Munc18-1 is a molecular chaperone for α-synuclein, controlling its self-replicating aggregation

Munc18-1 heterozygous mutations are associated with developmental diseases, including early infantile epileptic encephalopathy (EIEE). Chai et al. report that Munc18-1 acts as a chaperone for α-synuclein and controls its aggregative propensity. Munc18-1 EIEE-associated mutations promote the aggregation of endogenous α-synuclein in neurons, leading to a neurodegenerative phenotype.

[1]  A. Kania,et al.  Normal Molecular Specification and Neurodegenerative Disease-Like Death of Spinal Neurons Lacking the SNARE-Associated Synaptic Protein Munc18-1 , 2016, The Journal of Neuroscience.

[2]  D. Geschwind,et al.  Evidence for α-synuclein prions causing multiple system atrophy in humans with parkinsonism , 2015, Proceedings of the National Academy of Sciences.

[3]  J. Danielsson,et al.  SOD1 aggregation in ALS mice shows simplistic test tube behavior , 2015, Proceedings of the National Academy of Sciences.

[4]  Kirill Alexandrov,et al.  Gateway-compatible vectors for high-throughput protein expression in pro- and eukaryotic cell-free systems. , 2015, Journal of biotechnology.

[5]  Nancy T. Malintan,et al.  Increased polyubiquitination and proteasomal degradation of a Munc18-1 disease-linked mutant causes temperature-sensitive defect in exocytosis. , 2014, Cell reports.

[6]  T. Südhof,et al.  α-Synuclein assembles into higher-order multimers upon membrane binding to promote SNARE complex formation , 2014, Proceedings of the National Academy of Sciences.

[7]  Sally Martin,et al.  Secretagogue Stimulation of Neurosecretory Cells Elicits Filopodial Extensions Uncovering New Functional Release Sites , 2013, The Journal of Neuroscience.

[8]  David A. Scott,et al.  Genome engineering using the CRISPR-Cas9 system , 2013, Nature Protocols.

[9]  H. Saibil Chaperone machines for protein folding, unfolding and disaggregation , 2013, Nature Reviews Molecular Cell Biology.

[10]  Yujin E. Kim,et al.  Molecular chaperone functions in protein folding and proteostasis. , 2013, Annual review of biochemistry.

[11]  Nancy T. Malintan,et al.  The Munc18-1 domain 3a loop is essential for neuroexocytosis but not for syntaxin-1A transport to the plasma membrane , 2013, Journal of Cell Science.

[12]  Nancy T. Malintan,et al.  Myosin VI small insert isoform maintains exocytosis by tethering secretory granules to the cortical actin , 2013, The Journal of cell biology.

[13]  F. Scaglia,et al.  Molecular bases and clinical spectrum of early infantile epileptic encephalopathies. , 2012, European journal of medical genetics.

[14]  A. Levey,et al.  Analysis of a membrane‐enriched proteome from postmortem human brain tissue in Alzheimer's disease , 2012, Proteomics. Clinical applications.

[15]  Nancy T. Malintan,et al.  Munc18-1 domain-1 controls vesicle docking and secretion by interacting with syntaxin-1 and chaperoning it to the plasma membrane , 2011, Molecular biology of the cell.

[16]  C. Depienne,et al.  STXBP1‐related encephalopathy presenting as infantile spasms and generalized tremor in three patients , 2011, Epilepsia.

[17]  Kirill Alexandrov,et al.  Leishmania cell-free protein expression system. , 2011, Methods.

[18]  J. Lacaille,et al.  Intellectual disability without epilepsy associated with STXBP1 disruption , 2011, European Journal of Human Genetics.

[19]  Alex Groisman,et al.  Visualizing a one-way protein encounter complex by ultrafast single-molecule mixing , 2011, Nature Methods.

[20]  H. Ikeda,et al.  STXBP1 mutations cause not only Ohtahara syndrome but also West syndrome—Result of Japanese cohort study , 2010, Epilepsia.

[21]  H. Arai,et al.  STXBP1 mutations in early infantile epileptic encephalopathy with suppression‐burst pattern , 2010, Epilepsia.

[22]  I. Scheffer,et al.  Clinical spectrum of early-onset epileptic encephalopathies associated with STXBP1 mutations , 2010, Neurology.

[23]  Nancy T. Malintan,et al.  Rescue of Munc18-1 and -2 double knockdown reveals the essential functions of interaction between Munc18 and closed syntaxin in PC12 cells. , 2009, Molecular Biology of the Cell.

[24]  Kirill Alexandrov,et al.  Species-independent translational leaders facilitate cell-free expression , 2009, Nature Biotechnology.

[25]  Jason J. Lavinder,et al.  Direct single-molecule observation of a protein living in two opposed native structures , 2009, Proceedings of the National Academy of Sciences.

[26]  E. Fombonne,et al.  De novo STXBP1 mutations in mental retardation and nonsyndromic epilepsy , 2009, Annals of neurology.

[27]  Nancy T. Malintan,et al.  Abrogating Munc18-1-SNARE Complex Interaction Has Limited Impact on Exocytosis in PC12 Cells* , 2009, The Journal of Biological Chemistry.

[28]  D. Hall,et al.  An ALS-Linked Mutant SOD1 Produces a Locomotor Defect Associated with Aggregation and Synaptic Dysfunction When Expressed in Neurons of Caenorhabditis elegans , 2009, PLoS genetics.

[29]  C. Elger,et al.  Autoantibodies to Munc18, cerebral plasma cells and B-lymphocytes in Rasmussen encephalitis , 2008, Epilepsy Research.

[30]  Naomichi Matsumoto,et al.  De novo mutations in the gene encoding STXBP1 (MUNC18-1) cause early infantile epileptic encephalopathy , 2008, Nature Genetics.

[31]  Satoshi Orimo,et al.  Axonal α-synuclein aggregates herald centripetal degeneration of cardiac sympathetic nerve in Parkinson's disease , 2008 .

[32]  P. Francis,et al.  Cyclin-dependent kinase 5, Munc18a and Munc18-interacting protein 1/X11α protein up-regulation in Alzheimer’s disease , 2006, Neuroscience.

[33]  B. Hyman,et al.  Alpha-Synuclein and Chaperones in Dementia With Lewy Bodies , 2005, Journal of neuropathology and experimental neurology.

[34]  Jeffrey H. Kordower,et al.  The role of α-synuclein in Parkinson's disease: insights from animal models , 2003, Nature Reviews Neuroscience.

[35]  John Q. Trojanowski,et al.  Chaperone Suppression of α-Synuclein Toxicity in a Drosophila Model for Parkinson's Disease , 2001, Science.

[36]  E. Mori [Dementia with Lewy bodies]. , 2000, Nihon Ronen Igakkai zasshi. Japanese journal of geriatrics.

[37]  Richard H. Scheller,et al.  Three-dimensional structure of the neuronal-Sec1–syntaxin 1a complex , 2000, Nature.

[38]  T. Südhof,et al.  Synaptic assembly of the brain in the absence of neurotransmitter secretion. , 2000, Science.

[39]  J Q Trojanowski,et al.  Axon pathology in Parkinson's disease and Lewy body dementia hippocampus contains alpha-, beta-, and gamma-synuclein. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[40]  M. L. Schmidt,et al.  α-Synuclein in Lewy bodies , 1997, Nature.

[41]  Robert L. Nussbaum,et al.  Mutation in the α-Synuclein Gene Identified in Families with Parkinson's Disease , 1997 .

[42]  R. Scheller,et al.  n-Sec1: a neural-specific syntaxin-binding protein. , 1994, Proceedings of the National Academy of Sciences of the United States of America.

[43]  N. Boddaert,et al.  Early epileptic encephalopathies associated with STXBP1 mutations: Could we better delineate the phenotype? , 2014, European journal of medical genetics.

[44]  Kirill Alexandrov,et al.  Production of eukaryotic cell-free lysate from Leishmania tarentolae. , 2014, Methods in molecular biology.

[45]  Jeffrey H Kordower,et al.  The role of alpha-synuclein in Parkinson's disease: insights from animal models. , 2003, Nature reviews. Neuroscience.

[46]  M. Vidal,et al.  GATEWAY recombinational cloning: application to the cloning of large numbers of open reading frames or ORFeomes. , 2000, Methods in enzymology.

[47]  S E Ide,et al.  Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. , 1997, Science.