1
|
Merryweather D, Moxon SR, Capel AJ, Hooper NM, Lewis MP, Roach P. Impact of type-1 collagen hydrogel density on integrin-linked morphogenic response of SH-SY5Y neuronal cells. RSC Adv 2021; 11:33124-33135. [PMID: 35493559 PMCID: PMC9042137 DOI: 10.1039/d1ra05257h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/28/2021] [Indexed: 11/23/2022] Open
Abstract
Cellular metabolism and behaviour is closely linked to cytoskeletal tension and scaffold mechanics. In the developing nervous system functional connectivity is controlled by the interplay between chemical and mechanical cues that initiate programs of cell behaviour. Replication of functional connectivity in neuronal populations in vitro has proven a technical challenge due to the absence of many systems of biomechanical regulation that control directional outgrowth in vivo. Here, a 3D culture system is explored by dilution of a type I collagen hydrogel to produce variation in gel stiffness. Hydrogel scaffold remodelling was found to be linked to gel collagen concentration, with a greater degree of gel contraction occurring at lower concentrations. Gel mechanics were found to evolve over the culture period according to collagen concentration. Less concentrated gels reduced in stiffness, whilst a biphasic pattern of increasing and then decreasing stiffness was observed at higher concentrations. Analysis of these cultures by PCR revealed a program of shifting integrin expression and highly variable activity in key morphogenic signal pathways, such as mitogen-associated protein kinase, indicating genetic impact of biomaterial interactions via mechano-regulation. Gel contraction at lower concentrations was also found to be accompanied by an increase in average collagen fibre diameter. Minor changes in biomaterial mechanics result in significant changes in programmed cell behaviour, resulting in adoption of markedly different cell morphologies and ability to remodel the scaffold. Advanced understanding of cell-biomaterial interactions, over short and long-term culture, is of critical importance in the development of novel tissue engineering strategies for the fabrication of biomimetic 3D neuro-tissue constructs. Simple methods of tailoring the initial mechanical environment presented to SH-SY5Y populations in 3D can lead to significantly different programs of network development over time.
Collapse
Affiliation(s)
- D Merryweather
- Department of Chemistry, Loughborough University Leicestershire LE11 3TU UK
| | - S R Moxon
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre Manchester M13 9PL UK
| | - A J Capel
- National Centre for Sport and Exercise Medicine (NCSEM), School of Sport, Exercise and Health Sciences, Loughborough University Leicestershire LE11 3TU UK
| | - N M Hooper
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre Manchester M13 9PL UK
| | - M P Lewis
- National Centre for Sport and Exercise Medicine (NCSEM), School of Sport, Exercise and Health Sciences, Loughborough University Leicestershire LE11 3TU UK
| | - P Roach
- Department of Chemistry, Loughborough University Leicestershire LE11 3TU UK
| |
Collapse
|
2
|
Alizzi RA, Xu D, Tenenbaum CM, Wang W, Gavis ER. The ELAV/Hu protein Found in neurons regulates cytoskeletal and ECM adhesion inputs for space-filling dendrite growth. PLoS Genet 2020; 16:e1009235. [PMID: 33370772 PMCID: PMC7793258 DOI: 10.1371/journal.pgen.1009235] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 01/08/2021] [Accepted: 10/29/2020] [Indexed: 12/17/2022] Open
Abstract
Dendritic arbor morphology influences how neurons receive and integrate extracellular signals. We show that the ELAV/Hu family RNA-binding protein Found in neurons (Fne) is required for space-filling dendrite growth to generate highly branched arbors of Drosophila larval class IV dendritic arborization neurons. Dendrites of fne mutant neurons are shorter and more dynamic than in wild-type, leading to decreased arbor coverage. These defects result from both a decrease in stable microtubules and loss of dendrite-substrate interactions within the arbor. Identification of transcripts encoding cytoskeletal regulators and cell-cell and cell-ECM interacting proteins as Fne targets using TRIBE further supports these results. Analysis of one target, encoding the cell adhesion protein Basigin, indicates that the cytoskeletal defects contributing to branch instability in fne mutant neurons are due in part to decreased Basigin expression. The ability of Fne to coordinately regulate the cytoskeleton and dendrite-substrate interactions in neurons may shed light on the behavior of cancer cells ectopically expressing ELAV/Hu proteins. Different types of neurons have different sizes and shapes that are tailored to their particular functions. In the fruit fly larva, a set of sensory neurons called class IV da neurons have highly branched trees of dendrites that cover the epidermis to sense potentially harmful stimuli. Neurons whose dendrites completely fill the territory they sample are also found in zebrafish, worms, mice and humans. We show that an RNA-binding protein called Fne plays an important role in coordinating different contributions to dendrite branching in class IV da neurons by impacting the organization of the cytoskeleton within the neuron and the ability of the dendrite to contact the substratum outside of it. The identification of mRNAs that code for cytoskeleton regulators and adhesive proteins as targets of Fne using a genome-wide approach further supports these results. While the ability of Fne to exert control over such proteins is crucial to generating the space-filling growth of neurons, it can be deleterious if not properly employed, such as when the homologs of Fne are expressed in cancer cells.
Collapse
Affiliation(s)
- Rebecca A. Alizzi
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Derek Xu
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Conrad M. Tenenbaum
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Wei Wang
- Lewis-Sigler Institute, Princeton University, Princeton, New Jersey, United States of America
| | - Elizabeth R. Gavis
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
3
|
Abstract
The formation of correct synaptic structures and neuronal connections is paramount for normal brain development and a functioning adult brain. The integrin family of cell adhesion receptors and their ligands play essential roles in the control of several processes regulating neuronal connectivity - including neurite outgrowth, the formation and maintenance of synapses, and synaptic plasticity - that are affected in neurodevelopmental disorders, such as autism spectrum disorders (ASDs) and schizophrenia. Many ASD- and schizophrenia-associated genes are linked to alterations in the genetic code of integrins and associated signalling pathways. In non-neuronal cells, crosstalk between integrin-mediated adhesions and the actin cytoskeleton, and the regulation of integrin activity (affinity for extracellular ligands) are widely studied in healthy and pathological settings. In contrast, the roles of integrin-linked pathways in the central nervous system remains less well defined. In this Review, we will provide an overview of the known pathways that are regulated by integrin-ECM interaction in developing neurons and in adult brain. We will also describe recent advances in the identification of mechanisms that regulate integrin activity in neurons, and highlight the interesting emerging links between integrins and neurodevelopment.
Collapse
Affiliation(s)
- Johanna Lilja
- Turku Centre for Biotechnology, University of Turku, FIN-20520 Turku, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku, FIN-20520 Turku, Finland .,Department of Biochemistry, University of Turku, FIN-20500 Turku, Finland
| |
Collapse
|
4
|
Clarke JP, Mearow K. Autophagy inhibition in endogenous and nutrient-deprived conditions reduces dorsal root ganglia neuron survival and neurite growth in vitro. J Neurosci Res 2016; 94:653-70. [PMID: 27018986 DOI: 10.1002/jnr.23733] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/05/2016] [Accepted: 02/28/2016] [Indexed: 12/31/2022]
Abstract
Peripheral neuropathies can result in cytoskeletal changes in axons, ultimately leading to Wallerian degeneration and cell death. Recently, autophagy has been studied as a potential target for improving axonal survival and growth during peripheral nerve damage. This study investigates the influence of autophagy on adult dorsal root ganglia (DRG) neuron survival and axonal growth under control and nutrient deprivation conditions. Constitutive autophagy was modulated with pharmacological activators (rapamycin; Rapa) and inhibitors (3-methyladenine, bafilomycin A1) in conjunction with either a nutrient-stable environment (standard culture medium) or a nutrient-deprived environment (Hank's balanced salt solution + Ca(2+) /Mg(2+) ). The results demonstrated that autophagy inhibition decreased cell viability and reduced neurite growth and branching complexity. Although autophagy was upregulated with nutrient deprivation compared with the control, it was not further activated by rapamycin, suggesting a threshold level of autophagy. Overall, both cellular and biochemical approaches combined to show the influence of autophagy on adult DRG neuron survival and growth. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Joseph-Patrick Clarke
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Karen Mearow
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| |
Collapse
|
5
|
Turney SG, Ahmed M, Chandrasekar I, Wysolmerski RB, Goeckeler ZM, Rioux RM, Whitesides GM, Bridgman PC. Nerve growth factor stimulates axon outgrowth through negative regulation of growth cone actomyosin restraint of microtubule advance. Mol Biol Cell 2016; 27:500-17. [PMID: 26631553 PMCID: PMC4751601 DOI: 10.1091/mbc.e15-09-0636] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/23/2015] [Accepted: 11/24/2015] [Indexed: 01/19/2023] Open
Abstract
Nerve growth factor (NGF) promotes growth, differentiation, and survival of sensory neurons in the mammalian nervous system. Little is known about how NGF elicits faster axon outgrowth or how growth cones integrate and transform signal input to motor output. Using cultured mouse dorsal root ganglion neurons, we found that myosin II (MII) is required for NGF to stimulate faster axon outgrowth. From experiments inducing loss or gain of function of MII, specific MII isoforms, and vinculin-dependent adhesion-cytoskeletal coupling, we determined that NGF causes decreased vinculin-dependent actomyosin restraint of microtubule advance. Inhibition of MII blocked NGF stimulation, indicating the central role of restraint in directed outgrowth. The restraint consists of myosin IIB- and IIA-dependent processes: retrograde actin network flow and transverse actin bundling, respectively. The processes differentially contribute on laminin-1 and fibronectin due to selective actin tethering to adhesions. On laminin-1, NGF induced greater vinculin-dependent adhesion-cytoskeletal coupling, which slowed retrograde actin network flow (i.e., it regulated the molecular clutch). On fibronectin, NGF caused inactivation of myosin IIA, which negatively regulated actin bundling. On both substrates, the result was the same: NGF-induced weakening of MII-dependent restraint led to dynamic microtubules entering the actin-rich periphery more frequently, giving rise to faster elongation.
Collapse
Affiliation(s)
- Stephen G Turney
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Mostafa Ahmed
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110
| | - Indra Chandrasekar
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110
| | - Robert B Wysolmerski
- Department of Neurobiology and Anatomy, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Zoe M Goeckeler
- Department of Neurobiology and Anatomy, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Robert M Rioux
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
| | - George M Whitesides
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
| | - Paul C Bridgman
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
6
|
Wong AW, K P Yeung J, Payne SC, Keast JR, Osborne PB. Neurite outgrowth in normal and injured primary sensory neurons reveals different regulation by nerve growth factor (NGF) and artemin. Mol Cell Neurosci 2015; 65:125-34. [PMID: 25752731 DOI: 10.1016/j.mcn.2015.03.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/19/2015] [Accepted: 03/05/2015] [Indexed: 12/11/2022] Open
Abstract
Neurotrophic factors have been intensively studied as potential therapeutic agents for promoting neural regeneration and functional recovery after nerve injury. Artemin is a member of the glial cell line-derived neurotrophic factor (GDNF) family of ligands (GFLs) that forms a signalling complex with GFRα3 and the tyrosine kinase Ret. Systemic administration of artemin in rodents is reported to facilitate regeneration of primary sensory neurons following axotomy, improve recovery of sensory function, and reduce sensory hypersensitivity that is a cause of pain. However, the biological mechanisms that underlie these effects are mostly unknown. This study has investigated the biological significance of the colocalisation of GFRα3 with TrkA (neurotrophin receptor for nerve growth factor [NGF]) in the peptidergic type of unmyelinated (C-fibre) sensory neurons in rat dorsal root ganglia (DRG). In vitro neurite outgrowth assays were used to study the effects of artemin and NGF by comparing DRG neurons that were previously uninjured, or were axotomised in vivo by transecting a visceral or somatic peripheral nerve. We found that artemin could facilitate neurite initiation but in comparison to NGF had low efficacy for facilitating neurite elongation and branching. This low efficacy was not increased when a preconditioning in vivo nerve injury was used to induce a pro-regenerative state. Neurite initiation was unaffected by artemin when PI3 kinase and Src family kinase signalling were blocked, but NGF had a reduced effect.
Collapse
Affiliation(s)
- Agnes W Wong
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria 3010, Australia
| | - James K P Yeung
- School of Medical Sciences, UNSW, Randwick, NSW 2052, Australia; Pain Management Research Institute (Kolling Institute), Sydney Medical School - Northern, The University of Sydney NSW 2010, Australia
| | - Sophie C Payne
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Janet R Keast
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria 3010, Australia; Pain Management Research Institute (Kolling Institute), Sydney Medical School - Northern, The University of Sydney NSW 2010, Australia
| | - Peregrine B Osborne
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria 3010, Australia; Pain Management Research Institute (Kolling Institute), Sydney Medical School - Northern, The University of Sydney NSW 2010, Australia.
| |
Collapse
|
7
|
Epineural Tube Repair. Plast Reconstr Surg 2015. [DOI: 10.1007/978-1-4471-6335-0_57] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
8
|
Romano NH, Madl CM, Heilshorn SC. Matrix RGD ligand density and L1CAM-mediated Schwann cell interactions synergistically enhance neurite outgrowth. Acta Biomater 2015; 11:48-57. [PMID: 25308870 DOI: 10.1016/j.actbio.2014.10.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 10/01/2014] [Accepted: 10/04/2014] [Indexed: 11/26/2022]
Abstract
The innate biological response to peripheral nerve injury involves a complex interplay of multiple molecular cues to guide neurites across the injury gap. Many current strategies to stimulate regeneration take inspiration from this biological response. However, little is known about the balance of cell-matrix and Schwann cell-neurite dynamics required for regeneration of neural architectures. We present an engineered extracellular matrix (eECM) microenvironment with tailored cell-matrix and cell-cell interactions to study their individual and combined effects on neurite outgrowth. This eECM regulates cell-matrix interactions by presenting integrin-binding RGD (Arg-Gly-Asp) ligands at specified densities. Simultaneously, the addition or exclusion of nerve growth factor (NGF) is used to modulate L1CAM-mediated Schwann cell-neurite interactions. Individually, increasing the RGD ligand density from 0.16 to 3.2mM resulted in increasing neurite lengths. In matrices presenting higher RGD ligand densities, neurite outgrowth was synergistically enhanced in the presence of soluble NGF. Analysis of Schwann cell migration and co-localization with neurites revealed that NGF enhanced cooperative outgrowth between the two cell types. Interestingly, neurites in NGF-supplemented conditions were unable to extend on the surrounding eECM without the assistance of Schwann cells. Blocking studies revealed that L1CAM is primarily responsible for these Schwann cell-neurite interactions. Without NGF supplementation, neurite outgrowth was unaffected by L1CAM blocking or the depletion of Schwann cells. These results underscore the synergistic interplay between cell-matrix and cell-cell interactions in enhancing neurite outgrowth for peripheral nerve regeneration.
Collapse
|
9
|
Ribeiro A, Balasubramanian S, Hughes D, Vargo S, Powell EM, Leach JB. β1-Integrin cytoskeletal signaling regulates sensory neuron response to matrix dimensionality. Neuroscience 2013; 248:67-78. [PMID: 23764511 DOI: 10.1016/j.neuroscience.2013.05.057] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 05/30/2013] [Indexed: 10/26/2022]
Abstract
Neuronal differentiation, pathfinding and morphology are directed by biochemical cues that in vivo are presented in a complex scaffold of extracellular matrix. This microenvironment is three-dimensional (3D) and heterogeneous. Therefore, it is not surprising that more physiologically-relevant cellular responses are found in 3D culture environments rather than on two-dimensional (2D) flat substrates. One key difference between 2D and 3D environments is the spatial arrangement of cell-matrix interactions. Integrins and other receptor proteins link the various molecules presented in the extracellular environment to intracellular signaling cascades and thus influence a number of neuronal responses including the availability and activation of integrins themselves. We have previously reported that a 3D substrate induces an important morphological transformation of embryonic mouse dorsal root ganglion (DRG) neurons. Here, we investigate the hypothesis that β1-integrin signaling via focal adhesion kinase (FAK) and the RhoGTPases Rac and Rho influences neuronal morphology in 2D vs 3D environments. We report that β1-integrin activity and FAK phosphorylation at tyrosine 397 (FAKpY397) are linked to neuronal polarization as well as neurite outgrowth and branching. Rac and Rho expression are decreased in 3D vs 2D culture but not correlated with β1-integrin function. These results suggest that proper β1-integrin activity is required for the elaboration of physiologic DRG morphology and that 3D culture provides a more appropriate milieu to the mimic in vivo scenario. We propose that neuronal morphology may be directed during development and regeneration by factors that influence how β1-integrin, FAK and RhoGTPase molecules integrate substrate signals in the 3D microenvironment.
Collapse
Affiliation(s)
- A Ribeiro
- Department of Chemical, Biochemical & Environmental Engineering, UMBC, Eng 314, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - S Balasubramanian
- Department of Chemical, Biochemical & Environmental Engineering, UMBC, Eng 314, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - D Hughes
- Department of Chemical, Biochemical & Environmental Engineering, UMBC, Eng 314, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - S Vargo
- Department of Chemical, Biochemical & Environmental Engineering, UMBC, Eng 314, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - E M Powell
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, HSF II S251, 20 Penn Street, Baltimore, MD 21201, USA; Department of Psychiatry, University of Maryland School of Medicine, HSF II S251, 20 Penn Street, Baltimore, MD 21201, USA; Department of Bioengineering, University of Maryland School of Medicine, HSF II S251, 20 Penn Street, Baltimore, MD 21201, USA
| | - J B Leach
- Department of Chemical, Biochemical & Environmental Engineering, UMBC, Eng 314, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| |
Collapse
|
10
|
Chung TW, Lai DM, Chen SD, Lin YI. Poly (ε-caprolactone) scaffolds functionalized by grafting NGF and GRGD promote growth and differentiation of PC12 cells. J Biomed Mater Res A 2013; 102:315-23. [DOI: 10.1002/jbm.a.34693] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 02/22/2013] [Indexed: 01/19/2023]
Affiliation(s)
- Tze-Wen Chung
- Department of Chemical and Materials Engineering; National Yunlin University of Science and Technology; Dou-Liu Yun-Lin 640 Taiwan, ROC
| | - Dar-Ming Lai
- Department of Surgery; National Taiwan University Hospital; National Taiwan University College of Medicine; Taipei Taiwan, ROC
| | - Shin-Der Chen
- Department of Chemical and Materials Engineering; National Yunlin University of Science and Technology; Dou-Liu Yun-Lin 640 Taiwan, ROC
| | - Ya-I Lin
- Department of Chemical and Materials Engineering; National Yunlin University of Science and Technology; Dou-Liu Yun-Lin 640 Taiwan, ROC
| |
Collapse
|
11
|
Fudge NJ, Mearow KM. Extracellular matrix-associated gene expression in adult sensory neuron populations cultured on a laminin substrate. BMC Neurosci 2013; 14:15. [PMID: 23360524 PMCID: PMC3610289 DOI: 10.1186/1471-2202-14-15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 01/08/2013] [Indexed: 12/03/2022] Open
Abstract
Background In our previous investigations of the role of the extracellular matrix (ECM) in promoting neurite growth we have observed that a permissive laminin (LN) substrate stimulates differential growth responses in subpopulations of mature dorsal root ganglion (DRG) neurons. DRG neurons expressing Trk and p75 receptors grow neurites on a LN substrate in the absence of neurotrophins, while isolectin B4-binding neurons (IB4+) do not display significant growth under the same conditions. We set out to determine whether there was an expression signature of the LN-induced neurite growth phenotype. Using a lectin binding protocol IB4+ neurons were isolated from dissociated DRG neurons, creating two groups - IB4+ and IB4-. A small-scale microarray approach was employed to screen the expression of a panel of ECM-associated genes following dissociation (t=0) and after 24 hr culture on LN (t=24LN). This was followed by qRT-PCR and immunocytochemistry of selected genes. Results The microarray screen showed that 36 of the 144 genes on the arrays were consistently expressed by the neurons. The array analyses showed that six genes had lower expression in the IB4+ neurons compared to the IB4- cells at t=0 (CTSH, Icam1, Itgβ1, Lamb1, Plat, Spp1), and one gene was expressed at higher levels in the IB4+ cells (Plaur). qRT-PCR was carried out as an independent assessment of the array results. There were discrepancies between the two methods, with qRT-PCR confirming the differences in Lamb1, Plat and Plaur, and showing decreased expression of AdamTs1, FN, and Icam in the IB4+ cells at t=0. After 24 hr culture on LN, there were no significant differences detected by qRT-PCR between the IB4+ and IB4- cells. However, both groups showed upregulation of Itgβ1 and Plaur after 24 hr on LN, the IB4+ group also had increased Plat, and the IB4- cells showed decreased Lamb1, Icam1 and AdamTs1. Further, the array screen also detected a number of genes (not subjected to qRT-PCR) expressed similarly by both populations in relatively high levels but not detectably influenced by time in culture (Bsg, Cst3, Ctsb, Ctsd, Ctsl, Mmp14, Mmp19, Sparc. We carried out immunohistochemistry to confirm expression of proteins encoded by a number of these genes. Conclusions Our results show that 1B4+ and IB4- neurons differ in the expression of several genes that are associated with responsiveness to the ECM prior to culturing (AdamTs1, FN, Icam1, Lamb1, Plat, Plaur). The data suggest that the genes expressed at higher levels in the IB4- neurons could contribute to the initial growth response of these cells in a permissive environment and could also represent a common injury response that subsequently promotes axon regeneration. The differential expression of several extracellular matrix molecules (FN, Lamb1, Icam) may suggest that the IB4- neurons are capable of maintaining /secreting their local extracellular environment which could aid in the regenerative process. Overall, these data provide new information on potential targets that could be manipulated to enhance axonal regeneration in the mature nervous system.
Collapse
Affiliation(s)
- Neva J Fudge
- Division of BioMedical Sciences, Memorial University of Newfoundland, St, John's, NL, Canada
| | | |
Collapse
|
12
|
Santos TG, Beraldo FH, Hajj GNM, Lopes MH, Roffe M, Lupinacci FCS, Ostapchenko VG, Prado VF, Prado MAM, Martins VR. Laminin-γ1 chain and stress inducible protein 1 synergistically mediate PrPC-dependent axonal growth via Ca2+ mobilization in dorsal root ganglia neurons. J Neurochem 2012; 124:210-23. [PMID: 23145988 DOI: 10.1111/jnc.12091] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 11/01/2012] [Accepted: 11/01/2012] [Indexed: 12/01/2022]
Abstract
Prion protein (PrP(C)) is a cell surface glycoprotein that is abundantly expressed in nervous system. The elucidation of the PrP(C) interactome network and its significance on neural physiology is crucial to understanding neurodegenerative events associated with prion and Alzheimer's diseases. PrP(C) co-opts stress inducible protein 1/alpha7 nicotinic acetylcholine receptor (STI1/α7nAChR) or laminin/Type I metabotropic glutamate receptors (mGluR1/5) to modulate hippocampal neuronal survival and differentiation. However, potential cross-talk between these protein complexes and their role in peripheral neurons has never been addressed. To explore this issue, we investigated PrP(C)-mediated axonogenesis in peripheral neurons in response to STI1 and laminin-γ1 chain-derived peptide (Ln-γ1). STI1 and Ln-γ1 promoted robust axonogenesis in wild-type neurons, whereas no effect was observed in neurons from PrP(C) -null mice. PrP(C) binding to Ln-γ1 or STI1 led to an increase in intracellular Ca(2+) levels via distinct mechanisms: STI1 promoted extracellular Ca(2+) influx, and Ln-γ1 released calcium from intracellular stores. Both effects depend on phospholipase C activation, which is modulated by mGluR1/5 for Ln-γ1, but depends on, C-type transient receptor potential (TRPC) channels rather than α7nAChR for STI1. Treatment of neurons with suboptimal concentrations of both ligands led to synergistic actions on PrP(C)-mediated calcium response and axonogenesis. This effect was likely mediated by simultaneous binding of the two ligands to PrP(C). These results suggest a role for PrP(C) as an organizer of diverse multiprotein complexes, triggering specific signaling pathways and promoting axonogenesis in the peripheral nervous system.
Collapse
Affiliation(s)
- Tiago G Santos
- International Research Center, A.C. Camargo Hospital, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Zhang XF, Hyland C, Van Goor D, Forscher P. Calcineurin-dependent cofilin activation and increased retrograde actin flow drive 5-HT-dependent neurite outgrowth in Aplysia bag cell neurons. Mol Biol Cell 2012; 23:4833-48. [PMID: 23097492 PMCID: PMC3521690 DOI: 10.1091/mbc.e12-10-0715] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Neurite outgrowth in response to soluble growth factors often involves changes in intracellular Ca(2+); however, mechanistic roles for Ca(2+) in controlling the underlying dynamic cytoskeletal processes have remained enigmatic. Bag cell neurons exposed to serotonin (5-hydroxytryptamine [5-HT]) respond with a threefold increase in neurite outgrowth rates. Outgrowth depends on phospholipase C (PLC) → inositol trisphosphate → Ca(2+) → calcineurin signaling and is accompanied by increased rates of retrograde actin network flow in the growth cone P domain. Calcineurin inhibitors had no effect on Ca(2+) release or basal levels of retrograde actin flow; however, they completely suppressed 5-HT-dependent outgrowth and F-actin flow acceleration. 5-HT treatments were accompanied by calcineurin-dependent increases in cofilin activity in the growth cone P domain. 5-HT effects were mimicked by direct activation of PLC, suggesting that increased actin network treadmilling may be a widespread mechanism for promoting neurite outgrowth in response to neurotrophic factors.
Collapse
Affiliation(s)
- Xiao-Feng Zhang
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | | | | | | |
Collapse
|
14
|
Hodgkinson GN, Tresco PA, Hlady V. The role of well-defined patterned substrata on the regeneration of DRG neuron pathfinding and integrin expression dynamics using chondroitin sulfate proteoglycans. Biomaterials 2012; 33:4288-97. [PMID: 22436802 DOI: 10.1016/j.biomaterials.2012.02.046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 02/25/2012] [Indexed: 12/13/2022]
Abstract
Injured neurons intrinsically adapt to and partially overcome inhibitory proteoglycan expression in the central nervous system by upregulating integrin expression. It remains unclear however, to what extent varying proteoglycan concentrations influence the strength of this response, how rapidly neurons adapt to proteoglycans, and how pathfinding dynamics are altered over time as integrin expression is modulated in response to proteoglycan signals. To investigate these quandaries, we created well-defined substrata in which postnatal DRG neuron pathfinding dynamics and growth cone integrin expression were interrogated as a function of proteoglycan substrata density. DRGs responded by upregulating integrin expression in a proteoglycan dose dependent fashion and exhibited robust outgrowth over all proteoglycan densities at initial time frames. However, after prolonged proteoglycan exposure, neurons exhibited decreasing velocities associated with increasing proteoglycan densities, while neurons growing on low proteoglycan levels exhibited robust outgrowth at all time points. Additionally, DRG outgrowth over proteoglycan density step boundaries, and a brief β1 integrin functional block proved that regeneration was integrin dependent and that DRGs exhibit delayed slowing and loss in persistence after even transient encounters with dense proteoglycan boundaries. These findings demonstrate the complexity of proteoglycan regulation on integrin expression and regenerative pathfinding.
Collapse
Affiliation(s)
- Gerald N Hodgkinson
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA
| | | | | |
Collapse
|
15
|
Greene AC, Washburn CM, Bachand GD, James CD. Combined chemical and topographical guidance cues for directing cytoarchitectural polarization in primary neurons. Biomaterials 2011; 32:8860-9. [DOI: 10.1016/j.biomaterials.2011.08.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 08/04/2011] [Indexed: 11/28/2022]
|
16
|
Myers JP, Santiago-Medina M, Gomez TM. Regulation of axonal outgrowth and pathfinding by integrin-ECM interactions. Dev Neurobiol 2011; 71:901-23. [PMID: 21714101 PMCID: PMC3192254 DOI: 10.1002/dneu.20931] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Developing neurons use a combination of guidance cues to assemble a functional neural network. A variety of proteins immobilized within the extracellular matrix (ECM) provide specific binding sites for integrin receptors on neurons. Integrin receptors on growth cones associate with a number of cytosolic adaptor and signaling proteins that regulate cytoskeletal dynamics and cell adhesion. Recent evidence suggests that soluble growth factors and classic axon guidance cues may direct axon pathfinding by controlling integrin-based adhesion. Moreover, because classic axon guidance cues themselves are immobilized within the ECM and integrins modulate cellular responses to many axon guidance cues, interactions between activated receptors modulate cell signals and adhesion. Ultimately, growth cones control axon outgrowth and pathfinding behaviors by integrating distinct biochemical signals to promote the proper assembly of the nervous system. In this review, we discuss our current understanding how ECM proteins and their associated integrin receptors control neural network formation.
Collapse
Affiliation(s)
- Jonathan P Myers
- Department of Neuroscience, Neuroscience Training Program, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | | | |
Collapse
|
17
|
Gardiner NJ. Integrins and the extracellular matrix: Key mediators of development and regeneration of the sensory nervous system. Dev Neurobiol 2011; 71:1054-72. [DOI: 10.1002/dneu.20950] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
18
|
Ribeiro A, Vargo S, Powell EM, Leach JB. Substrate three-dimensionality induces elemental morphological transformation of sensory neurons on a physiologic timescale. Tissue Eng Part A 2011; 18:93-102. [PMID: 21910606 DOI: 10.1089/ten.tea.2011.0221] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The natural environment of a neuron is the three-dimensional (3D) tissue. In vivo, embryonic sensory neurons transiently express a bipolar morphology with two opposing neurites before undergoing cytoplasmic and cytoskeletal rearrangement to a more mature pseudo-unipolar axonal arbor before birth. The unipolar morphology is crucial in the adult for correct information transmission from the periphery to the central nervous system. On two-dimensional (2D) substrates this transformation is delayed significantly or absent. We report that a 3D culture platform can invoke the characteristic transformation to the unipolar axonal arbor within a time frame similar to in vivo, overcoming the loss of this essential milestone in 2D substrates. Additionally, 3D substrates alone provided an environment that promoted axonal branching features that reflect morphological patterns observed in vivo. We have also analyzed the involvement of soluble cues in these morphogenic processes by culturing the neurons in the presence and absence of nerve growth factor (NGF), a molecule that plays distinct roles in the development of the peripheral and central nervous systems. Without NGF, both 2D and 3D cultures had significant decreases in the relative population of unipolar neurons as well as shorter neurite lengths and fewer branch points compared to cultures with NGF. Interestingly, branching features of neurons cultured in 3D without NGF resemble those of neurons cultured in 2D with NGF. Therefore, neurons cultured in 3D without NGF lost the ability to differentiate into unipolar neurons, suggesting that this morphological hallmark requires not only presentation of soluble cues like NGF, but also the surrounding 3D presentation of adhesive ligands to allow for realization of the innate morphogenic program. We propose that in a 3D environment, various matrix and soluble cues are presented toward all surfaces of the cell; this optimized milieu allows neurons to elaborate their genuine phenotype and follow programmed instructions that are intrinsic to the neuron, but disrupted when cells were dissected from the embryo. Thus, this study presents quantitative data supporting that 3D substrates are critical for sustaining the in vivo ontogeny of neurons and deciphering signaling mechanisms necessary for designing biomaterial scaffolds for nerve generation and repair.
Collapse
Affiliation(s)
- Andreia Ribeiro
- Department of Chemical and Biochemical Engineering, UMBC, Baltimore, Maryland 21250, USA
| | | | | | | |
Collapse
|
19
|
Peripheral Nerve Defect Repair With Epineural Tubes Supported With Bone Marrow Stromal Cells. Ann Plast Surg 2011; 67:73-84. [DOI: 10.1097/sap.0b013e318223c2db] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
20
|
Marquardt L, Willits RK. Student award winner in the undergraduate's degree category for the Society for Biomaterials 35th Annual Meeting, Orlando, Florida, April 13-16, 2011. Neurite growth in PEG gels: effect of mechanical stiffness and laminin concentration. J Biomed Mater Res A 2011; 98:1-6. [PMID: 21538826 DOI: 10.1002/jbm.a.33044] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 01/07/2011] [Indexed: 01/18/2023]
Abstract
Within a 3D environment, the chemical and mechanical properties of a scaffold can significantly influence nerve behavior. How these properties influence with nerve cells is important for optimizing neurite extension within a scaffold. The purpose of this study was to investigate the effect of low concentration poly(ethylene glycol) (PEG) with added laminin on 3D growth of dissociated dorsal root ganglia. Because of its high affinity for neurite adhesion and ability to promote extension, laminin was conjugated to the PEG chain, as well as mixed in the gel, at various concentrations to provide chemical cues. Gel stiffness, as determined by G*, significantly decreased with decreasing PEG concentration and with increasing laminin conjugate. Extension within the gels increased as the concentration of laminin increased with no difference between how laminin was presented (mixed or conjugated) to the cells. For example, in 3% PEG, extension increased from 92.29 ± 5.27 μm to 146.35 ± 13.12 μm as laminin conjugate concentration increased from plain to 100 μg/ml. Results indicated that the chemical properties of the scaffold influenced neurite growth more than the mechanical properties as laminin concentration had a greater impact on growth than the stiffness of the gel over the range studied. Neurite length as a function of scaffold stiffness and adhesion properties was also characterized and demonstrated a positive linear relationship between rate of neurite extension and laminin concentration. This study further demonstrates the importance of characterizing interactions between cell behavior and the chemical and mechanical environment.
Collapse
Affiliation(s)
- Laura Marquardt
- Department of Biomedical Engineering, Saint Louis University, St. Louis, Missouri 63130, USA
| | | |
Collapse
|
21
|
Williams KL, Mearow KM. Phosphorylation status of heat shock protein 27 influences neurite growth in adult dorsal root ganglion sensory neurons in vitro. J Neurosci Res 2011; 89:1160-72. [DOI: 10.1002/jnr.22634] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 01/19/2011] [Accepted: 01/31/2011] [Indexed: 12/15/2022]
|
22
|
Nerve growth factor induces axonal filopodia through localized microdomains of phosphoinositide 3-kinase activity that drive the formation of cytoskeletal precursors to filopodia. J Neurosci 2010; 30:12185-97. [PMID: 20826681 DOI: 10.1523/jneurosci.1740-10.2010] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The initiation of axonal filopodia is the first step in the formation of collateral branches and synaptic structures. In sensory neurons, nerve growth factor (NGF) promotes the formation of axonal filopodia and branches. However, the signaling and cytoskeletal mechanisms of NGF-induced initiation of axonal filopodia are not clear. Axonal filopodia arise from precursor axonal cytoskeletal structures termed filamentous actin (F-actin) patches. Patches form spontaneously and are transient. Although filopodia emerge from patches, only a fraction of patches normally gives rise to filopodia. Using chicken sensory neurons and live imaging of enhanced yellow fluorescent protein (eYFP)-actin dynamics, we report that NGF promotes the formation of axonal filopodia by increasing the rate of F-actin patch formation but not the fraction of patches that give rise to filopodia. We also demonstrate that activation of the phosphatidylinositol 3-kinase (PI3K)-Akt pathway is sufficient and required for driving the formation of axonal F-actin patches, filopodia, and axon branches. Using the green fluorescent protein-plekstrin homology domain of Akt, which targets to PI3K-generated phosphatidylinositol-3,4,5-triphosphate (PIP(3)), we report localized microdomains of PIP(3) accumulation that form in synchrony with F-actin patches and that NGF promotes the formation of microdomains of PIP(3) and patches. Finally, we find that, in NGF, F-actin patches form in association with axonal mitochondria and oxidative phosphorylation is required for patch formation. This investigation demonstrates that surprisingly NGF promotes formation of axonal filopodia by increasing the formation of cytoskeletal filopodial precursors (patches) through localized microdomains of PI3K signaling but not the emergence of filopodia from patches.
Collapse
|
23
|
Gilardino A, Farcito S, Zamburlin P, Audisio C, Lovisolo D. Specificity of the second messenger pathways involved in basic fibroblast growth factor-induced survival and neurite growth in chick ciliary ganglion neurons. J Neurosci Res 2010; 87:2951-62. [PMID: 19405103 DOI: 10.1002/jnr.22116] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Basic fibroblast growth factor (bFGF) exerts multiple neurotrophic actions on cultured neurons from the ciliary ganglion of chick embryo, among them promotion of neuronal survival and of neurite outgrowth. To understand the specificity of the signal transduction cascades involved in the control of these processes, we used pharmacological inhibitors of the three main effectors known to act downstream of the bFGF receptor (FGFR): phospholipase Cgamma (PLCgamma), mitogen-activated protein kinase (MAPK), and phosphatidylinositol 3-kinase (PI3-K). Neuronal survival was assessed at 24 and 48 hr; neurite growth was analyzed both on dissociated neurons and on explants of whole ganglia. Our data show that only the PI3-K pathway is involved in the survival-promoting effect of bFGF; on the other hand, all three effectors converge on the enhancement of neurite outgrowth, both on isolated neurons and in whole ganglia.
Collapse
|
24
|
Jaehrling S, Thelen K, Wolfram T, Pollerberg GE. Nanopatterns biofunctionalized with cell adhesion molecule DM-GRASP offered as cell substrate: spacing determines attachment and differentiation of neurons. NANO LETTERS 2009; 9:4115-4121. [PMID: 19694460 DOI: 10.1021/nl9023325] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The density/spacing of plasma membrane proteins is thought to be crucial for their function; clear-cut experimental evidence, however, is still rare. We examined nanopatterns biofunctionalized with cell adhesion molecule DM-GRASP with respect to their impact on neuron attachment and neurite growth. Data analysis/modeling revealed that these cellular responses improve with increasing DM-GRASP density, with the exception of one spacing which does not allow for the anchorage of a cytoskeletal protein (spectrin) to three DM-GRASP molecules.
Collapse
Affiliation(s)
- Steffen Jaehrling
- Department of Developmental Neurobiology, Institute of Zoology, University of Heidelberg, 69120 Heidelberg, Im Neuenheimer Feld 232, Germany
| | | | | | | |
Collapse
|
25
|
King M, Nafar F, Clarke J, Mearow K. The small heat shock protein Hsp27 protects cortical neurons against the toxic effects of β-amyloid peptide. J Neurosci Res 2009; 87:3161-75. [DOI: 10.1002/jnr.22145] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
26
|
Ishitani T, Ishitani S, Matsumoto K, Itoh M. Nemo-like kinase is involved in NGF-induced neurite outgrowth via phosphorylating MAP1B and paxillin. J Neurochem 2009; 111:1104-18. [PMID: 19840224 DOI: 10.1111/j.1471-4159.2009.06400.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Nerve growth factor (NGF) promotes neurite outgrowth through regulating cytoskeletal organization and cell adhesion. These activities are modulated by protein phosphorylation. Nemo-like kinase (NLK) is an evolutionarily conserved MAP kinase-like kinase that phosphorylates several transcription factors. Although NLK is known to be expressed at relatively high levels in the nervous system, its function is not well understood. We found that NGF promotes the translocation of NLK to PC12 cells' leading edges, and triggers NLK kinase activity in them. Activated NLK directly phosphorylates microtubule-associated protein-1B (MAP1B) and the focal adhesion adaptor protein, paxillin. Knockdown of NLK attenuates the phosphorylation of both paxillin and MAP1B and inhibits both the NGF-induced re-distribution of F-actin and neurite outgrowth. We also discovered that NLK is a LiCl-sensitive kinase. LiCl is known to block NGF-induced neurite outgrowth and the phosphorylation of MAP1B and paxillin in PC12 cells. Therefore, the effects of LiCl are mediated in part by blocking NLK activity. These results suggest that NLK controls the dynamics of the cytoskeleton downstream of NGF signaling.
Collapse
Affiliation(s)
- Tohru Ishitani
- Division of Cell Regulation Systems, Department of Post-Genome Science Center, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka, Japan
| | | | | | | |
Collapse
|
27
|
Harper MM, Ye EA, Blong CC, Jacobson ML, Sakaguchi DS. Integrins contribute to initial morphological development and process outgrowth in rat adult hippocampal progenitor cells. J Mol Neurosci 2009; 40:269-83. [PMID: 19499350 DOI: 10.1007/s12031-009-9211-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2008] [Accepted: 05/19/2009] [Indexed: 10/20/2022]
Abstract
Adult rat hippocampal progenitor cells (AHPCs) are self-renewing, multipotent neural progenitor cells (NPCs) that can differentiate into neurons, oligodendrocytes, and astrocytes. AHPCs contact a variety of molecular cues within their surrounding microenvironment via integrins. We hypothesize that integrin receptors are important for NPCs. In this study, we have examined the distribution of integrins in neuronal-like, oligodendrocyte-like, and astrocyte-like AHPCs when grown on substrates that support integrin-mediated adhesion (laminin, fibronectin), and those that do not (poly-L: -ornithine, PLO) using immunocytochemistry as well as characterized the phenotypic differentiation of AHPCs plated on laminin and fibronectin. Focal adhesions were prominent in AHPCs plated on purified substrates, but were also found in AHPCs plated on PLO. The focal adhesions observed in AHPCs plated on PLO substrates may be formed by self-adhesion to the endogenously produced laminin or fibronectin. We have demonstrated that integrins contribute to the initial morphological differentiation of AHPCs, as inhibition of fibronectin binding with the competitive inhibitor echistatin significantly decreased the number of processes and microspikes present in treated cells, and also decreased overall cell area. Finally, we have characterized the genetic profile of a subset of integrins and integrin-related genes in the AHPCs using reverse transcriptase polymerase chain reaction. These results demonstrate an important role of integrins, in vitro, for the initial morphological differentiation of AHPCs.
Collapse
|
28
|
Achyuta AKH, Cieri R, Unger K, Murthy SK. Synergistic effect of immobilized laminin and nerve growth factor on PC12 neurite outgrowth. Biotechnol Prog 2009; 25:227-34. [PMID: 19224594 DOI: 10.1002/btpr.58] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Immobilized extracellular matrix proteins and neurotrophins have been extensively studied to enhance neuronal adhesion and proliferation on surfaces for applications in nerve tissue engineering and neuroprosthetic devices. This article describes how the coimmobilization of laminin, an extracellular matrix protein and nerve growth factor (NGF), a neurotrophin can enhance neurite outgrowth observed separately with each type of molecule. In the absence of immobilized NGF, PC12 neurite outgrowth is influenced strongly by the presence of NGF in solution and unaffected by significant increases in laminin surface density (18.7-93.5 ng/mm(2)). However, when both laminin and NGF are immobilized together, the surface density of laminin is an important factor in determining whether or not the neurite outgrowth-promoting effect of NGF can be obtained. PC12 neurite outgrowth on surfaces with coimmobilized laminin and NGF with surface densities of 27.6 ng/mm(2) and 1.4 ng/mm(2), respectively, are similar to that observed on surfaces with immobilized laminin and dissolved NGF.
Collapse
|
29
|
Abstract
Regeneration following axonal injury of the adult peripheral sensory nervous system is heavily influenced by factors located in a neuron's extracellular environment. These factors include neurotrophins, such as Nerve Growth Factor (NGF) and the extracellular matrix, such as laminin. The presence of these molecules in the peripheral nervous system (PNS) is a major contributing factor for the dichotomy between regenerative capacities of central vs. peripheral neurons. Although PNS neurons are capable of spontaneous regeneration, this response is critically dependent on many different factors including the type, location and severity of the injury. In this article, we will focus on the plasticity of adult dorsal root ganglion (DRG) sensory neurons and how trophic factors and the extracellular environment stimulate the activation of intracellular signaling cascades that promote axonal growth in adult dorsal root ganglion neurons.
Collapse
|
30
|
Ichikawa N, Iwabuchi K, Kurihara H, Ishii K, Kobayashi T, Sasaki T, Hattori N, Mizuno Y, Hozumi K, Yamada Y, Arikawa-Hirasawa E. Binding of laminin-1 to monosialoganglioside GM1 in lipid rafts is crucial for neurite outgrowth. J Cell Sci 2009; 122:289-99. [PMID: 19118221 DOI: 10.1242/jcs.030338] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Laminin-1, an extracellular matrix molecule, promotes neurite outgrowth through the interaction of integrin and actin. Monosialoganglioside GM1 in the lipid rafts associates with and activates the NGF receptor TrkA, and enhances neurite outgrowth. However, the role of GM1 in laminin-1-induced neurite outgrowth was still unclear. Here, we describe that laminin-1 binds to GM1 through a carbohydrate moiety and a specific conformation of GM1, induces focal formation of large clusters of GM1, and enhances the relocation of TrkA in the membrane of dorsal root ganglion (DRG) and PC12 cells. We found that laminin-1-mediated clustering of GM1 causes the translocation and enrichment of beta1 integrin in lipid rafts--where TrkA colocalizes with beta1 integrin--and the activation of Lyn, Akt and MAPK to promote the outgrowth of neurites. Our results suggest that the binding of laminin-1 to GM1 facilitates the formation of a focal microdomain in the membrane, and enhances signal transduction that promotes neurite outgrowth by linking NGF-TrkA signaling with the laminin-integrin signaling pathways.
Collapse
Affiliation(s)
- Naoki Ichikawa
- Research Institute for Diseases of Old Age, Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Agnati LF, Leo G, Genedani S, Piron L, Rivera A, Guidolin D, Fuxe K. Common key-signals in learning and neurodegeneration: focus on excito-amino acids, beta-amyloid peptides and alpha-synuclein. J Neural Transm (Vienna) 2008; 116:953-74. [PMID: 19018448 DOI: 10.1007/s00702-008-0150-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2008] [Accepted: 10/20/2008] [Indexed: 01/11/2023]
Abstract
In this paper a hypothesis that some special signals ("key-signals" excito-amino acids, beta-amyloid peptides and alpha-synuclein) are not only involved in information handling by the neuronal circuits, but also trigger out substantial structural and/or functional changes in the Central Nervous System (CNS) is introduced. This forces the neuronal circuits to move from one stable state towards a new state, but in doing so these signals became potentially dangerous. Several mechanisms are put in action to protect neurons and glial cells from these potentially harmful signals. However, in agreement with the Red Queen Theory of Ageing (Agnati et al. in Acta Physiol Scand 145:301-309, 1992), it is proposed that during ageing these neuroprotective processes become less effective while, in the meantime, a shortage of brain plasticity occurs together with an increased need of plasticity for repairing the wear and tear of the CNS. The paper presents findings supporting the concept that such key-signals in instances such as ageing may favour neurodegenerative processes in an attempt of maximizing neuronal plasticity.
Collapse
Affiliation(s)
- L F Agnati
- Department of BioMedical Sciences, University of Modena, Via Campi 287, 41100 Modena, Italy.
| | | | | | | | | | | | | |
Collapse
|
32
|
Rankin SL, Guy CS, Mearow KM. Neurite outgrowth is enhanced by laminin-mediated down-regulation of the low affinity neurotrophin receptor, p75NTR. J Neurochem 2008; 107:799-813. [PMID: 18786176 DOI: 10.1111/j.1471-4159.2008.05663.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Laminin (LN), an extracellular matrix component, is a key factor in promoting axonal regeneration, coordinately regulating growth in conjunction with trophic signals provided by the neurotrophins, including nerve growth factor (NGF). This study investigated potential interactions between the LN and NGF-mediated signaling pathways in PC12 cells and primary neurons. Neurite outgrowth stimulated by NGF was enhanced on a LN substrate. Western blot analysis of pertinent signal transduction components revealed both enhanced phosphorylation of early signaling intermediates upon co-stimulation, and a LN-induced down-regulation of p75NTR which could be prevented by the addition of integrin inhibitory arginine-glycine-aspartate (RGD) peptides. This p75NTR down-regulation was associated with a LN-mediated up-regulation of PTEN and resulted in a decrease in Rho activity. Studies using over-expression or siRNA-mediated knock-down of PTEN demonstrate a consistent inverse relationship with p75NTR, and the over-expression of p75NTR impaired neurite outgrowth on a LN substrate, as well as resulting in sustained activation of Rho which is inhibitory to neurite outgrowth. p75NTR is documented for its role in the transduction of inhibitory myelin-derived signals, and our results point to extracellular matrix regulation of p75NTR as a potential mechanism to ameliorate inhibitory signaling leading to optimized neurite outgrowth.
Collapse
Affiliation(s)
- Sherri L Rankin
- Division of BioMedical Sciences, Memorial University of Newfoundland, St John's, NL, Canada
| | | | | |
Collapse
|
33
|
Ransome MI, Turnley AM. Erythropoietin promotes axonal growth in a model of neuronal polarization. Mol Cell Neurosci 2008; 38:537-47. [PMID: 18586515 DOI: 10.1016/j.mcn.2008.05.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Revised: 03/19/2008] [Accepted: 05/03/2008] [Indexed: 11/16/2022] Open
Abstract
Erythropoietin (EPO) enhances neurogenesis, neuroprotection and regeneration. Here, we examined the effects of EPO on axonal and dendritic growth in a model of neuronal polarization. EPO did not effect survival or the polarized morphology of hippocampal neurons but its effect on neurite outgrowth depended upon the stage of polarization. When added to neurons in the process of establishing polarity (0-2 days in vitro (DIV)), it enhanced axonal and dendritic growth, while EPO added to early polarized cultures at 3-4 DIV promoted the growth of axons but not dendrites. EPO stimulated the phosphorylation of Akt at serine-473 and co-incubation of the Akt/PI-3 kinase pathway inhibitor LY294002 with EPO abolished its effects on Akt phosphorylation and axonal growth. However, while Leukemia Inhibitory Factor (LIF) similarly stimulated phosphorylation of Akt, it had no effect on axonal or dendritic growth, indicating that AKT phosphorylation is necessary but not sufficient for neurite outgrowth in hippocampal neurons.
Collapse
Affiliation(s)
- Mark I Ransome
- Neural Regeneration Laboratory, Centre for Neuroscience, University of Melbourne, Melbourne, Victoria 3010, Australia
| | | |
Collapse
|
34
|
Hermann PM, Nicol JJ, Bulloch AGM, Wildering WC. RGD-dependent mechanisms in the endoneurial phagocyte response and axonal regeneration in the nervous system of the snail Lymnaea stagnalis. ACTA ACUST UNITED AC 2008; 211:491-501. [PMID: 18245625 DOI: 10.1242/jeb.013102] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Activation of phagocytic cells in the injury zone is a crucial step in the regeneration of peripheral axons. Many aspects of the mechanisms underlying the recruitment of active phagocytes remain, however, unclear. Notably, our understanding of the interactions between injury, extracellular matrix (ECM) degradation and phagocyte activation is limited. Most animal cell types, phagocytes included, interact with proteins of the ECM through one or more members of the integrin family, transmembrane cell adhesion receptors that typically bind their ligands through short linear amino acid sequences. This study focused on the role of one of the most common of such integrin recognition sequences, the Arg-Gly-Asp (RGD) motif in the recruitment and activation of endoneurial phagocytes in the injury response of the nervous system of the pond snail Lymnaea stagnalis. Like the mammalian nervous system, the Lymnaea nervous system responds to injury with recruitment and activation of endoneurial phagocytes (i.e. phagocytes residing in Lymnaea's nerves), a process involving substantial changes in the morphology, motility and adhesion status of these cells. Using synthetic water-soluble RGD-peptides, we investigated the relevance of RGD-dependent mechanisms in the activation of endoneurial phagocytes and injury response of the organ-cultured nervous system of Lymnaea. Our results show that RGD-peptides modulate various aspects of phagocyte activation (i.e. spreading response, particle engulfment, oxidative burst) in vitro and in situ and significantly affect nerve regeneration in this model system. Surprisingly, while linear RGD-analogues suppressed both phagocyte activation and axonal regeneration, a circularized RGD-peptide analogue modulated these parameters in a concentration-dependent, biphasic manner. Collectively, these results emphasize the significance of RGD-dependent mechanisms in the regenerative response of the Lymnaea nervous system and implicate regulation of the cellular immune response as one of the factors in this context.
Collapse
Affiliation(s)
- Petra M Hermann
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada, T2N 1N4
| | | | | | | |
Collapse
|
35
|
Targeting axon growth from neuronal transplants along preformed guidance pathways in the adult CNS. J Neurosci 2008; 28:340-8. [PMID: 18184776 DOI: 10.1523/jneurosci.3819-07.2008] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
To re-establish neuronal circuits lost after CNS injury, transplanted neurons must be able to extend axons toward their appropriate targets. Such growth is highly restricted within the adult CNS attributable to the expression of inhibitory molecules and general lack of guidance cues to direct axon growth. This environment typically induces random patterns of growth and aberrant innervation, if growth occurs at all. To target the growth of axons from neuronal transplants, we are using viral vectors to create guidance pathways before neuronal transplantation. In this study, we transplanted postnatal rat dorsal root ganglia neurons into the corpus callosum of adult rats. Replication-incompetent adenoviruses encoding growth or guidance factors were injected along the desired pathway 1 week before cell transplantation, allowing time for sufficient protein expression by host glial cells. With expression of nerve growth factor (NGF) and basic fibroblast growth factor, sensory axons were able to grow along the corpus callosum, across the midline, and toward an NGF-expressing target in either the contralateral striatum or cortex: a distance of 7-8 mm including a 90 degree turn from white matter into gray matter. Furthermore, expression of semaphorin 3A slightly dorsal and lateral to the turning point increased the number of axons turning into the striatal target. These results show that judicious expression of neuron-specific chemoattractant and chemorepellant molecules using viral vectors can support and target axon growth from neuronal transplants in the adult CNS.
Collapse
|
36
|
Wang Z, Gardiner NJ, Fernyhough P. Blockade of hexokinase activity and binding to mitochondria inhibits neurite outgrowth in cultured adult rat sensory neurons. Neurosci Lett 2008; 434:6-11. [PMID: 18308470 DOI: 10.1016/j.neulet.2008.01.057] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Revised: 12/19/2007] [Accepted: 01/10/2008] [Indexed: 01/03/2023]
Abstract
Hexokinase is known as the first enzyme and rate-limiting step in glycolysis. The role of hexokinase activity and localization in regulating the rate of axonal regeneration was studied in cultured adult sensory neurons of dorsal root ganglia (DRG). Immunofluorescent staining of DRG demonstrated that small-medium neurons and satellite cells exhibited high levels of expression of hexokinase I. Large neurons had negative staining for hexokinase I. Intracellular localization and biochemical studies in cultured adult rat sensory neurons revealed that hexokinase I was almost exclusively found in the mitochondrial compartment. The hypothesis that neurotrophic factor dependent activation of Akt would regulate hexokinase association with the mitochondria was tested and quantitative Western blotting showed no effect of blockade of the phosphoinositide 3-kinase (PI 3-kinase)/Akt pathway using the inhibitor LY294002, indicating this interaction of hexokinase with mitochondria was not neurotrophic factor or Akt-dependent. Finally, pharmacological blockade of hexokinase activity and inhibition of localization to the mitochondrial compartment with hexokinase II VDAC binding domain (Hxk2VBD) peptide caused a significant inhibition of neurotrophic factor-directed axon outgrowth. The results support a key role for hexokinase activity and/or localization to the mitochondria in the regulation of neurite outgrowth in cultured adult sensory neurons.
Collapse
Affiliation(s)
- Zuocheng Wang
- Division of Neurodegenerative Disorders, St Boniface Research Centre, Winnipeg, Manitoba, Canada
| | | | | |
Collapse
|
37
|
Duarte AS, Duarte EP, Correia A, Pires E, Barros MT. Cardosins improve neuronal regeneration after cell disruption: a comparative expression study. Cell Biol Toxicol 2008; 25:99-108. [DOI: 10.1007/s10565-008-9058-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2007] [Accepted: 01/03/2008] [Indexed: 11/30/2022]
|
38
|
Tucker BA, Rahimtula M, Mearow KM. Src and FAK are key early signalling intermediates required for neurite growth in NGF-responsive adult DRG neurons. Cell Signal 2007; 20:241-57. [PMID: 17999938 DOI: 10.1016/j.cellsig.2007.10.014] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Revised: 10/10/2007] [Accepted: 10/10/2007] [Indexed: 12/15/2022]
Abstract
Axonal regeneration is influenced by factors in the extracellular environment, including neurotrophins, such as NGF, and adhesion molecules, such as laminin. The provision of both NGF and a permissive substrate to cultured adult NGF-responsive DRG neurons results in enhanced levels of neurite growth not achievable by either factor alone. In this study, we have investigated the early signalling events that contribute to NGF and laminin-induced neurite growth. Adult NGF-responsive DRG neurons were plated on poly-d-lysine for 2 h then stimulated with NGF, laminin, or laminin plus NGF for 10 min, 1 h, or 6 h. Signalling pathways were subsequently analysed using Western blotting and pharmacological inhibition of specific signalling components. While activation of the various signalling intermediates (Src, FAK, Akt, MAPK) could be detected as early as 10 min-1 h after stimulation, significant neurite growth was observed mainly at the 6 h time point. The results of the time course experiments showed differential activation of the signalling intermediates. Src was activated by all treatments (NGF, laminin and the combination) at the earliest time point analysed, 10 min. NGF stimulation also resulted in detectable activation of FAK, Akt and MAPK by 10 min. However, laminin stimulation alone did not result in detectable activation of FAK, Akt or MAPK until the 1 h time point. Inhibition of either Src or FAK activity attenuated both laminin and/or NGF-induced PI 3-K/Akt and MEK/MAPK signalling pathways, as well as neurite growth. Downstream inhibition of Akt by Akt knockdown also blocked observed neurite growth, while inhibition of MEK/MAPK had no significant effect. Together, these results demonstrate that signalling underlying neurite growth can be detected within minutes of stimulation and provide a mechanism for the observed enhancement of neurite growth when both NGF and the permissive substrate, laminin, are provided.
Collapse
Affiliation(s)
- Budd A Tucker
- Schepens Eye Institute, Harvard University, Boston, MA, United States
| | | | | |
Collapse
|
39
|
Ding DC, Shyu WC, Chiang MF, Lin SZ, Chang YC, Wang HJ, Su CY, Li H. Enhancement of neuroplasticity through upregulation of β1-integrin in human umbilical cord-derived stromal cell implanted stroke model. Neurobiol Dis 2007; 27:339-53. [PMID: 17651977 DOI: 10.1016/j.nbd.2007.06.010] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Revised: 05/03/2007] [Accepted: 06/04/2007] [Indexed: 01/10/2023] Open
Abstract
Neuroplasticity subsequent to functional angiogenesis is an important goal for cell-based therapy of ischemic neural tissues. At present, the cellular and molecular mechanisms involved are still not well understood. In this study, we isolated mesenchymal stem cells (MSCs) from Wharton's jelly (WJ) to obtain clonally expanded human umbilical cord-derived mesenchymal stem cells (HUCMSCs) with multilineage differentiation potential. Experimental rats receiving intracerebral HUCMSC transplantation showed significantly improved neurological function compared to vehicle-treated control rats. Cortical neuronal activity, as evaluated by proton MR spectroscopy (1H-MRS), also increased considerably in the transplantation group. Transplanted HUCMSCs migrated towards the ischemic boundary zone and differentiated into glial, neuronal, doublecortin+, CXCR4+, and vascular endothelial cells to enhance neuroplasticity in the ischemic brain. In addition, HUCMSC transplantation promoted the formation of new vessels to increase local cortical blood flow in the ischemic hemisphere. Modulation by stem cell-derived macrophage/microglial interactions, and increased beta1-integrin expression, might enhance this angiogenic architecture within the ischemic brain. Inhibition of beta1-integrin expression blocked local angiogenesis and reduced recovery from neurological deficit. In addition, significantly increased modulation of neurotrophic factor expression was also found in the HUCMSC transplantation group. In summary, regulation of beta1-integrin expression plays a critical role in the plasticity of the ischemic brain after the implantation of HUCMSCs.
Collapse
Affiliation(s)
- Dah-Ching Ding
- Graduate Institute of Medical Science, School of Medicine, Buddhist Tzu Chi General Hospital, Tzu-Chi University, Department of Obstetrics and Gynecology, Hualien, Taiwan 970
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Paveliev M, Lume M, Velthut A, Phillips M, Arumäe U, Saarma M. Neurotrophic factors switch between two signaling pathways that trigger axonal growth. J Cell Sci 2007; 120:2507-16. [PMID: 17646673 DOI: 10.1242/jcs.003590] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Integration of multiple inputs from the extracellular environment, such as extracellular matrix molecules and growth factors, is a crucial process for cell function and information processing in multicellular organisms. Here we demonstrate that co-stimulation of dorsal root ganglion neurons with neurotrophic factors (NTFs) – glial-cell-line-derived neurotrophic factor, neurturin or nerve growth factor – and laminin leads to axonal growth that requires activation of Src family kinases (SFKs). A different, SFK-independent signaling pathway evokes axonal growth on laminin in the absence of the NTFs. By contrast, axonal branching is regulated by SFKs both in the presence and in the absence of NGF. We propose and experimentally verify a Boolean model of the signaling network triggered by NTFs and laminin. Our results demonstrate that NTFs provide an environmental cue that triggers a switch between separate pathways in the cell signaling network.
Collapse
Affiliation(s)
- Mikhail Paveliev
- Institute of Biotechnology, University of Helsinki, Helsinki FIN-00014, Finland.
| | | | | | | | | | | |
Collapse
|
41
|
Rex CS, Lin CY, Kramár EA, Chen LY, Gall CM, Lynch G. Brain-derived neurotrophic factor promotes long-term potentiation-related cytoskeletal changes in adult hippocampus. J Neurosci 2007; 27:3017-29. [PMID: 17360925 PMCID: PMC6672589 DOI: 10.1523/jneurosci.4037-06.2007] [Citation(s) in RCA: 255] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is an extremely potent, positive modulator of theta burst induced long-term potentiation (LTP) in the adult hippocampus. The present studies tested whether the neurotrophin exerts its effects by facilitating cytoskeletal changes in dendritic spines. BDNF caused no changes in phalloidin labeling of filamentous actin (F-actin) when applied alone to rat hippocampal slices but markedly enhanced the number of densely labeled spines produced by a threshold level of theta burst stimulation. Conversely, the BDNF scavenger TrkB-Fc completely blocked increases in spine F-actin produced by suprathreshold levels of theta stimulation. TrkB-Fc also blocked LTP consolidation when applied 1-2 min, but not 10 min, after theta trains. Additional experiments confirmed that p21 activated kinase and cofilin, two actin-regulatory proteins implicated in spine morphogenesis, are concentrated in spines in mature hippocampus and further showed that both undergo rapid, dose-dependent phosphorylation after infusion of BDNF. These results demonstrate that the influence of BDNF on the actin cytoskeleton is retained into adulthood in which it serves to positively modulate the time-dependent LTP consolidation process.
Collapse
Affiliation(s)
| | | | - Eniko A. Kramár
- Psychiatry and Human Behavior, University of California, Irvine, Irvine, California 92697-4292
| | | | - Christine M. Gall
- Departments of Neurobiology and Behavior
- Anatomy and Neurobiology, and
| | - Gary Lynch
- Psychiatry and Human Behavior, University of California, Irvine, Irvine, California 92697-4292
| |
Collapse
|
42
|
Yang T, Massa SM, Longo FM. LAR protein tyrosine phosphatase receptor associates with TrkB and modulates neurotrophic signaling pathways. ACTA ACUST UNITED AC 2007; 66:1420-36. [PMID: 17013927 DOI: 10.1002/neu.20291] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The identities of receptor protein tyrosine phosphatases (PTPs) that associate with Trk protein tyrosine kinase (PTK) receptors and modulate neurotrophic signaling are unknown. The leukocyte common antigen-related (LAR) receptor PTP is present in neurons expressing TrkB, and like TrkB is associated with caveolae and regulates survival and neurite outgrowth. We tested the hypothesis that LAR associates with TrkB and regulates neurotrophic signaling in embryonic hippocampal neurons. Coimmunoprecipitation and coimmunostaining demonstrated LAR interaction with TrkB that is increased by BDNF exposure. BDNF neurotrophic activity was reduced in LAR-/- and LAR siRNA-treated LAR+/+ neurons and was augmented in LAR-transfected neurons. In LAR-/- neurons, BDNF-induced activation of TrkB, Shc, AKT, ERK, and CREB was significantly decreased; while in LAR-transfected neurons, BDNF-induced CREB activation was augmented. Similarly, LAR+/+ neurons treated with LAR siRNA demonstrated decreased activation of Trk and AKT. LAR is known to activate the Src PTK by dephosphorylation of its negative regulatory domain and Src transactivates Trk. In LAR-/- neurons, or neurons treated with LAR siRNA, phosphorylation of the Src regulatory domain was increased (indicating Src inactivation), consistent with a role for Src in mediating LAR's ability to up-regulate neurotrophic signaling. Interactions between LAR, TrkB, and Src were further confirmed by the findings that Src coimmunoprecipitated with LAR, that the Src inhibitor PP2 blocked the ability of LAR to augment TrkB signaling, and that siRNA-induced depletion of Src decreased LAR interaction with TrkB. These studies demonstrate that receptor PTPs can associate with Trk complexes and promote neurotrophic signaling and point to receptor PTP-based strategies as a novel approach for modulating neurotrophin function.
Collapse
MESH Headings
- Animals
- Blotting, Western/methods
- Brain-Derived Neurotrophic Factor/metabolism
- Brain-Derived Neurotrophic Factor/pharmacology
- Cell Survival/drug effects
- Cells, Cultured
- Culture Media, Conditioned/pharmacology
- Dose-Response Relationship, Drug
- Embryo, Mammalian
- Enzyme Inhibitors/pharmacology
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/genetics
- Gene Expression Regulation/physiology
- Hippocampus/cytology
- Immunoprecipitation/methods
- Mice
- Mice, Knockout
- Mutation/physiology
- Nerve Tissue Proteins/immunology
- Nerve Tissue Proteins/physiology
- Neurons/drug effects
- Neurons/metabolism
- Phosphorylation/drug effects
- Protein Tyrosine Phosphatases/chemistry
- Protein Tyrosine Phosphatases/deficiency
- Protein Tyrosine Phosphatases/genetics
- Protein Tyrosine Phosphatases/immunology
- Protein Tyrosine Phosphatases/physiology
- Pyrimidines/pharmacology
- RNA, Small Interfering/pharmacology
- Receptor, trkB/physiology
- Receptor-Like Protein Tyrosine Phosphatases, Class 2
- Receptor-Like Protein Tyrosine Phosphatases, Class 4
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/deficiency
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/physiology
- Signal Transduction/physiology
- Time Factors
- Transfection/methods
- Tyrosine/metabolism
Collapse
Affiliation(s)
- Tao Yang
- Department of Neurology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | |
Collapse
|
43
|
Tucker BA, Rahimtula M, Mearow KM. Laminin and growth factor receptor activation stimulates differential growth responses in subpopulations of adult DRG neurons. Eur J Neurosci 2006; 24:676-90. [PMID: 16930399 DOI: 10.1111/j.1460-9568.2006.04963.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neurons in the adult rat dorsal root ganglion (DRG) can be classified into at least three separate subpopulations based on morphologic and phenotypic differences. In this study we have focused on the growth response of these specific subpopulations in vitro with respect to laminin (LN) and growth factor receptor activation. Using a cell selection approach we show that LN-induced neurite growth occurs in the absence of added trophic factors only in heavy-chain neurofilament-positive and calcitonin gene-related peptide-positive DRG neurons [nerve growth factor (NGF)-responsive population]. In contrast, LN alone is not sufficient to stimulate significant neurite growth from lectin Griffonia simplicifolia IB4-positive neurons (IB4+ve), although it is still required to elicit a growth response from these cells in the presence of glial-derived neurotrophic factor (GDNF, e.g. neurite growth occurred only when cells were plated on LN in the presence of GDNF). By using chemical inhibitors we demonstrate that only the phosphatidylinositol 3 kinase (PI 3-K)/Akt pathway is required for neurite growth from the NGF-responsive cell population. However, both the PI 3-K/Akt and MEK/mitogen-activated protein kinase signaling pathways are required for neurite growth from the IB4+ve cell population. Thus, we have identified specific signaling events and environmental requirements associated with neurite growth for different subpopulations of adult DRG neurons, pointing to potential therapeutic targets while identifying an inability for any one treatment alone to repair peripheral nerve damage.
Collapse
MESH Headings
- Animals
- Calcitonin Gene-Related Peptide/metabolism
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Cells, Cultured
- Enzyme Inhibitors/pharmacology
- Ganglia, Spinal/cytology
- Ganglia, Spinal/drug effects
- Ganglia, Spinal/metabolism
- Glial Cell Line-Derived Neurotrophic Factor/metabolism
- Glial Cell Line-Derived Neurotrophic Factor/pharmacology
- Growth Cones/drug effects
- Growth Cones/metabolism
- Growth Cones/ultrastructure
- Immunohistochemistry
- Laminin/metabolism
- Laminin/pharmacology
- MAP Kinase Signaling System/drug effects
- MAP Kinase Signaling System/physiology
- Nerve Regeneration/drug effects
- Nerve Regeneration/physiology
- Neurites/drug effects
- Neurites/metabolism
- Neurites/ultrastructure
- Neurofilament Proteins/drug effects
- Neurofilament Proteins/metabolism
- Neurons, Afferent/cytology
- Neurons, Afferent/drug effects
- Neurons, Afferent/metabolism
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphoinositide-3 Kinase Inhibitors
- Plant Lectins/metabolism
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Growth Factor/agonists
- Receptors, Growth Factor/metabolism
Collapse
Affiliation(s)
- Budd A Tucker
- Division of Basic Medical Sciences, Memorial University of Newfoundland, St. John's, NL, Canada A1B 3V6
| | | | | |
Collapse
|
44
|
Williams KL, Rahimtula M, Mearow KM. Heat shock protein 27 is involved in neurite extension and branching of dorsal root ganglion neurons in vitro. J Neurosci Res 2006; 84:716-23. [PMID: 16862544 DOI: 10.1002/jnr.20983] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Alteration of the cytoskeleton in response to growth factors and extracellular matrix proteins is necessary for neurite growth. The cytoskeletal components, such as actin and tubulin, can be modified through interaction with other cellular proteins, including the small heat shock protein Hsp27. Our previous work suggested that Hsp27 influences neurite growth, potentially via its phosphorylation state interactions with actin. To investigate further the role of Hsp27 in neurite outgrowth of adult dorsal root ganglion (DRG) neurons, we have both down-regulated endogenous Hsp27 and expressed exogenous Hsp27. Down-regulation of Hsp27 with Hsp27 siRNA resulted in a decrease of neuritic tree length and complexity. In contrast, expression of exogenous Hsp27 in these neurons resulted in an increase in neuritic tree length and branching. Collectively, these results demonstrate that Hsp27 may play a role in neuritic growth via modulation of the actin cytoskeleton.
Collapse
Affiliation(s)
- Kristy L Williams
- Division of Basic Medical Sciences, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | | | | |
Collapse
|
45
|
Arthur DB, Akassoglou K, Insel PA. P2Y2 and TrkA receptors interact with Src family kinase for neuronal differentiation. Biochem Biophys Res Commun 2006; 347:678-82. [PMID: 16842754 DOI: 10.1016/j.bbrc.2006.06.141] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2006] [Accepted: 06/22/2006] [Indexed: 10/24/2022]
Abstract
The crosstalk between the P2Y(2) G-protein-coupled receptor (GPCR) with TrkA receptor tyrosine kinase (RTK) is an important mechanism that regulates neuronal differentiation. We show that Src family kinases (SFK) regulate P2Y(2)-TrkA molecular crosstalk. SFK inhibitors block ATPgammaS/P2Y(2)-promoted enhancement of NGF/TrkA signaling and neuronal differentiation in PC12 cells, abrogate the enhancement by ATPgammaS of neurite outgrowth in primary cultures of dorsal root ganglion neurons, and block co-immunoprecipitation of TrkA, P2Y(2) receptors and SFK. These results identify SFK as mediating nucleotide-enhanced neurotrophin-dependent neuronal differentiation and thus, as a key convergence point for interaction between RTKs and GPCRs.
Collapse
Affiliation(s)
- David B Arthur
- Department of Pharmacology, University of California, San Diego, La Jolla, 92093, USA
| | | | | |
Collapse
|
46
|
Shyu WC, Lin SZ, Chiang MF, Su CY, Li H. Intracerebral peripheral blood stem cell (CD34+) implantation induces neuroplasticity by enhancing beta1 integrin-mediated angiogenesis in chronic stroke rats. J Neurosci 2006; 26:3444-53. [PMID: 16571751 PMCID: PMC6673856 DOI: 10.1523/jneurosci.5165-05.2006] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Although stem cell-based treatments for stroke and other neurodegenerative diseases have advanced rapidly, there are still few clinical treatments available. In this study, rats receiving intracerebral peripheral blood hematopoietic stem cell (CD34+) (PBSC) transplantation showed much more improvement in neurological function after chronic cerebral ischemia in comparison with vehicle-treated control rats. Using laser-scanning confocal microscopy, implanted PBSCs were seen to differentiate into glial cells [GFAP+ (glial fibrillary acidic protein-positive)], neurons [Nestin+, MAP-2+ (microtubule-associated protein 2-positive), Neu-N+ (neuronal nuclear antigen-positive)], and vascular endothelial cells [vWF+ (von Willebrand factor-positive)], thereby enhancing neuroplastic effects in the ischemic brain. Cortical neuronal activity, as evaluated by 1H-MRS (proton magnetic resonance spectroscopy), also increased considerably in PBSC-treated rats compared with a vehicle-treated control group. In addition, PBSC implantation promoted the formation of new vessels, thereby increasing the local cortical blood flow in the ischemic hemisphere. These observations may be explained by the involvement of stem cell-derived macrophage/microglial cells, and beta1 integrin expression, which might enhance this angiogenic architecture over the ischemic brain. Furthermore, quantitative reverse transcription-PCR analysis showed significantly increased modulation of neurotrophic factor expression in the ischemic hemisphere of the PBSC-transplanted rats compared with vehicle-treated control rats. Thus, intracerebral PBSC transplantation might have potential as a therapeutic strategy for treating cerebrovascular diseases.
Collapse
|
47
|
Drummond HA, Furtado MM, Myers S, Grifoni S, Parker KA, Hoover A, Stec DE. ENaC proteins are required for NGF-induced neurite growth. Am J Physiol Cell Physiol 2006; 290:C404-10. [PMID: 16192303 DOI: 10.1152/ajpcell.00210.2005] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neurite growth is required for nervous system development and repair. Multiple signals, including neurotrophic factors and intact mechanosensing mechanisms, interact to regulate neurite growth. Degenerin/epithelial Na+channel (DEG/ENaC) proteins have been identified as putative mechanosensors in sensory neurons. Recently, others have shown that the neurotrophic factor NGF stimulates expression of acid-sensing ion channel molecules, which are members of the DEG/ENaC family. However, it is unknown whether NGF regulates ENaC expression or whether ENaC expression is required for neurite formation. Therefore, the aims of the present study were to determine whether ENaC expression is 1) regulated by NGF and 2) required for NGF-induced neurite growth in pheochromocytoma PC-12 cells. We found NGF-induced expression of β- and γ-subunits of ENaC, but not α-ENaC. Tyrosine kinase A (TrkA) receptor blockade abolished NGF-induced β- and γ-ENaC expression and neurite formation. NGF-induced neurite formation was inhibited by disruption of ENaC expression using 1) pharmacological blockade with benzamil, a specific ENaC inhibitor; 2) small interfering RNA; and 3) dominant-negative ENaC molecules. These data indicate NGF-TrkA regulation of ENaC expression may be required for neurite growth and may suggest a novel role for DEG/ENaC proteins in neuronal remodeling and differentiation.
Collapse
Affiliation(s)
- Heather A Drummond
- Dept. of Physiology and Biophysics, Univ. of Mississippi Medical Center, 2500 North State St., N615, Jackson, MS 39216, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Yang T, Yin W, Derevyanny VD, Moore LA, Longo FM. Identification of an ectodomain within the LAR protein tyrosine phosphatase receptor that binds homophilically and activates signalling pathways promoting neurite outgrowth. Eur J Neurosci 2006; 22:2159-70. [PMID: 16262654 DOI: 10.1111/j.1460-9568.2005.04403.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Elucidation of mechanisms by which receptor protein tyrosine phosphatases (PTPs) regulate neurite outgrowth will require characterization of ligand-receptor interactions and identification of ligand-induced signalling components mediating neurite outgrowth. The first identified ligand of the leucocyte common antigen-related (LAR) receptor PTP consists of a 99-residue ectodomain isoform, termed LARFN5C, which undergoes homophilic binding to LAR and promotes neurite outgrowth. We employed peptide mapping of LARFN5C to identify an active neurite-promoting domain of LAR. A peptide mimetic consisting of 37 residues (L59) and corresponding to the fifth LAR fibronectin type III (FNIII) domain prevented LARFN5C homophilic binding, demonstrated homophilic binding to itself and promoted neurite outgrowth of mouse E16-17 hippocampal neurons and of dorsal root ganglia explants. Response to L59 was partially lost when using neurons derived from LAR-deficient (-/-) mice or neurons treated with LAR siRNA, consistent with homophilic interaction of L59 with LAR. L59 neurite-promoting activity was decreased in the presence of inhibitors of Src, Trk, PLCgamma, PKC, PI3K and MAPK. L59 activated Src (a known substrate of LAR), FAK and TrkB and also activated downstream signalling intermediates including PKC, ERK, AKT and CREB. BDNF augmented the maximal neurite-promoting activity of L59, a finding consistent with the presence of shared and distinct signalling pathways activated by L59 with BDNF and L59 with TrkB. These studies are the first to identify an ectodomain of LAR (located within the fifth FNIII domain) capable of promoting neurite outgrowth and point to novel approaches for promotion of neurite outgrowth.
Collapse
Affiliation(s)
- Tao Yang
- Department of Neurology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
49
|
Williams KL, Rahimtula M, Mearow KM. Hsp27 and axonal growth in adult sensory neurons in vitro. BMC Neurosci 2005; 6:24. [PMID: 15819993 PMCID: PMC1087488 DOI: 10.1186/1471-2202-6-24] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2004] [Accepted: 04/08/2005] [Indexed: 11/10/2022] Open
Abstract
Background Neurite growth can be elicited by growth factors and interactions with extracellular matrix molecules like laminin. Among the targets of the signalling pathways activated by these stimuli are cytoskeletal elements, such as actin, tubulin and neurofilaments. The cytoskeleton can also be modulated by other proteins, such as the small heat shock protein Hsp27. Hsp27 interacts with actin and tubulin in non-neuronal cells and while it has been suggested to play a role in the response of some neurons to injury, there have been no direct studies of its contribution to axonal regeneration. Results We have investigated neurite initiation and process extension using cultures of adult dorsal root ganglion (DRG) sensory neurons and a laminin stimulation paradigm. Employing confocal microscopy and biochemical analyses we have examined localization of Hsp27 at early and later stages of neurite growth. Our results show that Hsp27 is colocalized with actin and tubulin in lamellopodia, filopodia, focal contacts and mature neurites and growth cones. Disruption of the actin cytoskeleton with cytochalasin D results in aberrant neurite initiation and extension, effects which may be attributable to alterations in actin polymerization states. Inhibition of Hsp27 phosphorylation in our cultures results in an atypical growth pattern that may be attributable to an effect of pHsp27 on the stability of the actin cytoskeleton. Conclusion We observed colocalization of the phosphorylated and non-phosphorylated forms of Hsp27 with actin and tubulin in both very early and later stages of neurite growth from cultured adult DRG neurons. The colocalization of Hsp27 and pHsp27 with actin in lamellopodia and focal contacts at early stages of neurite growth, and in processes, branch points and growth cones at later stages, suggests that Hsp27 may play a role in neuritogenesis and subsequent neurite extension, and potentially in the patterning of this growth. Hsp27 has been reported to play a key role in modulating actin cytoskeletal dynamics as an actin-capping protein in non-neuronal cells. Our results suggest that this may also be the case in neurons and support a role for Hsp27 in neurite outgrowth via its phosphorylation state-dependent interactions with actin.
Collapse
Affiliation(s)
- Kristy L Williams
- Division of Basic Medical Sciences, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Masuma Rahimtula
- Division of Basic Medical Sciences, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Karen M Mearow
- Division of Basic Medical Sciences, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| |
Collapse
|