51
|
Solis GP, Hülsbusch N, Radon Y, Katanaev VL, Plattner H, Stuermer CAO. Reggies/flotillins interact with Rab11a and SNX4 at the tubulovesicular recycling compartment and function in transferrin receptor and E-cadherin trafficking. Mol Biol Cell 2013; 24:2689-702. [PMID: 23825023 PMCID: PMC3756921 DOI: 10.1091/mbc.e12-12-0854] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
In this study reggie-1/flotillin-2 is identified as a component of the tubulovesicular sorting and recycling compartment, where it interacts with and controls the activity of Rab11a and SNX4. Evidence is given that reggie-1 expression is necessary for the proper recycling of transferrin receptor and E-cadherin in HeLa and A431 cells, respectively. The lipid raft proteins reggie-1 and -2 (flotillins) are implicated in membrane protein trafficking but exactly how has been elusive. We find that reggie-1 and -2 associate with the Rab11a, SNX4, and EHD1–decorated tubulovesicular recycling compartment in HeLa cells and that reggie-1 directly interacts with Rab11a and SNX4. Short hairpin RNA–mediated down-regulation of reggie-1 (and -2) in HeLa cells reduces association of Rab11a with tubular structures and impairs recycling of the transferrin–transferrin receptor (TfR) complex to the plasma membrane. Overexpression of constitutively active Rab11a rescues TfR recycling in reggie-deficient HeLa cells. Similarly, in a Ca2+ switch assay in reggie-depleted A431 cells, internalized E-cadherin is not efficiently recycled to the plasma membrane upon Ca2+ repletion. E-cadherin recycling is rescued, however, by overexpression of constitutively active Rab11a or SNX4 in reggie-deficient A431 cells. This suggests that the function of reggie-1 in sorting and recycling occurs in association with Rab11a and SNX4. Of interest, impaired recycling in reggie-deficient cells leads to de novo E-cadherin biosynthesis and cell contact reformation, showing that cells have ways to compensate the loss of reggies. Together our results identify reggie-1 as a regulator of the Rab11a/SNX4-controlled sorting and recycling pathway, which is, like reggies, evolutionarily conserved.
Collapse
Affiliation(s)
- Gonzalo P Solis
- Department of Biology, University of Konstanz, 78467 Konstanz, Germany.
| | | | | | | | | | | |
Collapse
|
52
|
Monje FJ, Divisch I, Demit M, Lubec G, Pollak DD. Flotillin-1 is an evolutionary-conserved memory-related protein up-regulated in implicit and explicit learning paradigms. Ann Med 2013; 45:301-7. [PMID: 23631399 PMCID: PMC3681089 DOI: 10.3109/07853890.2013.770637] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Studies of synaptic plasticity using the marine mollusk Aplysia californica as model system have been successfully used to identify proteins involved in learning and memory. The importance of molecular elements regulated by the learning- related neurotransmitter serotonin in Aplysia can then be explored in rodent models and finally tested for their relevance for human physiology and pathology. MATERIALS AND METHODS Herein, 2-DE gel-based electrophoresis has been used to investigate protein level changes after treatment with serotonin in Aplysia abdominal ganglia. RESULTS Twenty-one proteins have been found to be regulated by serotonin, and protein level changes of actin depolymerizing factor (ADF), deleted in azoospermia associated protein (DAZAP-1), and Flotillin-1 have been verified by Western blotting. DISCUSSION Flotillin-1, a member of the flotillin/reggie family of scaffolding proteins, has been previously found to be involved in neuritic branching and synapse formation in hippocampal neurons in vitro. However, its importance for hippocampal- dependent learning and memory in the mouse has not been examined. Here, elevated levels of Flotillin-1 in hippocampal tissue of mice trained in the Morris water maze confirmed the relevance of Flotillin-1 for memory-related processes in a mammalian system. Thus, a translational approach-from invertebrates to rodents-led to the identification of Flotillin-1 as evolutionary-conserved memory-related protein.
Collapse
Affiliation(s)
- Francisco J Monje
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Austria
| | | | | | | | | |
Collapse
|
53
|
Koch JC, Solis GP, Bodrikov V, Michel U, Haralampieva D, Shypitsyna A, Tönges L, Bähr M, Lingor P, Stuermer CA. Upregulation of reggie-1/flotillin-2 promotes axon regeneration in the rat optic nerve in vivo and neurite growth in vitro. Neurobiol Dis 2013; 51:168-76. [DOI: 10.1016/j.nbd.2012.11.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 10/14/2012] [Accepted: 11/09/2012] [Indexed: 01/23/2023] Open
|
54
|
Wnt secretion and gradient formation. Int J Mol Sci 2013; 14:5130-45. [PMID: 23455472 PMCID: PMC3634490 DOI: 10.3390/ijms14035130] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 02/20/2013] [Accepted: 02/22/2013] [Indexed: 12/14/2022] Open
Abstract
Concentration gradients formed by the lipid-modified morphogens of the Wnt family are known for their pivotal roles during embryogenesis and adult tissue homeostasis. Wnt morphogens are also implicated in a variety of human diseases, especially cancer. Therefore, the signaling cascades triggered by Wnts have received considerable attention during recent decades. However, how Wnts are secreted and how concentration gradients are formed remains poorly understood. The use of model organisms such as Drosophila melanogaster has provided important advances in this area. For instance, we have previously shown that the lipid raft-associated reggie/flotillin proteins influence Wnt secretion and spreading in Drosophila. Our work supports the notion that producing cells secrete Wnt molecules in at least two pools: a poorly diffusible one and a reggie/flotillin-dependent highly diffusible pool which allows morphogen spreading over long distances away from its source of production. Here we revise the current views of Wnt secretion and spreading, and propose two models for the role of the reggie/flotillin proteins in these processes: (i) reggies/flotillins regulate the basolateral endocytosis of the poorly diffusible, membrane-bound Wnt pool, which is then sorted and secreted to apical compartments for long-range diffusion, and (ii) lipid rafts organized by reggies/flotillins serve as “dating points” where extracellular Wnt transiently interacts with lipoprotein receptors to allow its capture and further spreading via lipoprotein particles. We further discuss these processes in the context of human breast cancer. A better understanding of these phenomena may be relevant for identification of novel drug targets and therapeutic strategies.
Collapse
|
55
|
Kamioka Y, Fujikawa C, Ogai K, Sugitani K, Watanabe S, Kato S, Wakasugi K. Functional characterization of fish neuroglobin: zebrafish neuroglobin is highly expressed in amacrine cells after optic nerve injury and can translocate into ZF4 cells. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:1779-88. [PMID: 23481873 DOI: 10.1016/j.bbapap.2013.02.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Revised: 01/31/2013] [Accepted: 02/13/2013] [Indexed: 01/08/2023]
Abstract
Neuroglobin (Ngb) is a recently discovered vertebrate heme protein that is expressed in the brain and can reversibly bind oxygen. Mammalian Ngb is involved in neuroprotection under conditions of oxidative stress, such as ischemia and reperfusion. We previously found that zebrafish Ngb can penetrate the mammalian cell membrane. In the present study, we investigated the functional characteristics of fish Ngb by using the zebrafish cell line ZF4 and zebrafish retina. We found that zebrafish Ngb translocates into ZF4 cells, but cannot protect ZF4 cells against cell death induced by hydrogen peroxide. Furthermore, we demonstrated that a chimeric ZHHH Ngb protein, in which module M1 of human Ngb is replaced by that of zebrafish, is a cell-membrane-penetrating protein that can protect ZF4 cells against hydrogen peroxide exposure. Moreover, we investigated the localization of Ngb mRNA and protein in zebrafish retina and found that Ngb mRNA is expressed in amacrine cells in the inner nuclear layer and is significantly increased in amacrine cells 3days after optic nerve injury. Immunohistochemical studies clarified that Ngb protein levels were increased in both amacrine cells and presynaptic regions in the inner plexiform layer after nerve injury. Taken together, we hypothesize that fish Ngb, whose expression is upregulated in amacrine cells after optic nerve injury, might be released from amacrine cells, translocate into neighboring ganglion cells, and function in the early stage of optic nerve regeneration. This article is part of a Special Issue entitled: Oxygen Binding and Sensing Proteins.
Collapse
Affiliation(s)
- Yuki Kamioka
- Department of Life Sciences, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
56
|
Zou S, Tian C, Ge S, Hu B. Neurogenesis of retinal ganglion cells is not essential to visual functional recovery after optic nerve injury in adult zebrafish. PLoS One 2013; 8:e57280. [PMID: 23437359 PMCID: PMC3577741 DOI: 10.1371/journal.pone.0057280] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Accepted: 01/18/2013] [Indexed: 11/24/2022] Open
Abstract
Zebrafish central nervous system (CNS) possesses a strong neural regeneration ability to restore visual function completely after optic nerve injury (ONI). However, whether neurogenesis of retinal ganglion cell (RGC) contributes to functional recovery remains controversial. Our quantitative analysis of RGCs in different ONI models showed that almost all RGCs survived in optic nerve crush (ONC) model; while over 90% of RGCs survived in the first 2 weeks with 75% remaining after 7 weeks in optic nerve transection (ONT) model. Retrograde labeling from tectum revealed a surprising regeneration rate, with over 90% and over 50% of RGCs regrowing axons to tectum at the first week in ONC and ONT model respectively. In the latter one, the number of regenerative RGCs after 4 weeks had no significant difference from the control group. As for neurogenesis, newborn RGCs were rarely detected either by double retrograde labeling or BrdU marker. Since few RGCs died, microglia number showed a temporary increase at 3 days post injury (dpi) and a decrease at 14 dpi. Finally, myelin structure within retina kept integrity and optomotor response (OMR) test demonstrated visual functional restoration at 5 weeks post injury (wpi). In conclusion, our results have directly shown that RGC survival and axon regrowth are responsible for functional recovery after ONI in adult zebrafish.
Collapse
Affiliation(s)
- Suqi Zou
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Chen Tian
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Shuchao Ge
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Bing Hu
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, P. R. China
- * E-mail:
| |
Collapse
|
57
|
Thy-1-Interacting Molecules and Cellular Signaling in Cis and Trans. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 305:163-216. [DOI: 10.1016/b978-0-12-407695-2.00004-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
58
|
Banning A, Ockenga W, Finger F, Siebrasse P, Tikkanen R. Transcriptional regulation of flotillins by the extracellularly regulated kinases and retinoid X receptor complexes. PLoS One 2012; 7:e45514. [PMID: 23029064 PMCID: PMC3445523 DOI: 10.1371/journal.pone.0045514] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 08/21/2012] [Indexed: 11/25/2022] Open
Abstract
Flotillin-1 and flotillin-2 are important regulators of signal transduction pathways such as growth factor signaling. Flotillin expression is increased under pathological conditions such as neurodegenerative disorders and cancer. Despite their importance for signal transduction, very little is known about the transcriptional regulation of flotillins. Here, we analyzed the expression of flotillins at transcriptional level and identified flotillins as downstream targets of the mitogen activated kinases ERK1/2. The promoter activity of flotillins was increased upon growth factor stimulation in a MAPK dependent manner. Overexpression of serum response factor or early growth response gene 1 resulted in increased flotillin mRNA and protein expression. Furthermore, both promoter activity and expression of endogenous flotillins were increased upon treatment with retinoic acid or by overexpression of the retinoid X receptor and its binding partners RARα and PPARγ. Our data indicate that the expression of flotillins, which can be detected in all cultured cells, is fine-tuned in response to various external stimuli. This regulation may be critical for the outcome of signaling cascades in which flotillins are known to be involved.
Collapse
Affiliation(s)
- Antje Banning
- Institute of Biochemistry, Medical Faculty, University of Giessen, Giessen, Germany
| | - Wymke Ockenga
- Institute of Biochemistry, Medical Faculty, University of Giessen, Giessen, Germany
| | - Fabian Finger
- Institute of Biochemistry, Medical Faculty, University of Giessen, Giessen, Germany
| | - Philipp Siebrasse
- Institute of Biochemistry, Medical Faculty, University of Giessen, Giessen, Germany
| | - Ritva Tikkanen
- Institute of Biochemistry, Medical Faculty, University of Giessen, Giessen, Germany
- * E-mail:
| |
Collapse
|
59
|
Santiago JM, Torrado AI, Arocho LC, Rosas OR, Rodríguez AE, Toro FK, Salgado IK, Torres YA, Silva WI, Miranda JD. Expression profile of flotillin-2 and its pathophysiological role after spinal cord injury. J Mol Neurosci 2012; 49:347-59. [PMID: 22878913 DOI: 10.1007/s12031-012-9873-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 08/01/2012] [Indexed: 11/26/2022]
Abstract
Some receptors that block axonal regeneration or promote cell death after spinal cord injury (SCI) are localized in membrane rafts. Flotillin-2 (Flot-2) is an essential protein associated with the formation of these domains and the clustering of membranal proteins, which may have signaling activities. Our hypothesis is that trauma will change Flot-2 expression and interference of this lipid raft marker will promote functional locomotor recovery after SCI. Analyses were conducted to determine the spatiotemporal profile of Flot-2 expression in adult rats after SCI, using the MASCIS impactor device. Immunoblots showed that SCI produced a significant decrease in the level of Flot-2 at 2 days post-injury (DPI) that increased until 28 DPI. Confocal microscopy revealed Flot-2 expression in neurons, reactive astrocytes and oligodendrocytes specifically associated to myelin structures near or close to the axons of the cord. In the open field test and grid walking assays, to monitor locomotor recovery of injured rats infused intrathecally with Flot-2 antisense oligonucleotides for 28 days showed significant behavioral improvement at 14, 21 and 28 DPI. These findings suggest that Flot-2 has a role in the nonpermissive environment that blocks locomotor recovery after SCI by clustering unfavorable proteins in membrane rafts.
Collapse
Affiliation(s)
- José M Santiago
- Department of Natural Sciences, University of Puerto Rico Carolina Campus, Carolina, 00984, Puerto Rico
| | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Hicks DA, Nalivaeva NN, Turner AJ. Lipid rafts and Alzheimer's disease: protein-lipid interactions and perturbation of signaling. Front Physiol 2012; 3:189. [PMID: 22737128 PMCID: PMC3381238 DOI: 10.3389/fphys.2012.00189] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 05/21/2012] [Indexed: 12/16/2022] Open
Abstract
Lipid rafts are membrane domains, more ordered than the bulk membrane and enriched in cholesterol and sphingolipids. They represent a platform for protein-lipid and protein–protein interactions and for cellular signaling events. In addition to their normal functions, including membrane trafficking, ligand binding (including viruses), axonal development and maintenance of synaptic integrity, rafts have also been implicated in the pathogenesis of several neurodegenerative diseases including Alzheimer’s disease (AD). Lipid rafts promote interaction of the amyloid precursor protein (APP) with the secretase (BACE-1) responsible for generation of the amyloid β peptide, Aβ. Rafts also regulate cholinergic signaling as well as acetylcholinesterase and Aβ interaction. In addition, such major lipid raft components as cholesterol and GM1 ganglioside have been directly implicated in pathogenesis of the disease. Perturbation of lipid raft integrity can also affect various signaling pathways leading to cellular death and AD. In this review, we discuss modulation of APP cleavage by lipid rafts and their components, while also looking at more recent findings on the role of lipid rafts in signaling events.
Collapse
Affiliation(s)
- David A Hicks
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds Leeds, UK
| | | | | |
Collapse
|
61
|
Otto GP, Nichols BJ. The roles of flotillin microdomains--endocytosis and beyond. J Cell Sci 2012; 124:3933-40. [PMID: 22194304 DOI: 10.1242/jcs.092015] [Citation(s) in RCA: 195] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Flotillins are membrane proteins that form microdomains in the plasma membrane of all mammalian cell types studied to date. They span the evolutionary spectrum, with proteins related to flotillins present in bacteria, fungi, plants and metazoans, which suggests that they perform important, and probably conserved, functions. Flotillins have been implicated in myriad processes that include endocytosis, signal transduction and regulation of the cortical cytoskeleton, yet the molecular mechanisms that underlie flotillin function in these different cases are still poorly understood. In this Commentary, we will provide an introduction to these intriguing proteins, summarise their proposed functions and discuss in greater detail some recent insights into the role of flotillin microdomains in endocytosis that have been provided by several independent studies. Finally, we will focus on the questions that are raised by these new experiments and their implications for future studies.
Collapse
Affiliation(s)
- Grant P Otto
- Medical Research Council Laboratory of Molecular Biology, Hills Road, Cambridge CB2 0QH, UK
| | | |
Collapse
|
62
|
Antipsychotic induced alteration of growth and proteome of rat neural stem cells. Neurochem Res 2012; 37:1649-59. [PMID: 22528831 DOI: 10.1007/s11064-012-0768-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 01/19/2012] [Accepted: 03/27/2012] [Indexed: 01/19/2023]
Abstract
Neural stem cells (NSCs) play a crucial role in the development and maturation of the central nervous system and therefore have the potential to target by therapeutic agents for a wide variety of diseases including neurodegenerative and neuropsychiatric illnesses. It has been suggested that antipsychotic drugs have significant effects on NSC activities. However, the molecular mechanisms underlying antipsychotic-induced changes of NSC activities, particularly growth and protein expression, are largely unknown. NSCs were treated with either haloperidol (HD; 3 μM), risperidone (RS; 3 μM) or vehicle (DMSO) for 96 h. Protein expression profiles were studied through a proteomics approach. RS promoted and HD inhibited the growth of NSCs. Proteomics analysis revealed that 15 protein spots identified as 12 unique proteins in HD-, and 20 protein spots identified as 14 proteins in RS-treated groups, were differentially expressed relative to control. When these identified proteins were compared between the two drug-treated groups, 2 proteins overlapped leaving 10 HD-specific and 12 RS-specific proteins. Further comparison of the overlapped altered proteins of 96 h treatment with the neuroleptics-induced overlapped proteins at 24 h time interval (Kashem et al. [40] in Neurochem Int 55:558-565, 2009) suggested that overlapping altered proteins expression at 24 h was decreased (17 proteins i.e. 53 % of total expressed proteins) with the increase of time (96 h) (2 proteins; 8 % of total expressed proteins). This result indicated that at early stage both drugs showed common mode of action but the action was opposite to each other while administration was prolonged. The opposite morphological pattern of cellular growth at 96 h has been associated with dominant expression of oxidative stress and apoptosis cascades in HD, and activation of growth regulating metabolic pathways in RS treated cells. These results may explain RS induced repairing of neural damage caused by a wide variety of neural diseases including schizophrenia.
Collapse
|
63
|
Solis GP, Schrock Y, Hülsbusch N, Wiechers M, Plattner H, Stuermer CAO. Reggies/flotillins regulate E-cadherin-mediated cell contact formation by affecting EGFR trafficking. Mol Biol Cell 2012; 23:1812-25. [PMID: 22438585 PMCID: PMC3350547 DOI: 10.1091/mbc.e11-12-1006] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In epithelial cells, the reggie/flotillin proteins regulate—in association with PrP—the formation of E-cadherin adherens junctions (AJs) via the EGFR. Reggies control the EGF-mediated phosphorylation and internalization of EGFR. EGF signaling at the plasma membrane induces the macropinocytosis of E-cadherin and thus the formation of AJs. The reggie/flotillin proteins are implicated in membrane trafficking and, together with the cellular prion protein (PrP), in the recruitment of E-cadherin to cell contact sites. Here, we demonstrate that reggies, as well as PrP down-regulation, in epithelial A431 cells cause overlapping processes and abnormal formation of adherens junctions (AJs). This defect in cell adhesion results from reggie effects on Src tyrosine kinases and epidermal growth factor receptor (EGFR): loss of reggies reduces Src activation and EGFR phosphorylation at residues targeted by Src and c-cbl and leads to increased surface exposure of EGFR by blocking its internalization. The prolonged EGFR signaling at the plasma membrane enhances cell motility and macropinocytosis, by which junction-associated E-cadherin is internalized and recycled back to AJs. Accordingly, blockage of EGFR signaling or macropinocytosis in reggie-deficient cells restores normal AJ formation. Thus, by promoting EGFR internalization, reggies restrict the EGFR signaling involved in E-cadherin macropinocytosis and recycling and regulate AJ formation and dynamics and thereby cell adhesion.
Collapse
Affiliation(s)
- Gonzalo P Solis
- Department of Biology, University of Konstanz, 78467 Konstanz, Germany.
| | | | | | | | | | | |
Collapse
|
64
|
Stuermer CAO. How reggies regulate regeneration and axon growth. Cell Tissue Res 2012; 349:71-7. [DOI: 10.1007/s00441-012-1343-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 01/25/2012] [Indexed: 12/01/2022]
|
65
|
Takeshita N, Diallinas G, Fischer R. The role of flotillin FloA and stomatin StoA in the maintenance of apical sterol-rich membrane domains and polarity in the filamentous fungus Aspergillus nidulans. Mol Microbiol 2012; 83:1136-52. [PMID: 22329814 DOI: 10.1111/j.1365-2958.2012.07996.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Apical sterol-rich plasma membrane domains (SRDs), which can be viewed using the sterol-binding fluorescent dye filipin, are gaining attention for their important roles in polarized growth of filamentous fungi. The microdomain scaffolding protein flotillin/reggie and related stomatin were thought to be good candidates involved in the formation of SRDs. Here, we show that the flotillin/reggie orthologue FloA tagged with GFP localized as stable dots along the plasma membrane except hyphal tips. Deletion of floA reduced the growth rate, often resulted in irregularly shaped hyphae and impaired SRDs. In contrast, the stomatin orthologue StoA, tagged with GFP, localized at the cortex of young branch tips and at the subapical cortex in long hyphal tips, and was transported bi-directionally along microtubules on endosomes. Deletion of stoA resulted in irregular hyphal morphology and increased branching especially in young hyphae, but did not obviously affect SRDs. Double deletion of floA and stoA enhanced the defects of growth and hyphal morphology. Our data suggest that the plasma membrane of hyphal tips and in subapical regions are distinct and that FloA is involved in membrane compartmentalization and probably indirectly in SRD maintenance.
Collapse
Affiliation(s)
- Norio Takeshita
- Karlsruhe Institute of Technology, Institute for Applied Biosciences, Dept. of Microbiology, Hertzstrasse 16, D-76187 Karlsruhe, Germany.
| | | | | |
Collapse
|
66
|
Prion protein promotes growth cone development through reggie/flotillin-dependent N-cadherin trafficking. J Neurosci 2012; 31:18013-25. [PMID: 22159115 DOI: 10.1523/jneurosci.4729-11.2011] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The role of prion protein (PrP) is insufficiently understood partially because PrP-deficient (-/-) neurons from C57BL/6J mice seem to differentiate normally and are functionally mildly impaired. Here, we reassessed this notion and, unexpectedly, discovered that PrP(-/-) hippocampal growth cones were abnormally small and poor in filopodia and cargo-containing vesicles. Based on our findings that PrP-PrP trans-interaction recruits E-cadherin to cell contact sites and reggie microdomains, and that reggies are essential for growth by regulating membrane trafficking, we reasoned that PrP and reggie might promote cargo (N-cadherin) delivery via PrP-reggie-connected signaling upon PrP activation (by PrP-Fc-induced trans-interaction). In wild-type but not PrP(-/-) neurons, PrP activation led to (1) enhanced PrP-reggie cocluster formation, (2) reggie-associated fyn and MAP kinase activation, (3) Exo70 and N-cadherin (cargo) recruitment to reggie, (4) the preference of the growth cone for PrP-Fc as substrate, and (5) longer neurites. Conversely, PrP-reggie-induced N-cadherin recruitment was blocked by mutant TC10, the GTPase downstream of reggie, triggering exocyst-assisted cargo delivery. This implies that PrP functions in reggie-mediated signaling and cargo trafficking, thus promoting growth cone complexity and vitality and thereby growth cone elongation.
Collapse
|
67
|
Wan W, Liu Z, Wang X, Luo X. Dark rearing maintains tyrosine hydroxylase expression in retinal amacrine cells following optic nerve transection. Neural Regen Res 2012; 7:18-23. [PMID: 25806053 PMCID: PMC4354110 DOI: 10.3969/j.issn.1673-5374.2012.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 11/25/2011] [Indexed: 01/10/2023] Open
Abstract
The present study examined changes in retinal tyrosine hydroxylase (TH) expression in rats having undergone optic nerve transection and housed under a normal day/night cycle or in the dark. The aim was to investigate the effects of amacrine cells on axonal regeneration in retinal ganglion cells and on the synapses that transmit visual signals. The results revealed that retinal TH expression gradually decreased following optic nerve transection in rats housed under a normal day/night cycle, reaching a minimum at 5 days. In contrast, retinal TH expression decreased to a minimum at 1 day following optic nerve transection in dark reared rats, gradually increasing afterward and reaching a normal level at 5–7 days. The number of TH-positive synaptic particles correlated with the TH levels, indicating that dark rearing can help maintain TH expression during the synaptic degeneration stage (5–7 days after optic nerve injury) in retinal amacrine cells.
Collapse
Affiliation(s)
- Wei Wan
- Department of Human Anatomy and Neurobiology, Xiangya Medical College of Central South University, Changsha 410013, Hunan Province, China ; Department of Human Anatomy, University of South China, Hengyang 421001, Hunan Province, China
| | - Zhenghai Liu
- Department of Human Anatomy, University of South China, Hengyang 421001, Hunan Province, China
| | - Xiaosheng Wang
- Department of Human Anatomy and Neurobiology, Xiangya Medical College of Central South University, Changsha 410013, Hunan Province, China
| | - Xuegang Luo
- Department of Human Anatomy and Neurobiology, Xiangya Medical College of Central South University, Changsha 410013, Hunan Province, China
| |
Collapse
|
68
|
Tomasovic A, Traub S, Tikkanen R. Molecular networks in FGF signaling: flotillin-1 and cbl-associated protein compete for the binding to fibroblast growth factor receptor substrate 2. PLoS One 2012; 7:e29739. [PMID: 22235335 PMCID: PMC3250484 DOI: 10.1371/journal.pone.0029739] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 12/04/2011] [Indexed: 11/18/2022] Open
Abstract
Fibroblast growth factor receptor substrate 2 (FRS2α) is a signaling adaptor protein that regulates downstream signaling of many receptor tyrosine kinases. During signal transduction, FRS2 can be both tyrosine and threonine phosphorylated and forms signaling complexes with other adaptor proteins and tyrosine phosphatases. We have here identified flotillin-1 and the cbl-associated protein/ponsin (CAP) as novel interaction partners of FRS2. Flotillin-1 binds to the phosphotyrosine binding domain (PTB) of FRS2 and competes for the binding with the fibroblast growth factor receptor. Flotillin-1 knockdown results in increased Tyr phosphorylation of FRS2, in line with the inhibition of ERK activity in the absence of flotillin-1. CAP directly interacts with FRS2 by means of its sorbin homology (SoHo) domain, which has previously been shown to interact with flotillin-1. In addition, the third SH3 domain in CAP binds to FRS2. Due to the overlapping binding domains, CAP and flotillin-1 appear to compete for the binding to FRS2. Thus, our results reveal a novel signaling network containing FRS2, CAP and flotillin-1, whose successive interactions are most likely required to regulate receptor tyrosine kinase signaling, especially the mitogen activated protein kinase pathway.
Collapse
Affiliation(s)
- Ana Tomasovic
- Institute of Biochemistry, University of Giessen, Giessen, Germany
- Institute of Biochemistry II, University Clinic of Frankfurt, Frankfurt am Main, Germany
| | - Stephanie Traub
- Institute of Biochemistry II, University Clinic of Frankfurt, Frankfurt am Main, Germany
| | - Ritva Tikkanen
- Institute of Biochemistry, University of Giessen, Giessen, Germany
- Institute of Biochemistry II, University Clinic of Frankfurt, Frankfurt am Main, Germany
- * E-mail:
| |
Collapse
|
69
|
Li Y, Martin BR, Cravatt BF, Hofmann SL. DHHC5 protein palmitoylates flotillin-2 and is rapidly degraded on induction of neuronal differentiation in cultured cells. J Biol Chem 2012; 287:523-530. [PMID: 22081607 PMCID: PMC3249106 DOI: 10.1074/jbc.m111.306183] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 11/07/2011] [Indexed: 01/20/2023] Open
Abstract
Post-translational palmitoylation of intracellular proteins is mediated by protein palmitoyltransferases belonging to the DHHC family, which share a common catalytic Asp-His-His-Cys (DHHC) motif. Several members have been implicated in neuronal development, neurotransmission, and synaptic plasticity. We previously observed that mice homozygous for a hypomorphic allele of the ZDHHC5 gene are impaired in context-dependent learning and memory. To identify potentially relevant protein substrates of DHHC5, we performed a quantitative proteomic analysis of stable isotope-labeled neuronal stem cell cultures from forebrains of normal and DHHC5-GT (gene-trapped) mice using the bioorthogonal palmitate analog 17-octadecynoic acid. We identified ∼300 17-octadecynoic acid-modified and hydroxylamine-sensitive proteins, of which a subset was decreased in abundance in DHHC5-GT cells. Palmitoylation and oligomerization of one of these proteins (flotillin-2) was abolished in DHHC5-GT neuronal stem cells. In COS-1 cells, overexpression of DHHC5 markedly stimulated the palmitoylation of flotillin-2, strongly suggesting a direct enzyme-substrate relationship. Serendipitously, we found that down-regulation of DHHC5 was triggered within minutes following growth factor withdrawal from normal neural stem cells, a maneuver that is used to induce neural differentiation in culture. The effect was reversible for up to 4 h, and degradation was partially prevented by inhibitors of ubiquitin-mediated proteolysis. These findings suggest that protein palmitoylation can be regulated through changes in DHHC PAT levels in response to differentiation signals.
Collapse
Affiliation(s)
- Yi Li
- Hamon Center for Therapeutic Oncology Research and Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-8593
| | - Brent R Martin
- Skaggs Institute of Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037
| | - Benjamin F Cravatt
- Skaggs Institute of Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037
| | - Sandra L Hofmann
- Hamon Center for Therapeutic Oncology Research and Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-8593.
| |
Collapse
|
70
|
Blackmore MG. Molecular control of axon growth: insights from comparative gene profiling and high-throughput screening. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2012. [PMID: 23206595 DOI: 10.1016/b978-0-12-398309-1.00004-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Axon regeneration in the mammalian adult central nervous system (CNS) is limited by an intrinsically low capacity for axon growth in many CNS neurons. In contrast, embryonic, peripheral, and many nonmammalian neurons are capable of successful regeneration. Numerous studies have compared mammalian CNS neurons to their counterparts in regenerating systems in an effort to identify candidate genes that control regenerative ability. This review summarizes work using this comparative strategy and examines our current understanding of gene function in axon growth, highlighting the emergence of genome-wide expression profiling and high-throughput screening strategies to identify novel regulators of axon growth.
Collapse
Affiliation(s)
- Murray G Blackmore
- Department of Biomedical Sciences, Marquette University, Milwaukee, Wisconsin, USA.
| |
Collapse
|
71
|
Duox, Flotillin-2, and Src42A are required to activate or delimit the spread of the transcriptional response to epidermal wounds in Drosophila. PLoS Genet 2011; 7:e1002424. [PMID: 22242003 PMCID: PMC3248467 DOI: 10.1371/journal.pgen.1002424] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 10/31/2011] [Indexed: 12/24/2022] Open
Abstract
The epidermis is the largest organ of the body for most animals, and the first line of defense against invading pathogens. A breach in the epidermal cell layer triggers a variety of localized responses that in favorable circumstances result in the repair of the wound. Many cellular and genetic responses must be limited to epidermal cells that are close to wounds, but how this is regulated is still poorly understood. The order and hierarchy of epidermal wound signaling factors are also still obscure. The Drosophila embryonic epidermis provides an excellent system to study genes that regulate wound healing processes. We have developed a variety of fluorescent reporters that provide a visible readout of wound-dependent transcriptional activation near epidermal wound sites. A large screen for mutants that alter the activity of these wound reporters has identified seven new genes required to activate or delimit wound-induced transcriptional responses to a narrow zone of cells surrounding wound sites. Among the genes required to delimit the spread of wound responses are Drosophila Flotillin-2 and Src42A, both of which are transcriptionally activated around wound sites. Flotillin-2 and constitutively active Src42A are also sufficient, when overexpressed at high levels, to inhibit wound-induced transcription in epidermal cells. One gene required to activate epidermal wound reporters encodes Dual oxidase, an enzyme that produces hydrogen peroxide. We also find that four biochemical treatments (a serine protease, a Src kinase inhibitor, methyl-ß-cyclodextrin, and hydrogen peroxide) are sufficient to globally activate epidermal wound response genes in Drosophila embryos. We explore the epistatic relationships among the factors that induce or delimit the spread of epidermal wound signals. Our results define new genetic functions that interact to instruct only a limited number of cells around puncture wounds to mount a transcriptional response, mediating local repair and regeneration. An epidermal wound provides signals that initiate a variety of localized responses, some of which act to regenerate and repair the breach in the epidermal barrier. The Drosophila melanogaster embryonic epidermis provides an excellent system to discover new genes that regulate wound-healing processes. Using fluorescent epidermal “wound” reporters that are locally activated around wound sites, we have screened almost 5,000 Drosophila mutants for functions required to activate or delimit wound-induced transcriptional responses to a local zone of epidermal cells. Among the seven new genes required to delimit the spread of wound responses are Flotillin-2 and Src42A. These two genes are also sufficient, when overexpressed at high levels, to inhibit wound-induced transcription in epidermal cells. One new gene required to activate epidermal wound reporters encodes Dual oxidase, an enzyme that produces hydrogen peroxide. We also find that four biochemical treatments (a serine protease, a Src kinase inhibitor, methyl-ß-cyclodextrin, and hydrogen peroxide) are sufficient to globally activate epidermal wound response genes in Drosophila embryos. Our results define new genetic functions, and the interactions among them, which regulate the local transcriptional response to puncture wounds.
Collapse
|
72
|
Proteomics and bioinformatics analysis of lovastatin-induced differentiation in ARO cells. J Proteomics 2011; 75:1170-80. [PMID: 22086082 DOI: 10.1016/j.jprot.2011.10.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 10/22/2011] [Accepted: 10/28/2011] [Indexed: 01/05/2023]
Abstract
Lovastatin (lova), a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, can induce differentiation in cancer cells at low concentration, thus having potential to be used as an auxiliary agent in cancer therapy. However, biological networks associated with the differentiation effect of lova have not been elucidated. To investigate molecular mechanisms of lova, the present study was aimed at proteomics and bioinformatics analyses on anaplastic thyroid cancer cell line ARO differentiated with low concentration of lova. Thyroid differentiation was induced by treating ARO cells with 25 μM of lova and confirmed by checking upregulation of some thyroid differentiation markers. Gel-based proteomics analysis was then performed to identify proteins differentially expressed between undifferentiated and lova-differentiated ARO cells. Bioinformatics analysis was finally performed to estimate biological networks regulated by lova. Our results showed that lova impacted on proteins involved in protein folding, biomolecule metabolism, signal transduction, protein expression and protein degradation. Specifically, transfecting ARO cells with plasmid DNA encoding flotillin 1 (FLOT1) up-regulated the thyroid differentiation markers, indicating that FLOT1 might at least partially mediate the lova-induced thyroid differentiation. These data may shed light on the mechanism underlying lova-induced re-differentiation of thyroid cancer, and give a rationale for clinical use of lova as an auxiliary agent in cancer therapy.
Collapse
|
73
|
Zhao F, Zhang J, Liu YS, Li L, He YL. Research advances on flotillins. Virol J 2011; 8:479. [PMID: 22023811 PMCID: PMC3215287 DOI: 10.1186/1743-422x-8-479] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 10/25/2011] [Indexed: 08/15/2023] Open
Abstract
The proteins of flotillin-1 and flotillin-2 were originally discovered in axon regeneration of goldfish retinal ganglion cells. They are generally used as marker proteins of lipid rafts and considered to be scaffolding proteins of lipid microdomains. Although they are ubiquitously expressed and well-conserved from fly to man, their exact functions remain controversial. In this review, we summarize the structure of flotillins and some functions of them, such as regulating axon regeneration, endocytosis, T cell activation, insulin signaling, membrane protein recruitment, roles in the progression of some diseases and so on.
Collapse
Affiliation(s)
- Feng Zhao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046 Gansu, P.R. China
| | | | | | | | | |
Collapse
|
74
|
Chadwick W, Mitchell N, Caroll J, Zhou Y, Park SS, Wang L, Becker KG, Zhang Y, Lehrmann E, Wood WH, Martin B, Maudsley S. Amitriptyline-mediated cognitive enhancement in aged 3×Tg Alzheimer's disease mice is associated with neurogenesis and neurotrophic activity. PLoS One 2011; 6:e21660. [PMID: 21738757 PMCID: PMC3124550 DOI: 10.1371/journal.pone.0021660] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Accepted: 06/07/2011] [Indexed: 01/22/2023] Open
Abstract
Approximately 35 million people worldwide suffer from Alzheimer's disease (AD). Existing therapeutics, while moderately effective, are currently unable to stem the widespread rise in AD prevalence. AD is associated with an increase in amyloid beta (Aβ) oligomers and hyperphosphorylated tau, along with cognitive impairment and neurodegeneration. Several antidepressants have shown promise in improving cognition and alleviating oxidative stress in AD but have failed as long-term therapeutics. In this study, amitriptyline, an FDA-approved tricyclic antidepressant, was administered orally to aged and cognitively impaired transgenic AD mice (3×TgAD). After amitriptyline treatment, cognitive behavior testing demonstrated that there was a significant improvement in both long- and short-term memory retention. Amitriptyline treatment also caused a significant potentiation of non-toxic Aβ monomer with a concomitant decrease in cytotoxic dimer Aβ load, compared to vehicle-treated 3×TgAD controls. In addition, amitriptyline administration caused a significant increase in dentate gyrus neurogenesis as well as increases in expression of neurosynaptic marker proteins. Amitriptyline treatment resulted in increases in hippocampal brain-derived neurotrophic factor protein as well as increased tyrosine phosphorylation of its cognate receptor (TrkB). These results indicate that amitriptyline has significant beneficial actions in aged and damaged AD brains and that it shows promise as a tolerable novel therapeutic for the treatment of AD.
Collapse
Affiliation(s)
- Wayne Chadwick
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Nick Mitchell
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Jenna Caroll
- Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Yu Zhou
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Sung-Soo Park
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Liyun Wang
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Kevin G. Becker
- Genomics Unit, Research Resources Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Yongqing Zhang
- Genomics Unit, Research Resources Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Elin Lehrmann
- Genomics Unit, Research Resources Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - William H. Wood
- Genomics Unit, Research Resources Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Bronwen Martin
- Metabolism Unit, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Stuart Maudsley
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
75
|
Nagashima M, Fujikawa C, Mawatari K, Mori Y, Kato S. HSP70, the earliest-induced gene in the zebrafish retina during optic nerve regeneration: its role in cell survival. Neurochem Int 2011; 58:888-95. [PMID: 21338645 DOI: 10.1016/j.neuint.2011.02.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Revised: 01/31/2011] [Accepted: 02/12/2011] [Indexed: 12/22/2022]
Abstract
Fish retinal ganglion cells (RGCs) can survive and regrow their axons after optic nerve injury. Injured RGCs express anti-apoptotic proteins, such as Bcl-2, after nerve injury; however, upstream effectors of this anti-apoptotic protein are not yet fully understood. Heat shock proteins (HSPs) play a crucial role in cell survival against various stress conditions. In this study, we focused on HSP70 expression in the zebrafish retina after optic nerve injury. HSP70 mRNA and protein levels increased rapidly 2.3-fold in RGCs by 1-6 h after injury and returned to control levels by 1-3 days. HSP70 transcription is regulated by heat shock factor 1 (HSF1). HSF1 mRNA and phosphorylated-HSF1 protein rapidly increased by 2.2-fold in RGCs 0.5-6 h after injury. Intraocular injection of HSP inhibitor I significantly suppressed the induction of HSP70 expression after nerve injury. It also suppressed Bcl-2 protein induction and resulted in TUNEL-positive cell death of RGCs at 5 days post-injury. Zebrafish treated with HSP inhibitor I retarded axonal elongation or visual function after injury, as analyzed by GAP43 expression and behavioral analysis of optomotor response, respectively. These results strongly indicate that HSP70, the earliest induced gene in the zebrafish retina after optic nerve injury, is a crucial factor for RGCs survival and optic nerve regeneration in fish.
Collapse
Affiliation(s)
- Mikiko Nagashima
- Division of Health Sciences, Graduate School of Medicine, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan
| | | | | | | | | |
Collapse
|
76
|
Wang Z, Jin Y. Genetic dissection of axon regeneration. Curr Opin Neurobiol 2011; 21:189-96. [PMID: 20832288 PMCID: PMC3070659 DOI: 10.1016/j.conb.2010.08.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 07/28/2010] [Accepted: 08/15/2010] [Indexed: 01/19/2023]
Abstract
Axon regeneration has long been studied in vertebrate model organisms and neuronal cultures. Recent development of axon regeneration paradigms in genetic model organisms, such as Caenorhabditis elegans, Drosophila and zebrafish, has opened an exciting field for in vivo functional dissection of regeneration pathways. Studies in these organisms have discovered essential genes and pathways for axon regrowth. The conservation of these genes crossing animal phyla suggests mechanistic relevance to higher organisms. The power of genetic approaches in these organisms makes large-scale genetic and pharmacological screens feasible and can greatly accelerate the mechanistic understanding of axon regeneration.
Collapse
Affiliation(s)
- Zhiping Wang
- Division of Biological Sciences, Section of Neurobiology, University of California, San Diego, CA 92093
| | - Yishi Jin
- Division of Biological Sciences, Section of Neurobiology, University of California, San Diego, CA 92093
- Howard Hughes Medical Institute
| |
Collapse
|
77
|
Abstract
The two proteins reggie-1/flotillin-2 and reggie-2/flotillin-1 form microdomains at the plasma membrane and at intracellular compartments where src tyrosine kinases associate with them. Specific GPI-anchored proteins, in particular prion protein and Thy-1, co-cluster with reggie microdomains at the plasma membrane and elicit signal transduction in association with reggies which regulates the activation of several GTPases involved in the recruitment of specific membrane proteins from intracellular carriers to target sites of the cell membrane in a cell type-specific manner. For example, prion protein and reggie regulate the recruitment and targeted delivery of the T cell receptor complex to the T cell cap, of E-cadherin to cell-cell contact sites in epithelial cells, and of bulk membrane and growth receptors to the growth cone in developing neurons. Evidence is accumulating that reggies are involved in guiding the cell-type-specific membrane proteins from the intracellular compartments to their target sites at the cell membrane, a function required in all cells which explains why reggies are expressed in many or all cells in invertebrates and vertebrates.
Collapse
|
78
|
Stuermer CAO. Microdomain-forming proteins and the role of the reggies/flotillins during axon regeneration in zebrafish. Biochim Biophys Acta Mol Basis Dis 2010; 1812:415-22. [PMID: 21147218 DOI: 10.1016/j.bbadis.2010.12.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 11/30/2010] [Accepted: 12/02/2010] [Indexed: 01/08/2023]
Abstract
The two proteins reggie-1 and reggie-2 (flotillins) were identified in axon-regenerating neurons in the central nervous system and shown to be essential for neurite growth and regeneration in fish and mammals. Reggies/flotillins are microdomain scaffolding proteins sharing biochemical properties with lipid raft molecules, form clusters at the cytoplasmic face of the plasma membrane and interact with signaling molecules in a cell type specific manner. In this review, reggie microdomains, lipid rafts, related scaffolding proteins and caveolin-which, however, are responsible for their own microdomains and functions-are introduced. Moreover, the function of the reggies in axon growth is demonstrated: neurons fail to extend axons after reggie knockdown. Furthermore, our current concept of the molecular mechanism underlying reggie function is presented: the association of glycosyl-phophatidyl inositol (GPJ)-anchored surface proteins with reggie microdomains elicits signals which activate src tyrosine and mitogen-activated protein kinases, as well as small guanosine 5'-triphosphate-hydrolyzing enzymes. This leads to the mobilization of intracellular vesicles and to the recruitment of bulk membrane and specific cargo proteins, such as cadherin, to specific sites of the plasma membrane such as the growth cone of elongating axons. Thus, reggies regulate the targeted delivery of cargo-a process which is required for process extension and growth. This article is part of a Special Issue entitled Zebrafish Models of Neurological Diseases.
Collapse
|
79
|
Abstract
Adult somatic stem cells are central to homeostasis in tissues that present with a high cellular turnover like the skin, intestine, and the hematopoietic system. It is thought that polarity is particularly important with respect to fate decisions on stem cell division (symmetric or asymmetric) as well as for the maintenance of stem cell adhesion and quiescence (interaction with the niche). Consequently the failure to establish or regulate stem cell polarity might result in disease or tissue attrition. Members of the family of small RhoGTPases are known to exert an important role in regulating cell polarity. We summarize and discuss here recent views on the role of cell polarity in somatic stem cell function, aging, and disease, concluding that targeting cell polarity might be a novel approach to ameliorate or even revert aberrant somatic stem cell function. Stem Cells 2010; 28:1623–1629.
Collapse
|
80
|
Swanwick CC, Shapiro ME, Vicini S, Wenthold RJ. Flotillin-1 mediates neurite branching induced by synaptic adhesion-like molecule 4 in hippocampal neurons. Mol Cell Neurosci 2010; 45:213-25. [DOI: 10.1016/j.mcn.2010.06.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 05/21/2010] [Accepted: 06/15/2010] [Indexed: 11/25/2022] Open
|
81
|
Swanwick CC, Shapiro ME, Vicini S, Wenthold RJ. Flotillin-1 promotes formation of glutamatergic synapses in hippocampal neurons. Dev Neurobiol 2010; 70:875-83. [PMID: 20669324 PMCID: PMC4482238 DOI: 10.1002/dneu.20828] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Synapse malformation underlies numerous neurodevelopmental illnesses, including autism spectrum disorders. Here we identify the lipid raft protein flotillin-1 as a promoter of glutamatergic synapse formation. We cultured neurons from the hippocampus, a brain region important for learning and memory, and examined them at two weeks in vitro, a time period rich with synapse formation. Double-label immunocytochemistry of native flot-1 with glutamatergic and GABAergic synapse markers showed that flot-1 was preferentially colocalized with the glutamatergic presynaptic marker vesicular glutamate transporter 1 (VGLUT1), compared to the GABAergic presynaptic marker glutamic acid decarboxylase-65 (GAD-65). Triple-label immunocytochemistry of native flot-1, VGLUT1, and NR1, the obligatory subunit of NMDA receptors, indicates that Flot-1 was preferentially localized to synaptic rather than extrasynaptic NR1. Furthermore, electrophysiological results using whole-cell patch clamp showed that Flot-1 increased the frequency of miniature excitatory postsynaptic currents (mEPSCs) but not miniature inhibitory postsynaptic currents (mIPSCs), whereas amplitude and decay kinetics of either type of synaptic current was not affected. Corresponding immunocytochemical data confirmed that the number of glutamatergic synapses increased with flot-1 overexpression. Overall, our anatomical and physiological results show that flot-1 enhances the formation of glutamatergic synapses but not GABAergic synapses, suggesting that the role of flot-1 in neurodevelopmental disorders should be explored.
Collapse
Affiliation(s)
- Catherine Croft Swanwick
- Laboratory of Neurochemistry, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | |
Collapse
|
82
|
Fleisch VC, Fraser B, Allison WT. Investigating regeneration and functional integration of CNS neurons: lessons from zebrafish genetics and other fish species. Biochim Biophys Acta Mol Basis Dis 2010; 1812:364-80. [PMID: 21044883 DOI: 10.1016/j.bbadis.2010.10.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 10/05/2010] [Accepted: 10/21/2010] [Indexed: 12/21/2022]
Abstract
Zebrafish possess a robust, innate CNS regenerative ability. Combined with their genetic tractability and vertebrate CNS architecture, this ability makes zebrafish an attractive model to gain requisite knowledge for clinical CNS regeneration. In treatment of neurological disorders, one can envisage replacing lost neurons through stem cell therapy or through activation of latent stem cells in the CNS. Here we review the evidence that radial glia are a major source of CNS stem cells in zebrafish and thus activation of radial glia is an attractive therapeutic target. We discuss the regenerative potential and the molecular mechanisms thereof, in the zebrafish spinal cord, retina, optic nerve and higher brain centres. We evaluate various cell ablation paradigms developed to induce regeneration, with particular emphasis on the need for (high throughput) indicators that neuronal regeneration has restored sensory or motor function. We also examine the potential confound that regeneration imposes as the community develops zebrafish models of neurodegeneration. We conclude that zebrafish combine several characters that make them a potent resource for testing hypotheses and discovering therapeutic targets in functional CNS regeneration. This article is part of a Special Issue entitled Zebrafish Models of Neurological Diseases.
Collapse
Affiliation(s)
- Valerie C Fleisch
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, Alberta, Canada.
| | | | | |
Collapse
|
83
|
Franco M, Seyfried NT, Brand AH, Peng J, Mayor U. A novel strategy to isolate ubiquitin conjugates reveals wide role for ubiquitination during neural development. Mol Cell Proteomics 2010; 10:M110.002188. [PMID: 20861518 PMCID: PMC3098581 DOI: 10.1074/mcp.m110.002188] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Ubiquitination has essential roles in neuronal development and function. Ubiquitin proteomics studies on yeast and HeLa cells have proven very informative, but there still is a gap regarding neuronal tissue-specific ubiquitination. In an organism context, direct evidence for the ubiquitination of neuronal proteins is even scarcer. Here, we report a novel proteomics strategy based on the in vivo biotinylation of ubiquitin to isolate ubiquitin conjugates from the neurons of Drosophila melanogaster embryos. We confidently identified 48 neuronal ubiquitin substrates, none of which was yet known to be ubiquitinated. Earlier proteomics and biochemical studies in non-neuronal cell types had identified orthologs to some of those but not to others. The identification here of novel ubiquitin substrates, those with no known ubiquitinated ortholog, suggests that proteomics studies must be performed on neuronal cells to identify ubiquitination pathways not shared by other cell types. Importantly, several of those newly found neuronal ubiquitin substrates are key players in synaptogenesis. Mass spectrometry results were validated by Western blotting to confirm that those proteins are indeed ubiquitinated in the Drosophila embryonic nervous system and to elucidate whether they are mono- or polyubiquitinated. In addition to the ubiquitin substrates, we also identified the ubiquitin carriers that are active during synaptogenesis. Identifying endogenously ubiquitinated proteins in specific cell types, at specific developmental stages, and within the context of a living organism will allow understanding how the tissue-specific function of those proteins is regulated by the ubiquitin system.
Collapse
Affiliation(s)
- Maribel Franco
- CIC Biogune, Bizkaia Teknologi Parkea, 48160 Derio, Spain
| | | | | | | | | |
Collapse
|
84
|
No Nogo66- and NgR-mediated inhibition of regenerating axons in the zebrafish optic nerve. J Neurosci 2010; 29:15489-98. [PMID: 20007473 DOI: 10.1523/jneurosci.3561-09.2009] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In contrast to mammals, lesioned axons in the zebrafish (ZF) optic nerve regenerate and restore vision. This correlates with the absence of the NogoA-specific N-terminal domains from the ZF nogo/rtn-4 (reticulon-4) gene that inhibits regeneration in mammals. However, mammalian nogo/rtn-4 carries a second inhibitory C-terminal domain, Nogo-66, being 70% identical with ZF-Nogo66. The present study examines, (1) whether ZF-Nogo66 is inhibitory and effecting similar signaling pathways upon Nogo66-binding to the Nogo66 receptor NgR and its coreceptors, and (2) whether Rat-Nogo66 on fish, and ZF-Nogo66 on mouse neurons, cause inhibition via NgR. Our results from "outgrowth, collapse and contact assays" suggest, surprisingly, that ZF-Nogo66 is growth-permissive for ZF and mouse neurons, quite in contrast to its Rat-Nogo66 homolog which inhibits growth. The opposite effects of ZF- and Rat-Nogo66 are, in both fish and mouse, transmitted by GPI (glycosylphosphatidylinositol)-anchored receptors, including NgR. The high degree of sequence homology in the predicted binding site is consistent with the ability of ZF- and mammalian-Nogo66 to bind to NgRs of both species. Yet, Rat-Nogo66 elicits phosphorylation of the downstream effector cofilin whereas ZF-Nogo66 has no influence on cofilin phosphorylation--probably because of significantly different Rat- versus ZF-Nogo66 sequences outside of the receptor-binding region effecting, by speculation, recruitment of a different set of coreceptors or microdomain association of NgR. Thus, not only was the NogoA-specific domain lost in fish, but Nogo66, the second inhibitory domain in mammals, and its signaling upon binding to NgR, was modified so that ZF-Nogo/RTN-4 does not impair axon regeneration.
Collapse
|
85
|
Stuermer CA. The reggie/flotillin connection to growth. Trends Cell Biol 2010; 20:6-13. [DOI: 10.1016/j.tcb.2009.10.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 10/09/2009] [Accepted: 10/09/2009] [Indexed: 10/20/2022]
|