Synucleins Are Developmentally Expressed, and Α-Synuclein Regulates the Size of the Presynaptic Vesicular Pool in Primary Hippo

Total Page:16

File Type:pdf, Size:1020Kb

Synucleins Are Developmentally Expressed, and Α-Synuclein Regulates the Size of the Presynaptic Vesicular Pool in Primary Hippo The Journal of Neuroscience, May 1, 2000, 20(9):3214–3220 Synucleins Are Developmentally Expressed, and ␣-Synuclein Regulates the Size of the Presynaptic Vesicular Pool in Primary Hippocampal Neurons Diane D. Murphy, Susan M. Rueter, John Q. Trojanowski, and Virginia M.-Y. Lee Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania Medical School, Philadelphia, Pennsylvania 19104 ␣-, ␤-, and ␥-Synuclein, a novel family of neuronal proteins, has in the presynaptic terminal, whereas little ␥-synuclein was ex- become the focus of research interest because ␣-synuclein has pressed at all. To assess the function of ␣-synuclein, we sup- been increasingly implicated in the pathogenesis of Parkinson’s pressed expression of this protein with antisense oligonucleo- and Alzheimer’s disease. However, the normal functions of the tide technology. Morphometric ultrastructural analysis of the synucleins are still unknown. For this reason, we characterized ␣-synuclein antisense oligonucleotide-treated cultures revealed ␣-, ␤-, and ␥-synuclein expression in primary hippocampal a significant reduction in the distal pool of synaptic vesicles. neuronal cultures and showed that the onset of ␣- and These data suggest that one function of ␣-synuclein may be to ␤-synuclein expression was delayed after synaptic develop- regulate the size of distinct pools of synaptic vesicles in mature ment, suggesting that these synucleins may not be essential for neurons. synapse formation. In mature cultured primary neurons, ␣- and Key words: Lewy bodies; Parkinson’s disease; primary neu- ␤-synuclein colocalized almost exclusively with synaptophysin rons; synapse; synuclein; vesicle ␣-Synuclein was first isolated in 1988 from synaptic vesicles of sin during CNS development, it has been suggested to play a role Torpedo californica and rat brain (Maroteaux et al., 1988). Sub- in synaptogenesis (Hsu et al., 1998). However, in vitro studies sequently, other members of this family of neuronal proteins have show that the vesicle-associated protein synapsin I appears as been identified: ␤-synuclein, also found mainly in the CNS, early as 24 hr, whereas ␣-synuclein is not detected until day 5 ␥-synuclein, more abundant in the PNS than in the CNS, and (Withers et al., 1997). Thus, it is unclear whether ␣-synuclein is synoretin, present primarily in the retina (Nakajo et al., 1990; needed for the development of synapses or for the maturation and Jakes et al., 1994; Ji et al., 1997; Buchman et al., 1998; Surguchov modulation of previously existing ones. There is also evidence et al., 1999). ␣-Synuclein has been shown to be a primary com- that ␣-synuclein may associate with synaptic vesicles because ponent of Lewy bodies, the neuropathological hallmarks of spo- ␣-synuclein has been detected in vesicular fractions of human radic Parkinson’s disease (PD) (Spillantini et al., 1997; Baba et brain (Irizarry et al., 1996) and binds to both synthetic mem- al., 1998), and pathogenic ␣-synuclein gene mutations have been branes (Davidson et al., 1998) and vesicles isolated from rat brain identified in familial forms of PD (Polymeropoulos et al., 1997; (Jensen et al., 1998). Kruger et al., 1998). Moreover, ␣-synuclein has been detected in The present study assessed ␣-, ␤-, and ␥-synuclein expression in Lewy bodies that are diagnostic of the Lewy body variant of primary hippocampal neurons grown at a substantially increased Alzheimer’s Disease (LBVAD) as well as dementia with Lewy density. These high-density cultures exhibit a robust, rapid devel- bodies (DLB) (Spillantini et al., 1997; Baba et al., 1998; Tro- opment of synaptic contacts and spines (Papa et al., 1995) that janowski et al., 1998). Thus, insight into mechanisms of synuclein corresponds to those seen during postnatal hippocampal devel- pathology may be critical for understanding brain degeneration in opment. Moreover, the synaptic ultrastructure of high-density PD, LBVAD, and DLB (Trojanowski et al., 1998). cultures resembles that found in situ (Bartlett and Banker, 1984). The implication of ␣-synuclein in neurodegenerative disease We used light microscopic immunocytochemistry, electron mi- has stimulated efforts to elucidate the normal distribution and croscopy, and protein biochemistry to characterize the expression functions of ␣-, ␤-, and ␥-synuclein. For example, because of the synuclein family of proteins in hippocampal neurons up to ␣-synuclein is localized to presynaptic terminals throughout the 3 weeks in culture. We also examined changes in synaptic struc- adult mammalian brain and may appear earlier than synaptophy- ture after treatment of the neurons with antisense (AS) oligonu- cleotides to reduce ␣-synuclein expression. Here, we report that Received Nov. 11, 1999; revised Feb. 14, 2000; accepted Feb. 24, 2000. ␣- and ␤-synuclein were expressed after synaptophysin and local- This work was supported by grants from the National Institute on Aging. We acknowledge Drs. N. B. Cole, R.W. Doms, and V. Zhukareva for scientific contri- ized almost exclusively to presynaptic terminals of mature neu- butions, Dr. R. Balice-Gordon for use of the confocal microscope, and N. Shah and rons. In addition, downregulation of ␣-synuclein by AS oligonu- J. Sanzo of the electron microscopy facility for EM preparations and use of the cleotides caused a selective reduction in the size of the electron microscope. ␣ D.M. and S.R. contributed equally to this work. presynaptic vesicular pool. Our data suggest that -synuclein may Correspondence should be addressed to Dr. Virginia M.-Y. Lee, Department of interact with and regulate specific pools of synaptic vesicles, Pathology and Laboratory Medicine, University of Pennsylvania School of Medi- cine, Maloney 3, HUP, 3600 Spruce Street, Philadelphia, PA 19104-4283. E-mail: thereby modulating synaptic functions in the normal brain. These [email protected]. findings have important implications for the role ␣-synuclein may Copyright © 2000 Society for Neuroscience 0270-6474/00/203214-07$15.00/0 play in neurodegenerative disease. Murphy et al. • ␣-Synuclein Regulates Vesicular Pool J. Neurosci., May 1, 2000, 20(9):3214–3220 3215 Figure 1. Synuclein characterization by immunofluorescence. 1 (a)-,2(b)-, and 3 (c)-week-old cultures stained for ␣-synuclein (red) and synaptophysin ( green). Cytosolic ␣-synuclein staining is evident in a. d, Double labeling of ␣-synuclein ( green) and GAD (red) show colocalization in an inhibitory neuron and in GABAergic presynaptic terminals ( yellow). e, ␣- and ␤-synuclein are colocalized ( yellow) to the presynaptic terminal. f, ␥-synuclein (red) is poorly expressed in mature hippocampal neurons as compared to synaptophysin ( green). The nuclei in a–f are labeled by Hoechst (blue) dye. Scale bar, 10 ␮m. MATERIALS AND METHODS 0.1% Triton X-100, 0.5 mM PMSF, and 1 mM DTT, protease inhibitor ϫ Hippocampal cultures. High-density hippocampal neuronal cultures were cocktail). Harvested cells were spun at 100,000 g in a TL100 ultracen- trifuge (Beckman). Protein concentration in the extracts was determined prepared as previously described (Papa et al., 1995). Briefly, 20-d-old ␮ embryos were taken from anesthetized Sprague Dawley rats. The brains by the Bradford assay. Approximately 30 g of the supernatant was were removed and placed in ice-cold (4°C) L-15 medium supplemented fractionated by SDS-PAGE and transferred onto nitrocellulose. The ␮ blots were incubated in blocking solution containing 5% nonfat dry milk with 0.6% glucose and 15 g/ml gentamicin. The hippocampus was ϫ dissected and mechanically disaggregated by gentle trituration using a in 1 Tris-buffered saline (TBS) and then incubated overnight at 4°C Pasteur pipette. Dissociated cells (500,000 cells/well) were plated onto with the various primary antibodies described above diluted in 5% sterile 12 mm glass or Thermanox coverslips that were coated with milk/TBS. As a control, blots were also stained for neuron-specific ␮ ␮ enolase (NSE; Polysciences, Warrington, PA) to normalize the amount collagen (50 g/ml) and poly-L-lysine (15 g/ml). Cells were also pre- ϩ pared on six-well tissue culture dishes for Western blot analysis. Ther- of neuronal protein. The blots were washed in TBS-T (TBS 0.1% Tween 20) and incubated for 2 hr at room temperature with secondary manox coverslips were used for electron microscopy (see below). The 125 plating medium was Eagle’s MEM containing 5% heat-inactivated horse antibodies [ I] conjugated to either anti-mouse IgG or Protein A at a ␮ serum, 5% fetal calf serum, 2 mM glutamine, 0.6% glucose, and 15 ␮g/ml concentration of 1 Ci/ml of PTX buffer (10 mM sodium phosphate, pH 7.3, 1 mM EDTA, 150 mM NaCl, 0.2% Triton X-100, and 4% BSA). The gentamicin. Cells were incubated at 37°C with 5% CO2. The first change of medium, ϳ4–6 d after plating, included 50 ␮g/ml uridine and 20 blots were washed again with TBS-T and exposed in a PhosphorImager ␮g/ml deoxyuridine to prevent glial cell overgrowth. The cultures were cassette (Molecular Dynamics, Sunnyvale, CA) for 24–48 hr. The pro- fed thereafter 1–2 times a week, with Eagle’s MEM as above. tein levels were quantified using ImageQuant software (Molecular Dy- Immunocytochemistry. Cells were fixed in 4% paraformaldehyde in namics), each lane normalized to NSE levels. Ј PBS. After blocking and permeabilization in 5% goat serum with 0.1% Antisense treatments. Oligonucleotides were prepared from the 5 end ␣ saponin, cells were incubated with primary antibodies to the following:
Recommended publications
  • 1 BETA- and GAMMA-SYNUCLEINS MODULATE SYNAPTIC VESICLE-BINDING of ALPHA-SYNUCLEIN Kathryn E. Carnazza1#, Lauren Komer1#, André
    bioRxiv preprint doi: https://doi.org/10.1101/2020.11.19.390419; this version posted November 20, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. BETA- AND GAMMA-SYNUCLEINS MODULATE SYNAPTIC VESICLE-BINDING OF ALPHA-SYNUCLEIN Kathryn E. Carnazza1#, Lauren Komer1#, André Pineda1, Yoonmi Na1, Trudy Ramlall2, Vladimir L. Buchman3, David Eliezer2, Manu Sharma1, Jacqueline Burré1,* 1Helen and Robert Appel Alzheimer’s Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10021, USA. 2Department of Biochemistry, Weill Cornell Medicine, New York, New York 10021, USA. 3School of Biosciences, Cardiff University, Cardiff CF103AX, UK & Institute of Physiologically Active Compounds Russian Academy of Sciences (IPAC RAS), Chernogolovka 142432, Moscow Region, Russian Federation. #These authors contributed equally to this work. *Correspondence: [email protected] 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.11.19.390419; this version posted November 20, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. SUMMARY α-Synuclein (αSyn), β-synuclein (βSyn), and γ-synuclein (γSyn) are abundantly expressed in the vertebrate nervous system. αSyn functions in neurotransmitter release via binding to and clustering synaptic vesicles and chaperoning of SNARE-complex assembly. The functions of βSyn and γSyn are unknown. Functional redundancy of the three synucleins and mutual compensation when one synuclein is deleted have been proposed, but with conflicting evidence. Here, we demonstrate that βSyn and γSyn have a reduced affinity towards membranes compared to αSyn, and that direct interaction of βSyn or γSyn with αSyn results in reduced membrane binding of αSyn.
    [Show full text]
  • Analysis of Proteins That Rapidly Change Upon Mechanistic
    crossmark Research © 2016 by The American Society for Biochemistry and Molecular Biology, Inc. This paper is available on line at http://www.mcponline.org Analysis of Proteins That Rapidly Change Upon Mechanistic/Mammalian Target of Rapamycin Complex 1 (mTORC1) Repression Identifies Parkinson Protein 7 (PARK7) as a Novel Protein Aberrantly Expressed in Tuberous Sclerosis Complex (TSC)*□S Farr Niere‡§¶ʈ§§, Sanjeev Namjoshi‡§ §§, Ehwang Song**, Geoffrey A. Dilly‡§¶, Grant Schoenhard‡‡, Boris V. Zemelman‡§¶, Yehia Mechref**, and Kimberly F. Raab-Graham‡§¶ʈ‡‡¶¶ Many biological processes involve the mechanistic/mam- ally alters the expression of proteins associated with malian target of rapamycin complex 1 (mTORC1). Thus, epilepsy, Alzheimer’s disease, and autism spectrum the challenge of deciphering mTORC1-mediated func- disorder—neurological disorders that exhibit elevated tions during normal and pathological states in the central mTORC1 activity. Through a protein–protein interaction nervous system is challenging. Because mTORC1 is at the network analysis, we have identified common proteins core of translation, we have investigated mTORC1 func- shared among these mTORC1-related diseases. One such tion in global and regional protein expression. Activation protein is Parkinson protein 7, which has been implicated of mTORC1 has been generally regarded to promote in Parkinson’s disease, yet not associated with epilepsy, translation. Few but recent works have shown that sup- Alzheimers disease, or autism spectrum disorder. To ver- pression of mTORC1 can also promote local protein syn- ify our finding, we provide evidence that the protein ex- thesis. Moreover, excessive mTORC1 activation during pression of Parkinson protein 7, including new protein diseased states represses basal and activity-induced pro- synthesis, is sensitive to mTORC1 inhibition.
    [Show full text]
  • A Novel Method of Neural Differentiation of PC12 Cells by Using Opti-MEM As a Basic Induction Medium
    INTERNATIONAL JOURNAL OF MOLECULAR MEDICINE 41: 195-201, 2018 A novel method of neural differentiation of PC12 cells by using Opti-MEM as a basic induction medium RENDONG HU1*, QIAOYU CAO2*, ZHONGQING SUN3, JINYING CHEN4, QING ZHENG2 and FEI XIAO1 1Department of Pharmacology, School of Medicine, Jinan University; 2College of Pharmacy, Jinan University, Guangzhou, Guangdong 510632; 3Department of Anesthesia and Intensive Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, SAR; 4Department of Ophthalmology, The First Clinical Medical College of Jinan University, Guangzhou, Guangdong 510632, P.R. China Received April 5, 2017; Accepted October 11, 2017 DOI: 10.3892/ijmm.2017.3195 Abstract. The PC12 cell line is a classical neuronal cell model Introduction due to its ability to acquire the sympathetic neurons features when deal with nerve growth factor (NGF). In the present study, The PC12 cell line is traceable to a pheochromocytoma from the authors used a variety of different methods to induce PC12 the rat adrenal medulla (1-4). When exposed to nerve growth cells, such as Opti-MEM medium containing different concen- factor (NGF), PC12 cells present an observable change in trations of fetal bovine serum (FBS) and horse serum compared sympathetic neuron phenotype and properties. Neural differ- with RPMI-1640 medium, and then observed the neurite length, entiation of PC12 has been widely used as a neuron cell model differentiation, adhesion, cell proliferation and action poten- in neuroscience, such as in the nerve injury-induced neuro- tial, as well as the protein levels of axonal growth-associated pathic pain model (5) and nitric oxide-induced neurotoxicity protein 43 (GAP-43) and synaptic protein synapsin-1, among model (6).
    [Show full text]
  • Reducing Endogenous Α-Synuclein Mitigates the Degeneration of Selective Neuronal Populations in an Alzheimer's Disease Tran
    The Journal of Neuroscience, July 27, 2016 • 36(30):7971–7984 • 7971 Neurobiology of Disease Reducing Endogenous ␣-Synuclein Mitigates the Degeneration of Selective Neuronal Populations in an Alzheimer’s Disease Transgenic Mouse Model X Brian Spencer,1 Paula A. Desplats,1,2 Cassia R. Overk,1 Elvira Valera-Martin,1 Robert A. Rissman,1 Chengbiao Wu,1 Michael Mante,1 Anthony Adame,1 Jazmin Florio,1 Edward Rockenstein,1 and Eliezer Masliah1,2 1Department of Neurosciences and 2Department of Pathology, University of California–San Diego, La Jolla, California 92093 Alzheimer’s disease (AD) is characterized by the progressive accumulation of amyloid ␤ (A␤) and microtubule associate protein tau, leading to the selective degeneration of neurons in the neocortex, limbic system, and nucleus basalis, among others. Recent studies have shown that ␣-synuclein (␣-syn) also accumulates in the brains of patients with AD and interacts with A␤ and tau, forming toxic hetero-oligomers. Although the involvement of ␣-syn has been investigated extensively in Lewy body disease, less is known about the role of this synaptic protein in AD. Here, we found that reducing endogenous ␣-syn in an APP transgenic mouse model of AD prevented the degeneration of cholinergic neurons, ameliorated corresponding deficits, and recovered the levels of Rab3a and Rab5 proteins involved in intracellular transport and sorting of nerve growth factor and brain-derived neurotrophic factor. Together, these results suggest that ␣-syn might participate in mechanisms of vulnerability of selected neuronal populations in AD and that reducing ␣-syn might be a potential approach to protecting these populations from the toxic effects of A␤.
    [Show full text]
  • Fluid Biomarkers for Synaptic Dysfunction and Loss
    BMI0010.1177/1177271920950319Biomarker InsightsCamporesi et al 950319review-article2020 Biomarker Insights Fluid Biomarkers for Synaptic Dysfunction and Loss Volume 15: 1–17 © The Author(s) 2020 Elena Camporesi1 , Johanna Nilsson1, Ann Brinkmalm1, Article reuse guidelines: sagepub.com/journals-permissions 1,2 1,3,4,5 1,2 Bruno Becker , Nicholas J Ashton , Kaj Blennow DOI:https://doi.org/10.1177/1177271920950319 10.1177/1177271920950319 and Henrik Zetterberg1,2,6,7 1Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden. 2Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden. 3King’s College London, Institute of Psychiatry, Psychology & Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, London, UK. 4NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK. 5Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden. 6Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK. 7UK Dementia Research Institute at UCL, London, UK. ABSTRACT: Synapses are the site for brain communication where information is transmitted between neurons and stored for memory forma- tion. Synaptic degeneration is a global and early pathogenic event in neurodegenerative
    [Show full text]
  • Agrin Differentially Regulates the Rates of Axonal and Dendritic Elongation in Cultured Hippocampal Neurons
    The Journal of Neuroscience, September 1, 2001, 21(17):6802–6809 Agrin Differentially Regulates the Rates of Axonal and Dendritic Elongation in Cultured Hippocampal Neurons Kristina B. Mantych and Adriana Ferreira Institute for Neuroscience and Department of Cell and Molecular Biology, Northwestern University Medical School, Chicago, Illinois 60611 In the present study, we examined the role of agrin in axonal elongation and branching were paralleled by changes in the and dendritic elongation in central neurons. Dissociated hip- composition of the cytoskeleton. In the presence of agrin, there pocampal neurons were grown in the presence of either recom- was an upregulation of the expression of microtubule- binant agrin or antisense oligonucleotides designed to block associated proteins MAP1B, MAP2, and tau. In contrast, a agrin expression. Our results indicate that agrin differentially downregulation of the expression of these MAPs was detected regulates axonal and dendritic growth. Recombinant agrin de- in agrin-depleted cells. Taken collectively, these results suggest creased the rate of elongation of main axons but induced the an important role for agrin as a trigger of the transcription of formation of axonal branches. On the other hand, agrin induced neuro-specific genes involved in neurite elongation and branch- both dendritic elongation and dendritic branching. Conversely, ing in central neurons. cultured hippocampal neurons depleted of agrin extended longer, nonbranched axons and shorter dendrites when com- Key words: agrin; neurite outgrowth; microtubule-associated pared with controls. These changes in the rates of neurite proteins; CREB; antisense oligonucleotides; axons and dendrites Agrin, an extracellular matrix protein, is synthesized by motor Conversely, neurons depleted of agrin elongated longer axons neurons and transported to their terminals where it is released when compared with control ones (Ferreira 1999; Serpinskaya et (Magill-Solc and McMahan, 1988; Martinou et al., 1991; Ruegg et al., 1999).
    [Show full text]
  • Synapsin I in PC1 2 Cells. I. Characterization of the Phosphoprotein and Effect of Chronic NGF Treatment
    The Journal of Neuroscience, May 1987, 7(5): 1294-l 299 Synapsin I in PC1 2 Cells. I. Characterization of the Phosphoprotein and Effect of Chronic NGF Treatment Carmelo Romano, Robert A. Nichols, Paul Greengard, and Lloyd A. Greene Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York 10021; and Department of Pharmacology, New York University School of Medicine, New York, New York 10016 PC1 2 cells contain a synapsin l-like molecule. Several serum Adrenal chromaffin cells and sympathetic neurons share a and monoclonal antibodies raised against bovine brain syn- neural crest origin and many other similarities (Coupland, 1965; apsin I bind to and precipitate this molecule, demonstrating Weston, 1970). Nevertheless, normal rat adrenal chromaffin immunochemical similarity between the brain and PC12 cells do not, whereassympathetic neurons do, contain synapsin species. PC12 synapsin I, like brain synapsin I, is a phos- I (DeCamilli et al., 1979; Fried et al., 1982). To better under- phoprotein: It is phosphorylated in intact cells and, when stand the developmental regulation of synapsin I, it was there- partially purified, serves as a substrate for several synapsin fore of interest to study synapsin I in PC 12 cells and its possible I kinases. PC1 2 cell synapsin I is structurally similar to brain alteration upon treatment of the cells with NGF. This paper synapsin I as shown by peptide mapping of %-methionine- characterizes the synapsin I present in PC12 cells and demon- and 32P-phosphate-labeled molecules from the 2 sources. strates effects of long-term NGF treatment of the cells on the Chronic NGF treatment of the cells induces a significant phosphoprotein.The accompanyingpaper (Roman0 et al., 1987) increase in the amount of synapsin I relative to total cell demonstratesthat short-term NGF treatment of PC12 cells re- protein, measured either by immunolabeling or incorporation sults in the phosphorylation of synapsin I at a novel site.
    [Show full text]
  • Agrin Controls Synaptic Differentiation in Hippocampal Neurons
    The Journal of Neuroscience, December 15, 2000, 20(24):9086–9095 Agrin Controls Synaptic Differentiation in Hippocampal Neurons Christian M. Bo¨ se,1 Dike Qiu,1 Andrea Bergamaschi,3 Biagio Gravante,3 Mario Bossi,2 Antonello Villa,2 Fabio Rupp,1 and Antonio Malgaroli3 1Department of Neuroscience, The Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, 2Microscopy and Image Analysis, University of Milan School of Medicine, and 3Department of Biological and Technological Research, Scientific Institute San Raffaele, 20123 Milano, Italy Agrin controls the formation of the neuromuscular junction. agrin and could not be detected in cultures from agrin-deficient Whether it regulates the differentiation of other types of synapses animals. Interestingly, the few synapses formed in reduced agrin remains unclear. Therefore, we have studied the role of agrin in conditions displayed diminished vesicular turnover, despite a cultured hippocampal neurons. Synaptogenesis was severely normal appearance at the EM level. Thus, our results demon- compromised when agrin expression or function was sup- strate the necessity of agrin for synaptogenesis in hippocampal pressed by antisense oligonucleotides and specific antibodies. neurons. The effects of antisense oligonucleotides were found to be highly Key words: agrin; synaptogenesis; synapses; primary hip- specific because they were reversed by adding recombinant pocampal neurons; agrin antibodies; antisense oligonucleotides The fidelity of neural transmission depends on interactions of a can be detected in CNS neurons, where the temporal pattern of large number of presynaptic and postsynaptic molecules. Despite expression of agrin parallels synaptogenesis (Hoch et al., 1993; the importance of these processes for neural communication, the O’Connor et al., 1994; Stone and Nikolics, 1995; N.
    [Show full text]
  • Hsc70 Chaperone Activity Is Required for the Cytosolic Slow Axonal Transport of Synapsin
    Hsc70 chaperone activity is required for the cytosolic slow axonal transport of synapsin Archan Ganguly, Xuemei Han, Utpal Das, Lina Wang, Jonathan Loi, Jichao Sun, Daniel Gitler, Ghislaine Caillol, Christophe Leterrier, John R. Yates Ill, et al. To cite this version: Archan Ganguly, Xuemei Han, Utpal Das, Lina Wang, Jonathan Loi, et al.. Hsc70 chaperone activity is required for the cytosolic slow axonal transport of synapsin. Journal of Cell Biology, Rockefeller University Press, 2017, 216 (7), pp.2059-2074. 10.1083/jcb.201604028. hal-01701379 HAL Id: hal-01701379 https://hal.archives-ouvertes.fr/hal-01701379 Submitted on 20 Apr 2018 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Published May 30, 2017 JCB: Article Hsc70 chaperone activity is required for the cytosolic slow axonal transport of synapsin Archan Ganguly,1 Xuemei Han,2* Utpal Das,1* Lina Wang,5 Jonathan Loi,5 Jichao Sun,5 Daniel Gitler,3 Ghislaine Caillol,4 Christophe Leterrier,4 John R. Yates III,2 and Subhojit Roy5,6 1Department of Pathology, University of California, San Diego,
    [Show full text]
  • In Vitro Aggregation Assays for the Characterization of Α-Synuclein Prion-Like Properties
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by SISSA Digital Library REVIEW REVIEW Prion 8:1, 19–32; January/February 2014; © 2014 Landes Bioscience In vitro aggregation assays for the characterization of α-synuclein prion-like properties Joanna Narkiewicz1, Gabriele Giachin1, and Giuseppe Legname1,2,* 1Prion Biology Laboratory; Department of Neuroscience; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Trieste, Italy; 2ELETTRA Laboratory; Sincrotrone Trieste S.C.p.A., Trieste, Italy; Keywords: alpha-synuclein, aggregation, Parkinson disease, prion-like features 14 Aggregation of α-synuclein plays a crucial role in for the protein in protecting neuronal cells against damage. In the pathogenesis of synucleinopathies, a group of particular, α-synuclein may cooperate with the cysteine-string neurodegenerative diseases including Parkinson disease protein-α (CSPα)—a synaptic vesicle protein with co-chaperone (PD), dementia with Lewy bodies (DLB), diffuse Lewy body activity—in preventing neurodegeneration.15 disease (DLBD) and multiple system atrophy (MSA). The Structurally, α-synuclein features an N-terminal domain common feature of these diseases is a pathological deposition (residues 1–60), a central hydrophobic portion denoted as non-Aβ of protein aggregates, known as Lewy bodies (LBs) in the component of Alzheimer diseases amyloid (NAC, residues 61–95) central nervous system. The major component of these and a C-terminal negatively charged region (residues 96–140). aggregates is α-synuclein, a natively unfolded protein, which Both the N-terminal and NAC domains contain six highly may undergo dramatic structural changes resulting in the conserved hexameric (KTKEGV) motifs (Fig. 1A and B).16 In its formation of β-sheet rich assemblies.
    [Show full text]
  • Impairment of Axonal Development and of Synaptogenesis in Hippocampal Neurons of Synapsin I-Deficient Mice
    Proc. Natl. Acad. Sci. USA Vol. 92, pp. 9230-9234, September 1995 Neurobiology Impairment of axonal development and of synaptogenesis in hippocampal neurons of synapsin I-deficient mice LIH-SHEN CHIN*, LIAN LI*t, ADRIANA FERREIRAt, KENNETH S. KOSIKt, AND PAUL GREENGARD* *Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10021; and 1Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 Contributed by Paul Greengard, June 13, 1995 ABSTRACT Synapsin I, the most abundant of all neuro- 6-kb Sst I-Sma I genomic fragment containing the 5' flanking nal phosphoproteins, is enriched in synaptic vesicles. It has sequence of exon 1 and a 3-kb BamHI fragment containing the been hypothesized to regulate synaptogenesis and neurotrans- 3' flanking sequence of exon 1 into the pPNT vector, which mitter release from adult nerve terminals. The evidence for contains the thymidine kinase gene and the neomycin-resistance such roles has been highly suggestive but not compelling. To gene (11). E14 ES cells (12) were transfected with 50 ,ug of evaluate the possible involvement of synapsin I in synapto- linearized targeting construct by electroporation using a Bio- genesis and in the function of adult synapses, we have gener- Rad Gene Pulser at 800 V and 3 ,F. Selection with G418 at ated synapsin I-deficient mice by homologous recombination. 150 ,ug/ml and 0.2 ,uM 1-(2-deoxy, 2-fluoro-f3-D-arabino- We report herein that outgrowth of predendritic neurites and furanosyl)-5-iodouracil (FIAU, Bristol-Meyers) was made of axons was severely retarded in the hippocampal neurons of 24 h after electroporation, and double-resistant clones were embryonic synapsin I mutant mice.
    [Show full text]
  • Targeted De-Repression of Neuronal Nrf2 Inhibits Α-Synuclein Accumulation Paul S
    Baxter et al. Cell Death and Disease (2021) 12:218 https://doi.org/10.1038/s41419-021-03507-z Cell Death & Disease ARTICLE Open Access Targeted de-repression of neuronal Nrf2 inhibits α-synuclein accumulation Paul S. Baxter1,2,NóraM.Márkus1,2,OwenDando1,2,3,XinHe1,2,BashayerR.Al-Mubarak1,2, Jing Qiu1,2 and Giles E. Hardingham 1,2 Abstract Many neurodegenerative diseases are associated with neuronal misfolded protein accumulation, indicating a need for proteostasis-promoting strategies. Here we show that de-repressing the transcription factor Nrf2, epigenetically shut- off in early neuronal development, can prevent protein aggregate accumulation. Using a paradigm of α-synuclein accumulation and clearance, we find that the classical electrophilic Nrf2 activator tBHQ promotes endogenous Nrf2- dependent α-synuclein clearance in astrocytes, but not cortical neurons, which mount no Nrf2-dependent transcriptional response. Moreover, due to neuronal Nrf2 shut-off and consequent weak antioxidant defences, electrophilic tBHQ actually induces oxidative neurotoxicity, via Nrf2-independent Jun induction. However, we find that epigenetic de-repression of neuronal Nrf2 enables them to respond to Nrf2 activators to drive α-synuclein clearance. Moreover, activation of neuronal Nrf2 expression using gRNA-targeted dCas9-based transcriptional activation complexes is sufficient to trigger Nrf2-dependent α-synuclein clearance. Thus, targeting reversal of the developmental shut-off of Nrf2 in forebrain neurons may alter neurodegenerative disease trajectory by boosting proteostasis. 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; Introduction Importantly, Nrf2 has also recently been found to be The transcription factor Nrf2 is a widely expressed capable of regulating proteostasis, both directly and indir- stress-responsive regulator of several aspects of homo- ectly, through a number of gain- and loss-of-function stu- eostatic physiology.
    [Show full text]