD may interact and reinforce one another. Finally, we address current attempts to prevent and/or treat AD in light of advances in understanding pathogenetic mechanisms and suggest that studies in the DS population may provide unique insights into AD pathogenesis and treatment.
Collapse
Affiliation(s)
- Xu-Qiao Chen
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| | - William C. Mobley
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
10
|
Erice C, Calhan OY, Kisiswa L, Wyatt S, Davies AM. Regional Differences in the Contributions of TNF Reverse and Forward Signaling to the Establishment of Sympathetic Innervation. Dev Neurobiol 2019; 79:317-334. [PMID: 31004466 PMCID: PMC6563146 DOI: 10.1002/dneu.22680] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/05/2019] [Accepted: 04/07/2019] [Indexed: 01/24/2023]
Abstract
Members of the TNF and TNF receptor superfamilies acting by both forward and reverse signaling are increasingly recognized as major physiological regulators of axon growth and tissue innervation in development. Studies of the experimentally tractable superior cervical ganglion (SCG) neurons and their targets have shown that only TNF reverse signaling, not forward signaling, is a physiological regulator of sympathetic innervation. Here, we compared SCG neurons and their targets with prevertebral ganglion (PVG) neurons and their targets. Whereas all SCG targets were markedly hypoinnervated in both TNF-deficient and TNFR1-deficient mice, PVG targets were not hypoinnervated in these mice and one PVG target, the spleen, was significantly hyperinnervated. These in vivo regional differences in innervation density were related to in vitro differences in the responses of SCG and PVG neurons to TNF reverse and forward signaling. Though TNF reverse signaling enhanced SCG axon growth, it did not affect PVG axon growth. Whereas activation of TNF forward signaling in PVG axons inhibited growth, TNF forward signaling could not be activated in SCG axons. These latter differences in the response of SCG and PVG axons to TNF forward signaling were related to TNFR1 expression, whereas PVG axons expressed TNFR1, SCG axons did not. These results show that both TNF reverse and forward signaling are physiological regulators of sympathetic innervation in different tissues.
Collapse
Affiliation(s)
- Clara Erice
- School of BiosciencesCardiff UniversityMuseum AvenueCardiffCF10 3ATWales
- Present address:
Sandra Rotman Centre for Global HealthUniversity Health Network: Toronto General HospitalTorontoOntarioCanada
| | - O. Yipkin Calhan
- School of BiosciencesCardiff UniversityMuseum AvenueCardiffCF10 3ATWales
| | - Lilian Kisiswa
- School of BiosciencesCardiff UniversityMuseum AvenueCardiffCF10 3ATWales
- Present address:
Department of PhysiologyNational University of SingaporeSingapore117597Singapore
| | - Sean Wyatt
- School of BiosciencesCardiff UniversityMuseum AvenueCardiffCF10 3ATWales
| | - Alun M. Davies
- School of BiosciencesCardiff UniversityMuseum AvenueCardiffCF10 3ATWales
| |
Collapse
|
11
|
Choucry AM, Al-Shorbagy MY, Attia AS, El-Abhar HS. Pharmacological Manipulation of Trk, p75NTR, and NGF Balance Restores Memory Deficit in Global Ischemia/Reperfusion Model in Rats. J Mol Neurosci 2019; 68:78-90. [PMID: 30863991 DOI: 10.1007/s12031-019-01284-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 02/21/2019] [Indexed: 01/17/2023]
Abstract
Long-term memory impairment is reported in more than 50% of cardiac arrest survivors. Monosialoganglioside (GM1) provided neuroprotection in experimental models of stroke but failed to replicate its promise clinically for unknown reasons. GM1 stimulates the release of nerve growth factor (NGF), which is synthesized as a precursor protein (pro-NGF) that either mediates apoptosis through the p75 neurotrophin receptor (p75NTR) or is cleaved by the protease furin (FUR) to yield mature NGF, the latter supporting survival through tropomyosin kinase receptor (Trk). The flavanol epicatechin (EPI) inhibits p75NTR-mediated signaling and apoptosis by pro-NGF. The aim of the current work is to test whether these two drugs affect, or communicate with, each other in the setting of CNS injuries. Using the two-vessel occlusion model of global ischemia/reperfusion (I/R), we tested if pharmacological modulation of Trk, p75NTR, and NGF balance with GM1, EPI, and their combination, can correct the memory deficit that follows this insult. Finally, we tested if FUR insufficiency and/or p75NTR-mediated apoptosis negatively affect the neurotherapeutic effect of GM1. Key proteins for Trk and p75NTR, FUR, and both forms of NGF were assessed. All treatment regiments successfully improved spatial memory retention and acquisition. A week after the insult, most Trk and p75NTR proteins were normal, but pro/mature NGF ratio remained sharply elevated and was associated with the poorest memory performance. Pharmacological correction of this balance was achieved by reinforcing Trk and p75NTR signaling. GM1 increased FUR levels, while concomitant administration of EPI weakened GM1 effect on pro-survival Trk and p75NTR mediators. GM1 neuroprotection is therefore not limited by FUR but could be dependent on p75NTR. Graphical Abstract "."
Collapse
Affiliation(s)
- Ali Mohamed Choucry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini st., Cairo, 11562, Egypt.,Department of Biochemistry, Graduate School of Medicine and Pharmaceutical Sciences, Toyama University, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Muhammad Yusuf Al-Shorbagy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini st., Cairo, 11562, Egypt. .,School of Pharmacy, New Giza University, Giza, Egypt.
| | - Ahmed Sherif Attia
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo, 11562, Egypt
| | - Hanan Salah El-Abhar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini st., Cairo, 11562, Egypt
| |
Collapse
|
12
|
Howard L, Wosnitzka E, Okakpu D, White MA, Wyatt S, Davies AM. TWE-PRIL reverse signalling suppresses sympathetic axon growth and tissue innervation. Development 2018; 145:dev.165936. [PMID: 30337376 PMCID: PMC6262789 DOI: 10.1242/dev.165936] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 10/10/2018] [Indexed: 12/14/2022]
Abstract
TWE-PRIL is a naturally occurring fusion protein of components of two TNF superfamily members: the extracellular domain of APRIL; and the intracellular and transmembrane domains of TWEAK with no known function. Here, we show that April−/− mice (which lack APRIL and TWE-PRIL) exhibited overgrowth of sympathetic fibres in vivo, and sympathetic neurons cultured from these mice had significantly longer axons than neurons cultured from wild-type littermates. Enhanced axon growth from sympathetic neurons cultured from April−/− mice was prevented by expressing full-length TWE-PRIL in these neurons but not by treating them with soluble APRIL. Soluble APRIL, however, enhanced axon growth from the sympathetic neurons of wild-type mice. siRNA knockdown of TWE-PRIL but not siRNA knockdown of APRIL alone also enhanced axon growth from wild-type sympathetic neurons. Our work reveals the first and physiologically relevant role for TWE-PRIL and suggests that it mediates reverse signalling. Summary:In vivo and in vitro studies of superior cervical ganglion neurons of April−/− mice reveal that TWE-PRIL is a physiological regulator of NGF-promoted sympathetic axon growth, acting as a reverse signalling receptor.
Collapse
Affiliation(s)
- Laura Howard
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| | - Erin Wosnitzka
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| | - Darian Okakpu
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| | - Matthew A White
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| | - Sean Wyatt
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| | - Alun M Davies
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| |
Collapse
|
13
|
Cheng I, Jin L, Rose LC, Deppmann CD. Temporally restricted death and the role of p75NTR as a survival receptor in the developing sensory nervous system. Dev Neurobiol 2018; 78:701-717. [PMID: 29569362 PMCID: PMC6023755 DOI: 10.1002/dneu.22591] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/26/2018] [Accepted: 03/16/2018] [Indexed: 11/05/2022]
Abstract
The peripheral somatosensory system overproduces neurons early in development followed by a period of cell death during final target innervation. The decision to survive or die in somatosensory neurons of the dorsal root ganglion (DRG) is mediated by target-derived neurotrophic factors and their cognate receptors. Subsets of peripheral somatosensory neurons can be crudely defined by the neurotrophic receptors that they express: peptidergic nociceptors (TrkA+), nonpeptidergic nociceptors (Ret+), mechanoreceptors (Ret+ or TrkB+), and proprioceptors (TrkC+). A direct comparison of early developmental timing between these subsets has not been performed. Here we characterized the accumulation and death of TrkA, B, C, and Ret+ neurons in the DRG as a function of developmental time. We find that TrkB, TrkC, and Ret-expressing neurons in the DRG complete developmental cell death prior to TrkA-expressing neurons. Given the broadly defined roles of the neurotrophin receptor p75NTR in augmenting neurotrophic signaling in sensory neurons, we investigated its role in supporting the survival of these distinct subpopulations. We find that TrkA+, TrkB+, and TrkC+ sensory neuron subpopulations require p75NTR for survival, but proliferating progenitors do not. These data demonstrate how diverging sensory neurons undergo successive waves of cell death and how p75NTR represses the magnitude, but not developmental window of this culling. © 2018 Wiley Periodicals, Inc. Develop Neurobiol 78: 701-717, 2018.
Collapse
Affiliation(s)
- Irene Cheng
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA
| | - Lucy Jin
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Lucy C. Rose
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Christopher D. Deppmann
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22903, USA
- Department of Biomedical Engineering University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
14
|
Chen Z, Donnelly CR, Dominguez B, Harada Y, Lin W, Halim AS, Bengoechea TG, Pierchala BA, Lee KF. p75 Is Required for the Establishment of Postnatal Sensory Neuron Diversity by Potentiating Ret Signaling. Cell Rep 2018; 21:707-720. [PMID: 29045838 DOI: 10.1016/j.celrep.2017.09.037] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 07/14/2017] [Accepted: 09/10/2017] [Indexed: 01/17/2023] Open
Abstract
Producing the neuronal diversity required to adequately discriminate all elements of somatosensation is a complex task during organogenesis. The mechanisms guiding this process during dorsal root ganglion (DRG) sensory neuron specification remain poorly understood. Here, we show that the p75 neurotrophin receptor interacts with Ret and its GFRα co-receptor upon stimulation with glial cell line-derived neurotrophic factor (GDNF). Furthermore, we demonstrate that p75 is required for GDNF-mediated Ret activation, survival, and cell surface localization of Ret in DRG neurons. In mice in which p75 is deleted specifically within sensory neurons beginning at E12.5, we observe that approximately 20% of neurons are lost between P14 and adulthood, and these losses selectively occur within a subpopulation of Ret+ nonpeptidergic nociceptors, with neurons expressing low levels of Ret impacted most heavily. These results suggest that p75 is required for the development of the nonpeptidergic nociceptor lineage by fine-tuning Ret-mediated trophic support.
Collapse
Affiliation(s)
- Zhijiang Chen
- The Salk Institute, Peptide Biology Laboratories, La Jolla, CA 92037, USA
| | - Christopher R Donnelly
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bertha Dominguez
- The Salk Institute, Peptide Biology Laboratories, La Jolla, CA 92037, USA
| | - Yoshinobu Harada
- The Salk Institute, Peptide Biology Laboratories, La Jolla, CA 92037, USA; National Institute of Radiological Sciences and National Institutes for Quantum and Radiological Science and Technology, Chiba 243-8555, Japan
| | - Weichun Lin
- UT Southwestern Medical Center, Neuroscience, Dallas, TX 75390, USA
| | - Alan S Halim
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Tasha G Bengoechea
- The Salk Institute, Peptide Biology Laboratories, La Jolla, CA 92037, USA
| | - Brian A Pierchala
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Kuo-Fen Lee
- The Salk Institute, Peptide Biology Laboratories, La Jolla, CA 92037, USA.
| |
Collapse
|
15
|
Kisiswa L, Erice C, Ferron L, Wyatt S, Osório C, Dolphin AC, Davies AM. T-type Ca2+ channels are required for enhanced sympathetic axon growth by TNFα reverse signalling. Open Biol 2017; 7:rsob.160288. [PMID: 28100666 PMCID: PMC5303278 DOI: 10.1098/rsob.160288] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/12/2016] [Indexed: 12/28/2022] Open
Abstract
Tumour necrosis factor receptor 1 (TNFR1)-activated TNFα reverse signalling, in which membrane-integrated TNFα functions as a receptor for TNFR1, enhances axon growth from developing sympathetic neurons and plays a crucial role in establishing sympathetic innervation. Here, we have investigated the link between TNFα reverse signalling and axon growth in cultured sympathetic neurons. TNFR1-activated TNFα reverse signalling promotes Ca2+ influx, and highly selective T-type Ca2+ channel inhibitors, but not pharmacological inhibitors of L-type, N-type and P/Q-type Ca2+ channels, prevented enhanced axon growth. T-type Ca2+ channel-specific inhibitors eliminated Ca2+ spikes promoted by TNFα reverse signalling in axons and prevented enhanced axon growth when applied locally to axons, but not when applied to cell somata. Blocking action potential generation did not affect the effect of TNFα reverse signalling on axon growth, suggesting that propagated action potentials are not required for enhanced axon growth. TNFα reverse signalling enhanced protein kinase C (PKC) activation, and pharmacological inhibition of PKC prevented the axon growth response. These results suggest that TNFα reverse signalling promotes opening of T-type Ca2+ channels along sympathetic axons, which is required for enhanced axon growth.
Collapse
Affiliation(s)
- Lilian Kisiswa
- School Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Clara Erice
- School Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Laurent Ferron
- Department of Neuroscience, Physiology and Pharmacology, University College London, Andrew Huxley Building, Gower Street, London WC1E 6BT, UK
| | - Sean Wyatt
- School Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Catarina Osório
- School Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, Andrew Huxley Building, Gower Street, London WC1E 6BT, UK
| | - Alun M Davies
- School Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| |
Collapse
|
16
|
Omerbašić D, Smith ESJ, Moroni M, Homfeld J, Eigenbrod O, Bennett NC, Reznick J, Faulkes CG, Selbach M, Lewin GR. Hypofunctional TrkA Accounts for the Absence of Pain Sensitization in the African Naked Mole-Rat. Cell Rep 2017; 17:748-758. [PMID: 27732851 PMCID: PMC5081396 DOI: 10.1016/j.celrep.2016.09.035] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 06/23/2016] [Accepted: 09/13/2016] [Indexed: 12/21/2022] Open
Abstract
The naked mole-rat is a subterranean rodent lacking several pain behaviors found in humans, rats, and mice. For example, nerve growth factor (NGF), an important mediator of pain sensitization, fails to produce thermal hyperalgesia in naked mole-rats. The sensitization of capsaicin-sensitive TRPV1 ion channels is necessary for NGF-induced hyperalgesia, but naked mole-rats have fully functional TRPV1 channels. We show that exposing isolated naked mole-rat nociceptors to NGF does not sensitize TRPV1. However, the naked mole-rat NGF receptor TrkA displays a reduced ability to engage signal transduction pathways that sensitize TRPV1. Between one- and three-amino-acid substitutions in the kinase domain of the naked mole-rat TrkA are sufficient to render the receptor hypofunctional, and this is associated with the absence of heat hyperalgesia. Our data suggest that evolution has selected for a TrkA variant that abolishes a robust nociceptive behavior in this species but is still compatible with species fitness. TRPV1 ion channels in naked mole-rat nociceptors are not sensitized by NGF Naked mole-rat TRPV1 channels are sensitized by NGF in mouse nociceptors NGF activation of naked mole-rat TrkA receptors does not sensitize TRPV1 One to three amino acids in the naked mole-rat TrkA receptors may render it hypofunctional
Collapse
Affiliation(s)
- Damir Omerbašić
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany; Proteome Dynamics Group, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Ewan St J Smith
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany; Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, UK
| | - Mirko Moroni
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Johanna Homfeld
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Ole Eigenbrod
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Nigel C Bennett
- Department of Zoology and Entomology, University of Pretoria, Pretoria, Hatfield 0028, Republic of South Africa
| | - Jane Reznick
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Chris G Faulkes
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Matthias Selbach
- Proteome Dynamics Group, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Gary R Lewin
- Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany; Excellence Cluster Neurocure, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany.
| |
Collapse
|
17
|
Mollá B, Muñoz-Lasso DC, Riveiro F, Bolinches-Amorós A, Pallardó FV, Fernandez-Vilata A, de la Iglesia-Vaya M, Palau F, Gonzalez-Cabo P. Reversible Axonal Dystrophy by Calcium Modulation in Frataxin-Deficient Sensory Neurons of YG8R Mice. Front Mol Neurosci 2017; 10:264. [PMID: 28912677 PMCID: PMC5583981 DOI: 10.3389/fnmol.2017.00264] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/04/2017] [Indexed: 11/13/2022] Open
Abstract
Friedreich’s ataxia (FRDA) is a peripheral neuropathy involving a loss of proprioceptive sensory neurons. Studies of biopsies from patients suggest that axonal dysfunction precedes the death of proprioceptive neurons in a dying-back process. We observed that the deficiency of frataxin in sensory neurons of dorsal root ganglia (DRG) of the YG8R mouse model causes the formation of axonal spheroids which retain dysfunctional mitochondria, shows alterations in the cytoskeleton and it produces impairment of axonal transport and autophagic flux. The homogenous distribution of axonal spheroids along the neurites supports the existence of continues focal damages. This lead us to propose for FRDA a model of distal axonopathy based on axonal focal damages. In addition, we observed the involvement of oxidative stress and dyshomeostasis of calcium in axonal spheroid formation generating axonal injury as a primary cause of pathophysiology. Axonal spheroids may be a consequence of calcium imbalance, thus we propose the quenching or removal extracellular Ca2+ to prevent spheroids formation. In our neuronal model, treatments with BAPTA and o-phenanthroline reverted the axonal dystrophy and the mitochondrial dysmorphic parameters. These results support the hypothesis that axonal pathology is reversible in FRDA by pharmacological manipulation of intracellular Ca2+ with Ca2+ chelators or metalloprotease inhibitors, preventing Ca2+-mediated axonal injury. Thus, the modulation of Ca2+ levels may be a relevant therapeutic target to develop early axonal protection and prevent dying-back neurodegeneration.
Collapse
Affiliation(s)
- Belén Mollá
- CIBER de Enfermedades Raras (CIBERER)Valencia, Spain.,Instituto de Biomedicina de Valencia (IBV), CSICValencia, Spain
| | - Diana C Muñoz-Lasso
- CIBER de Enfermedades Raras (CIBERER)Valencia, Spain.,Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia-Instituto de Investigación Sanitaria (INCLIVA)Valencia, Spain.,Associated Unit for Rare Diseases INCLIVA-CIPFValencia, Spain.,VEDAS Corporación de Investigación e Innovación, VEDASCIIMedellín, Colombia
| | - Fátima Riveiro
- CIBER de Enfermedades Raras (CIBERER)Valencia, Spain.,Fundacion Publica Galega de Medicina Xenomica (FPGMX)-SERGAS, Grupo de Medicina Xenomica, Hospital Clínico UniversitarioSantiago de Compostela, Spain
| | - Arantxa Bolinches-Amorós
- CIBER de Enfermedades Raras (CIBERER)Valencia, Spain.,Cell Therapy Program, Prince Felipe Research Centre (CIPF)Valencia, Spain
| | - Federico V Pallardó
- CIBER de Enfermedades Raras (CIBERER)Valencia, Spain.,Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia-Instituto de Investigación Sanitaria (INCLIVA)Valencia, Spain.,Associated Unit for Rare Diseases INCLIVA-CIPFValencia, Spain
| | | | - María de la Iglesia-Vaya
- Regional Ministry of Health in Valencia, Hospital Sagunto (CEIB-CSUSP)Valencia, Spain.,Brain Connectivity Laboratory, Joint Unit FISABIO & Prince Felipe Research Centre (CIPF)Valencia, Spain.,CIBER de Salud Mental (CIBERSAM)Valencia, Spain
| | - Francesc Palau
- CIBER de Enfermedades Raras (CIBERER)Valencia, Spain.,Department of Genetic and Molecular Medicine, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de DéuBarcelona, Spain.,Department of Pediatrics, University of Barcelona School of MedicineBarcelona, Spain
| | - Pilar Gonzalez-Cabo
- CIBER de Enfermedades Raras (CIBERER)Valencia, Spain.,Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia-Instituto de Investigación Sanitaria (INCLIVA)Valencia, Spain.,Associated Unit for Rare Diseases INCLIVA-CIPFValencia, Spain
| |
Collapse
|
18
|
ProNGF, but Not NGF, Switches from Neurotrophic to Apoptotic Activity in Response to Reductions in TrkA Receptor Levels. Int J Mol Sci 2017; 18:ijms18030599. [PMID: 28282920 PMCID: PMC5372615 DOI: 10.3390/ijms18030599] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/03/2017] [Accepted: 03/07/2017] [Indexed: 12/13/2022] Open
Abstract
Nerve growth factor (NGF) promotes the survival and differentiation of neurons. NGF is initially synthesized as a precursor, proNGF, which is the predominant form in the central nervous system. NGF and proNGF bind to TrkA/p75NTR to mediate cell survival and to sortilin/p75NTR to promote apoptosis. The ratio of TrkA to p75NTR affects whether proNGF and mature NGF signal cell survival or apoptosis. The purpose of this study was to determine whether the loss of TrkA influences p75NTR or sortilin expression levels, and to establish whether proNGF and mature NGF have a similar ability to switch between cell survival and cell death. We systematically altered TrkA receptor levels by priming cells with NGF, using small interfering RNA, and using the mutagenized PC12nnr5 cell line. We found that both NGF and proNGF can support cell survival in cells expressing TrkA, even in the presence of p75NTR and sortilin. However, when TrkA is reduced, proNGF signals cell death, while NGF exhibits no activity. In the absence of TrkA, proNGF-induced cell death occurs, even when p75NTR and sortilin levels are reduced. These results show that proNGF can switch between neurotrophic and apoptotic activity in response to changes in TrkA receptor levels, whereas mature NGF cannot. These results also support the model that proNGF is neurotrophic under normal circumstances, but that a loss in TrkA in the presence of p75NTR and sortilin, as occurs in neurodegenerative disease or injury, shifts proNGF, but not NGF, signalling from cell survival to cell death.
Collapse
|
19
|
Abstract
Neurotrophin molecules have been shown to play important roles in the survival of neurons during devel opment. Most early studies concentrated on the initially discovered factor—nerve growth factor. Recent work has demonstrated that nerve growth factor belongs to a family of neurotrophins that include brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-4/5. These neurotrophins exert their action via high- affinity receptors known as trks ( trkA, trkB, and trkC). A major goal of present research is to identify the neuronal locus of different trks to permit inferences about the specificity of action of the different growth factors. However, recent evidence suggests complex relationships between neurotrophins and trk receptors. These issues are explored in the context of dorsal root ganglion cells and motor neurons. Functional studies of the effects of nerve growth factor on its target cells, the nociceptive afferents, illustrate that this neurotro phin plays a role in development and function, which goes well beyond its classical role in promoting the survival of neurons during the early phases of development. The Neuroscientist 1:26-34, 1995
Collapse
Affiliation(s)
- Lorne M. Mendell
- Department of Neurobiology and Behavior SUNY at Stony
Brook Stony Brook, New York
| |
Collapse
|
20
|
Djouhri L. PG110, A Humanized Anti-NGF Antibody, Reverses Established Pain Hypersensitivity in Persistent Inflammatory Pain, but not Peripheral Neuropathic Pain, Rat Models. PAIN MEDICINE 2016; 17:2082-2094. [PMID: 26917622 DOI: 10.1093/pm/pnw007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Chronic inflammatory and peripheral neuropathic pain (PNP) is a major health problem for which effective drug treatment is lacking. The pathophysiology of these debilitating conditions is incompletely understood, but nerve growth factor (NGF) is believed to play a major role. NGF-antagonism has previously been shown to prevent pain hypersensitivity in rodent models of acute inflammatory pain and PNP, but most of those animal studies did not address the more clinically relevant issue of whether NGF-antagonism provides relief of established chronic pain behavior. Therefore, the aim of this study was to investigate whether blocking NGF actions with a humanized anti-NGF monoclonal antibody (PG110) would reverse/attenuate established pain hypersensitivity in rat models of chronic/persistent inflammatory pain and PNP. METHODS The complete Freund's adjuvant (CFA) rat model of persistent inflammatory pain, and the L5 spinal nerve axotomy (SNA) model of PNP, were used in the present study. The effect of a single intravenous injection (10, 30, and 300 µg/kg) of an anti-NGF antibody PG110 on heat and mechanical hypersensitivity was assessed 5 and 7 days after CFA and SNA, respectively. RESULTS Compared to vehicle treated group, PG110 dose dependently attenuated established heat and mechanical hypersensitivity induced by CFA, but not that induced by SNA. The anti-allodynic and anti-hyperalgesic effects of PG110 in the CFA model were similar to those of the positive control naproxen (30 mg/kg, i.v.). CONCLUSION These findings suggest that therapies that target NGF or its receptors may be effective for treatment of persistent/chronic inflammatory pain, but probably not PNP.
Collapse
Affiliation(s)
- Laiche Djouhri
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11472, KSA
| |
Collapse
|
21
|
O'Keeffe GW, Gutierrez H, Howard L, Laurie CW, Osorio C, Gavaldà N, Wyatt SL, Davies AM. Region-specific role of growth differentiation factor-5 in the establishment of sympathetic innervation. Neural Dev 2016; 11:4. [PMID: 26878848 PMCID: PMC4755026 DOI: 10.1186/s13064-016-0060-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/08/2016] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Nerve growth factor (NGF) is the prototypical target-derived neurotrophic factor required for sympathetic neuron survival and for the growth and ramification of sympathetic axons within most but not all sympathetic targets. This implies the operation of additional target-derived factors for regulating terminal sympathetic axon growth and branching. RESULTS Here report that growth differentiation factor 5 (GDF5), a widely expressed member of the transforming growth factor beta (TGFβ) superfamily required for limb development, promoted axon growth from mouse superior cervical ganglion (SCG) neurons independently of NGF and enhanced axon growth in combination with NGF. GDF5 had no effect on neuronal survival and influenced axon growth during a narrow window of postnatal development when sympathetic axons are ramifying extensively in their targets in vivo. SCG neurons expressed all receptors capable of participating in GDF5 signaling at this stage of development. Using compartment cultures, we demonstrated that GDF5 exerted its growth promoting effect by acting directly on axons and by initiating retrograde canonical Smad signalling to the nucleus. GDF5 is synthesized in sympathetic targets, and examination of several anatomically circumscribed tissues in Gdf5 null mice revealed regional deficits in sympathetic innervation. There was a marked, highly significant reduction in the sympathetic innervation density of the iris, a smaller though significant reduction in the trachea, but no reduction in the submandibular salivary gland. There was no reduction in the number of neurons in the SCG. CONCLUSIONS These findings show that GDF5 is a novel target-derived factor that promotes sympathetic axon growth and branching and makes a distinctive regional contribution to the establishment of sympathetic innervation, but unlike NGF, plays no role in regulating sympathetic neuron survival.
Collapse
Affiliation(s)
- Gerard W O'Keeffe
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
- Dept. Anatomy/Neuroscience and Biosciences Institute, UCC, Cork, Ireland
| | - Humberto Gutierrez
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
- Current address, School of Life Sciences, University of Lincoln, Brayford Pool, Lincoln, LN6 7TS, UK
| | - Laura Howard
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| | | | - Catarina Osorio
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
- Current address, MRC Centre for Developmental Neurobiology, King's College London, New Hunt's House, 4th Floor, Guy's Hospital Campus, London, SE1 1UL, UK
| | - Núria Gavaldà
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
- Current address, SOM Innovation Biotech SL, c/Baldiri Reixac 4, 08028, Barcelona, Spain
| | - Sean L Wyatt
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| | - Alun M Davies
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK.
| |
Collapse
|
22
|
Overexpression of NTRK1 Promotes Differentiation of Neural Stem Cells into Cholinergic Neurons. BIOMED RESEARCH INTERNATIONAL 2015; 2015:857202. [PMID: 26509167 PMCID: PMC4609807 DOI: 10.1155/2015/857202] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 08/25/2015] [Accepted: 09/16/2015] [Indexed: 11/17/2022]
Abstract
Neurotrophic tyrosine kinase type 1 (NTRK1) plays critical roles in proliferation, differentiation, and survival of cholinergic neurons; however, it remains unknown whether enhanced expression of NTRK1 in neural stem cells (NSCs) can promote their differentiation into mature neurons. In this study, a plasmid encoding the rat NTRK1 gene was constructed and transfected into C17.2 mouse neural stem cells (NSCs). NTRK1 overexpression in C17.2 cells was confirmed by western blot. The NSCs overexpressing NTRK1 and the C17.2 NSCs transfected by an empty plasmid vector were treated with or without 100 ng/mL nerve growth factor (NGF) for 7 days. Expression of the cholinergic cell marker, choline acetyltransferase (ChAT), was detected by florescent immunocytochemistry (ICC). In the presence of NGF induction, the NSCs overexpressing NTRK1 differentiated into ChAT-immunopositive cells at 3-fold higher than the NSCs transfected by the plasmid vector (26% versus 9%, P < 0.05). The data suggest that elevated NTRK1 expression increases differentiation of NSCs into cholinergic neurons under stimulation of NGF. The approach also represents an efficient strategy for generation of cholinergic neurons.
Collapse
|
23
|
Matusica D, Alfonsi F, Turner BJ, Butler TJ, Shepheard SR, Rogers ML, Skeldal S, Underwood CK, Mangelsdorf M, Coulson EJ. Inhibition of motor neuron death in vitro and in vivo by a p75 neurotrophin receptor intracellular domain fragment. J Cell Sci 2015; 129:517-30. [PMID: 26503157 DOI: 10.1242/jcs.173864] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 10/22/2015] [Indexed: 12/14/2022] Open
Abstract
The p75 neurotrophin receptor (p75(NTR); also known as NGFR) can mediate neuronal apoptosis in disease or following trauma, and facilitate survival through interactions with Trk receptors. Here we tested the ability of a p75(NTR)-derived trophic cell-permeable peptide, c29, to inhibit p75(NTR)-mediated motor neuron death. Acute c29 application to axotomized motor neuron axons decreased cell death, and systemic c29 treatment of SOD1(G93A) mice, a common model of amyotrophic lateral sclerosis, resulted in increased spinal motor neuron survival mid-disease as well as delayed disease onset. Coincident with this, c29 treatment of these mice reduced the production of p75(NTR) cleavage products. Although c29 treatment inhibited mature- and pro-nerve-growth-factor-induced death of cultured motor neurons, and these ligands induced the cleavage of p75(NTR) in motor-neuron-like NSC-34 cells, there was no direct effect of c29 on p75(NTR) cleavage. Rather, c29 promoted motor neuron survival in vitro by enhancing the activation of TrkB-dependent signaling pathways, provided that low levels of brain-derived neurotrophic factor (BDNF) were present, an effect that was replicated in vivo in SOD1(G93A) mice. We conclude that the c29 peptide facilitates BDNF-dependent survival of motor neurons in vitro and in vivo.
Collapse
Affiliation(s)
- Dusan Matusica
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia Department of Anatomy & Histology, Centre for Neuroscience, Flinders University, GPO Box 2100, Adelaide, South Australia 5001, Australia
| | - Fabienne Alfonsi
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Bradley J Turner
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Victoria 3051, Australia
| | - Tim J Butler
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Stephanie R Shepheard
- Department of Human Physiology, Centre for Neuroscience, Flinders University, GPO Box 2100, Adelaide, South Australia 5001, Australia
| | - Mary-Louise Rogers
- Department of Human Physiology, Centre for Neuroscience, Flinders University, GPO Box 2100, Adelaide, South Australia 5001, Australia
| | - Sune Skeldal
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Clare K Underwood
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Marie Mangelsdorf
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Elizabeth J Coulson
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
24
|
Abstract
We have investigated how CaSR activation enhances sympathetic axon growth. CaSR activation promotes phosphorylation of ERK1 and ERK2. Inhibition of ERK1/ERK2 phosphorylation blocks CaSR-promoted axon growth. CaSR-promoted axon growth requires a discrete region of the cytoplasmic domain.
The extracellular calcium-sensing receptor (CaSR) is a G-protein coupled receptor that monitors the systemic extracellular free ionized calcium level ([Ca2+]o) in organs involved in systemic [Ca2+]o homeostasis. CaSR is widely expressed in the nervous system and its activation promotes axon and dendrite growth during development, but the mechanism by which it does this is not known. Here we show that enhanced axon growth and branching from cultured embryonic sympathetic neurons by activation of the endogenous CaSR depends on the presence of nerve growth factor (NGF). Our observation that activation of overexpressed CaSR promotes axon growth in NGF-free medium has enabled us to investigate CaSR downstream signaling contributing to axon growth in the absence of NGF signaling. We show that activation of overexpressed CaSR leads to activation of ERK1 and ERK2, and pharmacological inhibition of CaSR-dependent ERK1/ERK2 activation prevents CaSR-dependent axon growth. Analysis of axon growth from cultured neurons expressing deletion mutants of the CaSR cytoplasmic tail revealed that the region between alanine 877 and glycine 907 is required for promoting axon growth that is distinct from the high-affinity filamin-A binding site that has previously been implicated in ERK1/ERK2 activation.
Collapse
Affiliation(s)
- Thomas N Vizard
- School of Biosciences, Biomedical Building, Museum Avenue, Cardiff CF10 3US, UK.
| | - Michael Newton
- School of Biosciences, Biomedical Building, Museum Avenue, Cardiff CF10 3US, UK
| | - Laura Howard
- School of Biosciences, Biomedical Building, Museum Avenue, Cardiff CF10 3US, UK
| | - Sean Wyatt
- School of Biosciences, Biomedical Building, Museum Avenue, Cardiff CF10 3US, UK
| | - Alun M Davies
- School of Biosciences, Biomedical Building, Museum Avenue, Cardiff CF10 3US, UK
| |
Collapse
|
25
|
Caroleo MC, Carito V, Pingitore A, Perrotta ID, Perri M, Mancuso D, Russo A, Cione E. Human kidney podocyte cell population as a novel biological target of nerve growth factor. Growth Factors 2015; 33:14-22. [PMID: 25347785 DOI: 10.3109/08977194.2014.975799] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human podocytes are highly specialized cells with a key role in kidney physiology. Alteration of their structure as a consequence of injury or developmental failure leads to severe renal diseases. Although several studies have tried to elucidate the molecular framework of this cellular system, the functional bases for the maintenance of podocytes in their specialized state to sustain kidney barrier filtration are not completely understood. In this study, the capability of podocytes to produce and secrete the nerve growth factor (NGF) has been demonstrated via a validated in vitro model. During the process of cell differentiation, NGF and its receptors are modulated in human podocytes just as NGF-responsive neurons. Blockade of NGF biological activity results in severe changes of cell morphology. Collectively, our results outline a novel function of the neurotrophin and add a new cellular target in the complex biological framework of NGF.
Collapse
Affiliation(s)
- Maria Cristina Caroleo
- Department of Pharmacy, Nutrition and Health Sciences, University of Calabria , Cosenza , Italy
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Newbern JM. Molecular control of the neural crest and peripheral nervous system development. Curr Top Dev Biol 2015; 111:201-31. [PMID: 25662262 DOI: 10.1016/bs.ctdb.2014.11.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A transient and unique population of multipotent stem cells, known as neural crest cells (NCCs), generate a bewildering array of cell types during vertebrate development. An attractive model among developmental biologists, the study of NCC biology has provided a wealth of knowledge regarding the cellular and molecular mechanisms important for embryogenesis. Studies in numerous species have defined how distinct phases of NCC specification, proliferation, migration, and survival contribute to the formation of multiple functionally distinct organ systems. NCC contributions to the peripheral nervous system (PNS) are well known. Critical developmental processes have been defined that provide outstanding models for understanding how extracellular stimuli, cell-cell interactions, and transcriptional networks cooperate to direct cellular diversification and PNS morphogenesis. Dissecting the complex extracellular and intracellular mechanisms that mediate the formation of the PNS from NCCs may have important therapeutic implications for neurocristopathies, neuropathies, and certain forms of cancer.
Collapse
Affiliation(s)
- Jason M Newbern
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA.
| |
Collapse
|
27
|
Marlin MC, Li G. Biogenesis and function of the NGF/TrkA signaling endosome. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 314:239-57. [PMID: 25619719 DOI: 10.1016/bs.ircmb.2014.10.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Target-derived neurotrophin nerve growth factor (NGF) and its receptor TrkA are well known for retrograde signaling to promote survival and innervation of sympathetic and sensory neurons. In recent years, the signaling endosome model has been used to describe the sustained NGF/TrkA retrograde signaling as a process of endocytosis and retrograde transport of NGF/TrkA-containing endosomes from the axon terminal to the cell body for activation of NGF-inducible gene expression responsible for neuronal survival and development. Here, we review the biogenesis and function of NGF, TrkA, and the signaling endosome and discuss possible roles of Rab GTPases in the biogenesis and trafficking of signaling endosomes.
Collapse
Affiliation(s)
- M Caleb Marlin
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Guangpu Li
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
28
|
Wheeler MA, Heffner DL, Kim S, Espy SM, Spano AJ, Cleland CL, Deppmann CD. TNF-α/TNFR1 signaling is required for the development and function of primary nociceptors. Neuron 2014; 82:587-602. [PMID: 24811380 DOI: 10.1016/j.neuron.2014.04.009] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2014] [Indexed: 12/19/2022]
Abstract
Primary nociceptors relay painful touch information from the periphery to the spinal cord. Although it is established that signals generated by receptor tyrosine kinases TrkA and Ret coordinate the development of distinct nociceptive circuits, mechanisms modulating TrkA or Ret pathways in developing nociceptors are unknown. We have identified tumor necrosis factor (TNF) receptor 1 (TNFR1) as a critical modifier of TrkA and Ret signaling in peptidergic and nonpeptidergic nociceptors. Specifically, TrkA+ peptidergic nociceptors require TNF-α-TNFR1 forward signaling to suppress nerve growth factor (NGF)-mediated neurite growth, survival, excitability, and differentiation. Conversely, TNFR1-TNF-α reverse signaling augments the neurite growth and excitability of Ret+ nonpeptidergic nociceptors. The developmental and functional nociceptive defects associated with loss of TNFR1 signaling manifest behaviorally as lower pain thresholds caused by increased sensitivity to NGF. Thus, TNFR1 exerts a dual role in nociceptor information processing by suppressing TrkA and enhancing Ret signaling in peptidergic and nonpeptidergic nociceptors, respectively.
Collapse
Affiliation(s)
- Michael A Wheeler
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA
| | - Danielle L Heffner
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Suemin Kim
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Sarah M Espy
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Anthony J Spano
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Corey L Cleland
- Department of Biology, James Madison University, Harrisonburg, VA 22807, USA
| | - Christopher D Deppmann
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA; Department of Cell Biology, University of Virginia, Charlottesville, VA 22903, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903, USA.
| |
Collapse
|
29
|
Dedoni S, Olianas MC, Ingianni A, Onali P. Type I interferons up-regulate the expression and signalling of p75 NTR/TrkA receptor complex in differentiated human SH-SY5Y neuroblastoma cells. Neuropharmacology 2014; 79:321-34. [PMID: 24333329 DOI: 10.1016/j.neuropharm.2013.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Revised: 10/25/2013] [Accepted: 12/02/2013] [Indexed: 01/10/2023]
Abstract
Both type I interferons (IFNs) and neurotrophins regulate neuroadaptive responses, but relatively little is known on the interaction between these two classes of regulatory proteins. Here we investigated the effect of IFN-β on the expression and functional activity of the common neurotrophin receptor p75NTR and the nerve growth factor (NGF) receptor TrkA. In differentiated human SH-SY5Y neuroblastoma cells prolonged exposure to IFN-β up-regulated p75NTR and TrkA levels, failed to affect the content of sortilin, a p75NTR co-receptor, and, consistent with our previous finding, down-regulated the brain-derived neurotrophic factor receptor TrkB. Quantitative real time RT-PCR indicated that IFN-β increased p75NTR and TrkA mRNA levels. In control and IFN-β treated cells proNGF failed to induce c-Jun N-terminal kinase and nuclear factor/kB activation, two p75NTR/sortilin signalling pathways mediating neuronal death. On the other hand, IFN-β treatment enhanced TrkA autophosphorylation and signalling induced by NGF and proNGF. Knockdown of p75NTR by siRNA reduced TrkA activation by proNGF and a subnanomolar concentration of NGF, whereas co-immunoprecipitation indicated close association of p75NTR and TrkA. Co-treatment with either NGF or proNGF reduced IFN-β pro-apoptotic and anti-neurotrophic effects. Similarly, in primary mouse hippocampal neurons IFN-β increased p75NTR and TrkA expression, down-regulated TrkB and enhanced NGF-induced phosphorylation of the pro-survival protein kinase Akt. The data demonstrate that in neuronal cells IFN-β differentially affects the expression and signalling of neurotrophin receptors and suggest that the up-regulation of the p75NTR/TrkA signalling complex may constitute a novel mechanism by which this cytokine selectively attenuates its pro-apoptotic effect in NGF-responsive cells.
Collapse
Affiliation(s)
- Simona Dedoni
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences and Clinical Pharmacology, and Section of Applied Microbiology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Maria C Olianas
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences and Clinical Pharmacology, and Section of Applied Microbiology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Angela Ingianni
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences and Clinical Pharmacology, and Section of Applied Microbiology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Pierluigi Onali
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences and Clinical Pharmacology, and Section of Applied Microbiology, Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
30
|
Kraemer BR, Yoon SO, Carter BD. The biological functions and signaling mechanisms of the p75 neurotrophin receptor. Handb Exp Pharmacol 2014; 220:121-164. [PMID: 24668472 DOI: 10.1007/978-3-642-45106-5_6] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The p75 neurotrophin receptor (p75(NTR)) regulates a wide range of cellular functions, including programmed cell death, axonal growth and degeneration, cell proliferation, myelination, and synaptic plasticity. The multiplicity of cellular functions governed by the receptor arises from the variety of ligands and co-receptors which associate with p75(NTR) and regulate its signaling. P75(NTR) promotes survival through interactions with Trk receptors, inhibits axonal regeneration via partnerships with Nogo receptor (Nogo-R) and Lingo-1, and promotes apoptosis through association with Sortilin. Signals downstream of these interactions are further modulated through regulated intramembrane proteolysis (RIP) of p75(NTR) and by interactions with numerous cytosolic partners. In this chapter, we discuss the intricate signaling mechanisms of p75(NTR), emphasizing how these signals are differentially regulated to mediate these diverse cellular functions.
Collapse
Affiliation(s)
- B R Kraemer
- Department of Biochemistry, Vanderbilt University School of Medicine, 625 Light Hall, Nashville, TN, 37232, USA
| | | | | |
Collapse
|
31
|
Ceni C, Unsain N, Zeinieh MP, Barker PA. Neurotrophins in the regulation of cellular survival and death. Handb Exp Pharmacol 2014; 220:193-221. [PMID: 24668474 DOI: 10.1007/978-3-642-45106-5_8] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The neurotrophins play crucial roles regulating survival and apoptosis in the developing and injured nervous system. The four neurotrophins exert profound and crucial survival effects on developing peripheral neurons, and their expression and action is intimately tied to successful innervation of peripheral targets. In the central nervous system, they are dispensable for neuronal survival during development but support neuronal survival after lesion or other forms of injury. Neurotrophins also regulate apoptosis of both peripheral and central neurons, and we now recognize that there are regulatory advantages to having the same molecules regulate life and death decisions. This chapter examines the biological contexts in which these events take place and highlights the specific ligands, receptors, and signaling mechanisms that allow them to occur.
Collapse
Affiliation(s)
- Claire Ceni
- Centre for Neuronal Survival, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC, Canada, H3A 2B4
| | | | | | | |
Collapse
|
32
|
Kisiswa L, Osório C, Erice C, Vizard T, Wyatt S, Davies AM. TNFα reverse signaling promotes sympathetic axon growth and target innervation. Nat Neurosci 2013; 16:865-73. [PMID: 23749144 PMCID: PMC3785146 DOI: 10.1038/nn.3430] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 05/07/2013] [Indexed: 12/22/2022]
Abstract
Reverse signaling via members of the tumor necrosis factor (TNF) superfamily controls multiple aspects of immune function. Here we document TNFα reverse signaling in the nervous system to our knowledge for the first time and show that it has a crucial role in establishing sympathetic innervation. During postnatal development, sympathetic axons express TNFα as they grow and branch in their target tissues, which in turn express TNF receptor 1 (TNFR1). In culture, soluble forms of TNFR1 act directly on postnatal sympathetic axons to promote growth and branching by a mechanism that depends on membrane-integrated TNFα and on downstream activation of ERK. Sympathetic innervation density is substantially lower in several tissues in postnatal and adult mice lacking either TNFα or TNFR1. These findings reveal that target-derived TNFR1 acts as a reverse-signaling ligand for membrane-integrated TNFα to promote growth and branching of sympathetic axons.
Collapse
MESH Headings
- ADAM Proteins/pharmacology
- ADAM17 Protein
- Animals
- Animals, Newborn
- Axons/physiology
- Calcium/metabolism
- Cells, Cultured
- Chelating Agents/pharmacology
- Dose-Response Relationship, Drug
- Egtazic Acid/analogs & derivatives
- Egtazic Acid/pharmacology
- Embryo, Mammalian
- Gene Expression Regulation, Developmental/drug effects
- Gene Expression Regulation, Developmental/genetics
- Mice
- Mice, Transgenic
- Nerve Fibers/physiology
- Nerve Growth Factor/pharmacology
- Neurons/cytology
- RNA, Messenger/metabolism
- Receptors, Tumor Necrosis Factor, Type I/deficiency
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Signal Transduction/physiology
- Superior Cervical Ganglion/cytology
- Sympathetic Nervous System/cytology
- Sympathetic Nervous System/embryology
- Sympathetic Nervous System/growth & development
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
- Tyrosine 3-Monooxygenase/metabolism
Collapse
Affiliation(s)
- Lilian Kisiswa
- Division of Molecular Biosciences, School of Biosciences, Cardiff University, Cardiff, UK
| | | | | | | | | | | |
Collapse
|
33
|
Howard L, Wyatt S, Nagappan G, Davies AM. ProNGF promotes neurite growth from a subset of NGF-dependent neurons by a p75NTR-dependent mechanism. Development 2013; 140:2108-17. [PMID: 23633509 PMCID: PMC3640218 DOI: 10.1242/dev.085266] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2013] [Indexed: 01/19/2023]
Abstract
The somatosensory and sympathetic innervation of the vertebrate head is derived principally from the neurons of trigeminal and superior cervical ganglia (SCG), respectively. During development, the survival of both populations of neurons and the terminal growth and branching of their axons in the tissues they innervate is regulated by the supply of nerve growth factor (NGF) produced by these tissues. NGF is derived by proteolytic cleavage of a large precursor protein, proNGF, which is recognised to possess distinctive biological functions. Here, we show that proNGF promotes profuse neurite growth and branching from cultured postnatal mouse SCG neurons. In marked contrast, proNGF does not promote the growth of trigeminal neurites. Studies using compartment cultures demonstrated that proNGF acts locally on SCG neurites to promote growth. The neurite growth-promoting effect of proNGF is not observed in SCG neurons cultured from p75(NTR)-deficient mice, and proNGF does not phosphorylate the NGF receptor tyrosine kinase TrkA. These findings suggest that proNGF selectively promotes the growth of neurites from a subset of NGF-responsive neurons by a p75(NTR)-dependent mechanism during postnatal development when the axons of these neurons are ramifying within their targets in vivo.
Collapse
Affiliation(s)
- Laura Howard
- Molecular Biosciences Division, School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AT, Wales, UK.
| | | | | | | |
Collapse
|
34
|
Matusica D, Skeldal S, Sykes AM, Palstra N, Sharma A, Coulson EJ. An intracellular domain fragment of the p75 neurotrophin receptor (p75(NTR)) enhances tropomyosin receptor kinase A (TrkA) receptor function. J Biol Chem 2013; 288:11144-54. [PMID: 23471969 PMCID: PMC3630867 DOI: 10.1074/jbc.m112.436469] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 02/17/2013] [Indexed: 11/06/2022] Open
Abstract
Facilitation of nerve growth factor (NGF) signaling by the p75 neurotrophin receptor (p75(NTR)) is critical for neuronal survival and differentiation. However, the interaction between p75(NTR) and TrkA receptors required for this activity is not understood. Here, we report that a specific 29-amino acid peptide derived from the intracellular domain fragment of p75(NTR) interacts with and potentiates binding of NGF to TrkA-expressing cells, leading to increased neurite outgrowth in sympathetic neurons as a result of enhanced Erk1/2 and Akt signaling. An endogenous intracellular domain fragment of p75(NTR) (p75(ICD)) containing these 29 amino acids is produced by regulated proteolysis of the full-length receptor. We demonstrate that generation of this fragment is a requirement for p75(NTR) to facilitate TrkA signaling in neurons and propose that the juxtamembrane region of p75(ICD) acts to cause a conformational change within the extracellular domain of TrkA. This finding provides new insight into the mechanism by which p75(NTR) and TrkA interact to enhance neurotrophic signaling.
Collapse
Affiliation(s)
- Dusan Matusica
- From the Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Sune Skeldal
- From the Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Alex M. Sykes
- From the Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Nickless Palstra
- From the Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Aanchal Sharma
- From the Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Elizabeth J. Coulson
- From the Queensland Brain Institute, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
35
|
Gutierrez H, Kisiswa L, O'Keeffe GW, Smithen MJ, Wyatt S, Davies AM. Regulation of neurite growth by tumour necrosis superfamily member RANKL. Open Biol 2013; 3:120150. [PMID: 23303310 PMCID: PMC3603457 DOI: 10.1098/rsob.120150] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
RANKL (receptor-activator of NF-κB ligand, TNFSF11) is a member of the TNF superfamily that regulates bone remodelling and the development of the thymus, lymph nodes and mammary glands. While RANKL and its membrane bound receptor RANK (TNFRSF11A) are expressed in the adult central nervous system and have been implicated in thermoregulation, the potential function of RANK signalling in the developing nervous system remains unexplored. Here, we show that RANK is expressed by sympathetic and sensory neurons of the developing mouse peripheral nervous system and that activating RANK signalling in these neurons during perinatal development by either treating cultured neurons with soluble RANKL or overexpressing RANK in the neurons inhibited neurotrophin-promoted neurite growth without affecting neurotrophin-promoted neuronal survival. RANKL is expressed in tissues innervated by these neurons, and studies in compartment cultures demonstrated that RANKL is capable of acting directly on neurites to inhibit growth locally. Enhancing RANK signalling in cultured neurons resulted in NF-κB activation and phosphorylation of the p65 NF-κB subunit on serine 536. Transfecting neurons with a series of mutated signalling proteins showed that NF-κB activation and p65 phosphorylation occurred by an IKKβ-dependent mechanism and that blockade of this signalling pathway prevented neurite growth inhibition by RANKL. These findings reveal that RANKL is a novel negative regulator of neurite growth from developing PNS neurons and that it exerts its effects by IKKβ-dependent activation of NF-κB.
Collapse
Affiliation(s)
- Humberto Gutierrez
- Cardiff School of Biosciences, Cardiff University, Cardiff Wales CF10 3AX, UK
| | | | | | | | | | | |
Collapse
|
36
|
Chirivella L, Cano-Jaimez M, Pérez-Sánchez F, Herraez L, Carretero J, Fariñas I, Burks DJ, Kirstein M. IRS2 signalling is required for the development of a subset of sensory spinal neurons. Eur J Neurosci 2012; 35:341-52. [PMID: 22288475 DOI: 10.1111/j.1460-9568.2011.07959.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Insulin and insulin-like growth factor-I play important roles in the development and maintenance of neurons and glial cells of the nervous system. Both factors activate tyrosine kinase receptors, which signal through adapter proteins of the insulin receptor substrate (IRS) family. Although insulin and insulin-like growth factor-I receptors are expressed in dorsal root ganglia (DRG), the function of IRS-mediated signalling in these structures has not been studied. Here we address the role of IRS2-mediated signalling in murine DRG. Studies in cultured DRG neurons from different embryonic stages indicated that a subset of nerve growth factor-responsive neurons is also dependent on insulin for survival at very early time points. Consistent with this, increased apoptosis during gangliogenesis resulted in a partial loss of trkA-positive neurons in DRG of Irs2 mutant embryos. Analyses in adult Irs2(-/-) mice revealed that unmyelinated fibre afferents, which express calcitonin gene-related peptide/substance P and isolectin B4, as well as some myelinated afferents to the skin were affected by the mutation. The diminished innervation of glabrous skin in adult Irs2(-/-) mice correlated with longer paw withdrawal latencies in the hot-plate assay. Collectively, these findings indicate that IRS2 signalling is required for the proper development of spinal sensory neurons involved in the perception of pain.
Collapse
Affiliation(s)
- Laura Chirivella
- Departamento de Biología Celular and Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Universidad de Valencia, Doctor Moliner 50, 46100 Burjassot, Spain
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Greferath U, Trieu J, Barrett GL. The p75 neurotrophin receptor has nonapoptotic antineurotrophic actions in the basal forebrain. J Neurosci Res 2012; 90:278-87. [PMID: 21922519 DOI: 10.1002/jnr.22735] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 06/09/2011] [Indexed: 11/06/2022]
Abstract
Because of controversy about the role of the p75 neurotrophin receptor (p75(NTR) ) in the cholinergic basal forebrain (CBF), we investigated this region in p75(NTR) third exon knockout mice that were congenic with 129/Sv controls. They express a shortened intracellular form of p75(NTR) , permitting detection of p75(NTR) -expressing cells. We performed separate counts of choline acetyltransferase (ChAT)-expressing and p75(NTR) -expressing neurons. In agreement with past reports, the number of ChAT-immunoreactive neurons in knockout mice was greater than in wild-type mice, and this was evident in each of the main anatomical divisions of the CBF. In contrast, the number of p75(NTR) -immunoreactive neurons did not differ between genotypes. The biggest increase in ChAT neurons (27%) was in the horizontal limb of the diagonal band of Broca (HDB), in which region the number of p75(NTR) -positive neurons was unchanged. Double staining revealed that some neurons in wild-type mice expressed p75(NTR) but not ChAT. In the knockout mice, all p75(NTR) -expressing neurons expressed ChAT. The increase in cholinergic neurons, therefore, was at least partially attributable to a higher proportion of ChAT immunoreactivity within the population of p75(NTR) -expressing neurons. Cholinergic neurons were also larger in knockout mice than in controls. In the hippocampal CA1 region, knockout mice had a greater number of cholinergic fibers. There was a 77% increase in hippocampal ChAT activity in knockout mice and a 38% increase in heterozygotes. The data do not support an apoptotic role but indicate a broad antineurotrophic role of p75(NTR) in the cholinergic basal forebrain.
Collapse
Affiliation(s)
- Ursula Greferath
- Department of Physiology, University of Melbourne, Parkville, Victoria, Australia
| | | | | |
Collapse
|
38
|
BDNF-promoted increases in proximal dendrites occur via CREB-dependent transcriptional regulation of cypin. J Neurosci 2011; 31:9735-45. [PMID: 21715638 DOI: 10.1523/jneurosci.6785-10.2011] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Alterations in dendrite branching and morphology are present in many neurodegenerative diseases. These variations disrupt postsynaptic transmission and affect neuronal communication. Thus, it is important to understand the molecular mechanisms that regulate dendritogenesis and how they go awry during disease states. Previously, our laboratory showed that cypin, a mammalian guanine deaminase, increases dendrite number when overexpressed and decreases dendrite number when knocked down in cultured hippocampal neurons. Here, we report that exposure to brain-derived neurotrophic factor (BDNF), an important mediator of dendrite arborization, for 72 h but not for 24 h or less increases cypin mRNA and protein levels in rat hippocampal neurons. BDNF signals through cypin to regulate dendrite number, since knocking down cypin blocks the effects of BDNF. Furthermore, BDNF increases cypin levels via mitogen-activated protein kinase and transcription-dependent signaling pathways. Moreover, the cypin promoter region contains putative conserved cAMP response element (CRE) regions, which we found can be recognized and activated by CRE-binding protein (CREB). In addition, exposure of the neurons to BDNF increased CREB binding to the cypin promoter and, in line with these data, expression of a dominant negative form of CREB blocked BDNF-promoted increases in cypin protein levels and proximal dendrite branches. Together, these studies suggest that BDNF increases neuronal cypin expression by the activation of CREB, increasing cypin transcription leading to increased protein expression, thus identifying a novel pathway by which BDNF shapes the dendrite network.
Collapse
|
39
|
The MYCN oncogene and differentiation in neuroblastoma. Semin Cancer Biol 2011; 21:256-66. [PMID: 21849159 DOI: 10.1016/j.semcancer.2011.08.001] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 08/03/2011] [Indexed: 12/13/2022]
Abstract
Childhood neuroblastoma exhibits a heterogeneous clinical behavior ranging from low-risk tumors with the ability to spontaneously differentiate and regress, to high-risk tumors causing the highest number of cancer related deaths in infants. Amplification of the MYCN oncogene is one of the few prediction markers for adverse outcome. This gene encodes the MYCN transcriptional regulator predominantly expressed in the developing peripheral neural crest. MYCN is vital for proliferation, migration and stem cell homeostasis while decreased levels are associated with terminal neuronal differentiation. Interestingly, high-risk tumors without MYCN amplification frequently display increased c-MYC expression and/or activation of MYC signaling pathways. On the other hand, downregulation of MYCN leads to decreased proliferation and differentiation, emphasizing the importance of MYC signaling in neuroblastoma biology. Furthermore, expression of the neurotrophin receptor TrkA is associated with good prognosis, the ability to differentiate and spontaneous regression while expression of the related TrkB receptor is correlated with bad prognosis and MYCN amplification. Here we discuss the role of MYCN in neuroblastoma with a special focus on the contribution of elevated MYCN signaling for an aggressive and undifferentiated phenotype as well as the potential of using MYCN as a therapeutic target.
Collapse
|
40
|
Wyatt SL, Spori B, Vizard TN, Davies AM. Selective regulation of nerve growth factor expression in developing cutaneous tissue by early sensory innervation. Neural Dev 2011; 6:18. [PMID: 21529369 PMCID: PMC3113312 DOI: 10.1186/1749-8104-6-18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 04/30/2011] [Indexed: 11/10/2022] Open
Abstract
Background In the developing vertebrate peripheral nervous system, the survival of sympathetic neurons and the majority of sensory neurons depends on a supply of nerve growth factor (NGF) from tissues they innervate. Although neurotrophic theory presupposes, and the available evidence suggests, that the level of NGF expression is completely independent of innervation, the possibility that innervation may regulate the timing or level of NGF expression has not been rigorously investigated in a sufficiently well-characterized developing system. Results To address this important question, we studied the influence of innervation on the regulation of NGF mRNA expression in the embryonic mouse maxillary process in vitro and in vivo. The maxillary process receives its innervation from predominantly NGF-dependent sensory neurons of the trigeminal ganglion and is the most densely innervated cutaneous territory with the highest levels of NGF in the embryo. When early, uninnervated maxillary processes were cultured alone, the level of NGF mRNA rose more slowly than in maxillary processes cultured with attached trigeminal ganglia. In contrast to the positive influence of early innervation on NGF mRNA expression, the levels of brain-derived neurotrophic factor (BDNF) mRNA and neurotrophin-3 (NT3) mRNA rose to the same extent in early maxillary processes grown with and without trigeminal ganglia. The level of NGF mRNA, but not BDNF mRNA or NT3 mRNA, was also significantly lower in the maxillary processes of erbB3-/- mice, which have substantially fewer trigeminal neurons than wild-type mice. Conclusions This selective effect of initial innervation on target field NGF mRNA expression provokes a re-evaluation of a key assertion of neurotrophic theory that the level of NGF expression is independent of innervation.
Collapse
Affiliation(s)
- Sean L Wyatt
- Molecular Biosciences Research Division, Life Sciences Building, School of Biosciences, Museum Avenue, Cardiff CF10 3AT, UK
| | | | | | | |
Collapse
|
41
|
Ho R, Minturn JE, Simpson AM, Iyer R, Light JE, Evans AE, Brodeur GM. The effect of P75 on Trk receptors in neuroblastomas. Cancer Lett 2011; 305:76-85. [PMID: 21419569 DOI: 10.1016/j.canlet.2011.02.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 02/09/2011] [Accepted: 02/10/2011] [Indexed: 01/04/2023]
Abstract
Neuroblastomas (NBs) with favorable outcome usually express TrkA, whereas unfavorable NBs frequently express TrkB and its cognate ligand BDNF. P75 (p75(LNTR), NGFR, TNFRSF16) binds NGF-related neurotrophins with low affinity and usually is coexpressed with Trk receptors in NBs. Here, we investigated the importance of p75 coexpression with Trk receptors in NBs. We transfected p75 into two Trk-null NB cell lines, SH-SY5Y and NLF that were also engineered to stably express TrkA or TrkB. Cell numbers were compared between single (Trk alone) and double (Trk+p75) transfectants, and proliferation was assessed by flow cytometry. P75 coexpression had little effect on cell growth in Trk NB cells in the absence of ligand, but it increased sensitivity and greatly enhanced the effect of cognate ligand. Exogenous NGF induced greater phosphorylation of TrkA and AKT. This was associated with increased cell number in TrkA/p75 cells compared to TrkA cells (p<0.01), which was due to increased proliferation in TrkA/p75 cells (p<0.05), followed by differentiation. Exogenous BDNF also increased cell number in TrkB/p75 compared to TrkB cells (p<0.01), due to an increase in proliferation, but without differentiation. Coexpression of p75 also increased specificity of Trk-expressing cells to ligand. NT3-induced phosphorylation of TrkA and AKT was reduced in TrkA/p75 cells. NT3-induced phosphorylation of TrkB (as well as AKT and MAPK) was also reduced with p75 coexpression. Our results suggest that p75 plays an important role in enhancing both the sensitivity of Trk receptors to low levels of ligand, as well as increasing the specificity of Trks to their cognate ligands. It also enhances ligand-induced differentiation in TrkA/p75 but not TrkB/p75 cells.
Collapse
Affiliation(s)
- Ruth Ho
- Division of Oncology, The Children's Hospital of Philadelphia, PA 19104, United States
| | | | | | | | | | | | | |
Collapse
|
42
|
Kommaddi RP, Thomas R, Ceni C, Daigneault K, Barker PA. Trk-dependent ADAM17 activation facilitates neurotrophin survival signaling. FASEB J 2011; 25:2061-70. [PMID: 21411748 DOI: 10.1096/fj.10-173740] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Signaling by TrkA and TrkB receptor tyrosine kinase is required for peripheral neuron survival. TrkA and TrkB signaling is facilitated by the p75 neurotrophin receptor (p75NTR), a member of the tumor necrosis factor (TNF) receptor superfamily, through mechanisms that remain obscure. Here, we demonstrate that TrkA and TrkB induces MEK-dependent phosphorylation of the transmembrane cysteine protease ADAM17 (a disintegrin and metalloprotease 17) at the intracellular residue threonine 735. Phosphorylation at this site activates ADAM17 and causes cleavage of p75NTR and production of the receptors' intracellular domain (p75NTR(ICD)) in PC12 cells and in primary cerebellar granule neurons. We show that Trk-induced ADAM17 phosphorylation and generation of the p75NTR(ICD) is required for neurotrophin-induced Erk and Akt activation and for neurotrophin-dependent survival signaling. Survival of PC12 cells maintained in 10 ng/ml nerve growth factor drops by 47% in cells depleted of ADAM17; this survival deficit is resolved if the p75NTR(ICD) is overexpressed in the ADAM17 depleted cells. These studies identify a novel signaling circuit in which Trk activates ADAM17-dependent p75NTR(ICD) production to feedback to sustain Trk signaling and Trk-dependent survival.
Collapse
Affiliation(s)
- Reddy P Kommaddi
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, Canada, H3A 2B4
| | | | | | | | | |
Collapse
|
43
|
Ceni C, Kommaddi RP, Thomas R, Vereker E, Liu X, McPherson PS, Ritter B, Barker PA. The p75NTR intracellular domain generated by neurotrophin-induced receptor cleavage potentiates Trk signaling. J Cell Sci 2010; 123:2299-307. [PMID: 20530577 DOI: 10.1242/jcs.062612] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The p75 neurotrophin receptor (p75NTR) potentiates Trk signaling, but the underlying mechanisms remain uncertain. Here, we examine the relationship between p75NTR cleavage and Trk signaling. We found that, in PC12 cells, nerve growth factor (NGF) induces rapid and robust alpha-secretase- and gamma-secretase-dependent cleavage of p75NTR, releasing the resulting intracellular domain into the cytosol. Brain-derived neurotrophic factor similarly induces p75NTR cleavage in primary cerebellar granule neurons. p75NTR cleavage occurs by means of Trk-dependent activation of MEK-Erk signaling and induction of alpha-secretase activity, and is independent of ligand binding to p75NTR. Neurons and PC12 cells lacking p75NTR display defects in neurotrophin-dependent Akt activation. Normal Akt activation is rescued using full-length p75NTR or the p75 intracellular domain, but not cleavage-resistant p75NTR. We then demonstrate that NGF-dependent growth arrest of PC12 cells requires p75NTR cleavage and generation of the intracellular domain. We conclude that generation of the soluble p75NTR intracellular domain by Trk-induced cleavage plays a fundamental role in Trk-dependent signaling events.
Collapse
Affiliation(s)
- Claire Ceni
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Sahenk Z, Galloway G, Edwards C, Malik V, Kaspar BK, Eagle A, Yetter B, Forgie A, Tsao D, Lin JC. TrkB and TrkC agonist antibodies improve function, electrophysiologic and pathologic features in Trembler J mice. Exp Neurol 2010; 224:495-506. [PMID: 20553714 DOI: 10.1016/j.expneurol.2010.05.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Revised: 04/26/2010] [Accepted: 05/18/2010] [Indexed: 01/08/2023]
Abstract
Neurotrophic factors have been considered as potential therapeutics for peripheral neuropathies. Previously, we showed that neurotrophin-3 (NT-3) promotes nerve regeneration in Trembler(J) (Tr(J)) mice and in sural nerves from patients with Charcot-Marie-Tooth 1A (CMT1A). The relatively short plasma half-life of NT-3 and other neurotrophins, however, pose a practical difficulty in their clinical application. Therapeutic agonist antibodies (AAb) targeting the neurotrophic receptors may circumvent this obstacle due to their high specificity and long half-life. Using morphological, electrophysiological studies and functional motor testing, we assessed the efficacy of monoclonal TrkC AAb and TrkB AAb in the Tr(J) mice. Treatments of these AAbs individually or in combination over 20 weeks increased compound muscle action potential (CMAP) amplitude, which correlated with improved grip strength, as compared to the PBS control group. Improvements in CMAP amplitude were most prominent with TrkC AAb treatment. In all treatment groups, distal to the crush site of the sciatic nerves exhibited a significantly greater number of myelinated fibers (MFs) indicating improved regenerative response to injury. In the contralateral intact sciatic nerves, the number of MFs as well as the myelin thickness was also increased significantly by the AAb treatments, suggesting that the hypomyelination/amyelination state of the peripheral nerves in Tr(J) improved. Therapeutic response to AAb combination was often, albeit not always, the most prominent, indicating a non-redundant effect of TrkB and TrkC AAbs. An early functional recovery and the correlative morphological changes of enhanced regeneration were seen with TrkC AAb treatment. These results provide evidence for potential therapeutic use of monoclonal agonist antibodies for neurotrophin receptors in CMT1A and other neuropathies.
Collapse
Affiliation(s)
- Zarife Sahenk
- Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, OH 43205, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Moubarak RS, Solé C, Pascual M, Gutierrez H, Llovera M, Pérez-García MJ, Gozzelino R, Segura MF, Iglesias-Guimarais V, Reix S, Soler RM, Davies AM, Soriano E, Yuste VJ, Comella JX. The death receptor antagonist FLIP-L interacts with Trk and is necessary for neurite outgrowth induced by neurotrophins. J Neurosci 2010; 30:6094-105. [PMID: 20427667 PMCID: PMC6632611 DOI: 10.1523/jneurosci.0537-10.2010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 03/17/2010] [Indexed: 12/28/2022] Open
Abstract
FLICE-inhibitory protein (FLIP) is an endogenous inhibitor of the signaling pathway triggered by the activation of death receptors. Here, we reveal a novel biological function for the long form of FLIP (FLIP-L) in neuronal differentiation, which can be dissociated from its antiapoptotic role. We show that FLIP-L is expressed in different regions of the mouse embryonic nervous system. Immunohistochemistry of mouse brain sections at different stages reveals that, in neurons, FLIP is expressed early during the embryonic neuronal development (embryonic day 16) and decreases at later stages (postnatal days 5-15), when its expression is essentially detected in glial cells. FLIP-L overexpression significantly enhances neurotrophin-induced neurite outgrowth in motoneurons, superior cervical ganglion neurons, and PC12 cells. Conversely, the downregulation of FLIP-L protein levels by specific RNA interference significantly reduces neurite outgrowth, even in the presence of the appropriate neurotrophin stimulus. Moreover, NGF-dependent activation of two main intracellular pathways involved in the regulation of neurite outgrowth, extracellular signal-regulated kinases (ERKs) and nuclear factor kappaB (NF-kappaB), is impaired when endogenous FLIP-L is downregulated, although TrkA remains activated. Finally, we demonstrate that FLIP-L interacts with TrkA, and not with p75(NTR), in an NGF-dependent manner, and endogenous FLIP-L interacts with TrkB in whole-brain lysates from embryonic day 15 mice embryos. Altogether, we uncover a new role for FLIP-L as an unexpected critical player in neurotrophin-induced mitogen-activated protein kinase/ERK- and NF-kappaB-mediated control of neurite growth in developing neurons.
Collapse
Affiliation(s)
- Rana S. Moubarak
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Campus de Bellaterra (Edifici M), 08193 Bellaterra, Spain
- Institut de Recerca de l'Hospital Universitari de la Vall d'Hebron, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Carme Solé
- Cell Signaling and Apoptosis Group, Departament de Ciències Mèdiques Bàsiques, Institut de Recerca Biomèdica de Lleida/Universitat de Lleida, 25198 Lleida, Spain
| | - Marta Pascual
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
- Developmental Neurobiology and Regeneration Unit, Institute for Research in Biomedicine, Parc Cientific de Barcelona and Department of Cell Biology, University of Barcelona, Barcelona 08028, Spain, and
| | | | - Marta Llovera
- Cell Signaling and Apoptosis Group, Departament de Ciències Mèdiques Bàsiques, Institut de Recerca Biomèdica de Lleida/Universitat de Lleida, 25198 Lleida, Spain
| | - M. José Pérez-García
- Cell Signaling and Apoptosis Group, Departament de Ciències Mèdiques Bàsiques, Institut de Recerca Biomèdica de Lleida/Universitat de Lleida, 25198 Lleida, Spain
| | - Raffaella Gozzelino
- Cell Signaling and Apoptosis Group, Departament de Ciències Mèdiques Bàsiques, Institut de Recerca Biomèdica de Lleida/Universitat de Lleida, 25198 Lleida, Spain
| | - Miguel F. Segura
- Cell Signaling and Apoptosis Group, Departament de Ciències Mèdiques Bàsiques, Institut de Recerca Biomèdica de Lleida/Universitat de Lleida, 25198 Lleida, Spain
| | - Victoria Iglesias-Guimarais
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Campus de Bellaterra (Edifici M), 08193 Bellaterra, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Stéphanie Reix
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Campus de Bellaterra (Edifici M), 08193 Bellaterra, Spain
- Institut de Recerca de l'Hospital Universitari de la Vall d'Hebron, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Rosa M. Soler
- Cell Signaling and Apoptosis Group, Departament de Ciències Mèdiques Bàsiques, Institut de Recerca Biomèdica de Lleida/Universitat de Lleida, 25198 Lleida, Spain
| | | | - Eduardo Soriano
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
- Developmental Neurobiology and Regeneration Unit, Institute for Research in Biomedicine, Parc Cientific de Barcelona and Department of Cell Biology, University of Barcelona, Barcelona 08028, Spain, and
| | - Victor J. Yuste
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Campus de Bellaterra (Edifici M), 08193 Bellaterra, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Joan X. Comella
- Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Campus de Bellaterra (Edifici M), 08193 Bellaterra, Spain
- Cell Signaling and Apoptosis Group, Departament de Ciències Mèdiques Bàsiques, Institut de Recerca Biomèdica de Lleida/Universitat de Lleida, 25198 Lleida, Spain
- Institut de Recerca de l'Hospital Universitari de la Vall d'Hebron, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| |
Collapse
|
46
|
Pérez-Sánchez F, Milán M, Buendía P, Cano-Jaimez M, Ambrosio S, Rosenthal A, Fariñas I. Prosurvival effect of human wild-type alpha-synuclein on MPTP-induced toxicity to central but not peripheral catecholaminergic neurons isolated from transgenic mice. Neuroscience 2010; 167:261-76. [PMID: 20156526 DOI: 10.1016/j.neuroscience.2010.02.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 02/01/2010] [Accepted: 02/08/2010] [Indexed: 12/15/2022]
Abstract
In the present work we report the generation of a new line of alpha-synuclein (alpha-SYN) transgenic mice in which the human wild-type alpha-SYN cDNA is expressed under the control of a tyrosine hydroxylase (TH) promoter. We provide evidence that the ectopic protein is found in TH expressing neurons of both central and peripheral nervous systems. The transgene is expressed very early in development coinciding with the activity of the TH promoter and in the adult brain the human protein distributes normally to the nerve endings and cell bodies of dopaminergic nigral neurons without any evidence of abnormal aggregation. Our results indicate that expression of human wild-type alpha-SYN does not affect normal development or maintenance of TH immunoreactive nigral neurons, striatal dopamine content, or locomotor activity. Systemic administration of the parkinsonian neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induces a loss of TH immunoreactive nigral neurons and terminals and of dopamine levels to the same degree in both transgenic and non-transgenic adult mice. Intoxication also results in a similar loss of cardiac noradrenaline in both genotypes. Surprisingly, cultured transgenic ventral mesencephalic fetal dopaminergic neurons exhibit complete resistance to cell death induced by 1-methyl-4-phenylpyridinium ion (MPP(+)) intoxication, without changes in dopamine transporter (DAT) surface levels. Interestingly, this protection is not observed in other populations of catecholaminergic neurons such as peripheral sympathetic neurons, despite their high sensitivity to MPP(+)in vitro.
Collapse
Affiliation(s)
- F Pérez-Sánchez
- Departament de Biologia cellular and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat de València, 46100 Burjassot, València, Spain.
| | | | | | | | | | | | | |
Collapse
|
47
|
Vulnerability of peripheral catecholaminergic neurons to MPTP is not regulated by alpha-synuclein. Neurobiol Dis 2010; 38:92-103. [PMID: 20079841 DOI: 10.1016/j.nbd.2010.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 12/19/2009] [Accepted: 01/07/2010] [Indexed: 12/21/2022] Open
Abstract
Although generally considered a prototypical movement disorder, Parkinson's disease is commonly associated with a broad-spectrum of non-motor symptoms, including autonomic dysfunctions caused by significant alterations in catecholaminergic neurons of the peripheral sympathetic nervous system. Here we present evidence that alpha-synuclein is highly expressed by sympathetic ganglion neurons throughout embryonic and postnatal life and that it is found in tyrosine hydroxylase-positive sympathetic fibers innervating the heart of adult mice. However, mice deficient in alpha-synuclein do not exhibit any apparent alterations in sympathetic development. Sympathetic neurons isolated from mouse embryos and early postnatal mice are sensitive to the parkinsonian drug MPTP/MPP(+) and intoxication requires entry of the neurotoxin through the noradrenaline transporter. Furthermore, recovery of noradrenaline from cardiac sympathetic fibers is reduced in adult mice treated with MPTP systemically. However, MPP(+)-induced sympathetic neuron loss in vitro or MPTP-induced cardiac noradrenaline depletion in vivo is not modified in mice lacking alpha-synuclein. This is in clear contrast with the observation that dopaminergic neurons of the central nervous system are significantly less vulnerable to MPTP/MPP(+) in the absence of alpha-synuclein, suggesting different actions of this molecule in central and peripheral catecholaminergic neurons.
Collapse
|
48
|
BDNF is essentially required for the early postnatal survival of nociceptors. Dev Biol 2010; 339:465-76. [PMID: 20067784 DOI: 10.1016/j.ydbio.2010.01.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Revised: 12/13/2009] [Accepted: 01/05/2010] [Indexed: 01/19/2023]
Abstract
Neurotrophins promote the survival of specific types of neurons during development and ensure proper maintenance and function of mature responsive neurons. Significant effects of BDNF (Brain-Derived Neurotrophic Factor) on pain physiology have been reported but the contribution of this neurotrophin to the development of nociceptors has not been investigated. We present evidence that BDNF is required for the survival of a significant fraction of peptidergic and non-peptidergic nociceptors in dorsal root ganglia (DRG) postnatally. Bdnf homozygous mutant mice lose approximately half of all nociceptive neurons during the first 2 weeks of life and adult heterozygotes exhibit hypoalgesia and a loss of 25% of all nociceptive neurons. Our in vitro analyses indicate that BDNF-dependent nociceptive neurons also respond to NGF and GDNF. Expression analyses at perinatal times indicate that BDNF is predominantly produced within sensory ganglia and is more abundant than skin-derived NGF or GDNF. Function-blocking studies with BDNF specific antibodies in vitro or cultures of BDNF-deficient sensory neurons suggest that BDNF acts in an autocrine/paracrine way to promote the early postnatal survival of nociceptors that are also responsive to NGF and GDNF. Altogether, the data demonstrate an essential requirement for BDNF in the early postnatal survival of nociceptive neurons.
Collapse
|
49
|
Gavaldà N, Gutierrez H, Davies AM. Developmental switch in NF-kappaB signalling required for neurite growth. Development 2009; 136:3405-12. [PMID: 19762427 DOI: 10.1242/dev.035295] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
For a given cell type, particular extracellular signals generate characteristic patterns of activity in intracellular signalling networks that lead to distinctive cell-type specific responses. Here, we report the first known occurrence of a developmental switch in the intracellular signalling network required for an identical cellular response to the same extracellular signal in the same cell type. We show that although NF-kappaB signalling is required for BDNF-promoted neurite growth from both foetal and postnatal mouse sensory neurons, there is a developmental switch between these stages in the NF-kappaB activation mechanism and the phosphorylation status of the p65 NF-kappaB subunit required for neurite growth. Shortly before birth, BDNF activates NF-kappaB by an atypical mechanism that involves tyrosine phosphorylation of IkappaBalpha by Src family kinases, and dephosphorylates p65 at serine 536. Immediately after birth, BDNF-independent constitutive activation of NF-kappaB signalling by serine phosphorylation of IkappaBalpha and constitutive dephosphorylation of p65 at serine 536 are required for BDNF-promoted neurite growth. This abrupt developmental switch in NF-kappaB signalling in a highly differentiated cell type illustrates an unsuspected plasticity in signalling networks in the generation of identical cellular responses to the same extracellular signal.
Collapse
Affiliation(s)
- Núria Gavaldà
- Cardiff School of Biosciences, Biomedical Building, Wales, UK
| | | | | |
Collapse
|
50
|
Masoudi R, Ioannou MS, Coughlin MD, Pagadala P, Neet KE, Clewes O, Allen SJ, Dawbarn D, Fahnestock M. Biological activity of nerve growth factor precursor is dependent upon relative levels of its receptors. J Biol Chem 2009; 284:18424-33. [PMID: 19389705 PMCID: PMC2709390 DOI: 10.1074/jbc.m109.007104] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Indexed: 01/19/2023] Open
Abstract
Nerve growth factor (NGF) is produced as a precursor called pro-nerve growth factor (proNGF), which is secreted by many tissues and is the predominant form of NGF in the central nervous system. In Alzheimer disease brain, cholinergic neurons degenerate and can no longer transport NGF as efficiently, leading to an increase in untransported NGF in the target tissue. The protein that accumulates in the target tissue is proNGF, not the mature form. The role of this precursor is controversial, and both neurotrophic and apoptotic activities have been reported for recombinant proNGFs. Differences in the protein structures, protein expression systems, methods used for protein purification, and methods used for bioassay may affect the activity of these proteins. Here, we show that proNGF is neurotrophic regardless of mutations or tags, and no matter how it is purified or in which system it is expressed. However, although proNGF is neurotrophic under our assay conditions for primary sympathetic neurons and for pheochromocytoma (PC12) cells, it is apoptotic for unprimed PC12 cells when they are deprived of serum. The ratio of tropomyosin-related kinase A to p75 neurotrophin receptor is low in unprimed PC12 cells compared with primed PC12 cells and sympathetic neurons, altering the balance of proNGF-induced signaling to favor apoptosis. We conclude that the relative level of proNGF receptors determines whether this precursor exhibits neurotrophic or apoptotic activity.
Collapse
Affiliation(s)
| | | | - Michael D. Coughlin
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Promila Pagadala
- the Rosalind Franklin University of Medicine & Science, North Chicago, Illinois 60064, and
| | - Kenneth E. Neet
- the Rosalind Franklin University of Medicine & Science, North Chicago, Illinois 60064, and
| | - Oliver Clewes
- the University of Bristol, Bristol BS1 3NY, United Kingdom
| | | | - David Dawbarn
- the University of Bristol, Bristol BS1 3NY, United Kingdom
| | - Margaret Fahnestock
- From the Department of Biology
- Graduate Program in Neuroscience, and
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| |
Collapse
|