63
Views
8
CrossRef citations to date
0
Altmetric
Review

Identification of autoantigens in pediatric opsoclonus-myoclonus syndrome

, &
Pages 975-982 | Published online: 10 Jan 2014

References

  • Kinsbourne M. Myoclonic encephalopathy of infants. J. Neurol. Neurosurg. Psychiatr.25(2), 271–279 (1962).
  • Solomon G, Churtorian AM. Opsoclonus and occult neuroblastoma. New Engl. J. Med.279(9), 475–477 (1968).
  • Dyken P, Kolar O. Dancing eyes, dancing feet: infantile polymyoclonia. Brain91(2), 305–320 (1968).
  • Plantaz D, Michon J, Valteau-Couanet D et al. [Opsoclonus–myoclonus syndrome associated with non-metastatic neuroblastoma. Long-term survival. Study of the French Society of Pediatric Oncologists]. Arch. Pediatr.7(6), 621–628 (2000).
  • Mitchell WG, Davalos-Gonzalez Y, Brumm VL et al. Opsoclonus–ataxia caused by childhood neuroblastoma: developmental and neurologic sequelae. Pediatrics109(1), 86–98 (2002).
  • Tate ED, Allison TJ, Pranzatelli MR, Verhulst SJ. Neuroepidemiologic trends in 105 US cases of pediatric opsoclonus–myoclonus syndrome. J. Pediatr. Oncol. Nurs.22(1), 8–19 (2005).
  • Rudnick E, Khakoo Y, Antunes NL et al. Opsoclonus–myoclonus–ataxia syndrome in neuroblastoma: clinical outcome and antineuronal antibodies – a report from the Children’s Cancer Group Study. Med. Pediatr. Oncol.36(6), 612–622 (2001).
  • Altman AJ, Baehner RL. Favorable prognosis for survival in children with coincident opso–myoclonus and neuroblastoma. Cancer37(2), 846–852 (1976).
  • Pranzatelli MR. The immunopharmacology of the opsoclonus–myoclonus syndrome. Clin. Neuropharmacol.19(1), 1–47 (1996).
  • Rostasy K, Wilken B, Baumann M et al. High dose pulsatile dexamethasone therapy in children with opsoclonus–myoclonus syndrome. Neuropediatrics37(5), 291–295 (2006).
  • Ertle F, Behnisch W, Al Mulla NA et al. Treatment of neuroblastoma-related opsoclonus–myoclonus–ataxia syndrome with high-dose dexamethasone pulses. Pediatr. Blood Cancer DOI 10.1002/pbc.21107 (2007) (Epub ahead of print).
  • Pranzatelli MR, Tate ED, Travelstead AL et al. Rituximab (anti-CD20) adjunctive therapy for opsoclonus–myoclonus syndrome. J. Pediatr. Hematol. Oncol.28(9), 585–593 (2006).
  • Armstrong MB, Robertson PL, Castle VP. Delayed, recurrent opsoclonus–myoclonus syndrome responding to plasmapheresis. Pediatr. Neurol.33(5), 365–367 (2005).
  • Russo C, Cohn SL, Petruzzi MJ, de Alarcon PA. Long-term neurologic outcome in children with opsoclonus–myoclonus associated with neuroblastoma: a report from the Pediatric Oncology Group. Med. Pediatr. Oncol.28(4), 284–288 (1997).
  • Straube A, Leigh RJ, Bronstein A et al. EFNS task force – therapy of nystagmus and oscillopsia. Eur. J. Neurol.11(2), 83–89 (2004).
  • Caviness JN. Myoclonus. Mayo Clin. Proc.71(7), 679–688 (1996).
  • Ridley A, Kennard C, Scholtz CL et al. Omnipause neurons in two cases of opsoclonus associated with oat cell carcinoma of the lung. Brain110(Pt 6), 1699–1709 (1987).
  • Kaneko CR. Effect of ibotenic acid lesions of the omnipause neurons on saccadic eye movements in rhesus macaques. J. Neurophysiol.75(6), 2229–2242 (1996).
  • Wong AM, Musallam S, Tomlinson RD, Shannon P, Sharpe JA. Opsoclonus in three dimensions: oculographic, neuropathologic and modelling correlates. J. Neurol. Sci.189(1–2), 71–81 (2001).
  • Ramat S, Leigh RJ, Zee DS, Optican LM. What clinical disorders tell us about the neural control of saccadic eye movements. Brain130(Pt 1), 10–35 (2007).
  • Blain PG, Nightingale S, Stoddart JC. Strychnine poisoning: abnormal eye movements. J. Toxicol.19(2), 215–217 (1982).
  • Tuchman RF, Alvarez LA, Kantrowitz AB et al. Opsoclonus–myoclonus syndrome: correlation of radiographic and pathological observations. Neuroradiology31(3), 250–252 (1989).
  • Ziter FA, Bray PF, Cancilla PA. Neuropathologic findings in a patient with neuroblastoma and myoclonic encephalopathy. Arch. Neurol.36(1), 51 (1979).
  • Hayward K, Jeremy RJ, Jenkins S et al. Long-term neurobehavioral outcomes in children with neuroblastoma and opsoclonus–myoclonus–ataxia syndrome: relationship to MRI findings and anti-neuronal antibodies. J. Pediatr.139(4), 552–559 (2001).
  • Helmchen C, Rambold H, Sprenger A, Erdmann C, Binkofski F. Cerebellar activation in opsoclonus: an fMRI study. Neurology61(3), 412–415 (2003).
  • Oguro K, Kobayashi J, Aiba H, Hojo H. Opsoclonus–myoclonus syndrome with abnormal single photon emission computed tomography imaging. Pediatr. Neurol.16(4), 334–336 (1997).
  • van Toorn R, Rabie H, Warwick JM. Opsoclonus–myoclonus in an HIV-infected child on antiretroviral therapy – possible immune reconstitution inflammatory syndrome. Eur. J. Paediatr. Neurol.9(6), 423–426 (2005).
  • Bellur SN. Opsoclonus: its clinical value. Neurology25(6), 502–507 (1975).
  • Kostulas VK, Link H, Lefvert AK. Oligoclonal IgG bands in cerebrospinal fluid. Principles for demonstration and interpretation based on findings in 1114 neurological patients. Arch. Neurol.44(10), 1041–1044 (1987).
  • Pranzatelli MR, Travelstead AL, Tate ED et al. B- and T-cell markers in opsoclonus–myoclonus syndrome: immunophenotyping of CSF lymphocytes. Neurology62(9), 1526–1532 (2004).
  • Hero B, Berthold F. Neuroblastoma. Monatsschr. Kinderheilkund.150(6), 775–788 (2002).
  • Martin RF, Beckwith JB. Lymphoid infiltrates in neuroblastomas: their occurrence and prognostic significance. J. Pediatr. Surg.3(1), 161–164 (1968).
  • Cooper R, Khakoo Y, Matthay KK et al. Opsoclonus–myoclonus–ataxia syndrome in neuroblastoma: histopathologic features – a report from the Children’s Cancer Group. Med. Pediatr. Oncol.36(6), 623–629 (2001).
  • Gambini C, Conte M, Bernini G et al. Neuroblastic tumors associated with opsoclonus–myoclonus syndrome: histological, immunohistochemical and molecular features of 15 Italian cases. Virchows Arch.442(6), 555–562 (2003).
  • Ollert MW, David K, Vollmert C et al. Mechanisms of in vivo anti-neuroblastoma activity of human natural IgM. Eur. J. Cancer33(12), 1942–1948 (1997).
  • David K, Ehrhardt A, Ollert MW et al. Expression of a 260 kDa neuroblastoma surface antigen, the target of cytotoxic natural human IgM: correlation to MYCN amplification and effects of retinoic acid. Eur. J. Cancer33(12), 1937–1941 (1997).
  • Korfei M, Fuhlhuber V, Schmidt-Woll T et al. Functional characterisation of autoantibodies from patients with pediatric opsoclonus–myoclonus-syndrome. J. Neuroimmunol.170(1–2), 150–157 (2005).
  • Cohn SL, Salwen H, Herst CV et al. Single copies of the N-myc oncogene in neuroblastomas from children presenting with the syndrome of opsoclonus–myoclonus. Cancer62(4), 723–726 (1988).
  • Blaes F. Immunotherapeutic approaches to paraneoplastic neurological disorders. Expert Opin. Biol. Ther.2(4), 419–430 (2002).
  • Fisher PG, Wechsler DS, Singer HS. Anti-Hu antibody in a neuroblastoma-associated paraneoplastic syndrome. Pediatr. Neurol.10(4), 309–312 (1994).
  • Graus F, Keime-Guibert F, Rene R et al. Anti-Hu-associated paraneoplastic encephalomyelitis: analysis of 200 patients. Brain124(Pt 6), 1138–1148 (2001).
  • Dalmau J, Furneaux HM, Rosenblum MK, Graus F, Posner JB. Detection of the anti-Hu antibody in specific regions of the nervous system and tumor from patients with paraneoplastic encephalomyelitis/ sensory neuronopathy. Neurology41(11), 1757–1764 (1991).
  • Wang X, Tanaka Hall TM. Structural basis for recognition of AU-rich element RNA by the HuD protein. Nat. Struct. Biol.8(2), 141–145 (2001).
  • Mobarak CD, Anderson KD, Morin M et al. The RNA-binding protein HuD is required for GAP-43 mRNA stability, GAP-43 gene expression, and PKC-dependent neurite outgrowth in PC12 cells. Mol. Biol. Cell11(9), 3191–3203 (2000).
  • Deschenes-Furry J, Mousavi K, Bolognani F et al. The RNA-binding protein HuD binds acetylcholinesterase mRNA in neurons and regulates its expression after axotomy. J. Neurosci.27(3), 665–675 (2007).
  • Antunes NL, Khakoo Y, Matthay KK et al. Antineuronal antibodies in patients with neuroblastoma and paraneoplastic opsoclonus-myoclonus. J. Pediatr. Hematol. Oncol.22(4), 315–320 (2000).
  • Schafer KH, Klotz M, Mergner D et al. IgG-mediated cytotoxicity to myenteric plexus cultures in patients with paraneoplastic neurological syndromes. J. Autoimmun.15(4), 479–484 (2000).
  • Tanaka K, Tanaka M, Onodera O et al. Passive transfer and active immunization with the recombinant leucine-zipper (Yo) protein as an attempt to establish an animal model of paraneoplastic cerebellar degeneration. J. Neurol. Sci.127(2), 153–158 (1994).
  • Buckanovich RJ, Yang YY, Darnell RB. The onconeural antigen Nova-1 is a neuron-specific RNA-binding protein, the activity of which is inhibited by paraneoplastic antibodies. J. Neurosci.16(3), 1114–1122 (1996).
  • Jensen KB, Dredge BK, Stefani G et al. Nova-1 regulates neuron-specific alternative splicing and is essential for neuronal viability. Neuron25(2), 359–371 (2000).
  • Buckanovich RJ, Darnell RB. The neuronal RNA binding protein Nova-1 recognizes specific RNA targets in vitro and in vivo. Mol. Cell. Biol.17(6), 3194–3201 (1997).
  • Bataller L, Rosenfeld MR, Graus F et al. Autoantigen diversity in the opsoclonus–myoclonus syndrome. Ann. Neurol.53(3), 347–353 (2003).
  • Redlitz A, Fowler BJ, Plow EF, Miles LA. The role of an enolase-related molecule in plasminogen binding to cells. Eur. J. Biochem.227(1–2), 407–415 (1995).
  • Nakajima K, Hamanoue M, Takemoto N et al. Plasminogen binds specifically to α-enolase on rat neuronal plasma membrane. J. Neurochem.63(6), 2048–2057 (1994).
  • Bergmann S, Rohde M, Chhatwal GS, Hammerschmidt S. α-enolase of Streptococcus pneumoniae is a plasmin(ogen)-binding protein displayed on the bacterial cell surface. Mol. Microbiol.40(6), 1273–1287 (2001).
  • Dot C, Guigay J, Adamus G. Anti-α-enolase antibodies in cancer-associated retinopathy with small cell carcinoma of the lung. Am. J. Ophthalmol.139(4), 746–747 (2005).
  • Fujii A, Yoneda M, Ito T et al. Autoantibodies against the amino terminal of α-enolase are a useful diagnostic marker of Hashimoto’s encephalopathy. J. Neuroimmunol.162(1–2), 130–136 (2005).
  • Linker K, Pautz A, Fechir M et al. Involvement of KSRP in the post-transcriptional regulation of human iNOS expression–complex interplay of KSRP with TTP and HuR. Nucleic Acids Res.33(15), 4813–4827 (2005).
  • Gherzi R, Trabucchi M, Ponassi M et al. The RNA-binding protein KSRP promotes decay of β-catenin mRNA and is inactivated by PI3K–AKT signaling. PLoS Biol.5(1), e5 (2006).
  • Rehbein M, Wege K, Buck F et al. Molecular characterization of MARTA1, a protein interacting with the dendritic targeting element of MAP2 mRNAs. J. Neurochem.82(5), 1039–1046 (2002).
  • Boeckers TM. The postsynaptic density. Cell. Tissue Res.326(2), 409–422 (2006).
  • Noetzel MJ, Cawley LP, James VL, Minard BJ, Agrawal HC. Anti-neurofilament protein antibodies in opsoclonus–myoclonus. J. Neuroimmunol.15(2), 137–145 (1987).
  • Stefansson K, Marton LS, Dieperink ME et al. Circulating autoantibodies to the 200,000-dalton protein of neurofilaments in the serum of healthy individuals. Science228(4703), 1117–1119 (1985).
  • Braxton DB, Williams M, Kamali D et al. Specificity of human anti-neurofilament autoantibodies. J. Neuroimmunol.21(2–3), 193–203 (1989).
  • Salih AM, Nixon NB, Dawes PT, Mattey DL. Prevalence of antibodies to neurofilament polypeptides in patients with rheumatoid arthritis complicated by peripheral neuropathy. Clin. Exp. Rheumatol.16(6), 689–694 (1998).
  • Connolly AM, Pestronk A, Mehta S, Pranzatelli MR 3rd, Noetzel MJ. Serum autoantibodies in childhood opsoclonus–myoclonus syndrome: an analysis of antigenic targets in neural tissues. J. Pediatr.130(6), 878–884 (1997).
  • Candler PM, Dale RC, Griffin S et al. Post-streptococcal opsoclonus–myoclonus syndrome associated with anti-neuroleukin antibodies. J. Neurol. Neurosurg. Psychiatr.77(4), 507–512 (2006).
  • Blaes F, Beeson D, Plested P, Lang B, Vincent A. IgG from “seronegative” myasthenia gravis patients binds to a muscle cell line, TE671, but not to human acetylcholine receptor. Ann. Neurol.47(4), 504–510 (2000).
  • Lily O, Palace J, Vincent A. Serum autoantibodies to cell surface determinants in multiple sclerosis: a flow cytometric study. Brain127(Pt 2), 269–279 (2004).
  • Blaes F, Fuhlhuber V, Korfei M et al. Surface-binding autoantibodies to cerebellar neurons in opsoclonus syndrome. Ann. Neurol.58(2), 313–317 (2005).
  • Lee KH, Cho HJ, Kim HS et al. Activation of extracellular signal regulated kinase 1/2 in human dermal microvascular endothelial cells stimulated by anti-endothelial cell antibodies in sera of patients with Behcet’s disease. J. Dermatol. Sci.30(1), 63–72 (2002).
  • Beck S, Fuhlhuber V, Krasenbrink I et al. IgG subclass distribution of autoantibodies in pediatric opsoclonus–myoclonus syndrome. J. Neuroimmunol.185(1–2), 145–149 (2007).
  • Ichiba N, Miyake Y, Sato K, Oda M, Kimoto H. Mumps-induced opsoclonus–myoclonus and ataxia. Pediatr. Neurol.4(4), 224–227 (1988).
  • Kuban KC, Ephros MA, Freeman RL, Laffell LB, Bresnan MJ. Syndrome of opsoclonus-myoclonus caused by Coxsackie B3 infection. Ann. Neurol.13(1), 69–71 (1983).
  • Delreux V, Kevers L, Callewaert A, Sindic C. Opsoclonus secondary to an Epstein–Barr virus infection. Arch. Neurol.46(5), 480–481 (1989).
  • Khosla JS, Edelman MJ, Kennedy N, Reich SG. West Nile virus presenting as opsoclonus–myoclonus cerebellar ataxia. Neurology64(6), 1095 (2005).
  • Chemli J, Ketata S, Dalhoumi A et al. [Opsoclonus–myoclonus syndrome associated with Mycoplasma pneumoniae infection]. Arch. Pediatr.14(8), 1003–1006 (2007).
  • Wilfong AA, Parke JT, McCrary JA 3rd. Opsoclonus–myoclonus with Beckwith–Wiedemann syndrome and hepatoblastoma. Pediatr. Neurol.8(1), 77–79 (1992).
  • Anderson NE, Budde-Steffen C, Rosenblum MK et al. Opsoclonus, myoclonus, ataxia, and encephalopathy in adults with cancer: a distinct paraneoplastic syndrome. Medicine67(2), 100–109 (1988).
  • Averbuch-Heller L, Remler B. Opsoclonus. Semin. Neurol.16(1), 21–26 (1996).
  • Scharf D. Opsoclonus–myoclonus following the intranasal usage of cocaine. J. Neurol. Neurosurg. Psychiatr.52(12), 1447–1448 (1989).
  • Au WJ, Keltner JL. Opsoclonus with amitriptyline overdose. Ann. Neurol.6(1), 87 (1979).
  • Cohen WJ, Cohen NH. Lithium carbonate, haloperidol, and irreversible brain damage. JAMA230(9), 1283–1287 (1974).
  • Buckanovich RJ, Posner JB, Darnell RB. Nova, the paraneoplastic Ri antigen, is homologous to an RNA-binding protein and is specifically expressed in the developing motor system. Neuron11(4), 657–672 (1993).
  • Kirsten A, Beck S, Fühlhuber V et al. New autoantibodies in paediatric opsoclonus-myoclonus syndrome. Ann. NY Acad. Sci.1110, 256–260 (2007).
  • Deconinck N, Scaillon M, Segers V, Groswasser JJ, Dan B. Opsoclonus–myoclonus associated with celiac disease. Pediatr. Neurol.34(4), 312–314 (2006).
  • Blaes F, Jauss M, Kraus J et al. Adult paraneoplastic opsoclonus–myoclonus syndrome associated with antimitochondrial autoantibodies. J. Neurol. Neurosurg. Psychiatr.74(11), 1595–1596 (2003).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.