Skip to main content

Disorders Primarily Affecting White Matter

  • Chapter
Magnetic Resonance in Dementia

Abstract

There are several viral infections that can lead to dementia (Achim 1998). Some acute viral encephalitides can present with cognitive decline, but more typically chronic viral encephalitides lead to dementia. Apart from herpes simplex encephalitis and tick-borne encephalitis (TBE), most acute encephalitides are very uncommon (e.g., varicella zoster virus, human herpes virus type, other arboviruses). The major part of this section will deal with chronic viral encephalitides leading to dementia. The most common ones are subacute sclerosing panencephalitis (SSPE), HIV-1 encephalitis (HIVE) and progressive multifocal leukencephalopathy (PML). Other chronic virus infections are too obscure to warrant detailed description (e.g., Borna disease virus, HTLV-1 infection of the brain), or will be dealt with elsewhere (e.g., the subacute spongiform encephalitides, such as kuru and Creutzfeldt-Jakob disease; Sect. 4.7).

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 84.99
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 109.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References and Further Reading

  • Achim CL, Wiley CA. Virus-mediated dementias. In: Markesbery WR, ed. Neuropathology of dementing disorders. London: Edward Arnold, 1998: 312–339

    Google Scholar 

  • Ashikaga R, Araki Y, Ishida O. MR FLAIR imaging of herpes simplex encephalitis. Radiat Med, 1996; 14: 349–352

    CAS  PubMed  Google Scholar 

  • Bartolomei F, Gavaret M, Gastaut JA, Gambarelli D, Figarell-Branger D, Gastaut JL. Isolated, chronic, epilepsia partialis continua in an HIV-infected patient. Arch Neurol, 1999; 56: 111–114

    CAS  PubMed  Google Scholar 

  • Berger JR, Levy RM, Flomenhoft D, Dobbs M. Predictive factors for prolonged survival in AIDS: associated PML. J Neurovirol, 1998; 4: 342

    Google Scholar 

  • Budka H. Neuropathology of human immunodeficiency virus infection. Brain Pathol, 1991; 1: 163–175

    CAS  PubMed  Google Scholar 

  • Cattaneo R, Schmid R, Eschle D, Baczko K, ter Meurln V, Biller MA. Biased hypermutation and other genetic changes in defective measles viruses in human brain infections. Cell, 1988; 55: 255–265

    CAS  PubMed  Google Scholar 

  • Chang L, Ernst T. MR spectroscopy and diffusion-weighted MR imaging in focal brain lesions in AIDS. Neuroimaging Clin North Am, 1997; 7: 409–426

    CAS  Google Scholar 

  • Chong WK, Paley M, Wilkinson ID, Hall-Craggs MA, Sweeney B, Harrison MJ, Miller RF, Kendall BE. Localized cerebral proton MR spectroscopy in HIV infection and AIDS. AJNR Am J Neuroradiol, 1994; 15: 21–25

    CAS  PubMed  Google Scholar 

  • Domingues RB, Fink MC, Tsanaclis AM, de Castro CC, Cerri GG, Mayo MS, Lakeman FD. Diagnosis of herpes simplex encephalitis by magnetic resonance imaging and polymerase chain reaction assay of cerebrospinal fluid. J Neurol Sci, 1998; 157: 148–153

    CAS  PubMed  Google Scholar 

  • Dousset V, Armand JP, Huot P, Viaud B, Caille JM. Magnetization transfer imaging in AIDS-related brain diseases. Neuroimaging Clin North Am, 1997; 7: 447–460

    CAS  Google Scholar 

  • Ernst T, Chang L, Witt M, Walot I, Aronow H, Leonido-Yee M, Singer E. Progressive multifocal leukoencephalopathy and human immunodeficiency virus-associated white matter lesions in AIDS: magnetization transfer MR imaging. Radiology, 1999; 210: 539–543

    CAS  PubMed  Google Scholar 

  • Everall IP, Chong WK, Wilkinson ID, Paley MN, Chinn RJ, Hall-Craggs MA, Scaravilli F, Lantos PL, Luthert PJ, Harrison MJ. Correlation of MRI and neuropathology in AIDS. J Neurol Neurosurg Psychiatry, 1997; 62: 92–95

    CAS  PubMed  Google Scholar 

  • Gazzola P, Cocito L, Capello E, Roccatagliata L, Canepa M, Mancardi GL. Subacute measles encephalitis in a young man immunosuppressed for ankylosing spondylitis. Neurology, 1999; 52: 1074–1077

    CAS  PubMed  Google Scholar 

  • Kaiser R. The clinical and epidemiological profile of tickborne encephalitis in southern Germany 1994-98: a prospective study of 656 patients. Brian, 1999; 122: 2067–2078

    Google Scholar 

  • Kato T, Ishii C, Furusho J, Endo T, Tazaki I. Early diagnosis of herpes encephalopathy using fluid-attenuated inversion recovery pulse sequence. Pediatr Neurol, 1998; 19:58–61

    CAS  PubMed  Google Scholar 

  • Meyerhoff DJ, Bloomer C, Cardenas V, Norman D, Weiner MW, Fein G. Elevated subcortical choline metabolites in cognitively and clinically asymptomatic HIV + patients. Neurology, 1999; 52: 995–1003

    CAS  PubMed  Google Scholar 

  • Miller RF, Lucas SB, Hall-Craggs MA, Brink NS, Scaravilli F, Chinn RJ, Kendall BE, Williams IG, Harrison MJ. Comparison of magnetic resonance imaging with neuro-pathological findings in the diagnosis of HIV and CMV associated CNS disease in AIDS. J Neurol Neurosurg Psychiatry, 1997; 62: 346–351

    CAS  PubMed  Google Scholar 

  • Moller HE, Vermathen P, Lentschig MG, Schuierer G, Schwarz S, Wiedermann D, Evers S, Husstedt IW. Metabolic characterization of AIDS dementia complex by spectroscopic imaging. J Magn Reson Imaging, 1999; 9:10–18

    CAS  PubMed  Google Scholar 

  • Post MJ, Yiannoutsos C, Simpson D, Booss J, Clifford DB, Cohen B, McArthur JC, Hall CD. Progressive multifocal leukencephalopathy in AIDS: Are there any MR findings useful to patient management and predictive of patient survival? AJNR Am J Neuroradiol, 1999; 20: 1896–1906

    CAS  PubMed  Google Scholar 

  • Sawaishi Y, Yno T, Watanabe Y, Takada G. Migratory basal ganglia lesions in subacute sclerosing panencephalitis (SSPE): clinical implications of axonal spread. J Neurol Sci, 1999; 168: 137–140

    CAS  PubMed  Google Scholar 

  • Scatliff JH, Kwock L, Chancellor K, Bouldin TW, Kapoor CC, Castillo M. Postmortem MR imaging of the brains of patients with AIDS. Neuroimaging Clin North Am, 1997; 7:297–320

    CAS  Google Scholar 

  • Thurnher MM, Thurnher SA, Fleischmann D, Steuer A, Rieger A, Helbich T, Trattnig S, Schindler E, Hittmair K. Comparison of T2-weighted and fluid-attenuated inversion-recovery fast spin-echo MR sequences in intracerebral AIDS-associated disease. AJNR Am J Neuroradiol, 1997;18: 1601–1609

    CAS  PubMed  Google Scholar 

  • Tuncay R, Akman-Demir G, Gokyigit A, Eraksoy M, Barlas M, Tolun R, Gursoy G. MRI in subacute sclerosing panencephalitis. Neuroradiology, 1996; 38: 636–640

    CAS  PubMed  Google Scholar 

  • Arnett PA, Rao SM, Bernardin L, Grafman J, Yetkin FZ, Lobeck L. Relationship between frontal lobe lesions and Wisconsin Card Sorting Test performance in patients with multiple sclerosis. Neurology, 1994; 44: 420–425

    CAS  PubMed  Google Scholar 

  • Barkhof FJ, Elton M, Lindeboom J, Tas MW, Schmidt WF, Hommes OR, Polman CH, Kok A, Valk J. Functional correlates of callosal atrophy in relapsing-remitting multiple sclerosis patients: a preliminary study. J Neurol, 1998; 245: 153–188

    CAS  PubMed  Google Scholar 

  • Blinkenberg M, Rune K, Jensen CV, et al. Cortical cerebral metabolism correlates with MRI lesion load and cognitive dysfunction in MS. Neurology, 2000; 54: 558–564

    CAS  PubMed  Google Scholar 

  • Boggild MD, Williams R, Haq N, Hawkins CP. Cortical plaques visualised by fluid-attenuated inversion recovery imaging in relapsing multiple sclerosis. Neuroradiology, 1996; 38: 10–13

    Google Scholar 

  • Camp SJ, Stevenson VL, Thompson AJ, Miller DH, Borras C, Auriacombe S, Brochet B, Falautano M, Filippi M, Herisse-Dulo L, Montalban X, Parrcira E, Polman CH, De Sa J, Langdon DW. Cognitive function in primary progressive and transitional progressive multiple sclerosis: a controlled study with MRI correlates. Brain, 1999; 122:1341–1348

    PubMed  Google Scholar 

  • Comi G, Rovaris M, Falautano M, Santuccio G, Martinelli V, Rocca MA, Possa F, Leocani L, Paulesu E, Filippi M. A multiparametric study of frontal lobe dementia in multiple sclerosis. J Neurol Sci, 1999; 171: 135–141

    CAS  PubMed  Google Scholar 

  • Filippi M, Tortorella C, Rovaris M, Bozzali M, Possa F, Sormani MP, Iannucci G, Comi G. Changes in the normal appearing white brain tissue and cognitive impairment in multiple sclerosis. J Neurol Neurosurg Psychiatry, 2000; 68: 157–161

    CAS  PubMed  Google Scholar 

  • Fontaine B, Seilhean D, Tourbah A, Daumas-Duport C, Duyckaerts C, Benoit N, Devaux B, Hauw JJ, Rancurel G, Lyon-Caen O. Dementia in two histologically confirmed cases of multiple sclerosis: one case with isolated dementia and one case associated with psychiatric symptoms. J Neurol Neurosurg Psychiatry, 1994; 57: 353–359

    CAS  PubMed  Google Scholar 

  • Huber SJ, Paulson GW, Shuttleworth EC, Chakeres D, Clapp LE, Pakalnis A, Weiss K, Rammohan K. Magnetic resonance imaging correlates of dementia in multiple sclerosis. Arch Neurol, 1987; 44: 732–736

    CAS  PubMed  Google Scholar 

  • Janardhan V, Bakshi R. Quality of life and its relationship to brain lesions and atrophy on magnetic resonance images in 60 patients with multiple sclerosis. Arch Neurol, 2000; 57: 1485–1491

    CAS  PubMed  Google Scholar 

  • Kidd D, Barliliof F, McConnell R, Algra PR, Allen IV, Revesz T. Cortical lesions in multiple sclerosis. Brain, 1999; 122:17–26

    PubMed  Google Scholar 

  • Kits van Waveren L, Barkhof F, Scheltens P. Aggression and multiple sclerosis. Br J Psychiatry, 1991; 159: 736

    Google Scholar 

  • Lazeron RH, Langdon DW, Filippi M, van Waesberghe JH, Stevenson VL, Boringa JB, Origgi D, Thompson AJ, Falautano M, Polman CH, Barkhof F. Neuropsychological im-pairment in multiple sclerosis patients: the role of (juxta)-cortical lesion on FLAIR. Mult Scler, 2000; 6: 280–285

    CAS  PubMed  Google Scholar 

  • Lyoo IK, Seol HY, Byun HS, Renshaw PF. Unsuspected multiple sclerosis in patients with psychiatric disorders: a magnetic resonance imaging study. J Neuropsychiatry Clin Neurosci, 1996; 8: 54–59

    CAS  PubMed  Google Scholar 

  • Miki Y, Grossman RI, Udupa JK, Wei L, Kolson DL, Mannon LJ, Grossman M. Isolated U-fiber involvement in MS: preliminary observations. Neurology, 1998; 50: 1301–1306

    CAS  PubMed  Google Scholar 

  • Minden SL, Moes EJ, Orav J, Kaplan E, Reich P. Memory impairment in multiple sclerosis. J Clin Exp Neuropsychol, 1990;12:566–586

    CAS  PubMed  Google Scholar 

  • Pozzilli C, Bastianello S, Padovani A, Passafiume D, Millefiorini E, Bozzao L, Fieschi C. Anterior corpus callosum atrophy and verbal fluency in multiple sclerosis. Cortex, 1991;27:441–445

    CAS  PubMed  Google Scholar 

  • Rao SM, Leo GJ, Bernardin L, Unverzagt F. Cognitive dysfunction in multiple sclerosis. I. Frequency, patterns, and prediction. Neurology, 1991a; 41: 685–691

    CAS  Google Scholar 

  • Rao SM, Leo GJ, Ellington L, Nauertz T, Bernardin L, Unverzagt F. Cognitive dysfunction in multiple sclerosis. II. Impact on employment and social functioning. Neurology, 1991b; 41: 692–696

    CAS  Google Scholar 

  • Rovaris M, Filippi M, Minicucci L, Iannucci G, Santuccio G, Possa F, Comi G. Cortical/subcortical disease burden and cognitive impairment in patients with multiple sclerosis. AJNR Am J Neuroradiol, 2000; 21: 402–428

    CAS  PubMed  Google Scholar 

  • Sperling RA, Guttmann CR, Hohol MJ, Warfield SK, Jakab M, Parente M, Diamond EL, Daffner KR, Olek MJ, Orav EJ, Kikinis R, Jolesz FA, Weiner HL. Regional magnetic resonance imaging lesion burden and cognitive function in multiple sclerosis: a longitudinal study. Arch Neurol, 2001;58: 115–1121

    CAS  PubMed  Google Scholar 

  • Truyen L, Barkhof F, Frequin ST, Polman CH, Tobi H, Hommes OR, Valk J. Magnetic resonance imaging of epilepsy in multiple sclerosis: a case control study: implications for treatment trials with 4-aminopyridine. Mult Scler, 1996; 1: 213–217

    CAS  PubMed  Google Scholar 

  • van Buchem MA, Grossman RI, Armstrong C, Polansky M, Miki Y, Heyning FH, Boncoeur-Martel MP, Wei L, Udupa JK, Grossman M, Kolson DL, McGowan JC. Correlation of volumetric magnetization transfer imaging with clinical data in MS. Neurology, 1998; 50: 1609–1617

    PubMed  Google Scholar 

  • van Waesberghe JH, Kamphorst W, De Groot CJ, van Walderveen MA, Castelijns JA, Ravid R, Lycklama a Nijeholt GJ, van der Valk P, Polman CH, Thompson AJ, Barkhof F. Axonal loss in multiple sclerosis lesions: magnetic resonance imaging insights into substrates of disability. Ann Neurol, 1999;46:747–754

    PubMed  Google Scholar 

  • Vighetto A, Charles N, Salzmann M, Confavreux C, Aimard G. Korsakoff’s syndrome as the initial presentation of multiple sclerosis. J Neurol, 1991; 238: 351–354

    CAS  PubMed  Google Scholar 

  • Dragha R, Letourneur FV, Drugan C, Manicom J, Blanchot C, Kahn A, Poenaru L, Caillaud C. Metachromatic leukodystrophy: identification of the first deletion in Exon 1 and nine novel point mutations in arylsulfatase A gene. Hum Mutat, 1997; 9: 234–242

    Google Scholar 

  • Faerber EN, Melvin J, Smergel EM. MRI appearances of metachromatic leukodystrophy. Pediatr Radiol, 1999; 29:669–672

    CAS  PubMed  Google Scholar 

  • Fukutani Y, Noriki Y, Sasaki K, Isaki K, Kuriyama M, Kurosawa K, Ida H. Adult-type metachromatic leukodystrophy with a compound heterozygote mutation showing character change and dementia. Psychiatry Clin Neurosci, 1999; 53: 425–428

    CAS  PubMed  Google Scholar 

  • Landrieu P, Blanche S, Vanier MT, Metral S, Husson B, Sandhoff K, Fischer A. Bone marrow transplantation in metachromatic leukodystrophy caused by saposin-B deficiency: a case report with a 3-year follow-up period. J Pediatr, 1998; 133: 129–132

    CAS  PubMed  Google Scholar 

  • Malm G, Ringden O, Winiarski J, Grondahl E, Uyebrant P, Eriksson U, Hakansson H, Skjeldal O, Mansson JE. Clinical outcome in four children with metachromatic leukodystrophy treated by bone marrow transplantation. Bone Marrow Transplant, 1996; 17: 1003–1008

    CAS  PubMed  Google Scholar 

  • Minamikawa-Tachino R, Maeda Y, Fujishiro I, Itoh K, Satake A, Aoki S, Yamada H, Suzuki Y, Sakuraba H. Three-dimensional brain visualization for metachromatic leukodystrophy. Brain Dev, 1996; 18: 394–399

    CAS  PubMed  Google Scholar 

  • Reider-Gronwasser I, Bornstein N. CT and MRI in late onset metachromatic leukodystrophy. Acta Neurol Scand, 1987;75:64–69

    Google Scholar 

  • Salmon E, Van der Linden M, Maerfens Noordhout A, Brucher JJJM, Mouchette R, Waltregny A, Degueldre C, Franck G. Early thalamic and cortical hypometabolism in adult onset dementia due to metachromatic leukodystrophy. Acta Neurol Belg, 1999; 99: 185–188

    CAS  PubMed  Google Scholar 

  • Shapiro EG, Lockman LA, Knopman D, Krivit W. Characteristics of the dementia in late-onset metachromatic leukodystrophy. Neurology, 1994; 44: 662–665

    CAS  PubMed  Google Scholar 

  • Tylki-Szymanska AT, Czartoryska B, Lugowska A. Practical suggestions in diagnosing metachromatic leukodystrophy in probands and in testing family members. Eur Neurol, 1998;40:67–70

    CAS  PubMed  Google Scholar 

  • Al-Essa MA, Bakheet SM, Patay ZJ, Powe JE, Ozand PT. Clinical and cerebral FDG PET scan in a patient with Krabbe’s disease. Pediatr Neurol, 2000; 22: 44–47

    CAS  PubMed  Google Scholar 

  • Barone R, Bruhl K, Stoeter P, Fiumara A, Pavone L, Beck M. Clinical and neuroradiological findings in classic infantile and late-onset globoid-cell leukodystrophy (Krabbe disease). Am J Med Genet, 1996; 63: 209–217

    CAS  PubMed  Google Scholar 

  • Bernardini GL, Herrera DG, Carson D, DeGasperi R, Gama Sosa MA, Kolodny EH, Trifiletti R. Adult-onset Krabbe’s disease in siblings with novel mutations in the galactocerebroside gene. Ann Neurol, 1997; 41: 111–114

    CAS  PubMed  Google Scholar 

  • Datoh JI, Tokumoto H, Kurohara K, Yukitake M, Matsui M, Kuroda Y, Yamamoto T, Furuya H, Shinnoh N, Kobayashi T, Kukita Y, Hayashi K. Adult-onset Krabbe disease with homozygous T1853C mutation in the galactocerebrosidase gene: unusual MRI finding of corticospinal tract demyelination. Neurology; 49: 1392–1399

    Google Scholar 

  • De Stefano N, Dotti MT, Pappagallo E, Luzi P, Rafi MA, Formichi P, Inzitari D, Wenger A, Federico A. Evidence of diffuse brain pathology and unspecific genetic characterization in a patient with an atypical form of adult-onset Krabbe disease. J Neurol, 2000; 247: 226–228

    PubMed  Google Scholar 

  • Jardim LB, Giugliani R, Pires RF, Haussen S, Burin MG, Rafi MA, Wenger DA. Protracted course of Krabbe diseases in an adult patient: a novel mutation. Arch Neurol, 1999; 56:1014–1017

    CAS  PubMed  Google Scholar 

  • Loes DJ, Peters C, Krivit W. Globoid cell leukodystrophy: distinguishing early-onset from late-onset disease using a brain MR imaging scoring method. AJNR Am J Neuroradiol, 1999; 20: 316–323

    CAS  PubMed  Google Scholar 

  • Turazzini M, Beltramello A, Bassi R, Del Colle R, Silvestri M. Adult onset Krabbe’s leukodystrophy: a report of 2 cases. Acta Neurol Scand, 1997; 96: 413–415

    CAS  PubMed  Google Scholar 

  • Vasconcellos E, Smith M. MRI nerve root enhancement in Krabbe disease. Pediatr Neurol, 1998; 19: 151–152

    CAS  PubMed  Google Scholar 

  • Chu BC, Terae S, Takahashi C, Kikuchi Y, Miyasaka K, Abe S, Minowa K, Sawamura T. MRI of the brain in the Kearns-Sayre syndrome: a report of four cases and a review. Neuroradiology, 1999;41:759–764

    CAS  PubMed  Google Scholar 

  • De Coo IF, Gussinklo T, Arts PJ, Van Oost BA, Smeets HJ. PCR tests for progressive external ophthalmoplegia and Kearns-Sayre syndrome on DNA from blood samples. J Neurol Sci, 1997; 149:37–40

    PubMed  Google Scholar 

  • Desnuelle C, Pellissier J, Serratrice G, Pouget J, Turnbull D. Le syndrome de Kearns et Sayre: encéphalomyopathie mitochondriale par déficit de la chaine respiratoire. Rev Neurol, 1989; 145: 842–850

    CAS  PubMed  Google Scholar 

  • Holt IJ, Harding AE, Morgan-Hughes JA. Deletions of mitochondrial DNA in patients with mitochondrial myopathies. Nature, 1988; 331: 717–719

    CAS  PubMed  Google Scholar 

  • Kapeller P, Fazekas F, Offenbacher H, Stollberger R, Schmidt R, Bergloff J, Radner H, Fazekas G, Schafhalter-Zoppoth I. Magnetic resonance and spectroscopy of progressive cerebral involvement in Kearns-Sayre syndrome. J Neurol Sci, 1996; 135: 126–130

    CAS  PubMed  Google Scholar 

  • Kearns TP, Sayre GP. Retinitis pigmentosa, external ophthal-moplegia, and complete heart block. Arch Ophthalmol, 1958;60:280–289

    CAS  Google Scholar 

  • Kim SH, Chi JG, Reith A, Kadenbach B. Quantitative analysis of mitochondrial DNA deletion in paraffin embedded muscle tissues from patients with KSS and CPEO. Biochim Biophys Acta, 1997; 1360: 193–195

    CAS  PubMed  Google Scholar 

  • Larsson N-G, Holme E, Kristiansson B, Oldfors A, Tulinius M. Progressive increase of the mutated mitochondrial DNA fraction in Kearns-Sayre syndrome. Pediatr Res, 1990; 28: 131–136

    CAS  PubMed  Google Scholar 

  • Lestienne P, Ponsot G. Kearns-Sayre syndrome with muscle mitochondrial DNA deletion [letter]. Lancet, 1988; I: 885

    Google Scholar 

  • Nakagawa E, Osari S, Yamanouchi H, Matsuda H, Goto Y, Nonaka I. Long-term therapy with cytochrome c, flavin mononucleotide and thiamine diphosphate for a patient with Kearns-Sayre syndrome. Brain Dev, 1996; 18: 68–70

    CAS  PubMed  Google Scholar 

  • Oldfors A, Fyhr I-M, Larsson N-G, Tulinius M. Neuropathology in Kearns-Sayre syndrome. Acta Neuropathol, 1990; 80:541–546

    CAS  PubMed  Google Scholar 

  • Poulton J, Deadman ME, Gardiner RM. Duplications of mitochondrial DNA in mitochondrial myopathy. Lancet, 1989; I: 236–240

    Google Scholar 

  • Poulton J, O’Rahilly S, Morten KJ, Clark A. Mitochondrial DNA, diabetes and pancreatic pathology in Kearns-Sayre syndrome. Diabetologia, 1995; 38: 868–871

    CAS  PubMed  Google Scholar 

  • Schnitzler E, Robertson W. Familial Kearns-Sayre syndrome. Neurology, 1979; 29: 1172–1174

    CAS  PubMed  Google Scholar 

  • Schon EA, Rizzuto R, Moraes CT, Nakase H, Zeviani M, DiMauro S. A direct repeat is a hotspot for large-scale deletions of human mitochondrial DNA. Science, 1989; 244:346–349

    CAS  PubMed  Google Scholar 

  • Seigel R, Seeger J, Gabrielsen T, Allen R. Computed tomography in oculocraniosomatic disease (Kearns-Sayre syndrome). Neuroradiology, 1979; 130: 159–164

    CAS  Google Scholar 

  • Sparaco M, Bonilla E, DiMauro S, Powers JM. Neuropathology of mitochondrial encephalomyopathies due to mitochondrial DNA defects. J Neuropathol Exp Neurol, 1993; 52: 1–10

    CAS  PubMed  Google Scholar 

  • Wray SH, Provenzale JM, Johns DR, Thulborn KR. MR of the brain in mitochondrial myopathy. AJNR Am J Neuroradiol, 1995; 16: 1167–1173

    CAS  PubMed  Google Scholar 

  • Zeviani M, Servidei S, Gellera C, Bertini E, DiMauro S, DiDonato S. An autosomal dominant disorder with multiple deletions of mitochondrial DNA starting at the Dloop region. Nature, 1989; 339: 309–311

    CAS  PubMed  Google Scholar 

  • Abe K, Inui T, Hirono N, Mezaki T, Kobayashi Y, Kameyama M. Fluctuating MR images with mitochondrial encephalopathy, lactic acidosis, stroke-like syndrome (MELAS). Neuroradiology, 1990; 32: 77

    CAS  PubMed  Google Scholar 

  • Allard J, Tilak S, Carter A. CT and MR of MELAS syndrome. AJNR Am J Neuroradiol, 1988; 9: 1234–1238

    CAS  PubMed  Google Scholar 

  • Askanas V, King Engel W, Britton D, Aornato B, Eiben R. Reincarnation in cultured muscle of mitochondrial abnormalities. Arch Neurol, 1978; 35: 801–809

    CAS  PubMed  Google Scholar 

  • Brown WM, Prager EM, Wang A, Wilson AC. Mitochondrial DNA sequences of primates: tempo and mode of evolution. J Mol Evol, 1982; 18: 225–231

    CAS  PubMed  Google Scholar 

  • De Stefano N, Matthews PM, Arnold DL. Reversible decreases in N-acetylaspartate after acute brain injury. Magn Reson Med, 1995; 34: 721–727

    PubMed  Google Scholar 

  • DeVivo DC, Jackson A, Wade C, Altmann K, Stacpoole P, DiMauro S. Dichloroacetate treatment of MELAS-associated lactic acidosis. Ann Neurol, 1990; 28: 437

    Google Scholar 

  • Gilchrist JM, Sikirica M, Stopa E, Shanske S. Adult-onset MELAS: evidence for involvement of neurons a well as cerebral vasculature in strokelike episodes. Stroke, 1996; 27: 1420–1423

    CAS  PubMed  Google Scholar 

  • Goto Y-I, Nonaka I, Horai S. A new mtDNA mutation associated with mitochondrial myopathy, encephalopathy, lactic acidosis and stroke-like episodes (MELAS). Biochim Biophys Acta, 1991; 1097: 238–240

    CAS  PubMed  Google Scholar 

  • Goto Y-I, Tsugane K, Tanabe Y, Nonaka I, Horai S. A new point mutation at nucleotide 3291 of the mitochondrial tRNALeu(UUR) gene in a patient with mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS). Biochim Biophys Acta, 1994; 202: 1624–1630

    CAS  Google Scholar 

  • Hasegawa H, Matusoka T, Goto Y-I, Nonaka I. Strongly succinate dehydrogenase-reactive blood vessels in muscles from patients with mitochondrial myopathy, encephalopathy, lactic acidosis and stroke-like episodes. Ann Neurol, 1991; 29: 610–615

    Google Scholar 

  • Hirano M, Pavlakis S. MELAS: current concepts. Child Neurol, 1994; 9: 4–13

    CAS  Google Scholar 

  • Ikejiri Y, Mori E, Ishii K, Nishimoto K, Yasuda M, Sasaki M. Idebenone improves cerebral mitochondrial oxidative metabolism in a patient with MELAS. Neurology, 1996; 47:583–585

    CAS  PubMed  Google Scholar 

  • Johns DR, Stein AG, Wityk R. MELAS syndrome masquerading as herpes simplex encephalitis. Neurology, 1993; 43: 2471–2473

    CAS  PubMed  Google Scholar 

  • Manfredi G, Schon EA, Bonilla E, Moraes CT, Shanske S, DiMauro S. Identification of a mutation in the mitochondrial tRNACys gene associated with mitochondrial encephalopathy. Hum Mutat 1996; 7:158–163

    CAS  PubMed  Google Scholar 

  • Miyamoto A, Oki J, Takahashi S, Itoh J, Kusunoki Y, Cho K. Serial imaging in MELAS. Neuroradiology, 1997; 39:427–430

    CAS  PubMed  Google Scholar 

  • Moraes CT, Ricci E, Bonilla E, DiMauro S, Schon EA. The mitochondrial tRNALeu(UUR) mutation in mitochondrial encephalomyopathy, lactic acidosis, and strokelike episodes (MELAS): genetic, biochemical, and morphological correlations in skeletal muscles. Am J Hum Genet, 1992;50:934–939

    CAS  PubMed Central  PubMed  Google Scholar 

  • Morten KJ, Cooper JM, Brown GK, Lake BD, Pike D, Poulton J. A new point mutation associated with mitochondrial encephalomyopathy. Hum Mol Genet, 1993; 2: 2081–2087

    CAS  PubMed  Google Scholar 

  • Nishino I, Komatsu M, Kodama S, Horai S, Nonaka I, Goto Y-I. The 3260 mutation in mitochondrial DNA can cause mitochondrial myopathy, encephalopathy, lactic acidosis, and strokelike episodes (MELAS). Muscle Nerve, 1996; 19: 1603–1604

    CAS  PubMed  Google Scholar 

  • Oppenheim C, Galanaud D, Samson Y, Sahel M, Dormont D, Wechsler B, Marsault C. Can diffusion weighted magnetic resonance imaging help differentiate stroke from stroke-like events in MELAS? J Neurol Neurosurg Psychiatry, 2000; 69: 248–250

    CAS  PubMed  Google Scholar 

  • Pavlakis SG, Phillips PC, DiMauro S, DeVivo DC, Rowland LP. Mitochondrial myopathy, encephalopathy, lactic acidosis, and strokelike episodes: a distinctive clinical syndrome. Ann Neurol, 1984; 16: 481–488

    CAS  PubMed  Google Scholar 

  • Pavlakis SG, Kingsley PB, Kaplan GP, Stacpoole PW, O’Shea M, Lustbader D. Magnetic resonance spectroscopy: use in monitoring MELAS treatment. Arch Neurol, 1998; 55:849–852

    CAS  PubMed  Google Scholar 

  • Sharfstein SR, Gordon MF, Libman RB, Malkin ES. Adult-onset MELAS presenting as herpes encephalitis. Arch Neurol, 1999;56:241–243

    CAS  PubMed  Google Scholar 

  • Sparaco M, Bonilla E, DiMauro S, Powers J. Neuropathology of mitochondrial encephalomyopathies due to mitochondrial DNA defects. J Neuropathol Exp Neurol, 1993; 52: 1–10

    CAS  PubMed  Google Scholar 

  • Van der Knaap MS, Jakobs C, Valk J. Magnetic resonance imaging in lactic acidosis. J Inherit Metab Dis, 1996; 19:535–547

    PubMed  Google Scholar 

  • Barkhof F, Verrips A, Wesseling P, Van der Knaap MS, Van Engelen BGM, Gabreëls FJM, Keyser A, Wevers RA, Valk J. Cerbrotendinous xanthomatosis: the spectrum of imaging findings and the correlation with neuropathologic findings. Radiology, 2000; 217: 869–876

    CAS  PubMed  Google Scholar 

  • Bogaert L, Scherer H, Epstein E. Une forme cérebrale de la cholesterinose généralisée. Paris: Masson et Cie, 1937.

    Google Scholar 

  • Hokezu Y, Kuriyama M, Kubota R, Nakagawa M, Fujiyama J, Osame M. Cerebrotendinous xanthomatosis: cranial CT and MRI studies in eight patients. Neuroradiology, 1992; 34:308–312

    CAS  PubMed  Google Scholar 

  • Oftebro H, Björkhem I, Stormer FC, Pedersen JL. Cerebrotendinous xanthomatosis: defective liver mitochondrial hydroxylation of chenodeoxycholic acid precursors. J Lipid Res, 1981;22:632–640

    CAS  PubMed  Google Scholar 

  • Wolthers BG, Vollmer M, Van der Molen J, Koopman BJ, De Jager AE, Waterreus RJ. The diagnosis of cerebrotendinous xanthomatosis (CTX) and effect of chenodeoxycholic acid therapy by analysis of urine using capillary gas chromatography. Clin Chim Acta, 1983; 131: 53–65

    CAS  PubMed  Google Scholar 

  • Verrips A, Weyers RA, Van Engelen BG, Keyser A, Wolthers BG, Barkhof F, Stalenhoef A, De Graaf R, Janssen-Zijlstra F, Van Spreeken A, Gabreëls FJ. Effects of simvastatin in addition to chenodeoxycholic acid in patients with cerebrotendinous xanthomatosis. Metabolism, 1999; 48:233–238

    CAS  PubMed  Google Scholar 

  • Verrips A, Nijeholt GJ, Barkhof F, Van Engelen BBBG, Wesseling P, Luyten JA, Wevers RA, Stam J, Wokke JH, Van den Heuvel LP, Keyser A, Gabreëls FJ. Spinal xanthomatosis: a variant of cerebrotendinous xanthomatosis. Brain, 1999; 122: 1589–1595

    PubMed  Google Scholar 

  • Verrips A, Hoefsloiot LH, Steenbergen GC, Theelen JP, Wevers RA, Gabreëls FJ, Van Engelen BG, Van den Heuvel LP. Clinical and molecular genetic characteristics of patients with cerebrotendinous xanthomatosis. Brain, 2000; 123:908–919

    PubMed  Google Scholar 

  • Vanrietvelde F, Lemmerling M, Mespreuve M, Crevits L, De Reuck J, Kunnen M. MRI of the brain in cerebrotendinous xanthomatosis (Van Bogaert-Scherer-Epstein disease). Eur Radiol, 2000; 10: 576–578

    CAS  PubMed  Google Scholar 

  • Lachtermacher MB, Seuanez HN, Moser AB, Moser HW, Smith KD. Determination of 30 X-linked mutations, including 15 not previously described. Hum Mutat, 2000; 15:348–353

    CAS  PubMed  Google Scholar 

  • Melhem ER, Breiter SN, Ulug AM, Raymond GV, Moser HW. Improved tissue characterization in adrenoleukodystrophy using magnetic transfer imaging. AJR, 1996; 166:689–695

    CAS  PubMed  Google Scholar 

  • Powers JM, DeCiero DP, Ito M, Moser AB, Moser HW. Adrenomyeloneuropathy: a neuropathologic review featuring its noninflammatory myelopathy. J Neuropathol Exp Neurol, 2000; 59: 89–102

    CAS  PubMed  Google Scholar 

  • Tourbah A, Stievenart JL, Iba-Zizen MT, Lubetzki CCC, Baumann N, Eymard B, Moser HW, Lyon-Caen O, Cabanis EA. Localized proton magnetic resonance spectroscopy in patients with adult adrenoleukodystrophy: increase of choline compounds in normal appearing white matter. Arch Neurol, 1997; 54: 586–592

    CAS  PubMed  Google Scholar 

  • Van Geel BM, Assies J, Haverkort EB, Koelman JH, Verbeeten B Jr, Wanders R, Barth PG. Progression of abnormalities in adrenomyeloneuropathy and neurologically asymptomatic X-linked adrenoleukodystrophy despite treatment with “Lorenzo’s oil”. J Neurol Neurosurg Psychiatry, 1999; 67: 290–299

    PubMed  Google Scholar 

  • Van Geel BM, Assies J, Wanders RJ, Barth PG. X-linked adrenoleukodystrophy: clinical presentation, diagnosis, therapy. J Neurol Neurosurg Psychiatry, 1997; 63: 4–14

    PubMed  Google Scholar 

  • Van Geel BM, Assies J, Weverling GJ, Barth PG. Predominance of the adrenomyeloneuropathy phenotype of X-linked adrenoleukodystrophy in the Netherlands: a survey of 30 kindreds. Neurology, 1994; 12: 1243–1246

    Google Scholar 

  • Wichers M, Kohler W, Brennemann W, Boese V, Sokolowski P, Bidlingmaier F, Ludwig M. X-linked adrenomyeloneuropathy associated with 14 novel ALD-gene mutations: no correlation between type of mutation and age of onset. Hum Genet, 1999; 105: 116–119

    CAS  PubMed  Google Scholar 

  • Ashton-Prolla P, Tong B, Shabbeer J, Astrin KH, Eng CM, Desnick RJ. Fabry disease: twenty-two novel mutations in the alpha-galactosidase A gene and genotype/phenotype correlations in severely and mildly affected hemizygotes and heterozygotes. J Invest Med, 2000; 48: 227–235

    CAS  Google Scholar 

  • Crutchfield KE, Patronas NT, Dambrosia JM, Frei KP, Banerjee TK, Barton NW, Schiffmann R. Quantitative analysis of cerebral vasculopathy in patients with Fabry’s disease. Neurology, 1998; 50: 1746–1749

    CAS  PubMed  Google Scholar 

  • DeGraba T, Azhar S, Dignat-George F, Brown E, Boutiere B, Altarescu G, McCarron R, Schiffmann R. Profile of endothelial and leukocyte activation in Fabry patients. Ann Neurol, 2000;47:229–233

    CAS  PubMed  Google Scholar 

  • Kihiczak NI, Leevy CB, Krysicki MM, Janniger CK, Schneider I, Shriner DL, Lambert WC, Schwartz RA. Cutaneous signs of selected systemic diseases. J Med, 1999; 30: 3–12

    CAS  PubMed  Google Scholar 

  • Mendez MF, Stanley TM, Medel NM, Li Z, Tedesco DT. The vascular dementia of Fabry’s disease. Dement Geriatr Cogn Disord, 1997; 8: 252–257

    CAS  PubMed  Google Scholar 

  • Menkes DL, O’Neil TT, Saenz KK. Fabry’s disease presenting as syncope, angiokeratomas, and spoke-like cataracts in a young man: discussion of the differential diagnosis. Mil Med, 1997; 162: 773–776

    CAS  PubMed  Google Scholar 

  • Mitsias P, Levine SR. Cerebrovascular complications of Fabry’s disease. Ann Neurol, 1996;40:80–17

    Google Scholar 

  • Tedeschi G, Bonavita S, Banerjee TK, Virta A, Schiffmann R. Diffuse central neuronal involvement in Fabry disease: a proton MRS imaging study. Neurology, 1999; 12: 1663–1667

    Google Scholar 

  • Berkhoff M, Weiss J, Schroth G, Sturzenegger. Extensive white matter changes in case of adult polyglucosan body disease. Neuroradiology, 2001; 43: 234–236

    CAS  PubMed  Google Scholar 

  • Bigio EH, Weiner MF, Bonte FJ. White CL. Familial dementia due to adult polyglucosan body disease. Clin Neuropathol, 1997; 16: 227–234

    CAS  PubMed  Google Scholar 

  • Bruno C, Servidei S, Shanske S, Karpati G, Carpenter S, McKee D, Barohn RJ, Hirano M, Rofai Z, DiMauro S. Glycogen branching enzyme deficiency in adult polyglucosan body disease. Ann Neurol, 1993; 33: 88–94

    CAS  PubMed  Google Scholar 

  • Cavanagh JB. Corpora-amylacea and the family of polyglucosan diseases. Brain Res Brain Res Rev, 1999; 29: 265–295

    CAS  PubMed  Google Scholar 

  • Lossos A, Meiner Z, Barash V, Soffer D, Schlesinger I, Abramsky O, Argov Z, Shpitzen S, Meiner V. Adult polyglucosan bodr disease in Ashkenazi Jewish patients carrying the Tyr329Ser mutation in the glycogen branching enzyme. Ann Neurol, 1998; 44: 867–872

    CAS  PubMed  Google Scholar 

  • Rifai Z, Klizke M, Tawil R, Kazee Am, Shanke S, DiMauro S, Griggs RC. Dementia of adult polyglucosan body disease. Arch Neurol, 1994; 51: 90–94

    CAS  PubMed  Google Scholar 

  • Suzuki K, David E, Kutschman B. Presenile dementia with “Lafora-like” intraneuronal inclusions. Arch Neurol, 1971; 28: 69–80

    Google Scholar 

  • Ziemssen F, Sindern E, Schroder JM, Shin YS, Zange J, Kilimann MW, Malin JP, Vorgerd M. Novel missense mutations in the glycogen-branching enzyme gene in adult polyglucosan body disease. Ann Neurol, 2000; 47: 536–540

    CAS  PubMed  Google Scholar 

  • Hanefeld F, Holzbach U, Kruse B, Wilichowsky E, Christen HJ, Frahm J. Diffuse white matter disease in three children: an encephalopathy with unique features on magnetic resonance imaging and proton magnetic resonance spectroscopy. Neuropediatrics, 1993; 24: 244–248

    CAS  PubMed  Google Scholar 

  • Leegwater PAJ, Könst AAM, Kuyt B, Sandkuijl LA, Naidu S, Oudejans CBM, Schutgens RBH, Pronk JC, Van der Knaap MS. The gene for leukoencephalopathy with vanishing white matter is located on chromosome 3q27. Am J Hum Genet, 1999; 65: 728–734

    CAS  PubMed Central  PubMed  Google Scholar 

  • Schiffmann R, Moller JR, Trapp BD, Shoih HH, Farrer RG, Katz DA, Alger JR, Parker CC, Hauer PE, Kaneski CR, Heiss JD, Kaye EM, Quarles RH, Brady RO, Barton NW. Ann Neurol, 1994; 35: 331–340

    CAS  PubMed  Google Scholar 

  • Van der Knaap MS, Barth PG, Gabreels FJM, Franzoni E, Begeer JH, Stroink H, Rotteveel JJ, Valk J. A new leukoencephalopathy with vanishing white matter. Neurology, 1997;48:845–855

    PubMed  Google Scholar 

  • Van der Knaap MS, Kamphorst W, Barth PG, Kraaijeveld CL, Gut E, Valk J. Phenotypic variation in leukoencephalopathy with vanishing white matter. Neurology, 1998; 51:540–547

    PubMed  Google Scholar 

  • Aisen AM, Martel W, Gabrielsen TO, Glazer GM, Brewer G, Young AB, Hill G. Wilson disease of the brain: MR imaging. Radiology, 1985; 157: 137–141

    CAS  PubMed  Google Scholar 

  • Brewer GJ, Dick RD, Johnson V, Wang, Yuzbasiyan-Gurkan V, Kluin K, Fink JK, Aisen A. Treatment of Wilson’s disease with tetrathiomolybdate. I. Initial therapy in 17 neurologically affected patients. Arch Neurol, 1994; 51: 545–554

    CAS  Google Scholar 

  • Brewer GJ, Hill GM, Prasad AS, Dick RD. The treatment of Wilson’s disease with zinc. IV. Efficacy monitoring using urine and plasma copper. Proc Soc Exp Biol Med, 1987; 7:446–455

    Google Scholar 

  • Brewer GJ, Yuzbasiyan-Gurkan V, Johnson V, Dick RD, Wang Y. Treatment of Wilson’s disease with zinc. XII. Dose regimen requirements. Am J Med Sci, 1993; 305: 199–202

    CAS  Google Scholar 

  • Brewer GJ, Yuzbasiyan-Gurkan V. Wilson disease. Medicine, 1992; 71: 139–164

    CAS  PubMed  Google Scholar 

  • Cumings JN. The copper and iron content of brain and liver in the normal and in hepato-lenticular degeneration. Brain, 1948;71:410–415

    CAS  PubMed  Google Scholar 

  • Fleischer B. Zwei weitere Fälle von grünliche Verfärbung der Kornea. Klin Monatsbl Augenheilkd, 1903; 41: 489–491

    Google Scholar 

  • Hayek HW, Knoll E, Widhalm S. Über das organische Psychosyndrom bei morbus Wilson. Monatsschr Kinderheilkd, 1973;121:679–683

    CAS  PubMed  Google Scholar 

  • Hoogenraad TU, Loevoet R, De Ruyter Korver EGWM. Oral zinc sulfate as long-term treatment in Wilson’s disease (hepatolenticular degeneration). Eur Neurol, 1979; 18:205–211

    CAS  PubMed  Google Scholar 

  • Huang CC, Chu NS. Resolution of cerebral white matter lesions following long-term penicillamine therapy for Wilson disease: report of a case. J Formosan Med Assoc, 1992;91:627–629

    CAS  PubMed  Google Scholar 

  • Iyengar V, Brewer GJ, Dick RD, Owyang C. Studies of cholecystokinin-stimulated biliary secretions reveal a high molecular weight copper-binding substance in normal subjects that is absent in patients with Wilson’s disease. J Lab Clin Med, 1988; 111: 267–274

    CAS  PubMed  Google Scholar 

  • Kayser B. Über einen Fall von angeborener grünlicher Verfarbung der Kornea. Klin Monatsbl Augenheilkd, 1902; 40:22–25

    Google Scholar 

  • Mochikuzi H, Kamakura K, Masaki T, Okano M, Nagata N, Inui A, Fujisawa T, Kaji T. Atypical MRI features of Wilson’s disease: high signal in globus pallidus on T1-weighted images. Neuroradiology, 1997; 39: 171–174

    Google Scholar 

  • O’Reilly S, Weber PM, Oswald M, Shipley L. Abnormalities of the physiology of copper in Wilson’s disease. III. The excretion of copper. Arch Neurol, 1971; 25: 28–32

    Google Scholar 

  • Van Wassenaer-van Hall HN, Van den Heuvel AG, Algra A, Hoogenraad TU, Mali WPTM. Wilson disease: findings at MR imaging and CT of the brain with clinical correlation. Radiology, 1996; 198; 531–536

    Google Scholar 

  • Van Wassenaer-van Hall HN, Van den Heuvel AG, Jansen GH, Hoogenraad TU, Mali WPTM. Cranial MR in Wilson disease: abnormal white matter in extrapyramidal and pyramidal tracts. AJNR Am J Neuroradiol, 1995; 16:2021–2027

    Google Scholar 

  • Van Wassenaer-van Hall HN. Wilson disease: magnetic resonance findings. Thesis, University of Utrecht, 1996

    Google Scholar 

  • Wilson SAK. Progressive lenticular degeneration: a familial nervous disease associated with cirrhosis of the liver. PhD thesis, University of Edinburgh, 1911

    Google Scholar 

  • Wilson SAK. Progressive lenticular degeneration: a familial nervous disease associated with cirrhosis of the liver. Brain, 1912;34:295–507

    Google Scholar 

  • Yang XL, Miura N, Kawarada Y, Terada K, Petrukhin K, Gilliam TC, Sujgiyama N. Two forms of Wilson disease protein produced by alternative splicing are localized in distinct cellular compartments. Biochem J, 1997; 326: 897–902

    CAS  PubMed  Google Scholar 

  • Calandriello L, Matteucci C, Bertini E, Medalago Albani L, Antonelli A, Manfredi M, Palladini G. Biopsy diagnosis of a case of adult onset orthochromatic leukodystrophy: clinical and brain biopsy findings. Ital J Neurol Sci, 1992; 13: 787–792

    CAS  PubMed  Google Scholar 

  • Coffeen CM, McKenna CE, Koeppen AH, Plaster NM, Maragakis N, Mihalopoulos J, Schwankhaus JD, Flanigan KM, Gregg RG, Ptacek LJ, Fu YH. Genetic localization of an autosomal dominant leukodystrophy mimicking chronic progressive multiple sclerosis to chromosome 5q31. Hum Mol Genet, 2000; 22: 787–793

    Google Scholar 

  • Knopman D, Sung JH, Davis D. Progressive familial leukodystrophy of late onset. Neurology, 1996; 46: 429–434

    CAS  PubMed  Google Scholar 

  • Shannon P, Wherrett JR, Nag S. A rare form of adult onset leukodystrophy: orthochromatic leukodystrophy with pigmented glia. Can J Neurol Sci, 1997; 24: 146–150

    CAS  PubMed  Google Scholar 

  • Simon DK, Rodriguez ML, Frosh MP, Quackenbusch EJ, Feske SK, Natowicz MR. A unique familial leukodystrophy with adult onset dementia and abnormal glycolipid storage: a new lysosomal disease? J Neurol Neurosurg Psychiatry, 1998; 65: 251–254

    CAS  PubMed  Google Scholar 

  • Adams RD, Victor M, Mancall EL. Central pontine myelinolysis: a hitherto undescribed disease occurring in alcoholic and malnourished patients. Arch Neurol Psychiatry, 1959;81: 154–172

    CAS  Google Scholar 

  • Behse F, Buchthal F. Alcoholic neuropathy: clinical, electro-physiological and biopsy findings. Ann Neurol, 1977; 2:95–110

    Google Scholar 

  • Caine D, Halliday GM, Kril JJ, Harper CG. Operational criteria for the classification of chronic alcoholics: identification of Wernicke’s encephalopathy. J Neurol Neurosurg Psychiatry, 1997; 62: 51–60

    CAS  PubMed  Google Scholar 

  • Caparros-Lefebvre D, Pruvo JP, Josien E, Petuzon B, Clarisse J, Petit H. Marchiafava-Bignami disease: use of contrast media in CT and MR. Neuroradiology, 1994; 36: 509–511

    CAS  PubMed  Google Scholar 

  • Charness ME, Simon RP, Greenberg DA. Ethanol and the nervous system. N Engl J Med, 1989; 321: 442–454

    CAS  PubMed  Google Scholar 

  • Claus D, Eggers R, Engelhardt A, Neundorfer B, Warecka K. Ethanol and polyneuropathy. Acta Neurol Scand, 1985; 72:312–316

    CAS  PubMed  Google Scholar 

  • Diamond I, Messing RO. Neurologic effects of alcoholism. West J Med, 1994; 161: 279–287

    CAS  PubMed Central  PubMed  Google Scholar 

  • Fernandez-Sola J, Estruch R, Grau JM, Pare JC, Rubin E, Urban-Marquez A. The relation of alcoholic myopathy to cardiomyopathy. Ann Intern Med, 1994; 120: 529–536

    CAS  PubMed  Google Scholar 

  • Jennigan TL, Schafer K, Butters N, Cermak LS. Magnetic resonance imaging of alcoholic Korsakoff patients. Neuropsychopharmacology, 1991;4: 175–186

    Google Scholar 

  • Krabbendam L, Visser PJ, Derix MM, Verhey F, Hofman P, Verhoeven W, Tuinier S, Jolles J. Normal cognitive performance in patients with chronic alcoholism in contrast to patients with Korsakoff’s syndrome. J Neuropsychiatry Clin Neurosci, 2000; 12: 44–50

    CAS  PubMed  Google Scholar 

  • Mascalchi M, Simonelli P, Giangospero F, Petruzzi P, Bosincu L, Conti M, Sechi G, Salvi F. Do acute lesions of Wernicke’s encephalopathy enhance? Neuroradiology, 1999; 41:249–254

    CAS  PubMed  Google Scholar 

  • Monforte R, Estruch R, Valls-Sole J, Nicolas J, Villalta J, Urbano-Marquez A. Autonomic and peripheral neuropathies in patients with chronic alcoholism: a dose-related toxic effect of alcohol. Arch Neurol, 1995; 52: 45–51

    CAS  PubMed  Google Scholar 

  • Neiman J, Lang AE, Fornazzari L, Carlen PL. Movement disorders in alcoholism: a review. Neurology, 1990; 40: 741–746

    CAS  PubMed  Google Scholar 

  • Oishi M, Mochizuki Y, Shikata E. Corpus callosum atrophy and cerebral blood flow in chronic alcoholics. J Neurol Sci, 1999; 162: 51–55

    CAS  PubMed  Google Scholar 

  • Palliyath SK, Schwartz BD, Gant L. Peripheral nerve functions in chronic alcoholic patients on disulfiram: a six month follow up. J Neurol Neurosurg Psychiatry, 1990; 53:227–230

    CAS  PubMed  Google Scholar 

  • Pfefferbaum A, Sullivan E, Rosenbloom MJ, Mathalon DH, Lim KO. A controlled study of cortical gray matter and ventricular changes in alcoholic man over a 5-year interval. Arch Gen Psychiatry, 1998; 55: 905–912

    CAS  PubMed  Google Scholar 

  • Serdaru M, Hausser-Hauw C, Laplane D, Buge A, Castaigne P, Goulon M, Lhermitte F, Hauw JJ. The clinical spectrum of alcoholic pellagra encephalopathy: a retrospective analysis of 22 cases studied pathologically. Brain, 1988; 111: 829–842

    PubMed  Google Scholar 

  • Shiota JY, Nakano I, Kawamura M, Hirayama K. An autopsy case of Marchiafava-Bignami disease with peculiar chronological CT changes in the corpus callosum: neuroradiopathological correlations. J Neurol Sci, 1996; 136:90–93

    CAS  PubMed  Google Scholar 

  • Silver NC, Barker GJ, MacManus DG, Miller DH, Thorpe JW, Howard RS. Decreased magnetization transfer ratio due to demyelination in a case of central pontine myelinolysis. J Neurol Neurosurg Psychiatry 1996; 61: 208–209

    CAS  PubMed  Google Scholar 

  • Streissguth AP, Landesman-Dwyer S, Martin JC, Smith DW. Teratogenic effects of alcohol in humans and laboratory animals. Science, 1980; 209: 353–361

    CAS  PubMed  Google Scholar 

  • Urbano-Marquez A, Estruch R, Navarro-Lopez F, Grau JM, Mont L, Rubin E. The effects of alcoholism on skeletal and cardiac muscle. N Engl J Med, 1989; 320: 409–415

    CAS  PubMed  Google Scholar 

  • Victor M. Alcoholic dementia. Can J Neurol Sci, 1994; 21:88–99

    CAS  PubMed  Google Scholar 

  • Victor M, Brausch C. The role of abstinence in the genesis of alcoholic epilepsy. Epilepsia, 1967; 8: 1–20

    CAS  PubMed  Google Scholar 

  • Victor M, Adams RD, Mancall EL. A restricted form of cerebellar cortical degeneration occurring in alcoholic patients. Arch Neurol, 1959; 71: 579–688

    Google Scholar 

  • Victor M, Adams RD, Collins GH. The Wernicke-Korsakoff syndrome and related neurologic disorders due to alcoholism and malnutrition. Philadelphia: FA Davis, 1989

    Google Scholar 

  • Visser PJ, Krabbendam L, Verhey FR, Hofman PA, Verhoeven WM, Tuinier S, Wester A, Den Berg YW, Goessens LF, Werf YD, Jolles J. Brain correlates of memory dysfunction in alcoholic Korsakoff’s syndrome. J Neurol Neurosurg Psychiatry, 1999; 67: 774–778

    CAS  PubMed  Google Scholar 

  • Harada M, Nakahishi J, Konuma S, Ohno K, Kimura T, Yamaguchi H, Tsuruta K, Kizaki T, Okawara T, Ohno H. The present mercury contents of scalp hair and clinical symptoms in inhabitants of the Minamata area. Environ Res, 1998;77: 160–164

    CAS  PubMed  Google Scholar 

  • Harada M, Nakachi S, Cheu T, Hamada H, Ono Y, Tsuda T, Yanagida K, Kizaki T, Ohno H. Monitoring of mercury pollution in Tanzania: relation between head hair mercury and health. Sci Total Environ, 1999; 227: 249–256

    CAS  PubMed  Google Scholar 

  • Korogi Y, Takahashi M, Hirai T, Ikushima I, Kitajima M, Suguhara T, Shigematsu Y, Okajima T, Mukuno K. Representation of the Visual field in the striate cortex: comparison of MR findings with visual field deficits in organic mercury poisoning (Minamata disease). AJNR Am J Neuroradiol, 1997; 18: 1127–1130

    CAS  PubMed  Google Scholar 

  • Korogi Y, Takhashi M, Okajima T, Eto K. MR findings of Minamata disease — organic mercury poisoning. J Magn Reson Imaging, 1998; 8: 308–316

    CAS  PubMed  Google Scholar 

  • Valk J, Van der Knaap MS. Mercury intoxication. In: Valk J, Van der Knaap MS. Magnetic resonance of myelin, myelinatiOn and myelin disorders. Berlin Heidelberg New York: Springer 1989:270–271

    Google Scholar 

  • Barron TE, Devenyi AG, Mamourian AC. Symptomatic manganese neurotoxicity in a patient with chronic liver disease: correlation of clinical symptoms with MR findings. Pediatr Neurol, 1994; 10: 145–149

    CAS  PubMed  Google Scholar 

  • Cotzias GC, Papavasiliou S, Miller ST. Manganese in melanin. Nature 1964; 201: 1228–1229

    CAS  PubMed  Google Scholar 

  • Couper J. Sur les effets du peroxide de Mn. J Chim Med Pharm Toxicol, 1837; 3: 223–225

    Google Scholar 

  • Donaldson J. The physiologic significance of manganese in brain: its relation to schizophrenia and neurodegenerative disorders. Neurotoxicology, 1987; 3: 451–462

    Google Scholar 

  • Graham DG. Catecholamine toxicity: a proposal for the molecular pathogenesis of manganese toxicity and Parkinson’s disease. Neurotoxicology, 1984; 5: 83–96

    CAS  PubMed  Google Scholar 

  • Greenhouse AH. Heavy metals and the nervous system. Clin Neuropharmacol, 1982; 5: 45–92

    CAS  PubMed  Google Scholar 

  • Valk J, Van der Knaap MS. Toxic encephalopathy. AJNR Am J Neuroradiol, 1988; 13: 747–760

    Google Scholar 

  • Tan TP, Algra PR, Valk J, Walters EC. Toxic leukoencephalopathy after inhalation of poisoned heroin: MR findings. AJNR Am J Neuroradiol, 1994; 15: 175–178

    CAS  PubMed  Google Scholar 

  • Wolters EC, Van Wijngaarden GK, Stam PC. Leukoencephalopathy after inhaling heroin pyrolysite. Lancet, 1982; I:1233–1236

    Google Scholar 

  • Burg JR, Gist GL. Health effects of environmental contaminant exposure: an intrafile comparison of the Trichloroethylene Subregistry. Arch Environ Health, 1999; 54:231–241

    CAS  PubMed  Google Scholar 

  • Caldemeyer KS, Pascuzzi RM, Moran CC, Smith RR. Toluene abuse causing reduced MR signal intensity in the brain. AJR Am J Radiol, 1993; 161: 1259–1261

    CAS  Google Scholar 

  • Donoghe AM, Dryson EW, Wynn-Williams G. Contrast sensitivity in organic solvent induced chronic toxic encephalopathy. J Occup Environ Med, 1998; 37: 1357–1363

    Google Scholar 

  • Escobar A, Aruffo C. Chronic thinner intoxication: clinico-pathologic report of human case. J Neurol Neurosurg Psychiatry, 1980; 43: 986–994

    CAS  PubMed  Google Scholar 

  • Homes JT, Filey CM, Rosenberg NL. Neurologic sequelae of chronic solvent vapor abuse. Neurology, 1986; 36: 698–702

    Google Scholar 

  • Ikeda M, Tsukagoshi H. Encephalopathy due to toluene sniffing: report of a case with magnetic resonance imaging. Eur Neurol, 1990; 30: 347–349

    CAS  PubMed  Google Scholar 

  • Kornfeld M, Moser AB, Moser HW, Kleinschmidt-de Masters BK, Nolte K, Phelps A. Solvent vapor abuse leukoencephalopathy: comparison to adrenoleukodystrophy. J Neuropathol Exp Neurol, 1994; 53: 389–398

    CAS  PubMed  Google Scholar 

  • Palmer K, Inskip H, Martyn C, Coggon D. Dementia and occupational exposure to organic solvents. Occup Environ Med, 1998;55:712–715

    CAS  PubMed  Google Scholar 

  • Rosenberg NL, Kleinschmidt-Demasters KK, Davis KA, Dreisbach JN, Hormes JT, Filley CM. Toluene abuse causes diffuse central nervous system white matter changes. Ann Neurol, 1988; 23: 611–614

    CAS  PubMed  Google Scholar 

  • Ackrill P, Ralston AJ, Day JP, Hodge KC. Successful removal of aluminum from a patient with dialysis encephalopathy. Lancet, 1980; II: 692–693

    Google Scholar 

  • Alfrey AC, Miskell JM, Barks J, Contiguglia SR, Rudoph H, Lewin E, Holmes JH. Syndrome of dyspraxia and multifocal seizures associated with chronic hemodialysis. Trans Am Soc Artific Organs 1972; 18: 257–261

    CAS  Google Scholar 

  • Alfrey AC, Le Gendre GR, Kaehny WD. The dialysis encephalopathy syndrome: possible aluminum intoxication. N Engl J Med, 1976;294: 184–188

    CAS  PubMed  Google Scholar 

  • Banks WA, Kastin AJ. Aluminum increases permeability of the blood-brain barrier to labeled DSIP and beta-endorphin: possible implications for senile and dialysis dementia. Lancet, 1983; II: 1227–1229

    Google Scholar 

  • Bolla KI, Milstien S, Briefel G, Wieler L, Kaufman S. Dihydropteridine reductase activity: lack of association with serum aluminum levels and cognitive functioning in patients with end-stage renal disease. Neurology, 1991; 41:1806–1809

    CAS  PubMed  Google Scholar 

  • Boyce N, Wood C, Holdsworth S, Thomson NM, Atkins RC. Life-threatening sepsis complicating heavy metal chelation therapy with desferrioxamine. Aust N Z J Med, 1985; 15:654–655

    CAS  PubMed  Google Scholar 

  • Chui HC, Damasio AR. Progressive dialysis encephalopathy (“dialysis dementia”). J Neurol, 1980; 222: 145–157

    CAS  PubMed  Google Scholar 

  • Davison AM, Walker GS, Oli H, Lewins AM. Water supply aluminum concentration, dialysis dementia, and effect of reverse-osmosis water treatment. Lancet, 1982; II:785–787

    Google Scholar 

  • English A, Savage RD, Britton PG, Ward MK, Kerr DN. Intel-lectual impairment in chronic renal failure. BMI, 1978; I:888–890

    Google Scholar 

  • Etheridge WB, O’Neill WM Jr. The “dialysis encephalopathy syndrome” without dialysis. Clin Nephrol, 1978; 10: 250–252

    CAS  PubMed  Google Scholar 

  • Fraser CL. Neurologic manifestations of the uremic state. In: Arieff AI, Griggs RC, eds. Metabolic brain dysfunction in systemic disorders. Boston: Little Brown, 1992:139–166

    Google Scholar 

  • Jackson M, Warrington EK, Roe CJ, Baker LR. Cognitive function in haemodialysis patients. Clin Nephrol, 1987; 27:26–30

    CAS  PubMed  Google Scholar 

  • Kiley JE. Neurological aspects of dialysis. In: Nissenson AR, Fine RN, Gentile DE, eds. Clinical dialysis. Norwalk, NI: Appleton & Lange, 1990: 535–558

    Google Scholar 

  • Lai JC, Blass JP. Inhibition of brain glycolysis by aluminum. J Neurochem, 1984; 42: 438–446

    CAS  PubMed  Google Scholar 

  • Mahoney CA, Arieff AI. Uremic encephalopathies: clinical, biochemical, and experimental features. Am J Kidney Dis, 1982;2:324–336

    CAS  PubMed  Google Scholar 

  • Mahurkar SD, Salta R, Smith EC, Dhar SK, Meyers L Jr, Dunea G. Dialysis dementia. Lancet 1973; I: 1412–1415

    Google Scholar 

  • Matsubara O, Nakagawa S, Shinoda T, Iwamoto H, Kanno J, Okeda R, Kasuga T. Progressive multifocal leukoencephalopathy associated with prolonged hemodialysis treatment. Virchows Arch A Pathol Anat Histopathol, 1984;403:301–305

    CAS  PubMed  Google Scholar 

  • Molitoris BA, Alfrey AC, Alfrey PS, Miller NL. Rapid removal of DFO-chelated aluminum during hemodialysis using polysulfone dialyzers. Kidney Int, 1988; 34: 98–101

    CAS  PubMed  Google Scholar 

  • Rivera-Vazquez AB, Noriega-Sanchez A, Ramirez-Gonzales R, Martinez-Maldonado M. Acute hypercalcemia in hemodialysis patients: distinctive form dialysis dementia. Nephron, 1980;25:243–246

    CAS  PubMed  Google Scholar 

  • Rosati G, De Bastiani P, Gilli P, Paolino E. Oral aluminum and neuropsychological functioning: a study of dialysis patients receiving aluminum hydroxide gels. J Neurol, 1980;223:251–257

    CAS  PubMed  Google Scholar 

  • Scholtz CL, Swash M, Gray A, Kogeorgos J, Marsh F. Neuro-fibrillary neuronal degeneration in dialysis dementia: a feature of aluminum toxicity. Clin Neuropathol, 1987; 6: 93–97

    CAS  PubMed  Google Scholar 

  • Sherrard DJ, Walker JV, Boykin JL. Precipitation of dialysis dementia by deferoxamine treatment of aluminum-related bone disease. Am J Kidney Dis, 1988; 12: 126–130

    CAS  PubMed  Google Scholar 

  • Sideman S, Manor D. The dialysis dementia syndrome and aluminum intoxication. Nephron, 1982; 31: 1–10

    CAS  PubMed  Google Scholar 

  • Walker JA, Sherman RA, Eisinger RP. Thrombocytopenia associated with intravenous desferrioxamine. Am J Kidney Dis, 1985; 6: 254–256

    CAS  PubMed  Google Scholar 

  • White DM, Longstreth WT Jr, Rosenstock L, Claypoole KH, Brodkin CA, Townes BD. Neurologic syndrome in 25 workers from an aluminum smelting plant. Arch Intern Med, 1992; 152: 1443–1448

    CAS  PubMed  Google Scholar 

  • Ahern GL, O’Connor M, Dalmau J, Coleman A, Posner JB. Paraneoplastic temporal lobe epilepsy with testicular neoplasm and atypical amnesia. Neurology, 1994; 44:1270–1274

    CAS  PubMed  Google Scholar 

  • Alamowitch S, Graus F, Uchuya M, Rene R, Bescansa E, Delattre JY. Limbic encephalitis and small cell lung cancer: clinical and immunological features. Brain, 1997; 120:923–928

    PubMed  Google Scholar 

  • Antoine JC, Absi L, Honnorat J, et al. Antiamphiphysin anti-bodies are associated with various paraneoplastic neurological syndromes and tumors. Arch Neurol, 1999; 56:172–177

    CAS  PubMed  Google Scholar 

  • Antoine JC, Honnorat J, Anterion CT, Aguera M, Absi L, Fournel P. Limbic encephalitis and immunological perturbations in two patients with thymoma. J Neurol Neurosurg Psychiatry, 1995; 58: 706–710

    CAS  PubMed  Google Scholar 

  • Aydiner A, Gurvit H, Baral I. Paraneoplastic limbic encephalitis with immature ovarian teratoma. J Neurooncol, 1998;37:63–66

    CAS  PubMed  Google Scholar 

  • Bakheit AM, Kennedy PG, Behan PO. Paraneoplastic limbic encephalitis: clinicopathologic correlations. J Neurol Neurosurg Psychiatry, 1990; 53: 1084–1088

    CAS  PubMed  Google Scholar 

  • Bennett JL, Galetta SL, Frohman LP, et al. Neuro-ophthalmologic manifestations of a paraneoplastic syndrome and testicular carcinoma. Neurology, 1999; 52: 864–867

    CAS  PubMed  Google Scholar 

  • Brennan LV, Craddock PR. Limbic encephalopathy as a non-metastatic complication of oat cell lung cancer: its reversal after treatment of the primary lung lesion. Am J Med, 1983;75:518–520

    CAS  PubMed  Google Scholar 

  • Brierley JB, Corsellis JA, Hierons R, Nevin S. Subacute encephalitis of later adult life mainly affecting the limbic areas. Brain, 1960;83:357–368

    Google Scholar 

  • Burton GV, Bullard DE, Walther PJ, Burger PC. Paraneoplastic limbic encephalopathy with testicular carcinoma: a reversible neurologic syndrome. Cancer, 1988; 62: 2248–2251

    CAS  PubMed  Google Scholar 

  • Corsellis JA, Goldberg GJ, Norton AR. “Limbic encephalitis” and its association with carcinoma. Brain, 1968; 91: 481–496

    CAS  PubMed  Google Scholar 

  • Cunningham JD, Burt ME. Limbic encephalitis secondary to malignant thymoma. Ann Thorac Surg, 1994; 58: 250–251

    CAS  PubMed  Google Scholar 

  • Dalmau J, Graus F, Rosenblum MK, Posner JB. Anti-Hu-associated paraneoplastic encephalomyelitis/sensory neuronopathy: a clinical study of 71 patients. Medicine (Baltimore), 1992; 71: 59–72

    CAS  Google Scholar 

  • Daniel SE, Love S, Scaravilli F, Harding AE. Encephalomyeloneuropathy in the absence of a detectable neoplasm. Acta Neuropathol, 1985; 66: 311–317

    CAS  PubMed  Google Scholar 

  • Den Hollander AM, Van Hulst AM, Meerwaldt JD, Haasjes JG. Limbic encephalitis: a rare presentation of the small-cell lung carcinoma. Gen Hosp Psychiatry, 1989; 11: 3883–92

    Google Scholar 

  • Deodhare S, O’Connor P, Ghazarian D, Bilbao JM. Paraneo-plastic limbic encephalitis in Hodgkin’s disease. Can J Neurol Sci, 1996; 23: 138–140

    CAS  PubMed  Google Scholar 

  • Dirr LY, Elster AD, Donofrio PD, Smith M. Evolution of brain MRI abnormalities in limbic encephalitis. Neurology, 1990;40: 1304–1306

    CAS  PubMed  Google Scholar 

  • Dropcho EJ. Neurologic paraneoplastic syndromes. J Neurol Sci, 1998; 153: 264–278

    CAS  PubMed  Google Scholar 

  • Duyckaerts C, Derouesne C, Signoret JL, et al. Bilateral and limited amygdalohippocampal lesions causing a pure amnestic syndrome. Ann Neurol, 1985; 18: 314–319

    CAS  PubMed  Google Scholar 

  • Evoli A, Lo Monaco M, Maria R, Lino MM, Batocchi AP, Tonali PA. Multiple paraneoplastic diseases associated with thymoma. Neuromusc Disord, 1999; 9: 601–603

    CAS  PubMed  Google Scholar 

  • Gascon GG, Gilles F. Limbic dementia. J Neurol Neurosurg Psychiatry, 1973; 36: 421–430

    CAS  PubMed  Google Scholar 

  • Glaser GH, Pincus JH. Limbic encephalitis. J Nerv Ment Dis, 1969;149:59–67

    CAS  PubMed  Google Scholar 

  • Graus F, Vega F, Delattre JY, et al. Plasmapheresis and antineoplastic treatment in CNS paraneoplastic syndromes with antineuronal autoantibodies. Neurology, 1992; 42:536–540

    CAS  PubMed  Google Scholar 

  • Johnson PC, Rolak LA, Hamilton RH, Laguna JF. Paraneo-plastic vasculitis of nerve: a remote effect of cancer. Ann Neurol, 1979; 5: 437–444

    CAS  PubMed  Google Scholar 

  • Kaniecki R, Morris JC. Reversible paraneoplastic limbic encephalitis. Neurology, 1993; 43: 2418–2419

    CAS  PubMed  Google Scholar 

  • Kaplan AM, Itabashi HH. Encephalitis associated with carcinoma: central hypoventilation syndrome and cytoplasmic inclusion bodies. J Neurol Neurosurg Psychiatry, 1974;37: 1166–1176

    CAS  PubMed  Google Scholar 

  • Kodama T, Numaguchi Y, Gellad FE, Dwyer BA, Kristt DA. Magnetic resonance imaging of limbic encephalitis. Neuroradiology, 1991;33:520–523

    CAS  PubMed  Google Scholar 

  • Kohler J, Hufschmidt A, Hermle L, Volk B, Lucking CH. Limbic encephalitis: two cases. J Neurooncol, 1988; 20: 177–178

    CAS  Google Scholar 

  • Lacomis D, Khoshbin S, Schick RM. MR imaging of paraneoplastic limbic encephalitis. J Comput Assist Tomogr, 1990; 14: 115–117

    CAS  PubMed  Google Scholar 

  • Lovblad HE, Boucraut J, Steck AJ. Paraneoplastic subacute sensory Denny-Brown neuropathy or anti-Hu syndrome: a classical neoplastic syndrome. Rev Med Suisse Romande 1995; 115: 421–426

    CAS  PubMed  Google Scholar 

  • McArdle JP, Millingen KS. Limbic encephalitis associated with malignant thymoma. Pathology, 1988; 20: 292–295

    CAS  PubMed  Google Scholar 

  • Newman NJ, Bell IR, McKee AC. Paraneoplastic limbic encephalitis: neuropsychiatric presentation. Biol Psychiatry, 1990;27:529–542

    CAS  PubMed  Google Scholar 

  • Nokura K, Yamamoto H, Okawara Y, Koga H, Osawa H, Sakai K. Reversible limbic encephalitis caused by ovarian teratoma. Acta Neurol Scand, 1997; 95: 367–373

    CAS  PubMed  Google Scholar 

  • Petit T, Janser JC, Achour NR, Borel C, Haegele P. Paraneoplastic temporal lobe epilepsy and anti-Yo autoantibody. Ann Oncol, 1997; 8: 919

    CAS  PubMed  Google Scholar 

  • Provenzale JM, Barboriak DP, Coleman RE. Limbic encephalitis: comparison of FDG, PET and MR imaging findings. AJR Am J Roentgenol, 1998; 170: 1659–1660

    CAS  PubMed  Google Scholar 

  • Rosenbaum T, Gartner J, Korholz D, Janssen G, Schneider D, Engelbrecht V, Gobel U, Lenard HG. Paraneoplastic limbic encephalitis in two teenage girls. Neuropediatrics, 1998;29: 159–162

    CAS  PubMed  Google Scholar 

  • Stern RC, Hulette CM. Paraneoplastic limbic encephalitis associated with small cell carcinoma of the prostate. Mod Pathol, 1999; 12: 814–818

    CAS  PubMed  Google Scholar 

  • Sutton RC, Lipper MH, Brashear HR. Limbic encephalitis occurring in association with Alzheimer’s disease. J Neurol Neurosurg Psychiatry, 1993; 56: 808–811

    CAS  PubMed  Google Scholar 

  • Bodensteiner JB, Schaefer GB. Dementia pugilistica and ca-vum septi pellucidi: born to box? Sports Med, 1997; 24: 361–365

    CAS  PubMed  Google Scholar 

  • Carlock KS, Williams JP, Graves GC. MRI findings in headbangers. Clin Imaging, 1997; 21: 411–413

    CAS  PubMed  Google Scholar 

  • Geddes JF, Vowles GH, Nicoll JA, Revesz T. Neuronal cytoskeletal changes are an early consequence of repetitive head injury. Acta Neuropathol, 1999; 98: 171–178

    CAS  PubMed  Google Scholar 

  • Jordan BD, Relkin NR, Ravdin LD, Jacobs AR, Bennett A, Gandy S. Apolipoprotein E epsilon 4 associated with chronic traumatic brain injury in boxing. JAMA, 1997; 278:136–140

    CAS  PubMed  Google Scholar 

  • Jordan SE, Green GA, Galanty HL, Mandelbaum BR, Jabour BA. Acute and chronic brain injury in United States National Team soccer players. Am J Sports Med, 1996; 24:205–210

    CAS  PubMed  Google Scholar 

  • Mawdsley C, Ferguson FR, Isherwood I. Pneumencephalo-graphic changes in boxers. Act Radiol 1966; 5: 654–657

    Google Scholar 

  • McKenzie JE, Roberts GW, Royston MC. Comparative investigation of neurofibrillary damage in the temporal lobe in Alzheimer’s disease, Down’s syndrome and dementia pugilistica. Neurodegeneration, 1996; 5: 259–264

    CAS  PubMed  Google Scholar 

  • Moseley IF. The neuroimaging evidence for chronic brain damage due to boxing. Neuroradiology, 2000; 42: 1–8

    CAS  PubMed  Google Scholar 

  • Chan YL, Leung SF, King AD, Choi PH, Metreweli C. Late radiation injury to the temporal lobes: morphologic evaluation at MR imaging. Radiology, 1999; 213: 800–807

    CAS  PubMed  Google Scholar 

  • Chong VF, Fan YF, Mukherji SK. Radiation-induced temporal lobe changes: CT and MR imaging characteristics. AJR Am J Roentgenol, 2000; 175: 431–436

    CAS  PubMed  Google Scholar 

  • Chong VF, Rumpel H, Aw YS, Ho GL, Fan YF, Chua EJ. Temporal lobe necrosis following radiotherapy for nasopharyngeal carcinoma: 1H MR spectroscopic findings. Int J Radiat Oncol Biol Phys, 1999; 45: 699–705

    CAS  PubMed  Google Scholar 

  • Cirafisi C, Verderame F. Radiation-induced rhombencephalopathy. Ital J Neurol Sci, 1999; 20: 55–58

    CAS  PubMed  Google Scholar 

  • Meyers CA, Geara F, Wong PF, Morrison WHo Neurocognitive effects of therapeutic irradiation for base of skull tumors. Int J Radiat Oncol Biol Phys, 2000; 46: 51–55

    CAS  PubMed  Google Scholar 

  • Plowman PN. Stereotactic radiosurgery. VIII. The classification of postradiation reactions. Br J Neurosurg, 1999; 13: 256–264

    CAS  PubMed  Google Scholar 

  • Sugahara T, Korogi Y, Tomiguchi S, Shigematsu V, Ikushima I, Kira T, Liang L, Ushio Y, Takahashi M. Posttherapeutic intraaxial brain tumor: the value of perfusion sensitive contrast-enhanced MR imaging for differentiating tumor recurrence from nonneoplasic contrast-enhancing tissue. AJNR Am J Neuroradiol, 2000; 21: 901–909

    CAS  PubMed  Google Scholar 

  • Thompson TP, Lunsford LD, Kondziolka D. Distinguishing tumor and radiation necrosis with positron emission tomography versus stereotactic biopsy. Stereotact Funct Neurosurg, 1999;73:9–14

    CAS  PubMed  Google Scholar 

  • Tsui EY, Chan JH, Leung TW, Yuen MK, Cheung YK, Luk SH, Tung SY. Radionecrosis of the temporal lobe: dynamic susceptibility contrast MRI. Neuroradiology, 2000;42: 149–152

    CAS  PubMed  Google Scholar 

  • Vigliani MC, Duyckaerts C, Hauw JJ, Poisson M, Magdelenat H, Delattre JV. Dementia following treatment of brain tumors with radiotherapy administered alone or in combination with nitrosurea-based chemotherapy: a clinical and pathological study. J Neurooncol, 1999; 41: 137–149

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

Copyright information

© 2002 Springer-Verlag Berlin Heidelberg

About this chapter

Cite this chapter

Valk, J., Barkhof, F., Scheltens, P. (2002). Disorders Primarily Affecting White Matter. In: Magnetic Resonance in Dementia. Springer, Berlin, Heidelberg. https://doi.org/10.1007/978-3-642-56269-3_5

Download citation

  • DOI: https://doi.org/10.1007/978-3-642-56269-3_5

  • Publisher Name: Springer, Berlin, Heidelberg

  • Print ISBN: 978-3-642-62597-8

  • Online ISBN: 978-3-642-56269-3

  • eBook Packages: Springer Book Archive

Publish with us

Policies and ethics