Skip to main content
Top
Published in: Acta Neuropathologica 5/2014

01-05-2014 | Original Paper

Myopathy in Marinesco–Sjögren syndrome links endoplasmic reticulum chaperone dysfunction to nuclear envelope pathology

Authors: Andreas Roos, Stephan Buchkremer, Laxmikanth Kollipara, Thomas Labisch, Christian Gatz, Manuela Zitzelsberger, Eva Brauers, Kay Nolte, J. Michael Schröder, Janbernd Kirschner, Christopher Marvin Jesse, Hans Hilmar Goebel, Anand Goswami, Richard Zimmermann, René Peiman Zahedi, Jan Senderek, Joachim Weis

Published in: Acta Neuropathologica | Issue 5/2014

Login to get access

Abstract

Marinesco–Sjögren syndrome (MSS) features cerebellar ataxia, mental retardation, cataracts, and progressive vacuolar myopathy with peculiar myonuclear alterations. Most MSS patients carry homozygous or compound heterozygous SIL1 mutations. SIL1 is a nucleotide exchange factor for the endoplasmic reticulum resident chaperone BiP which controls a plethora of essential processes in the endoplasmic reticulum. In this study we made use of the spontaneous Sil1 mouse mutant woozy to explore pathomechanisms leading to Sil1 deficiency-related skeletal muscle pathology. We found severe, progressive myopathy characterized by alterations of the sarcoplasmic reticulum, accumulation of autophagic vacuoles, mitochondrial changes, and prominent myonuclear pathology including nuclear envelope and nuclear lamina alterations. These abnormalities were remarkably similar to the myopathy in human patients with MSS. In particular, the presence of perinuclear membranous structures which have been reported as an ultrastructural hallmark of MSS-related myopathy could be confirmed in woozy muscles. We found that these structures are derived from the nuclear envelope and nuclear lamina and associate with proliferations of the sarcoplasmic reticulum. In line with impaired function of BiP secondary to loss of its nucleotide exchange factor Sil1, we observed activation of the unfolded protein response and the endoplasmic-reticulum-associated protein degradation-pathway. Despite initiation of the autophagy–lysosomal system, autophagic clearance was found ineffective which is in agreement with the formation of autophagic vacuoles. This report identifies woozy muscle as a faithful phenocopy of the MSS myopathy. Moreover, we provide a link between two well-established disease mechanisms in skeletal muscle, dysfunction of chaperones and nuclear envelope pathology.
Appendix
Available only for authorised users
Literature
1.
go back to reference Agbulut O, Destombes J, Thiesson D, Butler-Browne G (2000) Age-related appearance of tubular aggregates in the skeletal muscle of almost all male inbred mice. Histochem Cell Biol 114(6):477–481PubMed Agbulut O, Destombes J, Thiesson D, Butler-Browne G (2000) Age-related appearance of tubular aggregates in the skeletal muscle of almost all male inbred mice. Histochem Cell Biol 114(6):477–481PubMed
3.
go back to reference Anttonen AK, Mahjneh I, Hamalainen RH, Lagier-Tourenne C, Kopra O, Waris L, Anttonen M, Joensuu T, Kalimo H, Paetau A, Tranebjaerg L, Chaigne D, Koenig M, Eeg-Olofsson O, Udd B, Somer M, Somer H, Lehesjoki AE (2005) The gene disrupted in Marinesco–Sjogren syndrome encodes SIL1, an HSPA5 cochaperone. Nat Genet 37(12):1309–1311. doi:10.1038/ng1677 PubMedCrossRef Anttonen AK, Mahjneh I, Hamalainen RH, Lagier-Tourenne C, Kopra O, Waris L, Anttonen M, Joensuu T, Kalimo H, Paetau A, Tranebjaerg L, Chaigne D, Koenig M, Eeg-Olofsson O, Udd B, Somer M, Somer H, Lehesjoki AE (2005) The gene disrupted in Marinesco–Sjogren syndrome encodes SIL1, an HSPA5 cochaperone. Nat Genet 37(12):1309–1311. doi:10.​1038/​ng1677 PubMedCrossRef
5.
6.
go back to reference Bjorkoy G, Lamark T, Johansen T (2006) p62/SQSTM1: a missing link between protein aggregates and the autophagy machinery. Autophagy 2(2):138–139. (pii:2405)PubMed Bjorkoy G, Lamark T, Johansen T (2006) p62/SQSTM1: a missing link between protein aggregates and the autophagy machinery. Autophagy 2(2):138–139. (pii:2405)PubMed
8.
go back to reference Dechat T, Korbei B, Vaughan OA, Vlcek S, Hutchison CJ, Foisner R (2000) Lamina-associated polypeptide 2alpha binds intranuclear A-type lamins. J Cell Sci 113(Pt 19):3473–3484PubMed Dechat T, Korbei B, Vaughan OA, Vlcek S, Hutchison CJ, Foisner R (2000) Lamina-associated polypeptide 2alpha binds intranuclear A-type lamins. J Cell Sci 113(Pt 19):3473–3484PubMed
10.
go back to reference Dubowitz V, Sewry CA, Oldfors A (2013) Muscle biopsy. A practical approach, Saunders Dubowitz V, Sewry CA, Oldfors A (2013) Muscle biopsy. A practical approach, Saunders
12.
go back to reference Eskelinen EL, Schmidt CK, Neu S, Willenborg M, Fuertes G, Salvador N, Tanaka Y, Lullmann-Rauch R, Hartmann D, Heeren J, von Figura K, Knecht E, Saftig P (2004) Disturbed cholesterol traffic but normal proteolytic function in LAMP-1/LAMP-2 double-deficient fibroblasts. Mol Biol Cell 15(7):3132–3145. doi:10.1091/mbc.E04-02-0103 PubMedCentralPubMedCrossRef Eskelinen EL, Schmidt CK, Neu S, Willenborg M, Fuertes G, Salvador N, Tanaka Y, Lullmann-Rauch R, Hartmann D, Heeren J, von Figura K, Knecht E, Saftig P (2004) Disturbed cholesterol traffic but normal proteolytic function in LAMP-1/LAMP-2 double-deficient fibroblasts. Mol Biol Cell 15(7):3132–3145. doi:10.​1091/​mbc.​E04-02-0103 PubMedCentralPubMedCrossRef
15.
go back to reference Goto Y, Komiyama A, Tanabe Y, Katafuchi Y, Ohtaki E, Nonaka I (1990) Myopathy in Marinesco–Sjogren syndrome: an ultrastructural study. Acta Neuropathol 80(2):123–128PubMedCrossRef Goto Y, Komiyama A, Tanabe Y, Katafuchi Y, Ohtaki E, Nonaka I (1990) Myopathy in Marinesco–Sjogren syndrome: an ultrastructural study. Acta Neuropathol 80(2):123–128PubMedCrossRef
16.
18.
go back to reference Harting I, Blaschek A, Wolf NI, Seitz A, Haupt M, Goebel HH, Rating D, Sartor K, Ebinger F (2004) T2-hyperintense cerebellar cortex in Marinesco–Sjogren syndrome. Neurology 63(12):2448–2449. doi:63/12/2448 PubMedCrossRef Harting I, Blaschek A, Wolf NI, Seitz A, Haupt M, Goebel HH, Rating D, Sartor K, Ebinger F (2004) T2-hyperintense cerebellar cortex in Marinesco–Sjogren syndrome. Neurology 63(12):2448–2449. doi:63/​12/​2448 PubMedCrossRef
19.
go back to reference Herva R, von Wendt L, von Wendt G, Saukkonen AL, Leisti J, Dubowitz V (1987) A syndrome with juvenile cataract, cerebellar atrophy, mental retardation and myopathy. Neuropediatrics 18(3):164–169. doi:10.1055/s-2008-1052473 PubMedCrossRef Herva R, von Wendt L, von Wendt G, Saukkonen AL, Leisti J, Dubowitz V (1987) A syndrome with juvenile cataract, cerebellar atrophy, mental retardation and myopathy. Neuropediatrics 18(3):164–169. doi:10.​1055/​s-2008-1052473 PubMedCrossRef
20.
go back to reference Hoyer-Hansen M, Bastholm L, Szyniarowski P, Campanella M, Szabadkai G, Farkas T, Bianchi K, Fehrenbacher N, Elling F, Rizzuto R, Mathiasen IS, Jaattela M (2007) Control of macroautophagy by calcium, calmodulin-dependent kinase kinase-beta, and Bcl-2. Mol Cell 25(2):193–205. doi:10.1016/j.molcel.2006.12.009 PubMedCrossRef Hoyer-Hansen M, Bastholm L, Szyniarowski P, Campanella M, Szabadkai G, Farkas T, Bianchi K, Fehrenbacher N, Elling F, Rizzuto R, Mathiasen IS, Jaattela M (2007) Control of macroautophagy by calcium, calmodulin-dependent kinase kinase-beta, and Bcl-2. Mol Cell 25(2):193–205. doi:10.​1016/​j.​molcel.​2006.​12.​009 PubMedCrossRef
21.
go back to reference Izawa I, Nishizawa M, Ohtakara K, Ohtsuka K, Inada H, Inagaki M (2000) Identification of Mrj, a DnaJ/Hsp40 family protein, as a keratin 8/18 filament regulatory protein. J Biol Chem 275(44):34521–34527. doi:10.1074/jbc.M003492200 PubMedCrossRef Izawa I, Nishizawa M, Ohtakara K, Ohtsuka K, Inada H, Inagaki M (2000) Identification of Mrj, a DnaJ/Hsp40 family protein, as a keratin 8/18 filament regulatory protein. J Biol Chem 275(44):34521–34527. doi:10.​1074/​jbc.​M003492200 PubMedCrossRef
22.
go back to reference Jarosch E, Taxis C, Volkwein C, Bordallo J, Finley D, Wolf DH, Sommer T (2002) Protein dislocation from the ER requires polyubiquitination and the AAA-ATPase Cdc48. Nat Cell Biol 4(2):134–139. doi:10.1038/ncb746 PubMedCrossRef Jarosch E, Taxis C, Volkwein C, Bordallo J, Finley D, Wolf DH, Sommer T (2002) Protein dislocation from the ER requires polyubiquitination and the AAA-ATPase Cdc48. Nat Cell Biol 4(2):134–139. doi:10.​1038/​ncb746 PubMedCrossRef
24.
go back to reference Kobayashi T, Ohta Y (2003) Enforced expression of oxygen-regulated protein, ORP150, induces vacuolar degeneration in mouse myocardium. Transgenic Res 12(1):13–22PubMedCrossRef Kobayashi T, Ohta Y (2003) Enforced expression of oxygen-regulated protein, ORP150, induces vacuolar degeneration in mouse myocardium. Transgenic Res 12(1):13–22PubMedCrossRef
26.
go back to reference Kollmann K, Damme M, Markmann S, Morelle W, Schweizer M, Hermans-Borgmeyer I, Rochert AK, Pohl S, Lubke T, Michalski JC, Kakela R, Walkley SU, Braulke T (2012) Lysosomal dysfunction causes neurodegeneration in mucolipidosis II ‘knock-in’ mice. Brain 135(Pt 9):2661–2675. doi:10.1093/brain/aws209 PubMedCentralPubMedCrossRef Kollmann K, Damme M, Markmann S, Morelle W, Schweizer M, Hermans-Borgmeyer I, Rochert AK, Pohl S, Lubke T, Michalski JC, Kakela R, Walkley SU, Braulke T (2012) Lysosomal dysfunction causes neurodegeneration in mucolipidosis II ‘knock-in’ mice. Brain 135(Pt 9):2661–2675. doi:10.​1093/​brain/​aws209 PubMedCentralPubMedCrossRef
27.
go back to reference Komatsu M, Waguri S, Koike M, Sou YS, Ueno T, Hara T, Mizushima N, Iwata J, Ezaki J, Murata S, Hamazaki J, Nishito Y, Iemura S, Natsume T, Yanagawa T, Uwayama J, Warabi E, Yoshida H, Ishii T, Kobayashi A, Yamamoto M, Yue Z, Uchiyama Y, Kominami E, Tanaka K (2007) Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice. Cell 131(6):1149–1163. doi:10.1016/j.cell.2007.10.035 PubMedCrossRef Komatsu M, Waguri S, Koike M, Sou YS, Ueno T, Hara T, Mizushima N, Iwata J, Ezaki J, Murata S, Hamazaki J, Nishito Y, Iemura S, Natsume T, Yanagawa T, Uwayama J, Warabi E, Yoshida H, Ishii T, Kobayashi A, Yamamoto M, Yue Z, Uchiyama Y, Kominami E, Tanaka K (2007) Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice. Cell 131(6):1149–1163. doi:10.​1016/​j.​cell.​2007.​10.​035 PubMedCrossRef
28.
go back to reference Komiyama A, Nonaka I, Hirayama K (1989) Muscle pathology in Marinesco–Sjogren syndrome. J Neurol Sci 89(1):103–113PubMedCrossRef Komiyama A, Nonaka I, Hirayama K (1989) Muscle pathology in Marinesco–Sjogren syndrome. J Neurol Sci 89(1):103–113PubMedCrossRef
29.
go back to reference Krieger M, Roos A, Stendel C, Claeys KG, Sonmez FM, Baudis M, Bauer P, Bornemann A, de Goede C, Dufke A, Finkel RS, Goebel HH, Häussler M, Kingston H, Kirschner J, Medne L, Muschke P, Rivier F, Rudnik-Schöneborn S, Spengler S, Inzana F, Stanzial F, Benedicenti F, Synofzik M, Taratuto AL, Pirra L, Tay SK-H, Topaloglu H, Uyanik G, Wand D, Williams D, Zerres K, Weis J, Senderek J (2013) SIL1 mutations and clinical spectrum in patients with Marinesco–Sjögren syndrome. Brain Accepted for publication Krieger M, Roos A, Stendel C, Claeys KG, Sonmez FM, Baudis M, Bauer P, Bornemann A, de Goede C, Dufke A, Finkel RS, Goebel HH, Häussler M, Kingston H, Kirschner J, Medne L, Muschke P, Rivier F, Rudnik-Schöneborn S, Spengler S, Inzana F, Stanzial F, Benedicenti F, Synofzik M, Taratuto AL, Pirra L, Tay SK-H, Topaloglu H, Uyanik G, Wand D, Williams D, Zerres K, Weis J, Senderek J (2013) SIL1 mutations and clinical spectrum in patients with Marinesco–Sjögren syndrome. Brain Accepted for publication
30.
go back to reference Kudlow BA, Kennedy BK, Monnat RJ Jr (2007) Werner and Hutchinson–Gilford progeria syndromes: mechanistic basis of human progeroid diseases. Nat Rev Mol Cell Biol 8(5):394–404. doi:10.1038/nrm2161 PubMedCrossRef Kudlow BA, Kennedy BK, Monnat RJ Jr (2007) Werner and Hutchinson–Gilford progeria syndromes: mechanistic basis of human progeroid diseases. Nat Rev Mol Cell Biol 8(5):394–404. doi:10.​1038/​nrm2161 PubMedCrossRef
31.
34.
35.
go back to reference Liu GH, Qu J, Suzuki K, Nivet E, Li M, Montserrat N, Yi F, Xu X, Ruiz S, Zhang W, Wagner U, Kim A, Ren B, Li Y, Goebl A, Kim J, Soligalla RD, Dubova I, Thompson J, Yates J 3rd, Esteban CR, Sancho-Martinez I, Izpisua Belmonte JC (2012) Progressive degeneration of human neural stem cells caused by pathogenic LRRK2. Nature 491(7425):603–607. doi:10.1038/nature11557 PubMedCentralPubMedCrossRef Liu GH, Qu J, Suzuki K, Nivet E, Li M, Montserrat N, Yi F, Xu X, Ruiz S, Zhang W, Wagner U, Kim A, Ren B, Li Y, Goebl A, Kim J, Soligalla RD, Dubova I, Thompson J, Yates J 3rd, Esteban CR, Sancho-Martinez I, Izpisua Belmonte JC (2012) Progressive degeneration of human neural stem cells caused by pathogenic LRRK2. Nature 491(7425):603–607. doi:10.​1038/​nature11557 PubMedCentralPubMedCrossRef
36.
41.
go back to reference Marinescu G, Draganesco S, Vasiliu D (1931) Nouvelle maladie familiale caractérisée par une cataracte congénitale et un arrêt du développement somato-neuro-psychique. Encéphale Marinescu G, Draganesco S, Vasiliu D (1931) Nouvelle maladie familiale caractérisée par une cataracte congénitale et un arrêt du développement somato-neuro-psychique. Encéphale
42.
go back to reference Munro S, Pelham HR (1986) An Hsp70-like protein in the ER: identity with the 78 kd glucose-regulated protein and immunoglobulin heavy chain binding protein. Cell 46(2):291–300. doi:0092-8674(86)90746-4 PubMedCrossRef Munro S, Pelham HR (1986) An Hsp70-like protein in the ER: identity with the 78 kd glucose-regulated protein and immunoglobulin heavy chain binding protein. Cell 46(2):291–300. doi:0092-8674(86)90746-4 PubMedCrossRef
44.
go back to reference Nijholt DA, de Graaf TR, van Haastert ES, Oliveira AO, Berkers CR, Zwart R, Ovaa H, Baas F, Hoozemans JJ, Scheper W (2011) Endoplasmic reticulum stress activates autophagy but not the proteasome in neuronal cells: implications for Alzheimer’s disease. Cell Death Differ 18(6):1071–1081. doi:10.1038/cdd.2010.176 PubMedCentralPubMedCrossRef Nijholt DA, de Graaf TR, van Haastert ES, Oliveira AO, Berkers CR, Zwart R, Ovaa H, Baas F, Hoozemans JJ, Scheper W (2011) Endoplasmic reticulum stress activates autophagy but not the proteasome in neuronal cells: implications for Alzheimer’s disease. Cell Death Differ 18(6):1071–1081. doi:10.​1038/​cdd.​2010.​176 PubMedCentralPubMedCrossRef
45.
go back to reference Nishikawa S, Brodsky JL, Nakatsukasa K (2005) Roles of molecular chaperones in endoplasmic reticulum (ER) quality control and ER-associated degradation (ERAD). J Biochem 137(5):551–555. doi:10.1093/jb/mvi068 PubMedCrossRef Nishikawa S, Brodsky JL, Nakatsukasa K (2005) Roles of molecular chaperones in endoplasmic reticulum (ER) quality control and ER-associated degradation (ERAD). J Biochem 137(5):551–555. doi:10.​1093/​jb/​mvi068 PubMedCrossRef
47.
go back to reference Oh SH, Lim SC (2009) Endoplasmic reticulum stress-mediated autophagy/apoptosis induced by capsaicin (8-methyl-N-vanillyl-6-nonenamide) and dihydrocapsaicin is regulated by the extent of c-Jun NH2-terminal kinase/extracellular signal-regulated kinase activation in WI38 lung epithelial fibroblast cells. J Pharmacol Exp Ther 329(1):112–122. doi:10.1124/jpet.108.144113 PubMedCrossRef Oh SH, Lim SC (2009) Endoplasmic reticulum stress-mediated autophagy/apoptosis induced by capsaicin (8-methyl-N-vanillyl-6-nonenamide) and dihydrocapsaicin is regulated by the extent of c-Jun NH2-terminal kinase/extracellular signal-regulated kinase activation in WI38 lung epithelial fibroblast cells. J Pharmacol Exp Ther 329(1):112–122. doi:10.​1124/​jpet.​108.​144113 PubMedCrossRef
48.
go back to reference Orenstein SJ, Kuo SH, Tasset I, Arias E, Koga H, Fernandez-Carasa I, Cortes E, Honig LS, Dauer W, Consiglio A, Raya A, Sulzer D, Cuervo AM (2013) Interplay of LRRK2 with chaperone-mediated autophagy. Nat Neurosci 16(4):394–406. doi:10.1038/nn.3350 PubMedCentralPubMedCrossRef Orenstein SJ, Kuo SH, Tasset I, Arias E, Koga H, Fernandez-Carasa I, Cortes E, Honig LS, Dauer W, Consiglio A, Raya A, Sulzer D, Cuervo AM (2013) Interplay of LRRK2 with chaperone-mediated autophagy. Nat Neurosci 16(4):394–406. doi:10.​1038/​nn.​3350 PubMedCentralPubMedCrossRef
49.
go back to reference Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, Overvatn A, Bjorkoy G, Johansen T (2007) p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem 282(33):24131–24145. doi:10.1074/jbc.M702824200 PubMedCrossRef Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, Overvatn A, Bjorkoy G, Johansen T (2007) p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem 282(33):24131–24145. doi:10.​1074/​jbc.​M702824200 PubMedCrossRef
50.
go back to reference Park YE, Hayashi YK, Bonne G, Arimura T, Noguchi S, Nonaka I, Nishino I (2009) Autophagic degradation of nuclear components in mammalian cells. Autophagy 5(6):795–804. doi:8901 PubMed Park YE, Hayashi YK, Bonne G, Arimura T, Noguchi S, Nonaka I, Nishino I (2009) Autophagic degradation of nuclear components in mammalian cells. Autophagy 5(6):795–804. doi:8901 PubMed
52.
go back to reference Rabinovich E, Kerem A, Frohlich KU, Diamant N, Bar-Nun S (2002) AAA-ATPase p97/Cdc48p, a cytosolic chaperone required for endoplasmic reticulum-associated protein degradation. Mol Cell Biol 22(2):626–634PubMedCentralPubMedCrossRef Rabinovich E, Kerem A, Frohlich KU, Diamant N, Bar-Nun S (2002) AAA-ATPase p97/Cdc48p, a cytosolic chaperone required for endoplasmic reticulum-associated protein degradation. Mol Cell Biol 22(2):626–634PubMedCentralPubMedCrossRef
55.
go back to reference Sarparanta J, Jonson PH, Golzio C, Sandell S, Luque H, Screen M, McDonald K, Stajich JM, Mahjneh I, Vihola A, Raheem O, Penttila S, Lehtinen S, Huovinen S, Palmio J, Tasca G, Ricci E, Hackman P, Hauser M, Katsanis N, Udd B (2012) Mutations affecting the cytoplasmic functions of the co-chaperone DNAJB6 cause limb-girdle muscular dystrophy. Nat Genet 44(4):450–455. doi:10.1038/ng.1103 S451–452PubMedCentralPubMedCrossRef Sarparanta J, Jonson PH, Golzio C, Sandell S, Luque H, Screen M, McDonald K, Stajich JM, Mahjneh I, Vihola A, Raheem O, Penttila S, Lehtinen S, Huovinen S, Palmio J, Tasca G, Ricci E, Hackman P, Hauser M, Katsanis N, Udd B (2012) Mutations affecting the cytoplasmic functions of the co-chaperone DNAJB6 cause limb-girdle muscular dystrophy. Nat Genet 44(4):450–455. doi:10.​1038/​ng.​1103 S451–452PubMedCentralPubMedCrossRef
56.
go back to reference Sasaki K, Suga K, Tsugawa S, Sakuma K, Tachi N, Chiba S, Imamura S (1996) Muscle pathology in Marinesco–Sjogren syndrome: a unique ultrastructural feature. Brain Dev 18(1):64–67. doi:0387760495000968 PubMedCrossRef Sasaki K, Suga K, Tsugawa S, Sakuma K, Tachi N, Chiba S, Imamura S (1996) Muscle pathology in Marinesco–Sjogren syndrome: a unique ultrastructural feature. Brain Dev 18(1):64–67. doi:0387760495000968​ PubMedCrossRef
57.
go back to reference Schröder JM (1982) Pathologie der Muskulatur. Spezielle pathologische Anatomie, vol 15. Springer Verlag, Berlin Schröder JM (1982) Pathologie der Muskulatur. Spezielle pathologische Anatomie, vol 15. Springer Verlag, Berlin
59.
go back to reference Senderek J, Garvey SM, Krieger M, Guergueltcheva V, Urtizberea A, Roos A, Elbracht M, Stendel C, Tournev I, Mihailova V, Feit H, Tramonte J, Hedera P, Crooks K, Bergmann C, Rudnik-Schoneborn S, Zerres K, Lochmuller H, Seboun E, Weis J, Beckmann JS, Hauser MA, Jackson CE (2009) Autosomal-dominant distal myopathy associated with a recurrent missense mutation in the gene encoding the nuclear matrix protein, matrin 3. Am J Hum Genet 84(4):511–518PubMedCentralPubMedCrossRef Senderek J, Garvey SM, Krieger M, Guergueltcheva V, Urtizberea A, Roos A, Elbracht M, Stendel C, Tournev I, Mihailova V, Feit H, Tramonte J, Hedera P, Crooks K, Bergmann C, Rudnik-Schoneborn S, Zerres K, Lochmuller H, Seboun E, Weis J, Beckmann JS, Hauser MA, Jackson CE (2009) Autosomal-dominant distal myopathy associated with a recurrent missense mutation in the gene encoding the nuclear matrix protein, matrin 3. Am J Hum Genet 84(4):511–518PubMedCentralPubMedCrossRef
60.
go back to reference Senderek J, Krieger M, Stendel C, Bergmann C, Moser M, Breitbach-Faller N, Rudnik-Schoneborn S, Blaschek A, Wolf NI, Harting I, North K, Smith J, Muntoni F, Brockington M, Quijano-Roy S, Renault F, Herrmann R, Hendershot LM, Schroder JM, Lochmuller H, Topaloglu H, Voit T, Weis J, Ebinger F, Zerres K (2005) Mutations in SIL1 cause Marinesco–Sjogren syndrome, a cerebellar ataxia with cataract and myopathy. Nat Genet 37(12):1312–1314PubMedCrossRef Senderek J, Krieger M, Stendel C, Bergmann C, Moser M, Breitbach-Faller N, Rudnik-Schoneborn S, Blaschek A, Wolf NI, Harting I, North K, Smith J, Muntoni F, Brockington M, Quijano-Roy S, Renault F, Herrmann R, Hendershot LM, Schroder JM, Lochmuller H, Topaloglu H, Voit T, Weis J, Ebinger F, Zerres K (2005) Mutations in SIL1 cause Marinesco–Sjogren syndrome, a cerebellar ataxia with cataract and myopathy. Nat Genet 37(12):1312–1314PubMedCrossRef
63.
go back to reference Sjogren T (1950) Hereditary congenital spinocerebellar ataxia accompanied by congenital cataract and oligophrenia; a genetic and clinical investigation. Confin Neurol 10(5):293–308PubMed Sjogren T (1950) Hereditary congenital spinocerebellar ataxia accompanied by congenital cataract and oligophrenia; a genetic and clinical investigation. Confin Neurol 10(5):293–308PubMed
64.
go back to reference Su TR, Tsai FJ, Lin JJ, Huang HH, Chiu CC, Su JH, Yang YT, Chen JY, Wong BS, Wu YJ (2012) Induction of apoptosis by 11-dehydrosinulariolide via mitochondrial dysregulation and ER stress pathways in human melanoma cells. Mar Drugs 10(8):1883–1898. doi:10.3390/md10081883 PubMedCentralPubMedCrossRef Su TR, Tsai FJ, Lin JJ, Huang HH, Chiu CC, Su JH, Yang YT, Chen JY, Wong BS, Wu YJ (2012) Induction of apoptosis by 11-dehydrosinulariolide via mitochondrial dysregulation and ER stress pathways in human melanoma cells. Mar Drugs 10(8):1883–1898. doi:10.​3390/​md10081883 PubMedCentralPubMedCrossRef
65.
go back to reference Superneau DW, Wertelecki W, Zellweger H, Bastian F (1987) Myopathy in Marinesco–Sjogren syndrome. Eur Neurol 26(1):8–16PubMedCrossRef Superneau DW, Wertelecki W, Zellweger H, Bastian F (1987) Myopathy in Marinesco–Sjogren syndrome. Eur Neurol 26(1):8–16PubMedCrossRef
66.
go back to reference Suzuki Y, Murakami N, Goto Y, Orimo S, Komiyama A, Kuroiwa Y, Nonaka I (1997) Apoptotic nuclear degeneration in Marinesco–Sjogren syndrome. Acta Neuropathol 94(5):410–415PubMedCrossRef Suzuki Y, Murakami N, Goto Y, Orimo S, Komiyama A, Kuroiwa Y, Nonaka I (1997) Apoptotic nuclear degeneration in Marinesco–Sjogren syndrome. Acta Neuropathol 94(5):410–415PubMedCrossRef
67.
go back to reference Taylor MR, Slavov D, Gajewski A, Vlcek S, Ku L, Fain PR, Carniel E, Di Lenarda A, Sinagra G, Boucek MM, Cavanaugh J, Graw SL, Ruegg P, Feiger J, Zhu X, Ferguson DA, Bristow MR, Gotzmann J, Foisner R, Mestroni L (2005) Thymopoietin (lamina-associated polypeptide 2) gene mutation associated with dilated cardiomyopathy. Hum Mutat 26(6):566–574. doi:10.1002/humu.20250 PubMedCrossRef Taylor MR, Slavov D, Gajewski A, Vlcek S, Ku L, Fain PR, Carniel E, Di Lenarda A, Sinagra G, Boucek MM, Cavanaugh J, Graw SL, Ruegg P, Feiger J, Zhu X, Ferguson DA, Bristow MR, Gotzmann J, Foisner R, Mestroni L (2005) Thymopoietin (lamina-associated polypeptide 2) gene mutation associated with dilated cardiomyopathy. Hum Mutat 26(6):566–574. doi:10.​1002/​humu.​20250 PubMedCrossRef
68.
go back to reference Tirasophon W, Lee K, Callaghan B, Welihinda A, Kaufman RJ (2000) The endoribonuclease activity of mammalian IRE1 autoregulates its mRNA and is required for the unfolded protein response. Genes Dev 14(21):2725–2736PubMedCentralPubMedCrossRef Tirasophon W, Lee K, Callaghan B, Welihinda A, Kaufman RJ (2000) The endoribonuclease activity of mammalian IRE1 autoregulates its mRNA and is required for the unfolded protein response. Genes Dev 14(21):2725–2736PubMedCentralPubMedCrossRef
71.
go back to reference Villa A, Podini P, Nori A, Panzeri MC, Martini A, Meldolesi J, Volpe P (1993) The endoplasmic reticulum-sarcoplasmic reticulum connection. II. Postnatal differentiation of the sarcoplasmic reticulum in skeletal muscle fibers. Exp Cell Res 209(1):140–148. doi:S0014482783712942 PubMedCrossRef Villa A, Podini P, Nori A, Panzeri MC, Martini A, Meldolesi J, Volpe P (1993) The endoplasmic reticulum-sarcoplasmic reticulum connection. II. Postnatal differentiation of the sarcoplasmic reticulum in skeletal muscle fibers. Exp Cell Res 209(1):140–148. doi:S001448278371294​2 PubMedCrossRef
72.
go back to reference Villa A, Podini P, Panzeri MC, Soling HD, Volpe P, Meldolesi J (1993) The endoplasmic-sarcoplasmic reticulum of smooth muscle: immunocytochemistry of vas deferens fibers reveals specialized subcompartments differently equipped for the control of Ca2+ homeostasis. J Cell Biol 121(5):1041–1051PubMedCrossRef Villa A, Podini P, Panzeri MC, Soling HD, Volpe P, Meldolesi J (1993) The endoplasmic-sarcoplasmic reticulum of smooth muscle: immunocytochemistry of vas deferens fibers reveals specialized subcompartments differently equipped for the control of Ca2+ homeostasis. J Cell Biol 121(5):1041–1051PubMedCrossRef
73.
go back to reference Vlcek S, Dechat T, Foisner R (2001) Nuclear envelope and nuclear matrix: interactions and dynamics. Cell Mol Life Sci 58(12–13):1758–1765PubMedCrossRef Vlcek S, Dechat T, Foisner R (2001) Nuclear envelope and nuclear matrix: interactions and dynamics. Cell Mol Life Sci 58(12–13):1758–1765PubMedCrossRef
75.
go back to reference Weis J, Dimpfel W, Schroder JM (1995) Nerve conduction changes and fine structural alterations of extra- and intrafusal muscle and nerve fibers in streptozotocin diabetic rats. Muscle Nerve 18(2):175–184PubMedCrossRef Weis J, Dimpfel W, Schroder JM (1995) Nerve conduction changes and fine structural alterations of extra- and intrafusal muscle and nerve fibers in streptozotocin diabetic rats. Muscle Nerve 18(2):175–184PubMedCrossRef
79.
80.
82.
go back to reference Zhao L, Longo-Guess C, Harris BS, Lee JW, Ackerman SL (2005) Protein accumulation and neurodegeneration in the woozy mutant mouse is caused by disruption of SIL1, a cochaperone of BiP. Nat Genet 37(9):974–979. doi:10.1038/ng1620 PubMedCrossRef Zhao L, Longo-Guess C, Harris BS, Lee JW, Ackerman SL (2005) Protein accumulation and neurodegeneration in the woozy mutant mouse is caused by disruption of SIL1, a cochaperone of BiP. Nat Genet 37(9):974–979. doi:10.​1038/​ng1620 PubMedCrossRef
Metadata
Title
Myopathy in Marinesco–Sjögren syndrome links endoplasmic reticulum chaperone dysfunction to nuclear envelope pathology
Authors
Andreas Roos
Stephan Buchkremer
Laxmikanth Kollipara
Thomas Labisch
Christian Gatz
Manuela Zitzelsberger
Eva Brauers
Kay Nolte
J. Michael Schröder
Janbernd Kirschner
Christopher Marvin Jesse
Hans Hilmar Goebel
Anand Goswami
Richard Zimmermann
René Peiman Zahedi
Jan Senderek
Joachim Weis
Publication date
01-05-2014
Publisher
Springer Berlin Heidelberg
Published in
Acta Neuropathologica / Issue 5/2014
Print ISSN: 0001-6322
Electronic ISSN: 1432-0533
DOI
https://doi.org/10.1007/s00401-013-1224-4

Other articles of this Issue 5/2014

Acta Neuropathologica 5/2014 Go to the issue

Acknowledgement to Referees

Acknowledgement to referees