1
|
Stanga S, Boido M, Kienlen-Campard P. How to Build and to Protect the Neuromuscular Junction: The Role of the Glial Cell Line-Derived Neurotrophic Factor. Int J Mol Sci 2020; 22:ijms22010136. [PMID: 33374485 PMCID: PMC7794999 DOI: 10.3390/ijms22010136] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/07/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
The neuromuscular junction (NMJ) is at the crossroad between the nervous system (NS) and the muscle. Following neurotransmitter release from the motor neurons (MNs), muscle contraction occurs and movement is generated. Besides eliciting muscle contraction, the NMJ represents a site of chemical bidirectional interplay between nerve and muscle with the active participation of Schwann cells. Indeed, signals originating from the muscle play an important role in synapse formation, stabilization, maintenance and function, both in development and adulthood. We focus here on the contribution of the Glial cell line-Derived Neurotrophic Factor (GDNF) to these processes and to its potential role in the protection of the NMJ during neurodegeneration. Historically related to the maintenance and survival of dopaminergic neurons of the substantia nigra, GDNF also plays a fundamental role in the peripheral NS (PNS). At this level, it promotes muscle trophism and it participates to the functionality of synapses. Moreover, compared to the other neurotrophic factors, GDNF shows unique peculiarities, which make its contribution essential in neurodegenerative disorders. While describing the known structural and functional changes occurring at the NMJ during neurodegeneration, we highlight the role of GDNF in the NMJ–muscle cross-talk and we review its therapeutic potential in counteracting the degenerative process occurring in the PNS in progressive and severe diseases such as Alzheimer’s disease (AD), Amyotrophic Lateral Sclerosis (ALS) and Spinal Muscular Atrophy (SMA). We also describe functional 3D neuromuscular co-culture systems that have been recently developed as a model for studying both NMJ formation in vitro and its involvement in neuromuscular disorders.
Collapse
Affiliation(s)
- Serena Stanga
- Department of Neuroscience Rita Levi Montalcini, University of Turin, 10126 Turin, Italy;
- Laboratory of Brain Development and Disease, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano, Italy
- National Institute of Neuroscience (INN), 10125 Turin, Italy
- Correspondence:
| | - Marina Boido
- Department of Neuroscience Rita Levi Montalcini, University of Turin, 10126 Turin, Italy;
- Laboratory of Brain Development and Disease, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano, Italy
- National Institute of Neuroscience (INN), 10125 Turin, Italy
| | - Pascal Kienlen-Campard
- Institute of Neuroscience (IoNS), Université Catholique de Louvain (UCLouvain), 1200 Bruxelles, Belgium;
| |
Collapse
|
2
|
Kallestrup M, Møller HJ, Tankisi H, Andersen H. Soluble CD163 levels are elevated in cerebrospinal fluid and serum in people with Type 2 diabetes mellitus and are associated with impaired peripheral nerve function. Diabet Med 2015; 32:54-61. [PMID: 25156085 DOI: 10.1111/dme.12568] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 06/26/2014] [Accepted: 08/20/2014] [Indexed: 12/13/2022]
Abstract
AIMS To measure soluble CD163 levels in the cerebrospinal fluid and serum of people with Type 2 diabetes, with and without polyneuropathy, and to relate the findings to peripheral nerve function. METHODS A total of 22 people with Type 2 diabetes and 12 control subjects without diabetes were included in this case-control study. Participants with diabetes were divided into those with neuropathy (n = 8) and those without neuropathy (n = 14) based on clinical examination, vibratory perception thresholds and nerve conduction studies. Serum and cerebrospinal fluid soluble CD163 levels were analysed using an enzyme-linked immunosorbent assay. RESULTS Soluble CD163 levels were significantly higher in the cerebrospinal fluid and serum of the participants with Type 2 diabetes compared with the control participants [cerebrospinal fluid: median (range) 107 (70-190) vs 84 (54-115) μg/l, P < 0.01 and serum: 2305 (920-7060) vs 1420 (780-2740) μg/l, P < 0.01). Cerebrospinal fluid soluble CD163 was positively related to impaired peripheral nerve conduction (nerve conduction study rank score: r = 0.42; P = 0.0497) and there was a trend for higher levels of soluble CD163 in the cerebrospinal fluid and serum in participants with neuropathy than in those without neuropathy [cerebrospinal fluid: median (range) 131 (86-173) vs 101 (70-190) μg/l, P = 0.08 and serum: 3725 (920-7060) vs 2220 (1130-4780), P = 0.06). CONCLUSIONS Cerebrospinal fluid soluble CD163 level is associated with impaired peripheral nerve function. Higher levels of soluble CD163 in people with diabetic polyneuropathy suggest that inflammation plays a role in the development of neural impairment. The relationship between cerebrospinal fluid soluble CD163 level and peripheral nerve conduction indicates that soluble CD163 may be a potential biomarker for the severity of diabetic polyneuropathy.
Collapse
MESH Headings
- Adult
- Aged
- Antigens, CD/blood
- Antigens, CD/cerebrospinal fluid
- Antigens, Differentiation, Myelomonocytic/blood
- Antigens, Differentiation, Myelomonocytic/cerebrospinal fluid
- Biomarkers/blood
- Biomarkers/cerebrospinal fluid
- Body Mass Index
- Case-Control Studies
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/cerebrospinal fluid
- Diabetes Mellitus, Type 2/physiopathology
- Diabetic Neuropathies/blood
- Diabetic Neuropathies/cerebrospinal fluid
- Diabetic Neuropathies/physiopathology
- Enzyme-Linked Immunosorbent Assay
- Female
- Humans
- Inflammation/blood
- Inflammation/cerebrospinal fluid
- Inflammation/physiopathology
- Male
- Middle Aged
- Neural Conduction
- Receptors, Cell Surface/blood
Collapse
Affiliation(s)
- M Kallestrup
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | |
Collapse
|
3
|
Spooren A, Kolmus K, Laureys G, Clinckers R, De Keyser J, Haegeman G, Gerlo S. Interleukin-6, a mental cytokine. ACTA ACUST UNITED AC 2011; 67:157-83. [PMID: 21238488 DOI: 10.1016/j.brainresrev.2011.01.002] [Citation(s) in RCA: 271] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 12/21/2010] [Accepted: 01/08/2011] [Indexed: 12/18/2022]
Abstract
Almost a quarter of a century ago, interleukin-6 (IL-6) was discovered as an inflammatory cytokine involved in B cell differentiation. Today, IL-6 is recognized to be a highly versatile cytokine, with pleiotropic actions not only in immune cells, but also in other cell types, such as cells of the central nervous system (CNS). The first evidence implicating IL-6 in brain-related processes originated from its dysregulated expression in several neurological disorders such as multiple sclerosis, Alzheimer's disease and Parkinson's disease. In addition, IL-6 was shown to be involved in multiple physiological CNS processes such as neuron homeostasis, astrogliogenesis and neuronal differentiation. The molecular mechanisms underlying IL-6 functions in the brain have only recently started to emerge. In this review, an overview of the latest discoveries concerning the actions of IL-6 in the nervous system is provided. The central position of IL-6 in the neuroinflammatory reaction pattern, and more specifically, the role of IL-6 in specific neurodegenerative processes, which accompany Alzheimer's disease, multiple sclerosis and excitotoxicity, are discussed. It is evident that IL-6 has a dichotomic action in the CNS, displaying neurotrophic properties on the one hand, and detrimental actions on the other. This is in agreement with its central role in neuroinflammation, which evolved as a beneficial process, aimed at maintaining tissue homeostasis, but which can become malignant when exaggerated. In this perspective, it is not surprising that 'well-meant' actions of IL-6 are often causing harm instead of leading to recovery.
Collapse
Affiliation(s)
- Anneleen Spooren
- Laboratory of Eukaryotic Signal Transduction and Gene Expression, University of Ghent, K.L. Ledeganckstraat 35, 9000 Gent, Belgium.
| | | | | | | | | | | | | |
Collapse
|
4
|
Wernicke C, Hellmann J, Zieba B, Kuter K, Ossowska K, Frenzel M, Dencher NA, Rommelspacher H. 9-Methyl-beta-carboline has restorative effects in an animal model of Parkinson's disease. Pharmacol Rep 2010; 62:35-53. [PMID: 20360614 DOI: 10.1016/s1734-1140(10)70241-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Revised: 02/08/2009] [Indexed: 12/19/2022]
Abstract
In a previous study, a primary culture of midbrain cells was exposed to 9-methyl-beta-carboline for 48 h, which caused an increase in the number of tyrosine hydroxylase-positive cells. Quantitative RT-PCR revealed increased transcription of genes participating in the maturation of dopaminergic neurons. These in vitro findings prompted us to investigate the restorative actions of 9-methyl-beta-carboline in vivo. The compound was delivered for 14 days into the left cerebral ventricle of rats pretreated with the neurotoxin 1-methyl-4-phenyl-pyridinium ion (MPP+) for 28 days applying a dose which lowered dopamine by approximately 50%. Interestingly, 9-methyl-beta-carboline reversed the dopamine-lowering effect of the neurotoxin in the left striatum. Stereological counts of tyrosine hydroxylase-immunoreactive cells in the substantia nigra revealed that the neurotoxin caused a decrease in the number of those cells. However, when treated subsequently with 9-methyl-beta-carboline, the number reached normal values. In search of an explanation for the restorative activity, we analyzed the complexes that compose the respiratory chain in striatal mitochondria by 2-dimension gel electrophoresis followed by MALDI-TOF peptide mass fingerprinting.We found no changes in the overall composition of the complexes. However, the activity of complex I was increased by approximately 80% in mitochondria from rats treated with MPP+ and 9-methyl-beta-carboline compared to MPP+ and saline and to sham-operated rats, as determined by measurements of nicotinamide adenine dinucleotide dehydrogenase activity. Microarray technology and single RT-PCR revealed the induction of neurotrophins: brain-derived neurotrophic factor, conserved dopamine neurotrophic factor, cerebellin 1 precursor protein, and ciliary neurotrophic factor. Selected western blots yielded consistent results. The findings demonstrate restorative effects of 9-methyl-beta-carboline in an animal model of Parkinson's disease that improve the effectiveness of the respiratory chain and promote the transcription and expression of neurotrophin-related genes.
Collapse
Affiliation(s)
- Catrin Wernicke
- Department of Psychiatry, CCM, Charité-University Medicine Berlin, Dorotheenstr. 94, 10117 Berlin, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Zorina Y, Iyengar R, Bromberg KD. Cannabinoid 1 receptor and interleukin-6 receptor together induce integration of protein kinase and transcription factor signaling to trigger neurite outgrowth. J Biol Chem 2009; 285:1358-70. [PMID: 19861414 DOI: 10.1074/jbc.m109.049841] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of the G(o/i)-coupled cannabinoid 1 receptor (CB1R) has been shown to induce neurite outgrowth in Neuro2A cells through activation of Src kinase and STAT3 transcription factor. Signaling by the interleukin 6 receptor (IL-6R) also activates STAT3 through Jak kinase. We studied if signals from the two pathways could be integrated in a synergistic manner to trigger neurite outgrowth in Neuro2A cells. At low concentrations, when agonist at either receptor by itself has no effect, we found that CB1R and IL-6R stimulation together induced synergistic neurite outgrowth. Signal integration requires activation of transcription factors by Src, Jak, and mitogen-activated protein kinases. Mitogen-activated protein kinase can be activated by both receptors and shows enhanced early activation in the presence of both ligands. CREB and STAT3 transcription factors are required for synergy and show enhanced DNA-binding activity when both receptors are activated. STAT3 plays a critical role in integration of the signals downstream of the two receptors. When both pathways are activated, STAT3 phosphorylation is sustained for 6 h. This prolonged activation of STAT3 requires deactivation of SHP2 phosphatase. Reduction of SHP2 levels by RNA interference results in greater synergy in neurite outgrowth. Simultaneous knockdown of both SHP2 and STAT3 blocks the synergistic triggering of neurite outgrowth, indicating that STAT3 is downstream of SHP2. CB1R and IL-6R co-stimulation enhanced the differentiation of rat cortical neuron primary cultures. These results provide a mechanism where multiple protein kinases and transcription factors interact to integrate signals from G protein-coupled and cytokine receptor to evoke neurite outgrowth in Neuro2A cells.
Collapse
Affiliation(s)
- Yana Zorina
- Department of Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | |
Collapse
|
6
|
Neuroprotective properties of ciliary neurotrophic factor for cultured adult rat dorsal root ganglion neurons. Histochem Cell Biol 2008; 130:669-79. [PMID: 18679704 DOI: 10.1007/s00418-008-0484-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2008] [Indexed: 12/15/2022]
Abstract
We observed that recombinant ciliary neurotrophic factor (CNTF) enhanced survival and neurite outgrowth of cultured adult rat dorsal root ganglion (DRG) neurons. Among other neurotrophic factors (NGF and GDNF) and interleukin (IL)-6 cytokine members [IL-6, LIF, cardiotrophin-1, and oncostatin M (OSM)] at the same concentration (50 ng/ml), CNTF, as well as LIF and OSM, displayed high efficacy for the promotion of the number of viable neurons and neurite-bearing cells. CNTF enhanced the number of neurite-bearing cells in both small neurons (soma diameter <30 microm) and large neurons (soma diameter > or =30 microm), whereas NGF and GDNF promoted that in only small neurons. Western blot analysis revealed that CNTF induced phosphorylation of STAT3, Akt, and ERK1/2 in the neurons. Furthermore, the neurite outgrowth-promoting activity of CNTF was diminished by co-treatment with Janus kinase (JAK) 2 inhibitor, AG490; STAT3 inhibitor, STA-21; phosphatidyl inositol-3'-phosphate-kinase (PI3K) inhibitor, LY294002; and mitogen-activated protein kinase kinase (MEK) inhibitor, PD98059, in a concentration-dependent manner. Its survival-promoting activity was also affected by AG490, STA-21, and LY294002 at higher concentrations, but not by PD98059. These findings suggest the involvement of JAK2/STAT3, PI3K/Akt, and MEK/ERK signaling pathways in CNTF-induced neurite outgrowth, where the former two pathways are thought to play major roles in mediating the survival response of neurons to CNTF.
Collapse
|
7
|
Doherty GH, Oldreive C, Harvey J. Neuroprotective actions of leptin on central and peripheral neurons in vitro. Neuroscience 2008; 154:1297-307. [PMID: 18550292 DOI: 10.1016/j.neuroscience.2008.04.052] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2008] [Revised: 04/23/2008] [Accepted: 04/25/2008] [Indexed: 11/16/2022]
Abstract
Neuronal cell death and its regulation have been extensively studied as an essential process of both neurodevelopment and neurodegenerative conditions. However it is not clear how circulating hormones influence such processes. Therefore we aimed to determine whether the anti-obesity hormone leptin could promote the survival of murine central and peripheral neurons in vitro. Thus we established primary neuronal cultures of dopaminergic midbrain neurons and trigeminal sensory neurons and induced cell death via either toxic insult or growth factor withdrawal. We demonstrate that leptin promotes the survival of developing peripheral and central neurons via activation of mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3-kinase)/Akt/nuclear factor kappa B (NF-kappaB) -dependent signaling cascades. Specifically, leptin protects dopaminergic midbrain neurons from the apoptotic stimuli, tumor necrosis factor alpha (TNF-alpha) and 6-hydroxydopamine (6-OHDA). In addition, it promotes the survival of postnatal, but not embryonic, trigeminal sensory neurons following neurotrophin withdrawal. Our data reveal a novel neuroprotective role for leptin in the peripheral nervous system while expanding on the known anti-apoptotic role of leptin in the CNS. These findings have important implications for our understanding of neuronal viability.
Collapse
Affiliation(s)
- G H Doherty
- School of Biology, University of St Andrews, West Burn Lane, St Andrews, Fife, UK.
| | | | | |
Collapse
|
8
|
Pachuau J, Martin-Caraballo M. Extrinsic regulation of T-type Ca(2+) channel expression in chick nodose ganglion neurons. Dev Neurobiol 2008; 67:1915-31. [PMID: 17874459 DOI: 10.1002/dneu.20560] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Functional expression of T-type Ca(2+) channels is developmentally regulated in chick nodose neurons. In this study we have tested the hypothesis that extrinsic factors regulate the expression of T-type Ca(2+) channels in vitro. Voltage-gated Ca(2+) currents were measured using whole-cell patch clamp recordings in E7 nodose neurons cultured under various conditions. Culture of E7 nodose neurons for 48 h with a heart extract induced the expression of T-type Ca(2+) channels without any significant effect on HVA currents. T-type Ca(2+) channel expression was not stimulated by survival promoting factors such as BDNF. The stimulatory effect of heart extract was mediated by a heat-labile, trypsin-sensitive factor. Various hematopoietic cytokines including CNTF and LIF mimic the stimulatory effect of heart extract on T-type Ca(2+) channel expression. The stimulatory effect of heart extract and CNTF requires at least 12 h continuous exposure to reach maximal expression and is not altered by culture of nodose neurons with the protein synthesis inhibitor anisomycin, suggesting that T-type Ca(2+) channel expression is regulated by a posttranslational mechanism. Disruption of the Golgi apparatus with brefeldin-A inhibits the stimulatory effect of heart extract and CNTF suggesting that protein trafficking regulates the functional expression of T-type Ca(2+) channels. Heart extract- or CNTF-evoked stimulation of T-type Ca(2+) channel expression is blocked by the Jak/STAT and MAP kinase blockers, AG490 and U0126, respectively. This study provides new insights into the electrical differentiation of placode-derived sensory neurons and the role of extrinsic factors in regulating the functional expression of Ca(2+) channels.
Collapse
Affiliation(s)
- Judith Pachuau
- Department of Biology, University of Vermont, Burlington, VT 05405, USA
| | | |
Collapse
|
9
|
Analysis of leukemia inhibitory factor and leukemia inhibitory factor receptor in embryonic and adult zebrafish (Danio rerio). Dev Biol 2007; 314:250-60. [PMID: 18201692 DOI: 10.1016/j.ydbio.2007.10.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2007] [Revised: 10/07/2007] [Accepted: 10/12/2007] [Indexed: 01/13/2023]
Abstract
Leukemia inhibitory factor (LIF) is a member of the IL-6 cytokine family that functions in the survival, repair and formation of neurons as well as in the maintenance of neural and embryonic stem cells. The functions of LIF have been well documented in mammals, however until recently, the presence of IL-6 family cytokines in ectothermic vertebrates has only been speculated. We report on the identification of lif and lifr transcripts in the zebrafish and document the expression of these molecules in the developing embryos and tissues of adult zebrafish. We also examined the phylogenetic relationship between these molecules and other IL-6 cytokine family members known in mammals. In adult zebrafish, lif is expressed in the kidney and brain while lifr is expressed in the kidney, gill, brain, spleen and liver. During zebrafish embryogenesis, lif and lifr are both expressed as early as 12 hours postfertilization (hpf). In developing zebrafish, lif is expressed in the otic vesicle, retina and cranial sensory ganglia, and lifr is strongly expressed in the notochord, forebrain, otic vesicle, cranial ganglia and the retina. Morpholino knockdown of Lif and Lifr in developing embryos suggests that Lifr, but not Lif is required for proper neural development. lifr morpholino-injected embryos exhibit defects in the trigeminal, facial and vagal branchiomotor neurons, and improper axonal development as measured by acetylated tubulin staining. These embryos also display severe hydrocephaly by 48 hpf. This suggests that Lifrs are involved in proper neural development in zebrafish. This is the first evidence of the expression and role of an LIFR-like molecule in developing fish.
Collapse
|
10
|
Gallagher D, Gutierrez H, Gavalda N, O'Keeffe G, Hay R, Davies AM. Nuclear factor-kappaB activation via tyrosine phosphorylation of inhibitor kappaB-alpha is crucial for ciliary neurotrophic factor-promoted neurite growth from developing neurons. J Neurosci 2007; 27:9664-9. [PMID: 17804627 PMCID: PMC3512131 DOI: 10.1523/jneurosci.0608-07.2007] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The cytokine ciliary neurotrophic factor (CNTF) promotes the growth of neural processes from many kinds of neurons in the developing and regenerating adult nervous system, but the intracellular signaling mechanisms mediating this important function of CNTF are poorly understood. Here, we show that CNTF activates the nuclear factor-kappaB (NF-kappaB) transcriptional system in neonatal sensory neurons and that blocking NF-kappaB-dependent transcription inhibits CNTF-promoted neurite growth. Selectively blocking NF-kappaB activation by the noncanonical pathway that requires tyrosine phosphorylation of inhibitor kappaB-alpha (IkappaB-alpha), but not by the canonical pathway that requires serine phosphorylation of IkappaB-alpha, also effectively inhibits CNTF-promoted neurite growth. CNTF treatment activates spleen tyrosine kinase (SYK) whose substrates include IkappaB-alpha. CNTF-induced SYK phosphorylation is rapidly followed by increased tyrosine phosphorylation of IkappaB-alpha, and blocking SYK activation or tyrosine phosphorylation of IkappaB-alpha prevents CNTF-induced NF-kappaB activation and CNTF-promoted neurite growth. These findings demonstrate that NF-kappaB signaling by an unusual activation mechanism is essential for the ability of CNTF to promote the growth of neural processes in the developing nervous system.
Collapse
Affiliation(s)
- Denis Gallagher
- School of Biosciences, Cardiff CF10 3US, United Kingdom, and
| | | | - Nuria Gavalda
- School of Biosciences, Cardiff CF10 3US, United Kingdom, and
| | - Gerard O'Keeffe
- School of Biosciences, Cardiff CF10 3US, United Kingdom, and
| | - Ron Hay
- School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Alun M. Davies
- School of Biosciences, Cardiff CF10 3US, United Kingdom, and
| |
Collapse
|
11
|
Abstract
The ability of the skin to serve as a protective shield against environmental challenges and as a sensitive detector and responder to thermal, chemical, and mechanical stimuli speaks to its exquisite design. A central feature of this design is the diverse array of neuronal afferents that convey and respond to sensory stimuli that the skin encounters. Cutaneous neuron development, form, and function are highly dependent on communication with the skin through its production of multiple growth factor proteins that modulate afferent development, maturation, and function. Production by the skin of neurotrophin growth factors and members of the glial cell line-derived neurotrophic factor family are particularly important for support of specific subsets of sensory neurons with unique phenotypic and functional properties. Although these proteins have central roles in afferent development and function, challenges remain in identifying specific molecular mechanisms of growth factor communication and understanding how activation of signaling pathways direct neuron differentiation and function under normal and pathological conditions.
Collapse
Affiliation(s)
- Kathryn M Albers
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | | |
Collapse
|
12
|
Levy YS, Gilgun-Sherki Y, Melamed E, Offen D. Therapeutic potential of neurotrophic factors in neurodegenerative diseases. BioDrugs 2005; 19:97-127. [PMID: 15807629 DOI: 10.2165/00063030-200519020-00003] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
There is a vast amount of evidence indicating that neurotrophic factors play a major role in the development, maintenance, and survival of neurons and neuron-supporting cells such as glia and oligodendrocytes. In addition, it is well known that alterations in levels of neurotrophic factors or their receptors can lead to neuronal death and contribute to the pathogenesis of neurodegenerative diseases such as Parkinson disease, Alzheimer disease, Huntington disease, amyotrophic lateral sclerosis, and also aging. Although various treatments alleviate the symptoms of neurodegenerative diseases, none of them prevent or halt the neurodegenerative process. The high potency of neurotrophic factors, as shown by many experimental studies, makes them a rational candidate co-therapeutic agent in neurodegenerative disease. However, in practice, their clinical use is limited because of difficulties in protein delivery and pharmacokinetics in the central nervous system. To overcome these disadvantages and to facilitate the development of drugs with improved pharmacotherapeutic profiles, research is underway on neurotrophic factors and their receptors, and the molecular mechanisms by which they work, together with the development of new technologies for their delivery into the brain.
Collapse
Affiliation(s)
- Yossef S Levy
- Laboratory of Neuroscineces, Felsenstein Medical Research Center, Israel
| | | | | | | |
Collapse
|
13
|
Jelaso AM, DeLong C. NGF and IL-1beta are co-localized in the developing nervous system of the frog, Xenopus laevis. Int J Dev Neurosci 2005; 23:575-86. [PMID: 16143489 DOI: 10.1016/j.ijdevneu.2005.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2005] [Revised: 07/21/2005] [Accepted: 07/21/2005] [Indexed: 12/06/2022] Open
Abstract
NGF, a neurotrophic factor best known for its role in promoting cell survival, regulates many neurodevelopmental processes, including synaptic plasticity, neurite outgrowth and programmed cell death. Although there is a large amount of data regarding NGF in the developing nervous system of many species, there is little known about its regulation and role in the frog, Xenopus laevis. In this report, immunocytochemistry was used to characterize NGF protein expression in developing tadpoles. Protein expression was analyzed in tadpoles from stage 44/45 through stage 50, a period of development characterized by extensive neurite outgrowth, neuronal differentiation and an initial period of programmed cell death. Similar to other species, NGF was expressed in sensory cells and tissues, including the inner ear, eye, olfactory system, lateral line organs, papillae in the oral cavity, and gills tufts. In addition, NGF was expressed in specific cells in the central nervous system, cranial and dorsal root ganglia, spinal sensory and motoneurons, and muscle tissues in the tail and body cavity. In the mammalian nervous system, the cytokine, interleukin-1beta (IL-1beta) induces expression of NGF. In this report, double-label immunocytochemistry was used to determine the relationship between NGF and IL-1beta. Results showed most cell types and/or tissues that expressed NGF also expressed IL-1beta. However, NGF was typically associated with cellular and nuclear membranes, whereas IL-1beta appeared in the cytoplasm and nucleolus. The nuclear localization of IL-1beta supports the idea that it regulates gene transcription in the frog. The appearance of NGF and IL-1beta in the same cells suggests they may interact to influence neural development.
Collapse
Affiliation(s)
- Anna M Jelaso
- Environmental Institute, Western Michigan University, Kalamazoo, MI 49008, USA
| | | |
Collapse
|
14
|
|
15
|
Gould TW, Oppenheim RW. The function of neurotrophic factor receptors expressed by the developing adductor motor pool in vivo. J Neurosci 2004; 24:4668-82. [PMID: 15140938 PMCID: PMC6729401 DOI: 10.1523/jneurosci.0580-04.2004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We examined the spatio-temporal relationship between neurotrophic factor receptor (NTF-R) expression and motoneuron (MN) survival in the developing avian spinal cord and observed heterogeneity in the expression of NTF-Rs between, but not within, pools of MNs projecting to individual muscles. We then focused on the role of NTFs in regulating the survival of one motor pool of MNs, all of which innervate a pair of adductor muscles in the thigh and hence compete for survival during the period of programmed cell death (PCD). The complete NTF-R complement of these MNs was analyzed and found to include many, but not all, NTF-Rs. Treatment with exogenous individual NTFs rescued some, but not all, adductor MNs expressing appropriate NTF-Rs. In contrast, administration of multiple NTFs completely rescued adductor MNs from PCD. Additionally, adductor MNs were partially rescued from PCD by NTFs for which they failed to express receptors. NTF-Rs expressed by the nerve but not in the muscle target were capable of mediating survival signals to MNs in trans. Finally, the expression of some NTF-Rs by adductor MNs was not required for MN survival. These studies demonstrate the complexity in NTF regulation of a defined subset of competing MNs and suggest that properties other than NTF-R expression itself can play a role in mediating trophic responses to NTFs.
Collapse
Affiliation(s)
- Thomas W Gould
- Department of Neurobiology and Anatomy and Neuroscience Program, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | | |
Collapse
|
16
|
Gard AL, Gavin E, Solodushko V, Pennica D. Cardiotrophin-1 in choroid plexus and the cerebrospinal fluid circulatory system. Neuroscience 2004; 127:43-52. [PMID: 15219667 DOI: 10.1016/j.neuroscience.2004.03.065] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2004] [Indexed: 01/29/2023]
Abstract
There is a growing recognition of choroid plexus functioning as a source of neuropeptides, cytokines and growth factors in cerebrospinal fluid (CSF) with diffusional access into brain parenchyma. In this study, choroid plexus and other components of the CSF circulatory system were investigated by Western blotting, reverse transcriptase polymerase chain reaction and immunohistochemistry for production of interleukin-6-related cytokines characterized by neuroactivity [cardiotrophin-1 (CT-1), ciliary neurotrophic factor, leukemia inhibitory factor, oncostatin M] and signaling through the gp130/leukemia inhibitory factor receptor-beta receptor heterodimer. Western blot analysis showed that CT-1 was the only cytokine family member detectable in adult rat choroid plexus, as in leptomeninges. The specificity of detection was verified with blots of the same tissues from CT-1-deficient mice. Levels of both CT-1 mRNA and protein were constitutively high in rat from birth through adulthood in choroid plexus, up-regulated postnatally in leptomeninges and undetectable in brain parenchyma. Using antigen retrieval, CT-1 immunolocalized to choroid epithelial cells in all choroid plexuses in addition to leptomeninges (arachnoid and pial-glial membranes). Ependymal cells lining the ventricular neuroaxis, unlike the central canal, were also CT-1-immunoreactive. Western blots indicated rat choroid epithelial cells express and release CT-1 immunoreactivity under defined culture conditions and also revealed the presence of a CT-1-like protein in human choroid plexus and CSF. Previously, CT-1 has been conceptualized to function as a target-derived factor for PNS neurons. Our study clearly demonstrates production of CT-1 in the postnatal and adult CNS, specifically by cell types comprising the blood-CSF barrier, and its accumulation in ventricular ependyma. This finding has broad implications for CT-1 functioning apart from other leukemia inhibitory factor receptor ligands as a CSF-borne signal of brain homeostasis, one possibly involving regulation of the barrier itself, the ependyma or target cells in the surrounding parenchyma, including the subventricular zone. A rationale for studies examining CT-1-deficient mice in these respects is provided by the data.
Collapse
Affiliation(s)
- A L Gard
- Department of Cell Biology and Neuroscience, 2038 Medical Sciences Building, University of South Alabama College of Medicine, Mobile, AL 36688-0002, USA.
| | | | | | | |
Collapse
|
17
|
Aperghis M, Johnson IP, Patel N, Khadir A, Cannon J, Goldspink G. Age, diet and injury affect the survival of facial motoneurons. Neuroscience 2003; 117:97-104. [PMID: 12605896 DOI: 10.1016/s0306-4522(02)00762-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Using the model of facial nerve avulsion, we have compared the effects of injury, age and diet on motoneuronal survival. One to four weeks after nerve avulsion, 50-75% motoneuron loss was quantified in ad libitum-fed rats aged 7 days (neonate), 6 months (adult) and 24 months (aging) at the time of injury. Evidence of apoptosis was found for neonatal rats at 3 days post-injury, but not for neonates examined 7 days or adult or aging rats examined 1 month after injury. Non-operated, ad libitum-fed rats showed no significant loss of facial motoneurons by 24 months. Surprisingly, non-operated rats whose food intake was restricted to 15 g standard rat chow per day from the age of 6 months lost 50% of their motoneurons by 24 months. Facial nerve avulsion of 24-month-old rats raised on this restricted diet did not result in any additional loss of motoneurons one month after injury. These results challenge the common view that aging results in neuronal loss and that dietary restriction is universally beneficial.
Collapse
Affiliation(s)
- M Aperghis
- Department of Anatomy and Developmental Biology, Royal Free and University College Medical School, Rowland Hill Street, London NW3 2PF, UK
| | | | | | | | | | | |
Collapse
|
18
|
Lacroix S, Chang L, Rose-John S, Tuszynski MH. Delivery of hyper-interleukin-6 to the injured spinal cord increases neutrophil and macrophage infiltration and inhibits axonal growth. J Comp Neurol 2002; 454:213-28. [PMID: 12442313 DOI: 10.1002/cne.10407] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cytokine growth factors of the interleukin (IL)-6 family have recently been shown to play an important role in central nervous system (CNS) development, repair, and inflammation. These cytokines, which interact via specific membrane receptors, share a signal-transducing receptor subunit, glycoprotein 130 (gp130). Gp130 is expressed by motoneurons in the gray matter of the rat spinal cord and by several brainstem nuclei that project to the spinal cord including the red, reticular, and vestibular nuclei. In this study, we examined whether stimulation of gp130 signaling, with the use of grafts of fibroblasts genetically modified to deliver the fusion protein, hyper-IL-6 (H-IL-6), which consists of the cytokine growth factor, IL-6, and its alpha receptor, would elicit growth of injured spinal cord axons. Particular emphasis was placed on examining the potentially competing effects of growth factor versus proinflammatory influences of H-IL-6 in the context of spinal cord injury. Our results demonstrated that grafts delivering H-IL-6 induce a sixfold increase in the number of neutrophils (P < 0.05) and a twofold increase in the areas of spinal tissue occupied by macrophages and activated microglia (P < 0.01) at the site of the spinal cord injury when compared with control grafts. Of note, this augmentation in inflammatory cell infiltration correlated with a significant twofold increase in lesion size (P < 0.05) and a fourfold reduction in axonal growth (P < 0.01) at the lesion site. Thus, potential neurotrophic properties of this cytokine family of growth factors must be balanced against their inflammatory properties when considering therapeutic application to CNS injury.
Collapse
Affiliation(s)
- Steve Lacroix
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093, USA
| | | | | | | |
Collapse
|
19
|
Jüttler E, Tarabin V, Schwaninger M. Interleukin-6 (IL-6): a possible neuromodulator induced by neuronal activity. Neuroscientist 2002; 8:268-75. [PMID: 12061506 DOI: 10.1177/1073858402008003012] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
IL-6 and its receptor(s) are found in the CNS in health and disease. Cellular sources are glial cells and neurons. Glial production of IL-6 has intensively been studied, but comparatively little is known about the induction of IL-6 in neurons. Emerging evidence suggests that IL-6 possesses neurotrophic properties. Recent data show that neuronal IL-6 expression is induced by excitatory amino acids or membrane depolarization. This implicates that IL-6 is produced not only under pathological conditions but may play a critical role as a physiological neuromodulator that is induced by neuronal activity and regulates brain functions. In the following article, the authors review the current data on IL-6 expression in neurons, with special reference to the induction of IL-6 by neuronal activity. They discuss its direct and indirect effects as a neuromodulator and speculate about the possible function of IL-6 as a physiological regulatory molecule and as a neuroprotective agent in brain pathology.
Collapse
Affiliation(s)
- Eric Jüttler
- Department of Neurology, University of Heidelberg, Germany
| | | | | |
Collapse
|
20
|
Lacroix S, Tuszynski MH. Neurotrophic factors and gene therapy in spinal cord injury. Neurorehabil Neural Repair 2001; 14:265-75. [PMID: 11402877 DOI: 10.1177/154596830001400403] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Although it was once thought that the central nervous system (CNS) of mammals was incapable of substantial recovery from injury, it is now clear that the adult CNS remains responsive to various substances that can promote cell survival and stimulate axonal growth. Among these substances are growth factors, including the neurotrophins and cytokines, and growth-supportive cells such as Schwann cells, olfactory ensheathing glia, and stem cells. We review the effects of these substances on promoting axonal growth after spinal cord injury, placing particular emphasis on the genetic delivery of nervous system growth factors to specific sites of injury as a means of promoting axonal growth and, in limited instances, functional recovery.
Collapse
Affiliation(s)
- S Lacroix
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0626, USA
| | | |
Collapse
|
21
|
Alonzi T, Middleton G, Wyatt S, Buchman V, Betz UA, Müller W, Musiani P, Poli V, Davies AM. Role of STAT3 and PI 3-kinase/Akt in mediating the survival actions of cytokines on sensory neurons. Mol Cell Neurosci 2001; 18:270-82. [PMID: 11591128 DOI: 10.1006/mcne.2001.1018] [Citation(s) in RCA: 113] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The binding of cytokines to the gp130 receptor activates the STAT3, MEK/MAPK, and PI3K/Akt signalling pathways. To assess the relative importance of these pathways in promoting the survival of cytokine-dependent neurons, we conditionally inactivated STAT3 in mice and inhibited MEK, PI3K, and Akt in cultured neurons using pharmacological reagents and by expressing specific inhibitory proteins. Inactivation of STAT3 enhanced the death of the cytokine-dependent sensory neurons of the nodose ganglion in vivo and substantially reduced the response of these neurons to CNTF and LIF in vitro. LY294002, an inhibitor of PI3K, but not PD98059, an inhibitor of MEK, markedly reduced the response of these neurons to CNTF, as did dominant-negative PI3K, dominant-negative Akt, and overexpression of Ruk (a natural PI3K inhibitor). These results demonstrate that STAT3 and PI3K/Akt signalling play major roles in mediating the survival response of neurons to cytokines.
Collapse
Affiliation(s)
- T Alonzi
- School of Life Sciences, Wellcome Trust Biocentre, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Siegel GJ, Chauhan NB. Neurotrophic factors in Alzheimer's and Parkinson's disease brain. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2000; 33:199-227. [PMID: 11011066 DOI: 10.1016/s0165-0173(00)00030-8] [Citation(s) in RCA: 382] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The biomedical literature on the subject of neurotrophic growth factors has expanded prodigiously. This essay reviews neurotrophic factors (NTF) and their receptors in Alzheimer's disease (AD) and Parkinson's disease (PD) brain and recent updates on receptor signaling. The hypotheses for specific NTF involvement in neurodegenerative diseases in human and as potential therapy are based mainly on experimental animal and in vitro models. There are wide gaps in information on regional synthesis and cell contents of NTFs and their receptors in human brain. Observations on AD brain indicate increases in NGF and decreases in BDNF in surviving neurons of hippocampus and certain neocortical regions and decreases in TrkA in cortex and nucleus basalis. In PD brain, the few data available indicate decreases in neuronal content of GDNF and bFGF in surviving substantia nigra dopaminergic neurons. There are very few data regarding age-dependent effects on NTFs and on their receptors in human brain. Since NTFs in neurons are subject to retrograde and, in at least some cases, to anterograde transport from and to target neurons, their effects may be related to synthesis in local or remote sites or to changes in axoplasmic transport. Also, certain NTFs and their receptors are found to be expressed in activated glia. Thus, comparative in situ data for transcription levels and protein contents for NTFs and their receptors in both sites of neuronal origin and termination in human brain are needed to understand their potential roles in treating human diseases.
Collapse
Affiliation(s)
- G J Siegel
- Neurology Service (127), Edward Hines, Jr, Veterans Affairs Hospital, Bldg. #1, Rm#F-201, 60141, Hines, IL, USA.
| | | |
Collapse
|
23
|
Holcomb IN, Kabakoff RC, Chan B, Baker TW, Gurney A, Henzel W, Nelson C, Lowman HB, Wright BD, Skelton NJ, Frantz GD, Tumas DB, Peale FV, Shelton DL, Hébert CC. FIZZ1, a novel cysteine-rich secreted protein associated with pulmonary inflammation, defines a new gene family. EMBO J 2000; 19:4046-55. [PMID: 10921885 PMCID: PMC306596 DOI: 10.1093/emboj/19.15.4046] [Citation(s) in RCA: 476] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Bronchoalveolar lavage fluid from mice with experimentally induced allergic pulmonary inflammation contains a novel 9.4 kDa cysteine-rich secreted protein, FIZZ1 (found in inflammatory zone). Murine (m) FIZZ1 is the founding member of a new gene family including two other murine genes expressed, respectively, in intestinal crypt epithelium and white adipose tissue, and two related human genes. In control mice, FIZZ1 mRNA and protein expression occur at low levels in a subset of bronchial epithelial cells and in non-neuronal cells adjacent to neurovascular bundles in the peribronchial stroma, and in the wall of the large and small bowel. During allergic pulmonary inflammation, mFIZZ1 expression markedly increases in hypertrophic, hyperplastic bronchial epithelium and appears in type II alveolar pneumocytes. In vitro, recombinant mFIZZ1 inhibits the nerve growth factor (NGF)-mediated survival of rat embryonic day 14 dorsal root ganglion (DRG) neurons and NGF-induced CGRP gene expression in adult rat DRG neurons. In vivo, FIZZ1 may modulate the function of neurons innervating the bronchial tree, thereby altering the local tissue response to allergic pulmonary inflammation.
Collapse
Affiliation(s)
- I N Holcomb
- Department of Pathology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Arvanov VL, Seebach BS, Mendell LM. NT-3 evokes an LTP-like facilitation of AMPA/kainate receptor-mediated synaptic transmission in the neonatal rat spinal cord. J Neurophysiol 2000; 84:752-8. [PMID: 10938302 DOI: 10.1152/jn.2000.84.2.752] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neurotrophin-3 (NT-3) is a neurotrophic factor required for survival of muscle spindle afferents during prenatal development. It also acts postsynaptically to enhance the monosynaptic excitatory postsynaptic potential (EPSP) produced by these fibers in motoneurons when applied over a period of weeks to the axotomized muscle nerve in adult cats. Similar increases in the amplitude of the monosynaptic EPSP in motoneurons are observed after periodic systemic treatment of neonatal rats with NT-3. Here we show an acute action of NT-3 in enhancing the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA/kainate) receptor-mediated fast monosynaptic EPSP elicited in motoneurons by dorsal root (DR) stimulation in the in vitro hemisected neonatal rat spinal cord. The receptor tyrosine kinase inhibitor K252a blocks this action of NT-3 as does the calcium chelator bis-(o-aminophenoxy)-N,N,N',N'-tetraacetic acid (BAPTA) injected into the motoneuron. The effect of NT-3 resembles long-term potentiation (LTP) in that transient bath application of NT-3 to the isolated spinal cord produces a long-lasting increase in the amplitude of the monosynaptic EPSP. An additional similarity is that activation of N-methyl-D-aspartate (NMDA) receptors is required to initiate this increase but not to maintain it. The NMDA receptor blocker MK-801, introduced into the motoneuron through the recording microelectrode, blocks the effect of NT-3, indicating that NMDA receptors in the motoneuron membrane are crucial. The effect of NT-3 on motoneuron NMDA receptors is demonstrated by its enhancement of the depolarizing response of the motoneuron to bath-applied NMDA in the presence of tetrodotoxin (TTX). The potentiating effects of NT-3 do not persist beyond the first postnatal week. In addition, EPSPs with similar properties evoked in the same motoneurons by stimulation of descending fibers in the ventrolateral funiculus (VLF) are not modifiable by NT-3 even in the initial postnatal week. Thus, NT-3 produces synapse-specific and age-dependent LTP-like enhancement of AMPA/kainate receptor-mediated synaptic transmission in the spinal cord, and this action requires the availability of functional NMDA receptors in the motoneuron.
Collapse
Affiliation(s)
- V L Arvanov
- Department of Neurobiology and Behavior, State University of New York at Stony Brook, Stony Brook, New York 11794-5230, USA
| | | | | |
Collapse
|
25
|
Leukemia inhibitory factor requires concurrent p75LNTR signaling to induce apoptosis of cultured sympathetic neurons. J Neurosci 2000. [PMID: 10818155 DOI: 10.1523/jneurosci.20-11-04198.2000] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Apoptosis may result either from positive induction by ligand binding to a plasma membrane receptor or from negative induction attributable to loss of a suppressor signal. For example, apoptosis of developing sympathetic neurons may be induced in culture either by exposure to leukemia inhibitory factor (LIF) or by deprivation of nerve growth factor. This study compared the cell death pathways activated in sympathetic neurons by these two different stimuli. Both types of cell death were developmentally regulated; both were maximal in the immediate postnatal period and disappeared over the next 2 weeks. Both types of cell death were reduced by genetic deletion of Bax or by virally mediated overexpression of Bcl-2. Similarly both were reduced by inhibition of caspase activity or by inhibition of Nedd-2 synthesis with antisense oligonucleotides. Finally, both involved activation of c-Jun N-terminal kinase (JNK) signaling. Nedd-2 expression by sympathetic neurons declined in parallel with the developmental loss of LIF-mediated cell death, suggesting that downregulation of the caspase during development may underlie the loss of cytokine-mediated apoptosis. Treatment of sympathetic neurons with an antibody that blocks the function of the low-affinity neurotrophin receptor (p75(LNTR)) prevented LIF-induced cell death. Similarly genetic deletion of p75(LNTR) prevented apoptosis after LIF treatment. These observations suggest that concurrent p75(LNTR) signaling is necessary for LIF-induced cell death and that cytokine-mediated cell death and growth factor deprivation appear to activate the same intracellular pathways involving JNK signaling.
Collapse
|
26
|
Gama SAMD, Mattar Jr. R, Silva CFD, Lainetti RD. Estudo experimental comparativo da ação das neurocinas cardiotrofina-1 e oncostatina-m na regeneração nervosa periférica. ACTA ORTOPEDICA BRASILEIRA 2000. [DOI: 10.1590/s1413-78522000000200001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Os avanços das técnicas microcirúrgicas e o conhecimento detalhado do microambiente da regeneração podem contribuir significativamente na melhoria dos resultados das reparações nervosas periféricas. Nos últimos anos vários autores têm utilizado uma série de tecidos e substâncias interpostos entre os cotos de um nervo periférico seccionado, buscando estimular o crescimento axonal no local da lesão. Através da técnica de tubulização, os autores estudam o efeito de duas neurocinas, a cardiotrofina-1 (CT-1) e a oncostatina-M (OsM), no crescimento axonal e na sobrevida dos neurônios sensitivos nos gânglios da raiz dorsal de L5, após a lesão de nervos ciáticos em camundongos C57BL/6J. Utilizam 3 grupos de 7 animais que tiveram seus nervos seccionados e tubulizados com próteses de polietileno preenchidas com cardiotrofina-1, oncostatina-M e citocromo-C, associadas a um extrato de colágeno. Um quarto grupo de 3 animais, não operados, foi considerado por nós como grupo controle de normalidade. Após 4 semanas da cirurgia, os camundongos foram sacrificados, e realizada a contagem das fibras mielínicas nos cabos de regeneração retirados. Os gânglios das raizes dorsais de L5 também foram dissecados possibilitando a contagem dos neurônios sensitivos. Os dados foram analisados estatisticamente, permitindo concluir que as duas substâncias, utilizadas por nós, foram efetivas no estímulo ao brotamento axonal, porém, as mesmas não conseguiram impedir a morte dos neurônios sensitivos no gânglio da raiz dorsal de L5.
Collapse
|
27
|
Murphy PG, Borthwick LA, Altares M, Gauldie J, Kaplan D, Richardson PM. Reciprocal actions of interleukin-6 and brain-derived neurotrophic factor on rat and mouse primary sensory neurons. Eur J Neurosci 2000; 12:1891-9. [PMID: 10886330 DOI: 10.1046/j.1460-9568.2000.00074.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In low-density, serum-free cultures of neurons from embryonic rat dorsal root ganglia, interleukin-6 supports the survival of less than one third of the neurons yet virtually all of them bear interleukin-6 alpha-receptors. A finding that might explain this selectivity is that interleukin-6 acts on sensory neurons in culture through a mechanism requiring endogenous brain-derived neurotrophic factor. Antibodies or a trkB fusion protein that block the biological activity of brain-derived neurotrophic factor synthesized by dorsal root ganglion neurons also block the survival-promoting actions of interleukin-6 on these neurons. Two results indicate that interleukin-6 influences synthesis of brain-derived neurotrophic factor in adult dorsal root ganglion neurons. Intrathecal infusion of interleukin-6 in rats increases the concentration of brain-derived neurotrophic factor mRNA in rat lumbar dorsal root ganglia. The induction of brain-derived neurotrophic factor in dorsal root ganglion neurons that is seen after nerve injury in rats or wild-type mice is severely attenuated in mice with null mutation of the interleukin-6 gene. In brief, the ability of interleukin-6 to support the survival of embryonic sensory neurons in vitro depends upon the presence of endogenous brain-derived neurotrophic factor and the induction of brain-derived neurotrophic factor in injured adult sensory neurons depends upon the presence of endogenous interleukin-6.
Collapse
MESH Headings
- Age Factors
- Animals
- Antibodies
- Brain-Derived Neurotrophic Factor/analysis
- Brain-Derived Neurotrophic Factor/genetics
- Brain-Derived Neurotrophic Factor/immunology
- Cells, Cultured
- Fetus/cytology
- Ganglia, Spinal/cytology
- Gene Expression Regulation, Developmental
- In Situ Hybridization
- Interleukin-6/genetics
- Interleukin-6/pharmacology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neurons, Afferent/cytology
- Neurons, Afferent/drug effects
- Neurons, Afferent/physiology
- RNA, Messenger/analysis
- Rats
- Rats, Sprague-Dawley
- Receptors, Interleukin-6/metabolism
Collapse
Affiliation(s)
- P G Murphy
- Division of Neurosurgery, Montreal General Hospital and McGill University, Montreal, Canada
| | | | | | | | | | | |
Collapse
|
28
|
Turnley AM, Bartlett PF. Cytokines that signal through the leukemia inhibitory factor receptor-beta complex in the nervous system. J Neurochem 2000; 74:889-99. [PMID: 10693919 DOI: 10.1046/j.1471-4159.2000.0740889.x] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cytokines that signal through the leukemia inhibitory factor (LIF) receptor, such as LIF and ciliary neuronotrophic factor, have a wide range of roles within both the developing and mature nervous system. They play a vital role in the differentiation of neural precursor cells into astrocytes and can prevent or promote neuronal differentiation. One of the conundrums regarding signalling through the LIF receptor is how it can have multiple, often conflicting roles in different cell types, such as enhancing the differentiation of astrocytes while inhibiting the differentiation of some neuronal cells. Factors that can modulate signal transduction downstream of cytokine signalling, such as "suppressor of cytokine signalling" proteins, which inhibit the JAK/STAT but not the mitogen-activated protein kinase pathway, may therefore play an important role in determining how a given cell will respond to cytokine signalling. This review discusses the general effects of cytokine signalling within the nervous system. Special emphasis is placed on differentiation of neural precursor cells and the role that regulation of cytokine signalling may play in how a given precursor cell responds to cytokine stimulation.
Collapse
Affiliation(s)
- A M Turnley
- The Walter and Eliza Hall Institute of Medical Research, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | | |
Collapse
|
29
|
Middleton G, Hamanoue M, Enokido Y, Wyatt S, Pennica D, Jaffray E, Hay RT, Davies AM. Cytokine-induced nuclear factor kappa B activation promotes the survival of developing neurons. J Cell Biol 2000; 148:325-32. [PMID: 10648565 PMCID: PMC2174280 DOI: 10.1083/jcb.148.2.325] [Citation(s) in RCA: 114] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/1999] [Accepted: 12/02/1999] [Indexed: 11/22/2022] Open
Abstract
Ciliary neurotrophic factor (CNTF), leukemia inhibitory factor (LIF), cardiotrophin-1 (CT-1), and interleukin 6 (IL-6) comprise a group of structurally related cytokines that promote the survival of subsets of neurons in the developing peripheral nervous system, but the signaling pathways activated by these cytokines that prevent neuronal apoptosis are unclear. Here, we show that these cytokines activate NF-kappaB in cytokine-dependent developing sensory neurons. Preventing NF-kappaB activation with a super-repressor IkappaB-alpha protein markedly reduces the number of neurons that survive in the presence of cytokines, but has no effect on the survival response of the same neurons to brain-derived neurotrophic factors (BDNF), an unrelated neurotrophic factor that binds to a different class of receptors. Cytokine-dependent sensory neurons cultured from embryos that lack p65, a transcriptionally active subunit of NF-kappaB, have a markedly impaired ability to survive in response to cytokines, but respond normally to BDNF. There is increased apoptosis of cytokine- dependent neurons in p65(-/)- embryos in vivo, resulting in a reduction in the total number of these neurons compared with their numbers in wild-type embryos. These results demonstrate that NF-kappaB plays a key role in mediating the survival response of developing neurons to cytokines.
Collapse
Affiliation(s)
- Gayle Middleton
- School of Biomedical Sciences, Biomedical Science Building, University of St. Andrews, St. Andrews, Fife KY16 9AJ, Scotland
| | - Makoto Hamanoue
- School of Biomedical Sciences, Biomedical Science Building, University of St. Andrews, St. Andrews, Fife KY16 9AJ, Scotland
| | - Yasushi Enokido
- School of Biomedical Sciences, Biomedical Science Building, University of St. Andrews, St. Andrews, Fife KY16 9AJ, Scotland
| | - Sean Wyatt
- School of Biomedical Sciences, Biomedical Science Building, University of St. Andrews, St. Andrews, Fife KY16 9AJ, Scotland
| | - Diane Pennica
- Department of Molecular Oncology, Genentech Inc., South San Francisco, California 94080
| | - Ellis Jaffray
- School of Biomedical Sciences, Biomedical Science Building, University of St. Andrews, St. Andrews, Fife KY16 9AJ, Scotland
| | - Ronald T. Hay
- School of Biomedical Sciences, Biomedical Science Building, University of St. Andrews, St. Andrews, Fife KY16 9AJ, Scotland
| | - Alun M. Davies
- School of Biomedical Sciences, Biomedical Science Building, University of St. Andrews, St. Andrews, Fife KY16 9AJ, Scotland
| |
Collapse
|
30
|
Bordet T, Schmalbruch H, Pettmann B, Hagege A, Castelnau-Ptakhine L, Kahn A, Haase G. Adenoviral cardiotrophin-1 gene transfer protects pmn mice from progressive motor neuronopathy. J Clin Invest 1999; 104:1077-85. [PMID: 10525046 PMCID: PMC408570 DOI: 10.1172/jci6265] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cardiotrophin-1 (CT-1), an IL-6-related cytokine, causes hypertrophy of cardiac myocytes and has pleiotropic effects on various other cell types, including motoneurons. Here, we analyzed systemic CT-1 effects in progressive motor neuronopathy (pmn) mice that suffer from progressive motoneuronal degeneration, muscle paralysis, and premature death. Administration of an adenoviral CT-1 vector to newborn pmn mice leads to sustained CT-1 expression in the injected muscles and bloodstream, prolonged survival of animals, and improved motor functions. CT-1-treated pmn mice showed a significantly reduced degeneration of facial motoneuron cytons and phrenic nerve myelinated axons. The terminal innervation of skeletal muscle, grossly disturbed in untreated pmn mice, was almost completely preserved in CT-1-treated pmn mice. The remarkable neuroprotection conferred by CT-1 might become clinically relevant if CT-1 side effects, including cardiotoxicity, could be circumvented by a more targeted delivery of this cytokine to the nervous system.
Collapse
Affiliation(s)
- T Bordet
- Institut National de la Santé et de la Recherche Médicale (INSERM) U.129, Institut Cochin de Génétique Moléculaire, 75014 Paris, France
| | | | | | | | | | | | | |
Collapse
|
31
|
Nakashima K, Wiese S, Yanagisawa M, Arakawa H, Kimura N, Hisatsune T, Yoshida K, Kishimoto T, Sendtner M, Taga T. Developmental requirement of gp130 signaling in neuronal survival and astrocyte differentiation. J Neurosci 1999. [PMID: 10377352 DOI: 10.1002/(sici)1097-4695(19980915)3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
gp130 is a signal-transducing receptor component used in common by the interleukin-6 (IL-6) family of hematopoietic and neurotrophic cytokines, including IL-6, IL-11, leukemia-inhibitory factor, ciliary neurotrophic factor, oncostatin-M, and cardiotrophin-1. We have examined in this study a role of gp130 in the nervous system by analyzing developmental cell death of several neuronal populations and the differentiation of astrocytes in gp130-deficient mice. A significant reduction was observed in the number of sensory neurons in L5 dorsal root ganglia and motoneurons in the facial nucleus, the nucleus ambiguus, and the lumbar spinal cord in gp130 -/- mice on embryonic day 18.5. On the other hand, no significant neuronal loss was detectable on day 14.5, suggesting a physiological role of gp130 in supporting newly generated neurons during the late phase of development when naturally occurring cell death takes place. Moreover, expression of an astrocyte marker, GFAP, was severely reduced in the brain of gp130 -/- mice. Our data demonstrate that gp130 expression is essential for survival of subgroups of differentiated motor and sensory neurons and for the differentiation of major populations of astrocytes in vivo.
Collapse
Affiliation(s)
- K Nakashima
- Department of Molecular Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, 101-0062, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Developmental requirement of gp130 signaling in neuronal survival and astrocyte differentiation. J Neurosci 1999. [PMID: 10377352 DOI: 10.1523/jneurosci.19-13-05429.1999] [Citation(s) in RCA: 246] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
gp130 is a signal-transducing receptor component used in common by the interleukin-6 (IL-6) family of hematopoietic and neurotrophic cytokines, including IL-6, IL-11, leukemia-inhibitory factor, ciliary neurotrophic factor, oncostatin-M, and cardiotrophin-1. We have examined in this study a role of gp130 in the nervous system by analyzing developmental cell death of several neuronal populations and the differentiation of astrocytes in gp130-deficient mice. A significant reduction was observed in the number of sensory neurons in L5 dorsal root ganglia and motoneurons in the facial nucleus, the nucleus ambiguus, and the lumbar spinal cord in gp130 -/- mice on embryonic day 18.5. On the other hand, no significant neuronal loss was detectable on day 14.5, suggesting a physiological role of gp130 in supporting newly generated neurons during the late phase of development when naturally occurring cell death takes place. Moreover, expression of an astrocyte marker, GFAP, was severely reduced in the brain of gp130 -/- mice. Our data demonstrate that gp130 expression is essential for survival of subgroups of differentiated motor and sensory neurons and for the differentiation of major populations of astrocytes in vivo.
Collapse
|
33
|
Hamanoue M, Middleton G, Wyatt S, Jaffray E, Hay RT, Davies AM. p75-mediated NF-kappaB activation enhances the survival response of developing sensory neurons to nerve growth factor. Mol Cell Neurosci 1999; 14:28-40. [PMID: 10433815 DOI: 10.1006/mcne.1999.0770] [Citation(s) in RCA: 158] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
We have investigated whether the transcription factor NF-kappaB plays a role in regulating neuronal survival by manipulating NF-kappaB activation in the nerve growth factor (NGF)-dependent sensory neurons of the embryonic mouse trigeminal ganglion. Overexpression of either the p65 or the p50 NF-kappaB subunits resulted in NF-kappaB activation and promoted in vitro survival as effectively as NGF. Expression of a superrepressor IkappaB-alpha protein prevented NF-kappaB activation in p65/p50-overexpressing neurons and caused the neurons to die as rapidly as NGF-deprived neurons. NGF treatment also activated NF-kappaB, and preventing this activation with superrepressor IkappaB-alpha reduced the NGF survival response. Antibodies that block binding of NGF to the p75 receptor prevented NGF-induced NF-kappaB activation and reduced the NGF survival response to the same extent as superrepressor IkappaB-alpha. Trigeminal neurons cultured from p65(-/-) embryos showed a reduced survival response to NGF compared with neurons from wild-type embryos and there was increased apoptosis of neurons in the trigeminal ganglia of p65(-/-) embryos in vivo. However, as with p75-deficient sensory neurons, p65-deficient sensory neurons showed a normal survival response to BDNF. These results reveal a role for NF-kappaB in regulating neuronal survival during embryonic development and suggest that in addition to the well-established Trk receptor tyrosine kinase signaling cascade, NGF enhances neuronal survival by signaling via a p75-mediated pathway.
Collapse
Affiliation(s)
- M Hamanoue
- School of Biomedical Sciences, University of St. Andrews, Biomedical Science Building, St. Andrews, KY16 9AJ, Scotland
| | | | | | | | | | | |
Collapse
|
34
|
Huang EJ, Zang K, Schmidt A, Saulys A, Xiang M, Reichardt LF. POU domain factor Brn-3a controls the differentiation and survival of trigeminal neurons by regulating Trk receptor expression. Development 1999; 126:2869-82. [PMID: 10357931 PMCID: PMC2710123 DOI: 10.1242/dev.126.13.2869] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mice lacking the POU domain-containing transcription factor Brn-3a have several neuronal deficits. In the present paper, we show that Brn-3a plays two distinct roles during development of the trigeminal ganglion. In this ganglion, neurons expressing the neurotrophin receptors, TrkB and TrkC, are born between E9.5 and E11.5. In the absence of Brn-3a, very few neurons ever express TrkC, but TrkB-expressing neurons are present at E12.5 in elevated numbers, suggesting that Brn-3a may be a constituent of a regulatory circuit determining which Trk receptor is expressed by these early-born neurons. Most neurons expressing the neurotrophin receptor TrkA are generated between E11.5 and E13.5 in this ganglion and their initial generation is not prevented by absence of Brn-3a. However, after E12. 5, absence of Brn-3a results in a progressive loss in neuronal TrkA and TrkB expression, which leads to a massive wave of apoptosis that peaks at E15.5. Despite complete absence of the Trk receptors at E17. 5 and P0, approximately 30% of the normal complement of neurons survive to birth in Brn-3a mutants. Approximately 70% of these express the GDNF receptor subunit, c-ret; many can be sustained by GDNF, but not by NGF in culture. Thus, the vast majority of surviving neurons are probably sustained in vivo by trophic factor(s) whose receptors are not regulated by Brn-3a. In conclusion, our data indicate the specific functions of Brn-3a in controlling the survival and differentiation of trigeminal neurons by regulating expression of each of the three Trk receptors.
Collapse
Affiliation(s)
- Eric J. Huang
- Program in Neuroscience, Department of Physiology, and Howard Hughes Medical Institute, University of California, San Francisco, CA 94143-0723
| | - Keling Zang
- Program in Neuroscience, Department of Physiology, and Howard Hughes Medical Institute, University of California, San Francisco, CA 94143-0723
| | - Andrea Schmidt
- Program in Neuroscience, Department of Physiology, and Howard Hughes Medical Institute, University of California, San Francisco, CA 94143-0723
| | - Asta Saulys
- Program in Neuroscience, Department of Physiology, and Howard Hughes Medical Institute, University of California, San Francisco, CA 94143-0723
| | - Mengqing Xiang
- Center for Advanced Biotechnology and Medicine, Department of Pediatrics, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ 08854
| | - Louis F. Reichardt
- Program in Neuroscience, Department of Physiology, and Howard Hughes Medical Institute, University of California, San Francisco, CA 94143-0723
- Author for correspondence (e-mail: )
| |
Collapse
|
35
|
Abstract
Interleukin-6 (IL-6) is a multifunctional cytokine mediating inflammatory or immune reactions. Here we investigated the possible role of IL-6 in the intact or lesioned peripheral nervous system using adult IL-6 gene knockout (IL-6(-/-)) mice. Various sensory functions were tested by applying electrophysiological, morphological, biochemical, and behavioral methods. There was a 60% reduction of the compound action potential of the sensory branch of IL-6(-/-) mice as compared with the motor branch in the intact sciatic nerve. Cross sections of L5 DRG of IL-6(-/-) mice showed a shift in the relative size distribution of the neurons. The temperature sensitivity of IL-6(-/-) mice was also significantly reduced. After crush lesion of the sciatic nerve, its functional recovery was delayed in IL-6(-/-) mice as analyzed from a behavioral footprint assay. Measurements of compound action potentials 20 d after crush lesion showed that there was a very low level of recovery of the sensory but not of the motor branch of IL-6(-/-) mice. Similar results of sensory impairments were obtained with mice showing slow Wallerian degeneration (Wlds) and a delayed lesion-induced recruitment of macrophages. However, in contrast to WldS mice, in IL-6(-/-) mice we observed the characteristic lesion-induced invasion of macrophages and the upregulation of low-affinity neurotrophin receptor p75 (p75LNTR) mRNA levels identical to those of IL-6(+/+) mice. Thus, the mechanisms leading to the common sensory deficiencies were different between IL-6(-/-) and WldS mice. Altogether, the results suggest that interleukin-6 is essential to modulate sensory functions in vivo.
Collapse
|
36
|
Abstract
Advances in our understanding of the activation of peripheral damage-sensing neurons (nociceptors) over the past year have been complemented by electrophysiological and imaging studies of central nervous system pain-related centres. The manipulation of gene expression in a reversible and cell type specific way combined with imaging and electrophysiological studies holds promise for helping us to identify the spatial and molecular substrates of pain perception with increasing precision and gives hope for improved analgesic therapies.
Collapse
Affiliation(s)
- J N Wood
- Biology Department University College London, WC1E 6BT, UK. J.
| | | |
Collapse
|
37
|
Turnley AM, Bartlett PF. Nerve growth factor modulates myelin-associated glycoprotein binding to sensory neurons. Int J Dev Neurosci 1999; 17:109-19. [PMID: 10221670 DOI: 10.1016/s0736-5748(98)00068-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Myelin-associated glycoprotein (MAG) is a molecule expressed by myelinating cells at the myelin/axon interface, which binds to an as yet unidentified molecule on neurons. We have used a MAG-immunoglobulin Fc fusion protein to examine the expression and regulation of the MAG binding molecule on sensory neurons in culture. Binding of the MAG-Fc reached a maximum at 24-48 h and was higher on neurons which expressed high levels of neurofilament. Nerve growth factor (NGF) upregulated expression of the MAG binding molecule in a dose dependent manner. Schwann cells co-cultured with sensory neurons in serum-free medium stimulated maximal expression of the MAG binding molecule, which was decreased by addition of anti-NGF to the co-cultures. This indicated that Schwann cells can modulate expression of the MAG binding molecule via production of NGF and may represent a physiological mechanism for regulation of MAG-MAG binding molecule interactions during myelination and remyelination.
Collapse
Affiliation(s)
- A M Turnley
- The Walter and Eliza Hall Institute of Medical Research, PO Royal Melbourne Hospital, Victoria, Australia.
| | | |
Collapse
|
38
|
Davies AM. Developmental changes in the neurotrophic factor survival requirements of peripheral nervous system neurons. PROGRESS IN BRAIN RESEARCH 1999; 117:47-56. [PMID: 9932399 DOI: 10.1016/s0079-6123(08)64006-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Affiliation(s)
- A M Davies
- School of Biological and Medical Sciences, University of St. Andrews, Scotland, UK.
| |
Collapse
|