1
|
Li Y, Wei C, Wang W, Li Q, Wang Z. Tropomyosin receptor kinase B (TrkB) signalling: targeted therapy in neurogenic tumours. J Pathol Clin Res 2022; 9:89-99. [PMID: 36533776 PMCID: PMC9896160 DOI: 10.1002/cjp2.307] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
Tropomyosin receptor kinase B (TrkB), a transmembrane receptor protein, has been found to play a pivotal role in neural development. This protein is encoded by the neurotrophic receptor tyrosine kinase 2 (NTRK2) gene, and its abnormal activation caused by NTRK2 overexpression or fusion can contribute to tumour initiation, progression, and resistance to therapeutics in multiple types of neurogenic tumours. Targeted therapies for this mechanism have been designed and developed in preclinical and clinical studies, including selective TrkB inhibitors and pan-TRK inhibitors. This review describes the gene structure, biological function, abnormal TrkB activation mechanism, and current-related targeted therapies in neurogenic tumours.
Collapse
Affiliation(s)
- Yuehua Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Chengjiang Wei
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Wei Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Zhi‐Chao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| |
Collapse
|
2
|
Knockdown of NCOR2 Inhibits Cell Proliferation via BDNF/TrkB/ERK in NF1-Derived MPNSTs. Cancers (Basel) 2022; 14:cancers14235798. [PMID: 36497280 PMCID: PMC9738545 DOI: 10.3390/cancers14235798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022] Open
Abstract
(1) Background: malignant peripheral nerve sheath tumours (MPNSTs) are aggressive Schwann cell-derived sarcomas with dismal prognoses. Previous studies have shown that nuclear receptor corepressor 2 (NCOR2) plays a vital role in neurodevelopment and in various tumours. However, the impact of NCOR2 on the progression of MPNST remains unclear. (2) Methods: by GEO database, MPNST tissue microarray, and NF1-related tumour tissues and cell lines were used to explore NCOR2 expression level in the MPNSTs. The role and mechanism of NCOR2 in NF1-derived MPNSTs were explored by experiments in vivo and in vitro and by transcriptome high-throughput sequencing. (3) Results: NCOR2 expression is significantly elevated in NF1-derived MPNSTs and is associated with patient 10-year survival time. Knockdown of NCOR2 suppressed NF1-derived MPNST cell proliferation by blocking the cell cycle in the G0/G1 phase. Moreover, decreased NCOR2 expression could down-regulate MAPK signal activity through the BDNF/TrkB pathway. (4) Conclusions: our findings demonstrated that NCOR2 expression is significantly elevated in NF1-derived MPNSTs. NCOR2 knockdown can inhibit NF1-derived MPNST cell proliferation by weakened BDNF/TrkB/ERK signalling. Targeting NF1-derived MPNSTs with TrkB inhibitors, or in combination with ERK inhibitors, may be a novel therapeutic strategy for clinical trials.
Collapse
|
3
|
NGF-Dependent and BDNF-Dependent DRG Sensory Neurons Deploy Distinct Degenerative Signaling Mechanisms. eNeuro 2021; 8:ENEURO.0277-20.2020. [PMID: 33372032 PMCID: PMC7877462 DOI: 10.1523/eneuro.0277-20.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
The nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) are trophic factors required by distinct population of sensory neurons during development of the nervous system. Neurons that fail to receive appropriate trophic support are lost during this period of naturally occurring cell death. In the last decade, our understanding of the signaling pathways regulating neuronal death following NGF deprivation has advanced substantially. However, the signaling mechanisms promoting BDNF deprivation-induced sensory neuron degeneration are largely unknown. Using a well-established in vitro culture model of dorsal root ganglion (DRG), we have examined degeneration mechanisms triggered on BDNF withdrawal in sensory neurons. Our results indicate differences and similarities between the molecular signaling pathways behind NGF and BDNF deprivation-induced death. For instance, we observed that the inhibition of Trk receptors (K252a), PKC (Gö6976), protein translation (cycloheximide; CHX), or caspases (zVAD-fmk) provides protection from NGF deprivation-induced death but not from degeneration evoked by BDNF-withdrawal. Interestingly, degeneration of BDNF-dependent sensory neurons requires BAX and appears to rely on reactive oxygen species (ROS) generation rather than caspases to induce degeneration. These results highlight the complexity and divergence of mechanisms regulating developmental sensory neuron death.
Collapse
|
4
|
Chen S, Tian R, Li H, Chen M, Zhang H, Lin D. Optimized methods for rapidly dissecting spinal cords and harvesting spinal motor neurons with high survival and purity from rats at different embryonic stages. J Spinal Cord Med 2018; 41:281-291. [PMID: 28545340 PMCID: PMC6055952 DOI: 10.1080/10790268.2017.1329075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
STUDY DESIGN Experimental study, protocol optimization. OBJECTIVES To investigate and compare the isolation of spinal motor neurons from embryonic rats at different embryonic stages, and develop optimized methods for rapidly dissecting spinal cords and harvesting spinal motor neurons with high survival and purity. SETTING Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China. METHODS Embryonic rats at different embryonic stages (12-18 days) were used to isolate spinal motor neurons. Their shape and corresponding dissection procedures, time needed and skills were compared. After dissecting and dissociating spinal cords, cells were randomly divided into immunopanning group and control group, in which antibodies to p75NTR were used or not. After plating cells, different recipe were added at different stages in serum-free culture media. Morphological features of cells were observed during development. Immunoflurorescence assay was performed to indentify motor neurons and the proportion of motor neurons in both control and immunopanning group were evaluated and compared. RESULTS We summarized the operation essentials for rapid isolation of spinal cords, as well as compared anatomical features and dissection procedures of embryos at different embryonic stages, which help us to better evaluate the developmental profile and isolate cells by adopting corresponding skills. Through the fast isolation procedure and optimized culture media, cells grow in good viability. Moreover, compared with control group, the purity of spinal motor neurons in the immunopanning group was significantly increased, reaching a proportion of over 95%.
Collapse
Affiliation(s)
- Shudong Chen
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Ruimin Tian
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Hui Li
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meihui Chen
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Hu Zhang
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Dingkun Lin
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China,Guangzhou University of Chinese Medicine, Guangzhou, China,Correspondence to: Dingkun Lin, Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, No.111 Dade Road, Yuexiu District, Guangzhou, 510120, China.
| |
Collapse
|
5
|
Arbat-Plana A, Cobianchi S, Herrando-Grabulosa M, Navarro X, Udina E. Endogenous modulation of TrkB signaling by treadmill exercise after peripheral nerve injury. Neuroscience 2017; 340:188-200. [DOI: 10.1016/j.neuroscience.2016.10.057] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/18/2016] [Accepted: 10/22/2016] [Indexed: 12/20/2022]
|
6
|
Abstract
Peripheral axonal regeneration requires surface-expanding membrane addition. The continuous incorporation of new membranes into the axolemma allows the pushing force of elongating microtubules to drive axonal growth cones forwards. Hence, a constant supply of membranes and cytoskeletal building blocks is required, often for many weeks. In human peripheral nerves, axonal tips may be more than 1 m away from the neuronal cell body. Therefore, in the initial phase of regeneration, membranes are derived from pre-existing vesicles or synthesised locally. Only later stages of axonal regeneration are supported by membranes and proteins synthesised in neuronal cell bodies, considering that the fastest anterograde transport mechanisms deliver cargo at 20 cm/day. Whereas endocytosis and exocytosis of membrane vesicles are balanced in intact axons, membrane incorporation exceeds membrane retrieval during regeneration to compensate for the loss of membranes distal to the lesion site. Physiological membrane turnover rates will not be established before the completion of target reinnervation. In this review, the current knowledge on membrane traffic in axonal outgrowth is summarised, with a focus on endosomal vesicles as the providers of membranes and carriers of growth factor receptors required for initiating signalling pathways to promote the elongation and branching of regenerating axons in lesioned peripheral nerves.
Collapse
Affiliation(s)
- Barbara Hausott
- Division of Neuroanatomy, Department of Anatomy, Histology and Embryology, Medical University Innsbruck, 6020, Innsbruck, Austria
| | - Lars Klimaschewski
- Division of Neuroanatomy, Department of Anatomy, Histology and Embryology, Medical University Innsbruck, 6020, Innsbruck, Austria
| |
Collapse
|
7
|
Identification of a pharmacologically tractable Fra-1/ADORA2B axis promoting breast cancer metastasis. Proc Natl Acad Sci U S A 2013; 110:5139-44. [PMID: 23483055 DOI: 10.1073/pnas.1222085110] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Metastasis confronts clinicians with two major challenges: estimating the patient's risk of metastasis and identifying therapeutic targets. Because they are key signal integrators connecting cellular processes to clinical outcome, we aimed to identify transcriptional nodes regulating cancer cell metastasis. Using rodent xenograft models that we previously developed, we identified the transcription factor Fos-related antigen-1 (Fra-1) as a key coordinator of metastasis. Because Fra-1 often is overexpressed in human metastatic breast cancers and has been shown to control their invasive potential in vitro, we aimed to assess the implication and prognostic significance of the Fra-1-dependent genetic program in breast cancer metastasis and to identify potential Fra-1-dependent therapeutic targets. In several in vivo assays in mice, we demonstrate that stable RNAi depletion of Fra-1 from human breast cancer cells strongly suppresses their ability to metastasize. These results support a clinically important role for Fra-1 and the genetic program it controls. We show that a Fra-1-dependent gene-expression signature accurately predicts recurrence of breast cancer. Furthermore, a synthetic lethal drug screen revealed that antagonists of the adenosine receptor A2B (ADORA2B) are preferentially toxic to breast tumor cells expressing Fra-1. Both RNAi silencing and pharmacologic blockade of ADORA2B inhibited filopodia formation and invasive activity of breast cancer cells and correspondingly reduced tumor outgrowth in the lungs. These data show that Fra-1 activity is causally involved in and is a prognostic indicator of breast cancer metastasis. They suggest that Fra-1 activity predicts responsiveness to inhibition of pharmacologically tractable targets, such as ADORA2B, which may be used for clinical interference of metastatic breast cancer.
Collapse
|
8
|
Hausott B, Vallant N, Hochfilzer M, Mangger S, Irschick R, Haugsten EM, Klimaschewski L. Leupeptin enhances cell surface localization of fibroblast growth factor receptor 1 in adult sensory neurons by increased recycling. Eur J Cell Biol 2011; 91:129-38. [PMID: 22169219 DOI: 10.1016/j.ejcb.2011.09.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 09/15/2011] [Accepted: 09/15/2011] [Indexed: 01/13/2023] Open
Abstract
Fibroblast growth factors (FGFs) act as trophic factors during development and regeneration of the nervous system. FGFs mediate their responses by activation of four types of FGF receptors (FGFR1-4). FGFR1 is expressed in adult sensory neurons of dorsal root ganglia (DRG), and overexpression of FGFR1 enhances FGF-2-induced elongative axon growth in vitro. Ligand-induced activation of FGFR1 is followed by endocytosis and rapid lysosomal degradation. We previously reported that the lysosomal inhibitor leupeptin prevents degradation of FGFR1 and promotes FGF-2-induced elongative axon growth of DRG neurons overexpressing FGFR1. Therefore, we analyzed the effects of leupeptin on intracellular sorting of FGFR1 in PC12 pheochromocytoma cells and DRG neurons. Leupeptin increased colocalization of FGFR1 with lysosomes. Furthermore, leupeptin enhanced the cell surface localization of FGFR1 by increased receptor recycling and this effect was abolished by the recycling inhibitor monensin. In addition, a lysine mutant of FGFR1, which is preferentially recycled back to the cell surface, promoted elongative axon growth of DRG neurons similar to leupeptin. In contrast, the lysosomal inhibitor bafilomycin had no effect on surface localization of FGFR1, inhibited axon growth of DRG neurons and abolished the effects of leupeptin on receptor recycling. Together, our results strongly imply that increased recycling of FGFR1 promotes axon elongation, but not axonal branching, of adult DRG neurons in vitro.
Collapse
Affiliation(s)
- Barbara Hausott
- Division of Neuroanatomy, Innsbruck Medical University, Muellerstrasse 59, 6020 Innsbruck, Austria
| | | | | | | | | | | | | |
Collapse
|
9
|
Optimizing interfacial features to regulate neural progenitor cells using polyelectrolyte multilayers and brain derived neurotrophic factor. Biointerphases 2011; 6:189-99. [DOI: 10.1116/1.3656249] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
10
|
Cytoprotective effects of growth factors: BDNF more potent than GDNF in an organotypic culture model of Parkinson's disease. Brain Res 2011; 1378:105-18. [DOI: 10.1016/j.brainres.2010.12.090] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 12/29/2010] [Indexed: 01/19/2023]
|
11
|
Manns M, Bichler Z, Leske O, Heumann R. Neuronal Ras activation inhibits adult hippocampal progenitor cell division and impairs spatial short-term memory. GENES BRAIN AND BEHAVIOR 2010; 9:525-36. [PMID: 20398060 DOI: 10.1111/j.1601-183x.2010.00584.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A large number of endogenous and exogenous factors have been identified to upregulate and downregulate proliferation, differentiation and/or survival of newborn cells in the adult hippocampus. For studying neuronal mechanisms mediating the impact of those factors, we used a transgenic synRas mouse model expressing constitutively activated Valin12-Harvey Ras selectively in differentiated neurons. BrdU injections showed significantly reduced proliferation of new cells within the adult hippocampus of transgenic animals compared with their wild-type siblings. In contrast, the relative survival of newborn cells was increased in synRas mice, although this effect did not fully compensate for diminished proliferation. Inhibition of progenitor cell proliferation and enhancement of cellular survival were more pronounced in males compared with females. Double labelling and doublecortin immunostaining verified that specifically newborn neurons were decreased in synRas mice. Reduced cell generation was observed already 2 h after BrdU pulse injections, identifying an early precursor cell population as target of the inhibitory transgene effect. Differences in proliferation remained stable after 24 h and were specific for the subgranular zone of the dentate gyrus, as subventricular cell generation was not affected supporting a non-cell autonomous effect on neural hippocampal progenitors. Transgene expression only starts with synaptic differentiation and therefore reduced proliferation must represent an indirect secondary consequence of synRas activity in differentiated neurons. This was associated with impaired spatial short-term memory capacities as observed in a radial maze paradigm. Our data suggest that constantly high Ras activity in differentiated neurons downregulates hippocampal precursor cell generation in the neuronal lineage, but is modulated by sex-dependent factors.
Collapse
Affiliation(s)
- M Manns
- Molecular Neurobiochemistry, Faculty of Chemistry and Biochemistry, Ruhr-University-Bochum, Bochum, FRG.
| | | | | | | |
Collapse
|
12
|
Acute and gradual increases in BDNF concentration elicit distinct signaling and functions in neurons. Nat Neurosci 2010; 13:302-9. [PMID: 20173744 DOI: 10.1038/nn.2505] [Citation(s) in RCA: 263] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 01/22/2010] [Indexed: 12/16/2022]
Abstract
Extracellular factors may act on cells in two distinct modes: an acute increase in concentration as a result of regulated secretion, or a gradual increase in concentration when secreted constitutively or from a distant source. We found that cellular responses to brain-derived neurotrophic factor (BDNF) differed markedly depending on how BDNF was delivered. In cultured rat hippocampal neurons, acute and gradual increases in BDNF elicited transient and sustained activation of TrkB receptor and its downstream signaling, respectively, leading to differential expression of Homer1 and Arc. Transient TrkB activation promoted neurite elongation and spine head enlargement, whereas sustained TrkB activation facilitated neurite branch and spine neck elongation. In hippocampal slices, fast and slow increases in BDNF enhanced basal synaptic transmission and LTP, respectively. Thus, the kinetics of TrkB activation is critical for cell signaling and functions. This temporal dimension in cellular signaling may also have implications for the therapeutic drug design.
Collapse
|
13
|
Fu QL, Li X, Yip H, Shao Z, Wu W, Mi S, So KF. Combined effect of brain-derived neurotrophic factor and LINGO-1 fusion protein on long-term survival of retinal ganglion cells in chronic glaucoma. Neuroscience 2009; 162:375-82. [DOI: 10.1016/j.neuroscience.2009.04.075] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Revised: 04/22/2009] [Accepted: 04/26/2009] [Indexed: 12/09/2022]
|
14
|
Beck M, Karch C, Wiese S, Sendtner M. Motoneuron cell death and neurotrophic factors: Basic models for development of new therapeutic strategies in ALS. ACTA ACUST UNITED AC 2009. [DOI: 10.1080/146608201300079454] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
15
|
Davis MI. Ethanol-BDNF interactions: still more questions than answers. Pharmacol Ther 2008; 118:36-57. [PMID: 18394710 DOI: 10.1016/j.pharmthera.2008.01.003] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Accepted: 01/08/2008] [Indexed: 01/02/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) has emerged as a regulator of development, plasticity and, recently, addiction. Decreased neurotrophic activity may be involved in ethanol-induced neurodegeneration in the adult brain and in the etiology of alcohol-related neurodevelopmental disorders. This can occur through decreased expression of BDNF or through inability of the receptor to transduce signals in the presence of ethanol. In contrast, recent studies implicate region-specific up-regulation of BDNF and associated signaling pathways in anxiety, addiction and homeostasis after ethanol exposure. Anxiety and depression are precipitating factors for substance abuse and these disorders also involve region-specific changes in BDNF in both pathogenesis and response to pharmacotherapy. Polymorphisms in the genes coding for BDNF and its receptor TrkB are linked to affective, substance abuse and appetitive disorders and therefore may play a role in the development of alcoholism. This review summarizes historical and pre-clinical data on BDNF and TrkB as it relates to ethanol toxicity and addiction. Many unresolved questions about region-specific changes in BDNF expression and the precise role of BDNF in neuropsychiatric disorders and addiction remain to be elucidated. Resolution of these questions will require significant integration of the literature on addiction and comorbid psychiatric disorders that contribute to the development of alcoholism.
Collapse
Affiliation(s)
- Margaret I Davis
- Section on Synaptic Pharmacology, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
16
|
Rantamäki T, Hendolin P, Kankaanpää A, Mijatovic J, Piepponen P, Domenici E, Chao MV, Männistö PT, Castrén E. Pharmacologically diverse antidepressants rapidly activate brain-derived neurotrophic factor receptor TrkB and induce phospholipase-Cgamma signaling pathways in mouse brain. Neuropsychopharmacology 2007; 32:2152-62. [PMID: 17314919 DOI: 10.1038/sj.npp.1301345] [Citation(s) in RCA: 246] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Previous studies suggest that brain-derived neurotrophic factor and its receptor TrkB are critically involved in the therapeutic actions of antidepressant drugs. We have previously shown that the antidepressants imipramine and fluoxetine produce a rapid autophosphorylation of TrkB in the rodent brain. In the present study, we have further examined the biochemical and functional characteristics of antidepressant-induced TrkB activation in vivo. We show that all the antidepressants examined, including inhibitors of monoamine transporters and metabolism, activate TrkB rapidly in the rodent anterior cingulate cortex and hippocampus. Furthermore, the results indicate that acute and long-term antidepressant treatments induce TrkB-mediated activation of phospholipase-Cgamma1 (PLCgamma1) and increase the phosphorylation of cAMP-related element binding protein, a major transcription factor mediating neuronal plasticity. In contrast, we have not observed any modulation of the phosphorylation of TrkB Shc binding site, phosphorylation of mitogen-activated protein kinase or AKT by antidepressants. We also show that in the forced swim test, the behavioral effects of specific serotonergic antidepressant citalopram, but not those of the specific noradrenergic antidepressant reboxetine, are crucially dependent on TrkB signaling. Finally, brain monoamines seem to be critical mediators of antidepressant-induced TrkB activation, as antidepressants reboxetine and citalopram do not produce TrkB activation in the brains of serotonin- or norepinephrine-depleted mice. In conclusion, our data suggest that rapid activation of the TrkB neurotrophin receptor and PLCgamma1 signaling is a common mechanism for all antidepressant drugs.
Collapse
Affiliation(s)
- Tomi Rantamäki
- Neuroscience Center, University of Helsinki, PO box 56, 00014 Helsinki, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Neurotrophin stimulation of tropomyosin-related kinase (Trk) and p75 receptors influences cellular processes such as proliferation, growth, differentiation, and other cell-specific functions, as well as regeneration. In contrast to Trk receptors, which have a well-defined trophic role, p75 has activities ranging from trophism to apoptosis. Continued neurotrophin stimulation of differentiating neurons transforms the initially trophic character of p75 signaling into negative growth control and overstimulation leads to apoptosis. This function shift reflects the signaling effects of ceramide that is generated upon stimulation of p75. The use of ceramide signaling by p75 may provide a key to understanding the cell-biological role of p75. The review presents arguments that the control of cell shape formation and cell selection can serve as an organizing principle of p75 signaling. Concurrent stimulation by neurotrophins of p75 and Trk receptors constitutes a dual growth control with antagonistic and synergistic elements aimed at optimal morphological and functional integration of cells and cell populations into their context.
Collapse
Affiliation(s)
- A Blöchl
- Biochemie II, Fakultät Chemie und Biochemie, Ruhr-Universität Bochum, Bochum, Germany.
| | | |
Collapse
|
18
|
Terai K, Matsuda M. The amino-terminal B-Raf-specific region mediates calcium-dependent homo- and hetero-dimerization of Raf. EMBO J 2006; 25:3556-64. [PMID: 16858395 PMCID: PMC1538552 DOI: 10.1038/sj.emboj.7601241] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Accepted: 06/27/2006] [Indexed: 01/18/2023] Open
Abstract
B-Raf is a key regulatory molecule of the mitogen-activated protein kinase kinase (MEK). B-Raf differs from the other Raf isoforms in that it has a long amino-terminal region. By the use of probes based on the principle of fluorescence resonance energy transfer, we found that this amino-terminal B-Raf-specific region is essential for homo-dimerization of B-Raf and hetero-dimerization of B-Raf and c-Raf at the plasma membrane, followed by phosphorylation of Thr118 in the amino-terminal B-Raf-specific region. HeLa cells expressing B-Raf, but not c-Raf, or a B-Raf mutant lacking the B-Raf-specific region, showed enhanced MEK phosphorylation upon stimulation with a calcium agonist. Furthermore, increases in the intracellular calcium concentration were found to be necessary for dimerization and sufficient for the plasma membrane translocation of B-Raf. Notably, in calcium ionophore-stimulated HeLa cells, B-Raf could propagate signals to MEK under the basal level of GTP-Ras. Thus, we propose that the hitherto unidentified function of the B-Raf amino-terminal region is to mediate calcium-dependent activation of B-Raf and the following MEK activation, which may occur in the absence of Ras activation.
Collapse
Affiliation(s)
- Kenta Terai
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Michiyuki Matsuda
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Yoshida-Konoemachi, Sakyo-ku, Kyoto 606-8501, Japan. Tel.: +81 75 753 4421; Fax: +81 75 753 4698; E-mails: or
| |
Collapse
|
19
|
Franklin TB, Murphy JA, Myers TL, Clarke DB, Currie RW. Enriched environment during adolescence changes brain-derived neurotrophic factor and TrkB levels in the rat visual system but does not offer neuroprotection to retinal ganglion cells following axotomy. Brain Res 2006; 1095:1-11. [PMID: 16730677 DOI: 10.1016/j.brainres.2006.04.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2005] [Revised: 04/03/2006] [Accepted: 04/04/2006] [Indexed: 12/09/2022]
Abstract
The purpose of the present experiment was to characterize changes in TrkB signaling in the rat visual system resulting from exposure to enriched environment. Female Sprague-Dawley rats were placed in enriched or impoverished conditions for 1, 7 or 28 days. Levels of BDNF protein and its predominant receptor TrkB were examined in the retina, superior colliculus and visual cortex. In the retina, 1 day of enrichment increased full-length TrkB and after 28 days increased BDNF. In the superior colliculus, enrichment for 7 days reduced full-length TrkB and after 28 days increased BDNF and full-length TrkB. One day of enrichment significantly increased BDNF, reduced full-length TrkB and increased truncated TrkB in the visual cortex. Consequently, we further investigated whether exposure to enriched environment and the subsequent changes in BDNF and TrkB translates into a neuroprotective effect on retinal ganglion cells (RGCs) following transection of the optic nerve. Although exogenous intraocular application of BDNF provides neuroprotection to RGCs after axotomy, the endogenous increase in BDNF in the retina after 28 days of enrichment had no effect on RGC survival. While enriched housing conditions offer a model of non-invasive rehabilitation treatment for injury and modulates changes in BDNF and TrkB levels, these molecular changes did not translate into a neuroprotective effect on RGCs following transection of the optic nerve.
Collapse
Affiliation(s)
- Tamara B Franklin
- Laboratory of Molecular Neurobiology, Department of Anatomy and Neurobiology, Dalhousie University, Halifax, NS Canada B3H 1X5
| | | | | | | | | |
Collapse
|
20
|
Ohrtman JD, Stancik EK, Lovinger DM, Davis MI. Ethanol inhibits brain-derived neurotrophic factor stimulation of extracellular signal-regulated/mitogen-activated protein kinase in cerebellar granule cells. Alcohol 2006; 39:29-37. [PMID: 16938627 DOI: 10.1016/j.alcohol.2006.06.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2006] [Revised: 06/16/2006] [Accepted: 06/20/2006] [Indexed: 10/24/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) has emerged as a prominent mediator of neuronal development and synaptic plasticity. BDNF activates multiple signal transduction cascades that regulate cellular function through phosphorylation, transcription, and translation. Ethanol is known to inhibit neurotrophin signaling, but a thorough pharmacological analysis of the effect of ethanol on BDNF signaling in developing neurons has not been performed. These experiments were undertaken to determine the interactions between membrane depolarization, BDNF concentration, and ethanol concentration on extracellular signal-regulated protein kinase (ERK) activation in neurons. We examined cerebellar granule cells grown under physiological (5mM) or elevated (25mM) potassium culture conditions after 3 days in vitro. BDNF-stimulated ERK phosphorylation (pERK) within 10min and supported stimulation from 20 to 60min. Ethanol decreased basal pERK and reduced the magnitude of BDNF stimulation of ERK under both conditions. The NMDA receptor antagonist 2-amino-5-phosphonovalerate did not effect basal pERK or inhibit BDNF stimulation of ERK, suggesting that NMDA receptors do not modulate BDNF stimulation of ERK in short-term cultures. These data characterize the pharmacological effects of ethanol on growth factor signaling and provide the basis of a model for further characterization of the biochemical mechanisms of ERK inhibition by ethanol. Perturbation of BDNF signal transduction by ethanol may underlie some of the cognitive deficits and developmental abnormalities resulting from ethanol exposure.
Collapse
Affiliation(s)
- Joshua D Ohrtman
- Section on Synaptic Pharmacology, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fisher's Lane MSC 9411, Bethesda, MD 20892-9411, USA
| | | | | | | |
Collapse
|
21
|
Ferrer I, Ballabriga J, Martí E, Pérez E, Alberch J, Arenas E. BDNF up-regulates TrkB protein and prevents the death of CA1 neurons following transient forebrain ischemia. Brain Pathol 2006; 8:253-61. [PMID: 9546284 PMCID: PMC8098442 DOI: 10.1111/j.1750-3639.1998.tb00151.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The neurotrophin family of growth factors, which includes Nerve Growth Factor (NGF), Brain-Derived Neurotrophic Factor (BDNF), Neurotrophin-3 (NT3) and Neurotrophin-4/5 (NT4/5) bind and activate specific tyrosine kinase (Trk) receptors to promote cell survival and growth of different cell populations. For these reasons, growing attention has been paid to the use of neurotrophins as therapeutic agents in neurodegeneration, and to the regulation of the expression of their specific receptors by the ligands. BDNF expression, as revealed by immunohistochemistry, is found in the pre-subiculum, CA1, CA3, and dentate gyrus of the hippocampus. Strong TrkB immunoreactivity is present in most CA3 neurons but only in scattered neurons of the CA1 area. Weak TrkB immunoreactivity is found in the granule cell layer of the dentate gyrus. Unilateral grafting of BDNF-transfected fibroblasts into the hippocampus resulted in a marked increase in the intensity of the immunoreaction and in the number of TrkB-immunoreactive neurons in the granule cell layer of the dentate gyrus, pre-subiculum and CA1 area in the vicinity of the graft. No similar effects were produced after the injection of control mock-transfected fibroblasts. Delayed cell death in the CA1 area was produced following 5 min of forebrain ischemia in the gerbil. The majority of living cells in the CA1 area at the fourth day were BDNF/TrkB immunoreactive. Unilateral grafting of control mock-transfected or BDNF fibroblasts two days before ischemia resulted in a moderate non-specific protection of TrkB-negative, but not TrkB-positive cells, in the CA1 area of the grafted side. This finding is in line with a vascular and glial reaction, as revealed, by immunohistochemistry using astroglial and microglial cell markers. This astroglial response was higher in the grafted side than in the contralateral side in ischemic gerbils, but no differences were seen between BDNF-producing and non-BDNF-producing grafts. However, grafting of BDNF-producing fibroblasts two days before ischemia significantly and specifically prevented nerve cells from dying in the CA1 area of the ipsilateral hippocampus. Cell survival was associated with increased TrkB immunoreactivity as the majority of living cells were TrkB immunoreactive. Thus, our results show that BDNF is able to up-regulate the expression of TrkB in control and pathological states, and that BDNF prevention of neuronal death following transient forebrain ischemia is associated with increased expression of its specific receptor.
Collapse
Affiliation(s)
- I Ferrer
- Unitat de Neuropatologia, Servei d'Anatomia Patolïgica, Hospital Princeps d'Espanya, Spain
| | | | | | | | | | | |
Collapse
|
22
|
Kemi C, Grunewald J, Eklund A, Olgart Höglund C. Differential regulation of neurotrophin expression in human bronchial smooth muscle cells. Respir Res 2006; 7:18. [PMID: 16441896 PMCID: PMC1386667 DOI: 10.1186/1465-9921-7-18] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2005] [Accepted: 01/29/2006] [Indexed: 11/18/2022] Open
Abstract
Background Human bronchial smooth muscle cells (HBSMC) may regulate airway inflammation by secreting cytokines, chemokines and growth factors. The neurotrophins, including nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3), have been shown to be elevated during airway inflammation and evoke airway hyperresponsiveness. We studied if HBSMC may be a source of NGF, BDNF and NT-3, and if so, how inflammatory cytokines may influence their production. Methods Basal and cytokine (IL-1β, IFN-γ, IL-4)-stimulated neurotrophin expression in HBSMC cultured in vitro was quantified. The mRNA expression was quantified by real-time RT-PCR and the protein secretion into the cell culture medium by ELISA. Results We observed a constitutive NGF, BDNF and NT-3 expression. IL-1β stimulated a transient increase of NGF, while the increase of BDNF had a later onset and was more sustained. COX-inhibitors (indomethacin and NS-398) markedly decreased IL-1β-stimulated secretion of BDNF, but not IL-1β-stimulated NGF secretion. IFN-γ increased NGF expression, down-regulated BDNF expression and synergistically enhanced IL-1β-stimulated NGF expression. In contrast, IL-4 had no effect on basal NGF and BDNF expression, but decreased IL-1β-stimulated NGF expression. NT-3 was not altered by the tested cytokines. Conclusion Taken together, our data indicate that, in addition to the contractile capacity, HBSMC can express NGF, BDNF and NT-3. The expression of these neurotrophins may be differently regulated by inflammatory cytokines, suggesting a dynamic interplay that might have a potential role in airway inflammation.
Collapse
Affiliation(s)
- Cecilia Kemi
- Department of Medicine, Division of Respiratory Medicine, Lung Research Laboratory, Karolinska Institutet and Karolinska University Hospital Solna, 171 76 Stockholm, Sweden
| | - Johan Grunewald
- Department of Medicine, Division of Respiratory Medicine, Lung Research Laboratory, Karolinska Institutet and Karolinska University Hospital Solna, 171 76 Stockholm, Sweden
| | - Anders Eklund
- Department of Medicine, Division of Respiratory Medicine, Lung Research Laboratory, Karolinska Institutet and Karolinska University Hospital Solna, 171 76 Stockholm, Sweden
| | - Caroline Olgart Höglund
- Department of Medicine, Division of Respiratory Medicine, Lung Research Laboratory, Karolinska Institutet and Karolinska University Hospital Solna, 171 76 Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
23
|
Chen ZY, Ieraci A, Tanowitz M, Lee FS. A novel endocytic recycling signal distinguishes biological responses of Trk neurotrophin receptors. Mol Biol Cell 2005; 16:5761-72. [PMID: 16207814 PMCID: PMC1289419 DOI: 10.1091/mbc.e05-07-0651] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Endocytic trafficking of signaling receptors to alternate intracellular pathways has been shown to lead to diverse biological consequences. In this study, we report that two neurotrophin receptors (tropomyosin-related kinase TrkA and TrkB) traverse divergent endocytic pathways after binding to their respective ligands (nerve growth factor and brain-derived neurotrophic factor). We provide evidence that TrkA receptors in neurosecretory cells and neurons predominantly recycle back to the cell surface in a ligand-dependent manner. We have identified a specific sequence in the TrkA juxtamembrane region, which is distinct from that in TrkB receptors, and is both necessary and sufficient for rapid recycling of internalized receptors. Conversely, TrkB receptors are predominantly sorted to the degradative pathway. Transplantation of the TrkA recycling sequence into TrkB receptors reroutes the TrkB receptor to the recycling pathway. Finally, we link these divergent trafficking pathways to alternate biological responses. On prolonged neurotrophin treatment, TrkA receptors produce prolonged activation of phosphatidylinositol 3-kinase/Akt signaling as well as survival responses, compared with TrkB receptors. These results indicate that TrkA receptors, which predominantly recycle in signal-dependent manner, have unique biological properties dictated by its specific endocytic trafficking itinerary.
Collapse
Affiliation(s)
- Zhe-Yu Chen
- Department of Psychiatry, Weill Medical College of Cornell University, New York, NY 10021, USA.
| | | | | | | |
Collapse
|
24
|
Blöchl A, Blumenstein L, Ahmadian MR. Inactivation and activation of Ras by the neurotrophin receptor p75. Eur J Neurosci 2005; 20:2321-35. [PMID: 15525274 DOI: 10.1111/j.1460-9568.2004.03692.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The neurotrophin receptor p75 induces neurotrophic and/or apoptotic signalling pathways and can also cooperate with the neurotrophic Trk receptor tyrosine kinases. Its intracellular part encloses a so-called 'death domain' with a segment similar to the wasp venom mastoparan which binds small GTPases such as Rho. To study possible interactions of p75 and Ras (and Rho) we used wild-type and mutant genes of p75 stably expressed by MDCK cells which normally have neither Trk nor p75. We found that p75 can directly bind the GTPases Ras and Rho and that the unstimulated p75 inactivates total cellular Ras through a differential influence on the dissociation of GDP and GTP from Ras and an exchange of bound Ras.GDP for free Ras.GTP. These properties of p75 could also be demonstrated in vitro and should therefore be cell type-independent. Stimulation of p75 with nerve growth factor causes Ras activation via adapter proteins known from Trk signalling and induces rapid outgrowth of cellular processes. Both inactivation and activation of Ras by p75 are controlled by the phosphorylation state of the receptor's two intracellular tyrosines. p75 also influences Rho activation and inactivation, and the combined interactions of the receptor with the two GTPases Ras and Rho can regulate neurite formation in an efficient, synergistic way.
Collapse
Affiliation(s)
- Andrea Blöchl
- Molekulare Neurobiochemie, Fakultät Chemie, Molekulare Neurobiochemie, NC7/132, Ruhr-Universität Bochum, 44780 Bochum, Germany.
| | | | | |
Collapse
|
25
|
Barnea A, Roberts J, Croll SD. Continuous exposure to brain-derived neurotrophic factor is required for persistent activation of TrkB receptor, the ERK signaling pathway, and the induction of neuropeptide Y production in cortical cultures. Brain Res 2004; 1020:106-17. [PMID: 15312792 DOI: 10.1016/j.brainres.2004.06.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2004] [Indexed: 11/28/2022]
Abstract
We have previously demonstrated that brain-derived neurotrophic factor (BDNF) induces persistent neuropeptide Y (NPY) production in cortical cultures in an ERK1/2-dependent manner. In some studies, it was shown that BDNF leads to the downregulation of TrkB receptor and some of its downstream responses, whereas in others it does not. We examined whether the BDNF requirement for induction of persistent NPY production correlates with that for induction of phosphorylation of TrkB and ERK1/2. Continuous 24-h exposure to BDNF led to a 2- to 3-fold increase in NPY production (maximal level). While 1 h of BDNF exposure induced NPY production at a half maximal level, 8 h was required for induction of a maximal level. BDNF-induced NPY production was completely inhibited by co-exposure to TrkB-Fc fusion protein (TrkB extracellular domain fused to Fc) and partially inhibited by TrkB-Fc added 1 h after BDNF; TrkC-Fc did not do so. Activation of TrkB receptor was analyzed at two potential tyrosine phosphorylated sites, the activation loop and the Shc binding. BDNF led to coordinated phosphorylation of the two sites that persisted for 6-8 h, and this was not associated with changes in the content of TrkB protein. The presence of BDNF throughout the 6- to 8-h period was required for the persistent phosphorylation of TrkB and ERK1/2. Thus, continuous BDNF activation of TrkB is required for persistent activation of the ERK1/2 pathway and induction of NPY production. We propose that, within the time frame analyzed in this study, BDNF does not lead to the downregulation of TrkB receptor or of the biological responses leading to NPY production.
Collapse
Affiliation(s)
- Ayalla Barnea
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9032, USA.
| | | | | |
Collapse
|
26
|
Bonnet D, Garcia M, Vecino E, Lorentz JG, Sahel J, Hicks D. Brain-derived neurotrophic factor signalling in adult pig retinal ganglion cell neurite regeneration in vitro. Brain Res 2004; 1007:142-51. [PMID: 15064145 DOI: 10.1016/j.brainres.2004.02.023] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2004] [Indexed: 11/26/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) has been implicated in stimulating retinal ganglion cell (RGC) survival and axonal regeneration in rodent animal models in vivo and in vitro, but very little data are available on neurotrophin effects in higher mammals. We hence analysed BDNF signalling in primary cultures of adult pig RGC. As detected by immunohistochemistry, HPLC analysis and RT-PCR, BDNF protein and mRNA were present within pig retina in vivo and in vitro, where it may be involved in baseline RGC neuritogenesis. Initial dose-response studies established optimal effects were induced by 20 ng/ml BDNF, leading to an approximately threefold increase in neurite length. We analysed the respective contributions of phosphatidyl inositol 3 kinase (PI3K) and mitogen activated protein kinase (MAPK) cascades to BDNF-induced neurite regeneration. Addition of either the PI3K inhibitor wortmannin or the MAPK inhibitor U0126 blocked 50-100% BDNF-induced neurite elongation; U0126 also significantly reduced neurite regeneration below untreated control levels. The trk receptor inhibitor K252a had no observable effect on neurite regeneration or morphology. These data hence demonstrate that BDNF is a potent stimulator of neurite growth in RGC prepared from an adult large mammal retina, and that at least two signalling pathways are causally involved. BDNF-based therapy may be of potential use in treating RGC degeneration in humans.
Collapse
Affiliation(s)
- Delphine Bonnet
- Laboratoire de Physiopathologie Cellulaire et Moléculaire de la Rétine, INSERM U. 592, Clinique Médicale A, Centre Hospitalier Universitaire, BP 426, 67091 Strasbourg, France
| | | | | | | | | | | |
Collapse
|
27
|
Liu G, Nozell S, Xiao H, Chen X. DeltaNp73beta is active in transactivation and growth suppression. Mol Cell Biol 2004; 24:487-501. [PMID: 14701724 PMCID: PMC343790 DOI: 10.1128/mcb.24.2.487-501.2004] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
p73, a p53 family protein, shares significant sequence homolog and functional similarity with p53. However, unlike p53, p73 has at least seven alternatively spliced isoforms with different carboxyl termini (p73alpha-eta). Moreover, the p73 gene can be transcribed from a cryptic promoter located in intron 3, producing seven more proteins (DeltaNp73alpha-eta). DeltaNp73, which does not contain the N-terminal activation domain in p73, has been thought to be transcriptionally inactive and dominant negative over p53 or p73. To systemically analyze the activity of the DeltaN variant, we generated stable cell lines, which inducibly express DeltaNp73alpha, DeltaNp73beta, and various DeltaNp73beta mutants by using the tetracycline-inducible expression system. Surprisingly, we found that DeltaNp73beta is indeed active in inducing cell cycle arrest and apoptosis. Importantly, we found that, when DeltaNp73beta is expressed at a physiologically relevant level, it is capable of suppressing cell growth. We then demonstrated that these DeltaNp73beta activities are not cell type specific. We showed that the 13 unique residues at the N terminus are required for DeltaNp73beta to suppress cell growth. We also found that, among the 13 residues, residues 6 to 10 are critical to DeltaNp73beta function. Furthermore, we found that DeltaNp73beta is capable of inducing some p53 target genes, albeit to a lesser extent than does p73beta. Finally, we found that the 13 unique residues, together with the N-terminal PXXP motifs, constitute a novel activation domain. Like DeltaNp73beta, DeltaNp73gamma is active in transactivation. However, unlike DeltaNp73beta, DeltaNp73alpha is inactive in suppressing cell growth. Our data, together with others' previous findings, suggest that DeltaNp73beta may have distinct functions under certain cellular circumstances.
Collapse
Affiliation(s)
- Gang Liu
- Department of Cell Biology, The University of Alabama at Birmingham, 1530 3rd Avenue, Birmingham, AL 35294-0005, USA
| | | | | | | |
Collapse
|
28
|
Wiese S, Beck M, Karch C, Sendtner M. Signalling mechanisms for survival of lesioned motoneurons. ACTA NEUROCHIRURGICA. SUPPLEMENT 2004; 89:21-35. [PMID: 15335098 DOI: 10.1007/978-3-7091-0603-7_4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Mechanisms controlling neuronal survival play an important role both during development and after birth, in particular when the nervous system is lesioned. Isolated embryonic motoneurons and other types of primary neurons have been a useful tool for studying basic mechanisms underlying neuronal cell death during development and under pathophysiological conditions after neurotrauma. These studies have led to the identification of neurotrophic factors which under physiological conditions regulate survival and functional properties, and after neurotrauma promote regeneration and plasticity. Functional analysis of these molecules, in particular by generation of gene knockout mice, has led to a more detailed understanding of complex requirements of individual types of neurons for their survival and also paved the way for a better understanding of the signalling pathways in lesioned neurons which decide on cell death or survival after axotomy and other pathophysiological conditions. These findings could ultimately lead to a rational basis for therapeutic approaches aiming at improving neuronal survival and regeneration after neurotrauma.
Collapse
Affiliation(s)
- S Wiese
- Institute for Clinical Neurobiology, Würzburg, Germany
| | | | | | | |
Collapse
|
29
|
Abstract
Nerve growth factor was the first identified protein with anti-apoptotic activity on neurons. This prototypic neurotrophic factor, together with the three structurally and functionally related growth factors brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT3) and neurotrophin-4/5 (NT4/5), forms the neurotrophin protein family. Target T cells for neurotrophins include many neurons affected by neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and peripheral polyneuropathies. In addition, the neurotrophins act on neurons affected by other neurological and psychiatric pathologies including ischemia, epilepsy, depression and eating disorders. Work with cell cultures and animal models provided solid support for the hypothesis that neurotrophins prevent neuronal death. While no evidence exists that a lack of neurotrophins underlies the etiology of any neurodegenerative disease, these studies have spurred on hopes that neurotrophins might be useful symptomatic-therapeutic agents. However first clinical trials led to variable results and severe side effects were observed. For future therapeutic use of the neurotrophins it is therefore crucial to expand our knowledge about their physiological functions as well as their pharmacokinetic properties. A major challenge is to develop methods for their application in effective doses and in a precisely timed and localized fashion.
Collapse
Affiliation(s)
- Georg Dechant
- Neurobiochemistry, Max-Planck-Institute of Neurobiology, Am Klopferspitz 18a, 82152 Martinsried, Germany
| | | |
Collapse
|
30
|
Xu B, Michalski B, Racine RJ, Fahnestock M. Continuous infusion of neurotrophin-3 triggers sprouting, decreases the levels of TrkA and TrkC, and inhibits epileptogenesis and activity-dependent axonal growth in adult rats. Neuroscience 2003; 115:1295-308. [PMID: 12453498 DOI: 10.1016/s0306-4522(02)00384-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Neurotrophin-3 (NT-3), a member of the neurotrophin family of neurotrophic factors, is important for cell survival, axonal growth and neuronal plasticity. Epileptiform activation can regulate the expression of neurotrophins, and increases or decreases in neurotrophins can affect both epileptogenesis and seizure-related axonal growth. Interestingly, the expression of nerve growth factor and brain-derived neurotrophic factor is rapidly up-regulated following seizures, while NT-3 mRNA remains unchanged or undergoes a delayed down-regulation, suggesting that NT-3 might have a different function in epileptogenesis. In the present study, we demonstrate that continuous intraventricular infusion of NT-3 in the absence of kindling triggers mossy fiber sprouting in the inner molecular layer of the dentate gyrus and the stratum oriens of the CA3 region. Furthermore, despite this NT-3-related sprouting effect, continuous infusion of NT-3 retards the development of behavioral seizures and inhibits kindling-induced mossy fiber sprouting in the inner molecular layer of the dentate gyrus. We also show that prolonged infusion of NT-3 leads to a decrease in kindling-induced Trk phosphorylation and a down-regulation of the high-affinity Trk receptors, TrkA and TrkC, suggesting an involvement of both cholinergic nerve growth factor receptors and hippocampal NT-3 receptors in these effects. Our results demonstrate an important inhibitory role for NT-3 in seizure development and seizure-related synaptic reorganization.
Collapse
MESH Headings
- Animals
- Cell Count
- Cytochrome c Group/pharmacology
- Drug Administration Schedule
- Epilepsy/drug therapy
- Epilepsy/metabolism
- Epilepsy/physiopathology
- Growth Cones/drug effects
- Growth Cones/metabolism
- Kindling, Neurologic/drug effects
- Kindling, Neurologic/metabolism
- Male
- Molecular Weight
- Mossy Fibers, Hippocampal/drug effects
- Mossy Fibers, Hippocampal/growth & development
- Mossy Fibers, Hippocampal/metabolism
- Neuronal Plasticity/drug effects
- Neuronal Plasticity/physiology
- Neuropil/cytology
- Neuropil/drug effects
- Neurotrophin 3/metabolism
- Neurotrophin 3/pharmacology
- Phosphorylation/drug effects
- Rats
- Rats, Long-Evans
- Receptor Protein-Tyrosine Kinases/drug effects
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptor, trkA/drug effects
- Receptor, trkA/metabolism
- Receptor, trkB/drug effects
- Receptor, trkB/metabolism
- Receptor, trkC/drug effects
- Receptor, trkC/metabolism
- Seizures/drug therapy
- Seizures/metabolism
- Seizures/physiopathology
Collapse
Affiliation(s)
- B Xu
- Department of Psychology, McMaster University, L8S 4K1, Hamilton, ON, Canada
| | | | | | | |
Collapse
|
31
|
Haapasalo A, Sipola I, Larsson K, Akerman KEO, Stoilov P, Stamm S, Wong G, Castren E. Regulation of TRKB surface expression by brain-derived neurotrophic factor and truncated TRKB isoforms. J Biol Chem 2002; 277:43160-7. [PMID: 12202482 DOI: 10.1074/jbc.m205202200] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) signaling through its receptor TRKB modulates survival, differentiation, and activity of neurons. BDNF activates TRKB on the cell surface, which leads to the initiation of intracellular signaling cascades and different biological responses in neurons. Neuronal activity has been shown to regulate TRKB levels on the plasma membrane of neurons, but little is known about other factors affecting TRKB surface expression levels. We report here that BDNF regulates the cell surface levels of transfected or endogenously expressed full-length TRKB, depending on the exposure time in neuroblastoma cells and primary hippocampal neurons. BDNF rapidly increases TRKB surface expression levels in seconds, whereas treatment of cells with BDNF for a longer time (minutes to hours) leads to decreased TRKB surface levels. Coexpression of the full-length TRKB together with the truncated TRKB.T1 isoform results in decreased levels of full-length TRKB on the cell surface. This effect is specific to the T1 isoform, because coexpression of a kinase-dead TRKB mutant or another kinase domain-lacking TRKB form, truncated T-Shc, leads to increased TRKB surface levels. Our results suggest that regulation of TRKB surface expression levels by different factors is tightly controlled by complex mechanisms in active neurons.
Collapse
Affiliation(s)
- Annakaisa Haapasalo
- Department of Neurobiology, University of Kuopio, P. O. Box 1627, 70211 Kuopio, Finland
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Viegi A, Cotrufo T, Berardi N, Mascia L, Maffei L. Effects of dark rearing on phosphorylation of neurotrophin Trk receptors. Eur J Neurosci 2002; 16:1925-30. [PMID: 12453056 DOI: 10.1046/j.1460-9568.2002.02270.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Total lack of visual experience (dark rearing, DR) is known to affect development of mammalian visual cortex (VC) and to prolong the critical period of visual cortical plasticity. Neurotrophins (NTs) have been proposed to play a relevant role in activity dependent processes important for the final shaping of cortical visual connections. Neurotrophin supply or antagonism of endogenous NT action profoundly affect visual cortical development and plasticity; in particular, exogenous supply of NTs counteracts DR effects on VC development. However, the effects of DR on NT expression are still debated and mounting evidence reports a mismatch between BDNF mRNA and protein expression in DR animals. To gain insight into the effects of DR on expression of nerve growth factor (NGF) and the functional state of NT signalling pathways, we assessed the phosphorylation state of Trk receptors in light-reared animals (LR), in dark-reared animals (DR), in DR animals briefly exposed to light and in DR animals with exogenous supply of NTs [NGF, brain-derived neurotrophic factor (BDNF) and NT-4] in the VC. We report that DR increases the expression of NGF but reduces the phosphorylation of TrkA and TrkB receptors with respect to LR; normal phosphorylation is rapidly rescued by a brief exposure to light. Exogenous supply of NGF, BDNF or NT4 in DR animals also rescues the phosphorylation of their receptors.
Collapse
Affiliation(s)
- Alessandro Viegi
- Scuola Normale Superiore, p.zza Dei Cavalieri, 7, 56126, Pisa, Italy.
| | | | | | | | | |
Collapse
|
33
|
Mattingly RR, Felczak A, Chen CC, McCabe MJ, Rosenspire AJ. Low concentrations of inorganic mercury inhibit Ras activation during T cell receptor-mediated signal transduction. Toxicol Appl Pharmacol 2001; 176:162-8. [PMID: 11714248 DOI: 10.1006/taap.2001.9272] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mercury is widespread in the environment and consequently there are large populations that are currently exposed to low levels of mercury as a result of ubiquitous environmental factors. Whether these environmental levels of mercury are harmful is a matter of current debate, with epidemiological and animal studies suggesting detrimental effects on the immune and nervous systems. However, specific cellular effects of low concentrations of mercury have been hard to characterize. We now demonstrate that subtoxic concentrations of HgCl(2) can potently (maximal at 1 microM) increase Ras.GTP levels in Jurkat, a human T cell line. Remarkably, this activation of Ras occurs without a concomitant increase in MAP kinase activation, suggesting that mercury may direct Ras into a nonproductive state. In addition to its direct effect on Ras, concentrations of HgCl(2) as low as 0.6 microM inhibited the ability of the T cell receptor to activate Ras and MAP kinase. The inhibitory effect of mercury is selective, as activation of MAP kinase by phorbol diesters remain intact. Since the Ras/MAP kinase pathway is both highly conserved and central to signal transduction processes mediated by a myriad of diverse membrane receptor systems in a variety of cell types, these results suggest a mechanism for adverse health effects resulting from exposure to low levels of mercury. They also support a model for regulation of the Ras/MAP kinase pathway, whereby partial but unproductive activation of Ras can diminish signaling from cell surface receptors.
Collapse
Affiliation(s)
- R R Mattingly
- Department of Pharmacology, Wayne State University, Detroit, Michigan 48201, USA
| | | | | | | | | |
Collapse
|
34
|
Olson JM, Asakura A, Snider L, Hawkes R, Strand A, Stoeck J, Hallahan A, Pritchard J, Tapscott SJ. NeuroD2 is necessary for development and survival of central nervous system neurons. Dev Biol 2001; 234:174-87. [PMID: 11356028 DOI: 10.1006/dbio.2001.0245] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
NeuroD2 is sufficient to induce cell cycle arrest and neurogenic differentiation in nonneuronal cells. To determine whether this bHLH transcription factor was necessary for normal brain development, we used homologous recombination to replace the neuroD2 coding region with a beta-galactosidase reporter gene. The neuroD2 gene expressed the reporter in a subset of neurons in the central nervous system, including in neurons of the neocortex and hippocampus and cerebellum. NeuroD2(-/-) mice showed normal development until about day P14, when they began exhibiting ataxia and failure to thrive. Brain areas that expressed neuroD2 were smaller than normal and showed higher rates of apoptosis. Cerebella of neuroD2-null mice expressed reduced levels of genes encoding proteins that support cerebellar granule cell survival, including brain-derived neurotrophic factor (BDNF). Decreased levels of BDNF and higher rates of apoptosis in cerebellar granule cells of neuroD2(-/-) mice indicate that neuroD2 is necessary for the survival of specific populations of central nervous system neurons in addition to its known effects on cell cycle regulation and neuronal differentiation.
Collapse
Affiliation(s)
- J M Olson
- Clinical Research and Human Biology Divisions and Program in Developmental Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Blaha GR, Raghupathi R, Saatman KE, McIntosh TK. Brain-derived neurotrophic factor administration after traumatic brain injury in the rat does not protect against behavioral or histological deficits. Neuroscience 2001; 99:483-93. [PMID: 11029540 DOI: 10.1016/s0306-4522(00)00214-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Brain-derived neurotrophic factor has been shown to be neuroprotective in models of excitotoxicity, axotomy and cerebral ischemia. The present study evaluated the therapeutic potential of brain-derived neurotrophic factor following traumatic brain injury in the rat. Male Sprague-Dawley rats (N=99) were anesthetized and subjected to lateral fluid percussion brain injury of moderate severity (2.4-2.8 atm) or sham injury. Four hours after injury, the animals were reanesthetized, an indwelling, intraparenchymal cannula was implanted, and infusion of brain-derived neurotrophic factor or phosphate-buffered saline vehicle was initiated from a mini-osmotic pump and continued for two weeks. In Study 1 (N=48), vehicle or 12 microg/day of brain-derived neurotrophic factor was infused into the dorsal hippocampus. In Study 2 (N=51), vehicle or brain-derived neurotrophic factor at a high (12 microg/day) or low dose (1.2 microg/day) was infused into the injured parietal cortex. All animals were evaluated for neurological motor function at two days, one week and two weeks post-injury. Cognitive function (learning and memory) was assessed at two weeks post-injury using a Morris Water Maze. At two weeks post-injury, neuronal loss in the hippocampal CA3 and dentate hilus and in the injured cortex was evaluated. In Study 2, neuronal loss was also quantified in the thalamic medial geniculate nucleus. All of the above outcome measures demonstrated significant deleterious effects of brain injury (P<0.05 compared to sham). However, post-traumatic brain-derived neurotrophic factor infusion did not significantly affect neuromotor function, learning, memory or neuronal loss in the hippocampus, cortex or thalamus when compared to vehicle infusion in brain-injured animals, regardless of the infusion site or infusion dose (P>0.05 for each). In contrast to previous studies of axotomy, ischemia and excitotoxicity, our data indicate that brain-derived neurotrophic factor is not protective against behavioral or histological deficits caused by experimental traumatic brain injury using the delayed, post-traumatic infusion protocol examined in these studies.
Collapse
Affiliation(s)
- G R Blaha
- Department of Neurosurgery, University of Pennsylvania, Hayden Hall, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
36
|
Jezierski MK, Sohrabji F. Region- and peptide-specific regulation of the neurotrophins by estrogen. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2000; 85:77-84. [PMID: 11146109 DOI: 10.1016/s0169-328x(00)00244-8] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
We have previously shown that estrogen increases the expression of brain-derived neurotrophic factor (BDNF) mRNA in the olfactory bulb and cingulate cortex. Here we report that estrogen regulation of BDNF protein and the structurally related peptides nerve growth factor (NGF) and neurotrophin (NT)-4 is region- and peptide-specific. The olfactory bulb and cingulate cortex are both estrogen-sensitive targets and each receives a separate projection from neurons in the horizontal limb of the diagonal band of Broca (hlDBB). Furthermore, neurotrophins are retrogradely transported from the bulbar and cortical targets to the hlDBB. Four weeks of estrogen replacement to ovariectomized animals increased BDNF expression in the olfactory bulb, but decreased BDNF in the cingulate cortex. On the other hand, estrogen increased NT-4 expression in the cingulate cortex, but not in the olfactory bulb. NGF expression was not affected by estrogen in either region studied. In the hlDBB, estrogen increased BDNF but decreased NT-4, suggesting that estrogen differentially affects retrograde accumulation of these peptides. While both estrogen receptor alpha and beta have been identified in the olfactory bulb and cingulate cortex, our results indicate that estrogen receptor alpha expression is relatively higher in the olfactory bulb as compared to the cortex. Since the two estrogen receptors have been shown to stimulate different signaling pathways, we hypothesize that estrogen acting through specific receptors may differentially influence the extent and direction of neurotrophin expression.
Collapse
Affiliation(s)
- M K Jezierski
- Department of Human Anatomy and Medical Neurobiology, The Texas A&M University System Health Science Center, 228 Reynolds Medical Building, College Station, Texas, TX 77843-1114, USA
| | | |
Collapse
|
37
|
Pérez-Navarro E, Canudas AM, Akerund P, Alberch J, Arenas E. Brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-4/5 prevent the death of striatal projection neurons in a rodent model of Huntington's disease. J Neurochem 2000; 75:2190-9. [PMID: 11183872 DOI: 10.1046/j.1471-4159.2000.0752190.x] [Citation(s) in RCA: 143] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Intrastriatal injection of quinolinate has been proven to be a very useful animal model to study the pathogenesis and treatment of Huntington's disease. To determine whether growth factors of the neurotrophin family are able to prevent the degeneration of striatal projection neurons, cell lines expressing brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), or neurotrophin-4/5 (NT-4/5) were grafted in the adult rat striatum before quinolinate injection. Three days after lesioning, ongoing cell death was assessed by in situ detection of DNA fragmentation. In animals grafted with the control cell line, quinolinate injection induced a gradual cell loss that was differentially prevented by intrastriatal grafting of BDNF-, NT-3-, or NT-415-secreting cells. Seven days after lesioning, we characterized striatal projection neurons that were protected by neurotrophins. Quinolinate injection, alone or in combination with the control cell line, induced a selective loss of striatal projection neurons. Grafting of a BDNF-secreting cell line pre-vented the loss of all types of striatal projection neurons analyzed. Glutamic acid decarboxylase 67-, preproenkephalin-, and preprotachykinin A- but not prodynorphin-expressing neurons were protected by grafting of NT-3- or NT-4/5-secreting cells but with less efficiency than the BDNF-secreting cells. Our findings show that neurotrophins are able to promote the survival of striatal projection neurons in vivo and suggest that BDNF might be beneficial for the treatment of striatonigral degenerative disorders, including Huntington's disease.
Collapse
Affiliation(s)
- E Pérez-Navarro
- Departament de Biologia Cel.lular i Anatomia Patològica, Facultat de Medicina, IDIBAPS, Universitat de Barcelona, Spain
| | | | | | | | | |
Collapse
|
38
|
Pesheva P, Kuklinski S, Biersack HJ, Probstmeier R. Nerve growth factor-mediated expression of galectin-3 in mouse dorsal root ganglion neurons. Neurosci Lett 2000; 293:37-40. [PMID: 11065132 DOI: 10.1016/s0304-3940(00)01499-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Galectin-3, a member of the galectin family of beta-galactoside-specific lectins has been found to be expressed by subsets of dorsal root ganglion (DRG) neurons during development and in adulthood. Here we show that (i) after 3-7 days in vitro, DRG neurons derived from neonatal mice express galectin-3 intra- and extracellularly and (ii) lectin expression requires the presence of nerve growth factor (NGF). After 3 days in vitro, a higher number of DRG neurons expressed galectin-3 in the presence of NGF (65 +/- 7%) than in the presence of brain-derived neurotrophic factor (BDNF, 30 +/- 3%) or neurotrophin-3 (NT-3, 34 +/- 3%). After 7 days in vitro, these numbers dropped to 51 +/- 3% (for NGF), 0% (for BDNF) and 8 +/- 4% (for NT-3), respectively. Our findings provide first evidence for the contribution of a neurotrophin to the neuronal expression of galectins and suggest an NGF/TrkA-mediated expression of galectin-3 by early postnatal DRG neurons.
Collapse
Affiliation(s)
- P Pesheva
- Department of Nuclear Medicine, University of Bonn, Germany
| | | | | | | |
Collapse
|
39
|
Abstract
A variety of neurotrophic factors can influence the cell functions of the developing, mature and injured retinal ganglion cells. The discovery that retinal ganglion cell loss can be alleviated by neurotrophic factors has generated a great deal of interest in the therapeutic potential of these molecules. Recently, evidence has provided valuable information on the receptors that mediate these events and the intracellular signaling cascades after the binding of these ligands. Signaling by neurotrophic factors does not seem to restrict to retrograde messenger from the target but also includes local interactions with neighbouring cells along the axonal pathways, anterograde signaling from the afferents and autocrine signaling. More insight into the mechanisms of action of neurotrophic factors and the signal transduction pathway leading to the protection and regeneration of retinal ganglion cells may allow the design of new therapeutic strategies.
Collapse
Affiliation(s)
- H K Yip
- Department of Anatomy, Faculty of Medicine, The University of Hong Kong, Hong Kong.
| | | |
Collapse
|
40
|
Galuske RA, Kim DS, Castrén E, Singer W. Differential effects of neurotrophins on ocular dominance plasticity in developing and adult cat visual cortex. Eur J Neurosci 2000; 12:3315-30. [PMID: 10998115 DOI: 10.1046/j.1460-9568.2000.00213.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the present study we examine the influence of neurotrophins on experience-dependent synaptic rearrangement in developing and adult visual cortex. Brain-derived neurotrophic factor (BDNF) or nerve growth factor (NGF) was continuously infused into cortical area 18, and the functional architecture of the cortex was examined by use of optical and electrophysiological recording techniques. In kittens, BDNF infusion during monocular deprivation (MD) reversed the normally occurring ocular dominance (OD) shift towards the non-deprived eye so that the deprived eye dominated the BDNF-treated cortex after MD. Under conditions of equal activation of thalamocortical synapses, i.e. when animals were either subject to binocular deprivation (BD) or reared without deprivation, BDNF infusion did not disrupt binocularity of cortical units, but reversed the natural OD bias towards the contralateral eye in favour of the ipsilateral eye. In addition, BDNF treatment in kittens led to a loss of the orientation selectivity of cortical units irrespective of rearing conditions. In adult animals, BDNF influenced neither OD distributions nor orientation selectivity. The effect of NGF was markedly different. It was ineffective in kittens but in adult animals it caused a shift of OD towards the deprived eye when MD was combined with NGF infusion. However, in this case orientation selectivity was preserved. Thus, both neurotrophins have profound activity- and age-dependent effects on the functional architecture of the visual cortex. Moreover, our results indicate that simple substitution of neurotrophins in excess is unlikely to compensate for deprivation effects by preserving or restoring the normal functional architecture of the cortex.
Collapse
Affiliation(s)
- R A Galuske
- Max-Planck Institute for Brain Research, Frankfurt, Germany
| | | | | | | |
Collapse
|
41
|
Vogel KS, El-Afandi M, Parada LF. Neurofibromin negatively regulates neurotrophin signaling through p21ras in embryonic sensory neurons. Mol Cell Neurosci 2000; 15:398-407. [PMID: 10845775 DOI: 10.1006/mcne.2000.0836] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Embryonic sensory and sympathetic neurons that lack neurofibromin, the protein product of the neurofibromatosis type 1 (Nfl) gene, survive and extend neurites in the absence of neurotrophins. To determine whether neurofibromin negatively regulates neurotrophin signaling through its interaction with p21ras, we used Fab antibody fragments to block Ras function in DRG, trigeminal, nodose, and SCG neurons isolated from Nfl(-/-) and wild-type mouse embryos. We show that introduction of anti-Ras Fab fragments significantly reduces the ability of neurofibromin-deficient neurons to survive in the absence of neurotrophins. Moreover, addition of H-ras protein enhances the survival of Nfl(-/-), but not wild-type, DRG neurons. Our results are consistent with a major role for neurofibromin in modulating Trk signaling through p21ras during neuronal development.
Collapse
Affiliation(s)
- K S Vogel
- Center for Developmental Biology, University of Texas Southwestern Medical Center, Dallas 75235-9133, USA.
| | | | | |
Collapse
|
42
|
Frechilla D, Insausti R, Ruiz-Golvano P, García-Osta A, Rubio MP, Almendral JM, Del Río J. Implanted BDNF-producing fibroblasts prevent neurotoxin-induced serotonergic denervation in the rat striatum. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2000; 76:306-14. [PMID: 10762706 DOI: 10.1016/s0169-328x(00)00012-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Degeneration of serotonergic fibers in the rat striatum was produced by local administration of the serotonergic neurotoxin 5, 7-dihydroxytryptamine (5,7-DHT) or the dopaminergic neurotoxin 1-methyl-4-phenylpyridinium (MPP(+)), which is also toxic to serotonergic neurons. One week before neurotoxin administration, fibroblasts engineered to express the human BDNF gene were grafted into the mesencephalon, dorsal to the substantia nigra. Rats implanted with fibroblasts expressing the LacZ gene were used as controls, as well as sham-operated animals (not injected with any neurotoxin). After a survival period of 1 week, the serotonergic innervation of the striatum was assessed by measuring serotonin (5-HT) content and by immunohistochemical detection of 5-HT positive fibers. BDNF-producing cells prevented the striatal 5-HT loss induced by local administration of either 5,7-DHT or MPP(+), as well as the striatal dopamine (DA) loss induced by the latter neurotoxin. Grafting of fibroblasts carrying the BDNF or the Lac-Z gene did not modify striatal 5-HT or DA content in sham-operated animals. In 5, 7-DHT-lesioned rats, implanted or not with control Lac-Z fibroblasts, a striking reduction in the density of 5-HT immunoreactive fibers was observed. By contrast, the density of 5-HT fibers was similar in rats implanted with BDNF-producing fibroblasts as compared to sham-operated controls. The protective effect of BDNF on the damage to serotonergic terminals induced by the two neurotoxins suggests the interest of this neurotrophin in the treatment of behavioral disorders associated to neurodegenerative diseases.
Collapse
Affiliation(s)
- D Frechilla
- Department of Pharmacology, University of Navarra Medical School, c/ Irunlarrea 1, 31008-, Pamplona, Spain
| | | | | | | | | | | | | |
Collapse
|
43
|
Sommerfeld MT, Schweigreiter R, Barde YA, Hoppe E. Down-regulation of the neurotrophin receptor TrkB following ligand binding. Evidence for an involvement of the proteasome and differential regulation of TrkA and TrkB. J Biol Chem 2000; 275:8982-90. [PMID: 10722747 DOI: 10.1074/jbc.275.12.8982] [Citation(s) in RCA: 130] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
This study examines the mechanisms by which the tyrosine kinase receptor TrkB is down-regulated following binding of brain-derived neurotrophic factor (BDNF). In primary cultures of cerebellar granule neurons, BDNF-induced reduction of TrkB receptors was largely prevented by the addition of specific proteasome inhibitors. HN10 cells, a neuronal cell line that can be readily transfected, also showed a marked down-regulation of cell surface TrkB following BDNF exposure. In addition, we observed that prolonged exposure to nerve growth factor of TrkA-transfected cells did not lead to the down-regulation seen with BDNF and TrkB. TrkA and TrkB chimeric molecules were therefore expressed in HN10 cells and tested for ligand-induced regulation. These experiments led to the conclusion that the motives responsible for down-regulation are contained in the cytoplasmic domain of TrkB, and a short sequence in the juxtamembrane domain of TrkB was identified that confers nerve growth factor-induced down-regulation when inserted into TrkA.
Collapse
Affiliation(s)
- M T Sommerfeld
- Max-Planck Institute of Neurobiology, Department of Neurobiochemistry, Am Klopferspitz 18A, 82152 Planegg-Martinsried, Germany
| | | | | | | |
Collapse
|
44
|
Abstract
The retino-tectal system has been used to study developmental aspects of axon growth, synapse formation and the establishment of a precise topographic order as well as degeneration and regeneration of adult retinal ganglion cell (RGC) axons after axonal lesion. This paper reviews some novel findings that provide new insights into the mechanisms of developmental RGC axon growth, pathfinding, and target formation. It also focuses on the cellular and molecular cascades that underlie RGC degeneration following an axonal lesion and on some therapeutic strategies to enhance survival of axotomized RGCs in vivo. In addition, this review deals with problems related to the induction of regeneration after axonal lesion in the adult CNS using the retino-tectal system as model. Different therapeutic approaches to promote RGC regeneration and requirements for specific target formation of regenerating RGCs in vitro and in vivo are discussed.
Collapse
Affiliation(s)
- J Weise
- Neurologische Universitätsklinik, 72076 Tübingen, Germany
| | | | | |
Collapse
|
45
|
Karchewski LA, Kim FA, Johnston J, McKnight RM, Verge VM. Anatomical evidence supporting the potential for modulation by multiple neurotrophins in the majority of adult lumbar sensory neurons. J Comp Neurol 1999; 413:327-41. [PMID: 10524342 DOI: 10.1002/(sici)1096-9861(19991018)413:2<327::aid-cne11>3.0.co;2-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Neurotrophins exert effects on sensory neurons through receptor tyrosine kinases (trks) and a common neurotrophin receptor (p75). Quantitative in situ hybridization studies were performed on serial sections to identify neurons expressing single or multiple neurotrophin trk receptor mRNA(s) in adult lumbar dorsal root ganglion (DRG) in order to examine the possibility of multi-neurotrophin modulation of phenotype via different trk receptors or various trk isoforms. Expression of mRNA encoding trkA, trkB, trkC, or p75 is restricted to select subpopulations representing approximately 41%, 33%, 43%, and 79% of DRG neurons, respectively. Colocalization studies reveal that approximately 10% of DRG neurons coexpress trkA and trkB mRNA; 19% coexpress trkA and trkC mRNA; and 18% coexpress trkB and trkC mRNA. Trilocalization of all three trk mRNAs is rare, with approximately 3-4% of neurons in this category. Overall incidence of expression of more than one full length trk mRNA occurs in approximately 40% of DRG neurons, whereas expression of individual trk mRNA is found in approximately 34%. Full length trk receptor mRNA is rarely detected without p75, implicating the latter in neuronal response to neurotrophins. Examination of two full-length isoforms of trkA reveal that they are coexpressed with relative levels of expression positively correlated. TrkC mRNAs corresponding to 14- or 39-amino acid insert isoforms colocalize with the non-insert trkC isoform, but the converse is not necessarily true. The data suggest that substantial subpopulations of adult sensory neurons may be modulated through interactions with multiple neurotrophins, the consequences of which are largely unknown.
Collapse
MESH Headings
- Animals
- Base Sequence
- Ganglia, Spinal/cytology
- Gene Expression
- In Situ Hybridization
- Lumbosacral Region/anatomy & histology
- Male
- Molecular Sequence Data
- Nerve Tissue Proteins/analysis
- Nerve Tissue Proteins/genetics
- Neurons, Afferent/physiology
- Phenotype
- Protein Isoforms/analysis
- Protein Isoforms/genetics
- RNA, Messenger/analysis
- Rats
- Rats, Wistar
- Receptor, Nerve Growth Factor/analysis
- Receptor, Nerve Growth Factor/genetics
- Receptor, trkA/analysis
- Receptor, trkA/genetics
- Receptor, trkB/analysis
- Receptor, trkB/genetics
- Receptor, trkC/analysis
- Receptor, trkC/genetics
- Receptors, Nerve Growth Factor/analysis
- Receptors, Nerve Growth Factor/genetics
- Superior Cervical Ganglion/cytology
Collapse
Affiliation(s)
- L A Karchewski
- Department of Anatomy & Cell Biology, University of Saskatchewan, Canada
| | | | | | | | | |
Collapse
|
46
|
Wiese S, Metzger F, Holtmann B, Sendtner M. Mechanical and excitotoxic lesion of motoneurons: effects of neurotrophins and ciliary neurotrophic factor on survival and regeneration. ACTA NEUROCHIRURGICA. SUPPLEMENT 1999; 73:31-9. [PMID: 10494338 DOI: 10.1007/978-3-7091-6391-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Mechanical lesion of peripheral nerves leads to extensive death of corresponding motoneurons in newborn rodents. The extent of cell death can be significantly reduced by neurotrophic factors. These molecules are produced by glial and neuronal cells and play an important role in supporting survival and regeneration of various neuronal populations in the central nervous system, in particular after mechanical, excitotoxic and ischemic insults. In addition, factors such as ciliary neurotrophic factor and neurotrophin-3 influence glial cell proliferation and survival. We have investigated the role of neurotrophic factors on motoneurons, both in cell culture and after axotomy in vivo. Moreover, the role of excitatory neurotransmission in modulating dendritic architecture of these cells was analyzed. Our data suggest that motoneurons are a suitable model for investigating the complex functional and morphological changes after brain lesion and for the identification of new therapeutic strategies to influence survival and functional recovery under such circumstances.
Collapse
Affiliation(s)
- S Wiese
- Dept. of Neurology, University of Würzburg, Germany
| | | | | | | |
Collapse
|
47
|
Rabacchi SA, Kruk B, Hamilton J, Carney C, Hoffman JR, Meyer SL, Springer JE, Baird DH. BDNF and NT4/5 promote survival and neurite outgrowth of pontocerebellar mossy fiber neurons. ACTA ACUST UNITED AC 1999. [DOI: 10.1002/(sici)1097-4695(199908)40:2<254::aid-neu11>3.0.co;2-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
48
|
Osehobo P, Adams B, Sazgar M, Xu Y, Racine RJ, Fahnestock M. Brain-derived neurotrophic factor infusion delays amygdala and perforant path kindling without affecting paired-pulse measures of neuronal inhibition in adult rats. Neuroscience 1999; 92:1367-75. [PMID: 10426491 DOI: 10.1016/s0306-4522(99)00048-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Kindling is an animal model of human temporal lobe epilepsy in which excitability in limbic structures is permanently enhanced by repeated stimulations. Kindling also increases the expression of nerve growth factor, brain-derived neurotrophic factor, and brain-derived neurotrophic factor receptor messenger RNAs in both the hippocampus and cerebral cortex and causes structural changes in the hippocampus including hilar hypertrophy. We have recently shown that intraventricular nerve growth factor infusion enhances the development of kindling, whereas blocking nerve growth factor activity retards amygdaloid kindling. Furthermore, we have shown that nerve growth factor protects against kindling-induced hilar hypertrophy. The physiological role of brain-derived neurotrophic factor in kindling is not as clear. Acute injection of brain-derived neurotrophic factor increases neuronal excitability and causes seizures, whereas chronic brain-derived neurotrophic factor infusion in rats slows hippocampal kindling. In agreement with the latter, we show here that intrahilar brain-derived neurotrophic factor infusion delays amygdala and perforant path kindling. In addition, we show that brain-derived neurotrophic factor, unlike nerve growth factor, does not protect against kindling-induced increases in hilar area. To test the hypothesis that brain-derived neurotrophic factor suppresses kindling by increasing inhibition above normal levels, we performed paired-pulse measures in the perforant path-dentate gyrus pathway. Brain-derived neurotrophic factor infused into the hippocampus had no effect on the stimulus intensity function (input/output curves); there was also no significant effect on paired-pulse inhibition. We then kindled the perforant path 10 days after the end of brain-derived neurotrophic factor treatment. Once again, kindling was retarded, showing that the brain-derived neurotrophic factor effect is long-lasting. These results indicate that prolonged in vivo infusion of brain-derived neurotrophic factor reduces, rather than increases, excitability without increasing inhibitory neuron function, at least as assessed by paired-pulse protocols. This effect may be mediated by long-lasting effects on brain-derived neurotrophic factor receptor regulation.
Collapse
Affiliation(s)
- P Osehobo
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
The neurotrophin family of growth factors supports survival and differentiation of neurons in the developing vertebrate nervous system by binding activating receptor tyrosine kinases, the Trks. Activation of Trk receptors leads to stimulation of a number of intracellular signaling cascades including, among others, the ras/extracellular regulated kinase (erk) and the phosphatidylinositol-3 kinase (PI 3 kinase) cascades. Over the past several years, work in several neurotrophin responsive systems has begun to identify the role each of these signaling cascades plays in the cellular response to neurotrophins. It now appears that neurotrophins, in particular nerve growth factor (NGF), mediate their multiple effects through a number of distinct intracellular signaling cascades. In this review, we will overview the evidence implicating specific signaling cascades in aspects of the cellular response to the neurotrophins, specifically in response to activation of TrkA by NGF.
Collapse
Affiliation(s)
- L J Klesse
- Center for Developmental Biology, University of Texas, Southwestern Medical Center, Dallas 75235-9133, USA
| | | |
Collapse
|
50
|
Wiese S, Metzger F, Holtmann B, Sendtner M. The role of p75NTR in modulating neurotrophin survival effects in developing motoneurons. Eur J Neurosci 1999; 11:1668-76. [PMID: 10215920 DOI: 10.1046/j.1460-9568.1999.00585.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Neurotrophins exert their biological functions on neuronal cells through two types of receptors, the trk tyrosine kinases and the low-affinity neurotrophin receptor (p75NTR), which can bind all neurotrophins with similar affinity. The p75NTR is highly expressed in developing motoneurons and in adult motoneurons after axotomy, suggestive of a physiological role in mediating neurotrophin responses under such conditions. In order to characterize this specific function of p75NTR, we have tested the effects of nerve growth factor (NGF) on embryonic motoneurons from control and p75NTR-deficient mice. NGF antagonizes brain-derived neurotrophic factor (BDNF)- and neurotrophin-3 (NT-3)-mediated survival in control but not p75NTR-deficient motoneurons. Survival of cultured motoneurons in the presence of 0.5 ng/mL of either ciliary neurotrophic factor (CNTF) or glial-derived neurotrophic factor (GDNF) was not reduced by 20 ng/mL NGF. Dose-response investigations revealed that five times higher concentrations of BDNF are required for half-maximal survival of p75NTR-deficient motoneurons in comparison to motoneurons from wild-type controls. After facial nerve lesion in newborn wild-type mice, local administration of NGF reduced survival of corresponding motoneurons to less than 2% compared to the unlesioned control side. In p75NTR-deficient mice, the same treatment did not enhance facial motoneuron death on the lesioned side. In the facial nucleus of 1-week-old p75NTR -/- mice, a significant reduction of motoneurons was observed at the unlesioned side in comparison to p75NTR +/+ mice. The observation that motoneuron cell numbers are reduced in the facial nucleus of newborn p75NTR-deficient mice suggests that p75NTR might not function as a physiological cell death receptor in developing motoneurons.
Collapse
Affiliation(s)
- S Wiese
- Klinische Forschergruppe Neuroregeneration, Department of Neurology, University of Würzburg, Germany
| | | | | | | |
Collapse
|