Analysis of Proteins That Rapidly Change Upon Mechanistic

Total Page:16

File Type:pdf, Size:1020Kb

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. Using a tein synthesis. To determine the role of mTORC1 activa- mouse model of tuberous sclerosis complex, a disease tion in protein expression, we have used an unbiased, that displays both epilepsy and autism spectrum disorder large-scale proteomic approach. We provide evidence phenotypes and has overactive mTORC1 signaling, that a brief repression of mTORC1 activity in vivo by ra- we show that Parkinson protein 7 protein is elevated in pamycin has little effect globally, yet leads to a significant the dendrites and colocalizes with the postsynaptic remodeling of synaptic proteins, in particular those pro- marker postsynaptic density-95. Our work offers a com- teins that reside in the postsynaptic density. We have also prehensive view of mTORC1 and its role in regulating re- found that curtailing the activity of mTORC1 bidirection- gional protein expression in normal and diseased states. Molecular & Cellular Proteomics 15: 10.1074/ From the ‡Center for Learning and Memory, University of Texas, mcp.M115.055079, 426–444, 2016. Austin, 1 University Station C7000, Texas 78712; §Institute for Cell and Molecular Biology, University of Texas, Austin, Texas; ¶Institute for Neuroscience, University of Texas, Austin, Texas; ʈWaggoner The mechanistic/mammalian target of rapamycin complex Center for Alcohol and Addiction Research, University of Texas, Aus- 1 (mTORC1)1 is a serine/threonine protein kinase that is highly tin, Texas; **Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409; ‡‡Pain Therapeutics, Inc., 7801 N Capital of Texas Hwy, #260, Austin, Texas 78731 1 The abbreviations used are: mTORC1, mechanistic/mammalian Received August 28, 2015, and in revised form, September 15, target of rapamycin complex 1; AD, Alzheimer’s disease; AHA, azi- 2015 dohomoalanine; APP, amyloid precursor protein; ASD, autism spec- Published, MCP Papers in Press, September 29, 2015, DOI trum disorder; BONCAT, bioorthogonal noncanonical amino acid 10.1074/mcp.M115.055079 tagging; CI, confidence interval; cKO, conditional knockout; Cre, Cre- Author contributions: FN and KRG designed research. FN and SN recombinase; DAVID, database for annotation, visualization and inte- conducted experiments and analyzed data. SN performed bioinfor- grated discovery; DIV, day in vitro; Dlg4, discs large homolog 4; matics analyses. ES and YM conducted mass spectrometry analyses. DMSO, dimethyl sulfoxide; EASE, expression analysis systematic GS facilitated mass spectrometry experiments and provided technical explorer; FDA, Food and Drug Administration; GAP-43, growth-asso- advice. GAD and BVZ provided the virus and performed stereotaxic ciated protein-43; GO, gene ontology; GRIN, glutamate receptor, injections. FN, SN, and KRG wrote the manuscript. ionotropic NMDA subtype; ICC, immunocytochemistry; GluN, gluta- 426 Molecular & Cellular Proteomics 15.2 mTORC1 Rapidly Shifts Expression of Disease Proteomes expressed in many cell types (1). In the brain, mTORC1 tightly in protein expression. Herein, we provide evidence that coordinates different synaptic plasticities — long-term poten- mTORC1 activation bidirectionally regulates protein expres- tiation (LTP) and long-term depression (LTD) — the molecular sion, especially in the PSD where roughly an equal distribution correlates of learning and memory (2–5). Because mTORC1 is of proteins dynamically appear and disappear. Remarkably, at the core of many synaptic signaling pathways downstream using protein–protein interaction networks facilitated the of glutamate and neurotrophin receptors, many hypothesize novel discovery that PARK7, a protein thus far only implicated that dysregulated mTORC1 signaling underlies cognitive def- in Parkinson’s disease, (1) is up-regulated by increased icits observed in several neurodegenerative diseases (3, mTORC1 activity, (2) resides in the PSD only when mTORC1 6–17). For example, mTORC1 and its downstream targets are is active, and (3) is aberrantly expressed in a rodent model of hyperactive in human brains diagnosed with Alzheimer’s dis- TSC, an mTORC1-related disease that has symptoms of ep- ease (AD) (18–20). Additionally in animal models of autism ilepsy and autism. Collectively, these data provide the first spectrum disorder (ASD), altered mTORC1 signaling contrib- comprehensive list of proteins whose abundance or subcel- utes to the observed synaptic dysfunction and aberrant net- lular distributions are altered with acute changes in mTORC1 work connectivity (13, 15, 21–27). Furthermore, epilepsy, activity in vivo. which is common in AD and ASD, has enhanced mTORC1 activity (28–32). EXPERIMENTAL PROCEDURES Phosphorylation of mTORC1, considered the active form, is Sample Preparation—We used three sets of paired sibling male generally regarded to promote protein synthesis (33). Thus, Sprague-Dawley rats (seven to 9-weeks old) that were housed to- many theorize that diseases with overactive mTORC1 arise gether. Within each pair, one received rapamycin (10 mg/kg) and the from excessive protein synthesis (14). Emerging data, how- other received an equal volume of DMSO (carrier, control) via intra- ever, show that suppressing mTORC1 activation can trigger peritoneal (intraperitoneal) injection. After one hour, the animals were sacrificed. Cortices were homogenized, nuclei were pelleted by low local translation in neurons (34, 35). Pharmacological antag- speed centrifugation (100 ϫ g), and the resulting supernatant was onism of N-methyl-D-aspartate (NMDA) receptors, a subtype analyzed as cell lysates (L). To obtain synaptoneurosomes, homoge- of glutamate receptors that lies upstream of mTOR activation, nized cortices were processed as described (39). An aliquot of the promotes the synthesis of the voltage-gated potassium chan- synaptoneurosome fraction from each animal was solubilized in 1% ϫ nel, Kv1.1, in dendrites (34, 35). Consistent with these results, triton X-100 (10min) and centrifuged (12,000 g) to yield a triton X-100-soluble fraction (soluble, S) and a triton X-100-insoluble frac- in models of temporal lobe epilepsy there is a reduction in the tion (pellet, PSD). To prepare protein for LC-MS/MS, lysates, soluble, expression of voltage-gated ion channels including Kv1.1 (30, and PSD fractions were further solubilized in SDS-sample buffer. 31, 36). Interestingly in a model of focal neocortical epilepsy, SDS-solubilized fractions were run on a 10% SDS-polyacrylamide gel overexpression of Kv1.1 blocked seizure activity (37). Be- (4min). The migration was stopped as the ladder began to separate. cause both active and inactive mTORC1 permit protein syn- Gel plugs containing the sample were sectioned and sent for mass spectrometry analysis. Animal experiments were performed accord- thesis, we sought to determine the proteins whose expression ing to the National Institutes of Health’s Guide for the Care and Use is altered when mTORC1 phosphorylation is reduced in vivo. of Laboratory Animals and approved by the UT-Austin Institutional Rapamycin is an FDA-approved, immunosuppressive drug Care and Usage Committee. that inhibits mTORC1 activity (38). We capitalized on the Western Blot Analysis—Proteins were separated by SDS-poly- ability of rapamycin to reduce mTORC1 activity in vivo and the acrylamide gel electrophoresis (PAGE). To visualize the proteins, we used the following antibodies: rabbit anti-phospho-mTOR Ser2448 unbiased approach of mass spectrometry to identify changes (1:2000; Cell Signaling, Danvers, MA), mouse anti-mTOR (1:5000; Life Technologies, Grand Island, NY), mouse anti-Park7 (1:2000; Novus Biologicals, Littleton, CO) mouse anti-PSD-95 (1:10,000; NeuroMab, mate receptor, NMDA subtype; KAR, kainic
Recommended publications
  • RNA-Binding Protein Network Alteration Causes Aberrant Axon
    bioRxiv preprint doi: https://doi.org/10.1101/2020.08.26.268631; this version posted August 26, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 RNA-binding protein network alteration causes aberrant axon 2 branching and growth phenotypes in FUS ALS mutant motoneurons 3 4 Maria Giovanna Garone1, Nicol Birsa2,3, Maria Rosito4, Federico Salaris1,4, Michela Mochi1, Valeria 5 de Turris4, Remya R. Nair5, Thomas J. Cunningham5, Elizabeth M. C. Fisher2, Pietro Fratta2 and 6 Alessandro Rosa1,4,6,* 7 8 1. Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, P.le 9 A. Moro 5, 00185 Rome, Italy 10 2. UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, 11 UK 12 3. UK Dementia Research Institute, University College London, London, WC1E 6BT, UK 13 4. Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 14 Rome, Italy 15 5. MRC Harwell Institute, Oxfordshire, OX11 0RD, UK 16 6. Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of 17 Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Viale Regina Elena 18 291, 00161 Rome, Italy 19 20 * Corresponding author: [email protected]; Tel: +39-0649255218 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.08.26.268631; this version posted August 26, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.
    [Show full text]
  • 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]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [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]
  • DNA Methylation, Mechanisms of FMR1 Inactivation and Therapeutic Perspectives for Fragile X Syndrome
    biomolecules Review DNA Methylation, Mechanisms of FMR1 Inactivation and Therapeutic Perspectives for Fragile X Syndrome Veronica Nobile 1, Cecilia Pucci 1, Pietro Chiurazzi 1,2 , Giovanni Neri 1,3 and Elisabetta Tabolacci 1,* 1 Sezione di Medicina Genomica, Dipartimento Scienze della Vita e Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; [email protected] (V.N.); [email protected] (C.P.); [email protected] (P.C.); [email protected] (G.N.) 2 Fondazione Policlinico Universitario A. Gemelli IRCCS, UOC Genetica Medica, 00168 Rome, Italy 3 Greenwood Genetic Center, JC Self Research Institute, Greenwood, SC 29646, USA * Correspondence: [email protected]; Tel.: +39-06-30154606 Abstract: Among the inherited causes of intellectual disability and autism, Fragile X syndrome (FXS) is the most frequent form, for which there is currently no cure. In most FXS patients, the FMR1 gene is epigenetically inactivated following the expansion over 200 triplets of a CGG repeat (FM: full mutation). FMR1 encodes the Fragile X Mental Retardation Protein (FMRP), which binds several mRNAs, mainly in the brain. When the FM becomes methylated at 10–12 weeks of gestation, the FMR1 gene is transcriptionally silent. The molecular mechanisms involved in the epigenetic silencing are not fully elucidated. Among FXS families, there is a rare occurrence of males carrying a FM, which remains active because it is not methylated, thus ensuring enough FMRPs to allow for an intellectual development within normal range. Which mechanisms are responsible for sparing these individuals from being affected by FXS? In order to answer this critical question, which may have possible implications for FXS therapy, several potential epigenetic mechanisms have been described.
    [Show full text]
  • Therapeutic Strategies in Fragile X Syndrome: Dysregulated Mglur Signaling and Beyond
    Neuropsychopharmacology REVIEWS (2012) 37, 178–195 & 2012 American College of Neuropsychopharmacology All rights reserved 0893-133X/12 ............................................................................................................................................................... REVIEW 178 www.neuropsychopharmacology.org Therapeutic Strategies in Fragile X Syndrome: Dysregulated mGluR Signaling and Beyond 1 ,2 ,1,3 Christina Gross , Elizabeth M Berry-Kravis* and Gary J Bassell* 1 2 Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA; Departments of Pediatrics, Neurology, 3 and Biochemistry, Rush University Medical Center, Chicago, IL, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA Fragile X syndrome (FXS) is an inherited neurodevelopmental disease caused by loss of function of the fragile X mental retardation protein (FMRP). In the absence of FMRP, signaling through group 1 metabotropic glutamate receptors is elevated and insensitive to stimulation, which may underlie many of the neurological and neuropsychiatric features of FXS. Treatment of FXS animal models with negative allosteric modulators of these receptors and preliminary clinical trials in human patients support the hypothesis that metabotropic glutamate receptor signaling is a valuable therapeutic target in FXS. However, recent research has also shown that FMRP may regulate diverse aspects of neuronal signaling downstream of several cell surface receptors, suggesting a possible new route to more direct disease-targeted therapies. Here, we summarize promising recent advances in basic research identifying and testing novel therapeutic strategies in FXS models, and evaluate their potential therapeutic benefits. We provide an overview of recent and ongoing clinical trials motivated by some of these findings, and discuss the challenges for both basic science and clinical applications in the continued development of effective disease mechanism-targeted therapies for FXS.
    [Show full text]
  • Role of Phosphodiesterases in the Pathophysiology of Neurodevelopmental Disorders
    Molecular Psychiatry https://doi.org/10.1038/s41380-020-00997-9 REVIEW ARTICLE Role of phosphodiesterases in the pathophysiology of neurodevelopmental disorders 1 2 Sébastien Delhaye ● Barbara Bardoni Received: 4 July 2020 / Revised: 3 December 2020 / Accepted: 9 December 2020 © The Author(s), under exclusive licence to Springer Nature Limited part of Springer Nature 2021. This article is published with open access Abstract Phosphodiesterases (PDEs) are enzymes involved in the homeostasis of both cAMP and cGMP. They are members of a family of proteins that includes 11 subfamilies with different substrate specificities. Their main function is to catalyze the hydrolysis of cAMP, cGMP, or both. cAMP and cGMP are two key second messengers that modulate a wide array of intracellular processes and neurobehavioral functions, including memory and cognition. Even if these enzymes are present in all tissues, we focused on those PDEs that are expressed in the brain. We took into consideration genetic variants in patients affected by neurodevelopmental disorders, phenotypes of animal models, and pharmacological effects of PDE inhibitors, a class of drugs in rapid evolution and increasing application to brain disorders. Collectively, these data indicate the potential 1234567890();,: 1234567890();,: of PDE modulators to treat neurodevelopmental diseases characterized by learning and memory impairment, alteration of behaviors associated with depression, and deficits in social interaction. Indeed, clinical trials are in progress to treat patients with Alzheimer’s disease, schizophrenia, depression, and autism spectrum disorders. Among the most recent results, the application of some PDE inhibitors (PDE2A, PDE3, PDE4/4D, and PDE10A) to treat neurodevelopmental diseases, including autism spectrum disorders and intellectual disability, is a significant advance, since no specific therapies are available for these disorders that have a large prevalence.
    [Show full text]
  • Altered Gut Microbiota in a Fragile X Syndrome Mouse Model
    fnins-15-653120 May 26, 2021 Time: 10:27 # 1 ORIGINAL RESEARCH published: 26 May 2021 doi: 10.3389/fnins.2021.653120 Altered Gut Microbiota in a Fragile X Syndrome Mouse Model Francisco Altimiras1,2*, José Antonio Garcia1, Ismael Palacios-García3,4, Michael J. Hurley5, Robert Deacon6,7, Bernardo González8,9 and Patricia Cogram6,7* 1 Faculty of Engineering, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile, 2 Faculty of Engineering and Business, Universidad de las Américas, Santiago, Chile, 3 School of Psychology, Pontificia Universidad Católica de Chile, Santiago, Chile, 4 Centro de Estudios en Neurociencia Humana y Neuropsicología, Facultad de Psicología, Universidad Diego Portales, Santiago, Chile, 5 Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom, 6 Department of Genetics, Institute of Ecology and Biodiversity (IEB), Faculty of Sciences, Universidad de Chile, Santiago, Chile, 7 FRAXA-DVI, FRAXA Research Foundation, Santiago, Chile, 8 Faculty of Engineering and Sciences, Universidad Adolfo Ibáñez, Santiago, Chile, 9 Center of Applied Ecology and Sustainability (CAPES), Santiago, Chile The human gut microbiome is the ecosystem of microorganisms that live in the human digestive system. Several studies have related gut microbiome variants to metabolic, immune and nervous system disorders. Fragile X syndrome (FXS) is a neurodevelopmental disorder considered the most common cause of inherited intellectual disability and the leading monogenetic cause of autism.
    [Show full text]
  • Impaired Presynaptic Long-Term Potentiation in the Anterior Cingulate Cortex of Fmr1 Knock-Out Mice
    The Journal of Neuroscience, February 4, 2015 • 35(5):2033–2043 • 2033 Cellular/Molecular Impaired Presynaptic Long-Term Potentiation in the Anterior Cingulate Cortex of Fmr1 Knock-out Mice Kohei Koga,1,2* Ming-Gang Liu,1,3* Shuang Qiu,1,2* Qian Song,1,2 Gerile O’Den,2 Tao Chen,1,2,4 and Min Zhuo1,2 1Department of Physiology, Faculty of Medicine, University of Toronto, Medical Science Building, Toronto, Ontario, M5S 1A8, Canada, 2Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi’an Jiaotong University, Xi’an, Shanxi 710049, China, 3Department of Anatomy and Histology and Embryology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China, and 4Department of Anatomy and KK Leung Brain Research Center, Fourth Military Medical University, Xi’an, Shanxi 710032, China Fragile X syndrome is a common inherited form of mental impairment. Fragile X mental retardation protein (FMRP) plays important roles in the regulation of synaptic protein synthesis, and loss of FMRP leads to deficits in learning-related synaptic plasticity and behavioraldisability.Previousstudiesmostlyfocusonpostsynapticlong-termpotentiation(LTP)inFmr1knock-out(KO)mice.Here,we investigatetheroleofFMRPinpresynapticLTP(pre-LTP)intheadultmouseanteriorcingulatecortex(ACC).Low-frequencystimulation induced LTP in layer II/III pyramidal neurons under the voltage-clamp mode. Paired-pulse ratio, which is a parameter for presynaptic changes, was decreased after the low-frequency stimulation in Fmr1 wild-type (WT) mice. Cingulate pre-LTP was abolished in Fmr1 KO mice. We also used a 64-electrode array system for field EPSP recording and found that the combination of low-frequency stimulation paired with a GluK1-containing kainate receptor agonist induced NMDA receptor-independent and metabotropic glutamate receptor- dependentpre-LTPintheWTmice.ThispotentiationwasblockedinFmr1KOmice.BiochemicalexperimentsshowedthatFmr1KOmice displayed altered translocation of protein kinase A subunits in the ACC.
    [Show full text]
  • Npp2014167.Pdf
    Neuropsychopharmacology (2014) 39, 3100–3111 & 2014 American College of Neuropsychopharmacology. All rights reserved 0893-133X/14 www.neuropsychopharmacology.org Reduced Phenotypic Severity Following Adeno-Associated Virus-Mediated Fmr1 Gene Delivery in Fragile X Mice 1,3 1,3 1 1 Shervin Gholizadeh , Jason Arsenault , Ingrid Cong Yang Xuan , Laura K Pacey and David R Hampson*,1,2 1 2 Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada; Department of Pharmacology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by a trinucleotide repeat expansion in the FMR1 gene that codes for fragile X mental retardation protein (FMRP). To determine if FMRP expression in the central nervous system could reverse phenotypic deficits in the Fmr1 knockout (KO) mouse model of FXS, we used a single-stranded adeno-associated viral (AAV) vector with viral capsids from serotype 9 that contained a major isoform of FMRP. FMRP transgene expression was driven by the neuron-selective synapsin-1 promoter. The vector was delivered to the brain via a single bilateral intracerebroventricular injection into neonatal Fmr1 KO mice and transgene expression and behavioral assessments were conducted 22–26 or 50–56 days post injection. Western blotting and immunocytochemical analyses of AAV–FMRP-injected mice revealed FMRP expression in the striatum, hippocampus, retrosplenial B cortex, and cingulate cortex. Cellular expression was selective for neurons and reached 50% of wild-type levels in the hippocampus and cortex at 56 days post injection. The pathologically elevated repetitive behavior and the deficit in social dominance behavior seen in phosphate-buffered saline-injected Fmr1 KO mice were reversed in AAV–FMRP-injected mice.
    [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]