Skip to main content
Top
Published in: European Journal of Nuclear Medicine and Molecular Imaging 3/2017

01-03-2017 | Review Article

Positron emission tomography in amyotrophic lateral sclerosis: Towards targeting of molecular pathological hallmarks

Authors: Stefanie M. A. Willekens, Donatienne Van Weehaeghe, Philip Van Damme, Koen Van Laere

Published in: European Journal of Nuclear Medicine and Molecular Imaging | Issue 3/2017

Login to get access

Abstract

During the past decades, extensive efforts have been made to expand the knowledge of amyotrophic lateral sclerosis (ALS). However, clinical translation of this research, in terms of earlier diagnosis and improved therapy, remains challenging. Since more than 30% of motor neurons are lost when symptoms become clinically apparent, techniques allowing non-invasive, in vivo detection of motor neuron degeneration are needed in the early, pre-symptomatic disease stage. Furthermore, it has become apparent that non-motor signs play an important role in the disease and there is an overlap with cognitive disorders, such as frontotemporal dementia (FTD). Radionuclide imaging, such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT), form an attractive approach to quantitatively monitor the ongoing neurodegenerative processes. Although [18F]-FDG has been recently proposed as a potential biomarker for ALS, active targeting of the underlying pathologic molecular processes is likely to unravel further valuable disease information and may help to decipher the pathogenesis of ALS. In this review, we provide an overview of radiotracers that have already been applied in ALS and discuss possible novel targets for in vivo imaging of various pathogenic processes underlying ALS onset and progression.
Literature
3.
go back to reference Leigh PN, Abrahams S, Al-Chalabi A, Ampong MA, Goldstein LH, Johnson J. The management of motor neurone disease. J Neurol Neurosurg Psychiatry. 2003;74 Suppl 4:iv32–47.PubMedPubMedCentral Leigh PN, Abrahams S, Al-Chalabi A, Ampong MA, Goldstein LH, Johnson J. The management of motor neurone disease. J Neurol Neurosurg Psychiatry. 2003;74 Suppl 4:iv32–47.PubMedPubMedCentral
4.
7.
go back to reference Kretschmer BD, Kratzer U, Schmidt WJ. Riluzole, a glutamate release inhibitor, and motor behavior. Naunyn Schmiedebergs Arch Pharmacol. 1998;358(2):181–90.PubMedCrossRef Kretschmer BD, Kratzer U, Schmidt WJ. Riluzole, a glutamate release inhibitor, and motor behavior. Naunyn Schmiedebergs Arch Pharmacol. 1998;358(2):181–90.PubMedCrossRef
9.
go back to reference Lacomblez L, Bensimon G, Leigh PN, Guillet P, Meininger V. Dose-ranging study of riluzole in amyotrophic lateral sclerosis. Amyotrophic Lateral Sclerosis/Riluzole Study Group II. Lancet. 1996;347(9013):1425–31.PubMedCrossRef Lacomblez L, Bensimon G, Leigh PN, Guillet P, Meininger V. Dose-ranging study of riluzole in amyotrophic lateral sclerosis. Amyotrophic Lateral Sclerosis/Riluzole Study Group II. Lancet. 1996;347(9013):1425–31.PubMedCrossRef
13.
go back to reference Rosen DR. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature. 1993;364(6435):362. doi:10.1038/364362c0.PubMed Rosen DR. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature. 1993;364(6435):362. doi:10.​1038/​364362c0.PubMed
14.
go back to reference Kwiatkowski Jr TJ, Bosco DA, Leclerc AL, Tamrazian E, Vanderburg CR, Russ C, et al. Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science. 2009;323(5918):1205–8. doi:10.1126/science.1166066.PubMedCrossRef Kwiatkowski Jr TJ, Bosco DA, Leclerc AL, Tamrazian E, Vanderburg CR, Russ C, et al. Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science. 2009;323(5918):1205–8. doi:10.​1126/​science.​1166066.PubMedCrossRef
18.
go back to reference Chio A, Borghero G, Restagno G, Mora G, Drepper C, Traynor BJ, et al. Clinical characteristics of patients with familial amyotrophic lateral sclerosis carrying the pathogenic GGGGCC hexanucleotide repeat expansion of C9ORF72. Brain. 2012;135(Pt 3):784–93. doi:10.1093/brain/awr366.PubMedPubMedCentralCrossRef Chio A, Borghero G, Restagno G, Mora G, Drepper C, Traynor BJ, et al. Clinical characteristics of patients with familial amyotrophic lateral sclerosis carrying the pathogenic GGGGCC hexanucleotide repeat expansion of C9ORF72. Brain. 2012;135(Pt 3):784–93. doi:10.​1093/​brain/​awr366.PubMedPubMedCentralCrossRef
19.
go back to reference Millecamps S, Boillee S, Le Ber I, Seilhean D, Teyssou E, Giraudeau M, et al. Phenotype difference between ALS patients with expanded repeats in C9ORF72 and patients with mutations in other ALS-related genes. J Med Genet. 2012;49(4):258–63. doi:10.1136/jmedgenet-2011-100699.PubMedCrossRef Millecamps S, Boillee S, Le Ber I, Seilhean D, Teyssou E, Giraudeau M, et al. Phenotype difference between ALS patients with expanded repeats in C9ORF72 and patients with mutations in other ALS-related genes. J Med Genet. 2012;49(4):258–63. doi:10.​1136/​jmedgenet-2011-100699.PubMedCrossRef
20.
go back to reference Lomen-Hoerth C, Anderson T, Miller B. The overlap of amyotrophic lateral sclerosis and frontotemporal dementia. Neurology. 2002;59(7):1077–9.PubMedCrossRef Lomen-Hoerth C, Anderson T, Miller B. The overlap of amyotrophic lateral sclerosis and frontotemporal dementia. Neurology. 2002;59(7):1077–9.PubMedCrossRef
24.
go back to reference Neary D, Snowden JS, Mann DM. Cognitive change in motor neurone disease/amyotrophic lateral sclerosis (MND/ALS). J Neurol Sci. 2000;180(1-2):15–20.PubMedCrossRef Neary D, Snowden JS, Mann DM. Cognitive change in motor neurone disease/amyotrophic lateral sclerosis (MND/ALS). J Neurol Sci. 2000;180(1-2):15–20.PubMedCrossRef
28.
go back to reference Lyras L, Evans PJ, Shaw PJ, Ince PG, Halliwell B. Oxidative damage and motor neurone disease difficulties in the measurement of protein carbonyls in human brain tissue. Free Radic Res. 1996;24(5):397–406.PubMedCrossRef Lyras L, Evans PJ, Shaw PJ, Ince PG, Halliwell B. Oxidative damage and motor neurone disease difficulties in the measurement of protein carbonyls in human brain tissue. Free Radic Res. 1996;24(5):397–406.PubMedCrossRef
30.
go back to reference Simpson EP, Henry YK, Henkel JS, Smith RG, Appel SH. Increased lipid peroxidation in sera of ALS patients: a potential biomarker of disease burden. Neurology. 2004;62(10):1758–65.PubMedCrossRef Simpson EP, Henry YK, Henkel JS, Smith RG, Appel SH. Increased lipid peroxidation in sera of ALS patients: a potential biomarker of disease burden. Neurology. 2004;62(10):1758–65.PubMedCrossRef
31.
go back to reference Wiedemann FR, Manfredi G, Mawrin C, Beal MF, Schon EA. Mitochondrial DNA and respiratory chain function in spinal cords of ALS patients. J Neurochem. 2002;80(4):616–25.PubMedCrossRef Wiedemann FR, Manfredi G, Mawrin C, Beal MF, Schon EA. Mitochondrial DNA and respiratory chain function in spinal cords of ALS patients. J Neurochem. 2002;80(4):616–25.PubMedCrossRef
32.
go back to reference Kong J, Xu Z. Massive mitochondrial degeneration in motor neurons triggers the onset of amyotrophic lateral sclerosis in mice expressing a mutant SOD1. J Neurosci. 1998;18(9):3241–50.PubMed Kong J, Xu Z. Massive mitochondrial degeneration in motor neurons triggers the onset of amyotrophic lateral sclerosis in mice expressing a mutant SOD1. J Neurosci. 1998;18(9):3241–50.PubMed
34.
go back to reference Wang W, Wang L, Lu J, Siedlak SL, Fujioka H, Liang J, et al. The inhibition of TDP-43 mitochondrial localization blocks its neuronal toxicity. Nat Med. 2016. doi:10.1038/nm.4130. Wang W, Wang L, Lu J, Siedlak SL, Fujioka H, Liang J, et al. The inhibition of TDP-43 mitochondrial localization blocks its neuronal toxicity. Nat Med. 2016. doi:10.​1038/​nm.​4130.
36.
go back to reference Shaw PJ, Forrest V, Ince PG, Richardson JP, Wastell HJ. CSF and plasma amino acid levels in motor neuron disease: elevation of CSF glutamate in a subset of patients. Neurodegeneration. 1995;4(2):209–16.PubMedCrossRef Shaw PJ, Forrest V, Ince PG, Richardson JP, Wastell HJ. CSF and plasma amino acid levels in motor neuron disease: elevation of CSF glutamate in a subset of patients. Neurodegeneration. 1995;4(2):209–16.PubMedCrossRef
37.
go back to reference Okamoto K, Hirai S, Amari M, Watanabe M, Sakurai A. Bunina bodies in amyotrophic lateral sclerosis immunostained with rabbit anti-cystatin C serum. Neurosci Lett. 1993;162(1-2):125–8.PubMedCrossRef Okamoto K, Hirai S, Amari M, Watanabe M, Sakurai A. Bunina bodies in amyotrophic lateral sclerosis immunostained with rabbit anti-cystatin C serum. Neurosci Lett. 1993;162(1-2):125–8.PubMedCrossRef
38.
go back to reference Schmidt ML, Carden MJ, Lee VM, Trojanowski JQ. Phosphate dependent and independent neurofilament epitopes in the axonal swellings of patients with motor neuron disease and controls. Lab Invest. 1987;56(3):282–94.PubMed Schmidt ML, Carden MJ, Lee VM, Trojanowski JQ. Phosphate dependent and independent neurofilament epitopes in the axonal swellings of patients with motor neuron disease and controls. Lab Invest. 1987;56(3):282–94.PubMed
39.
go back to reference Zhang B, Tu P, Abtahian F, Trojanowski JQ, Lee VM. Neurofilaments and orthograde transport are reduced in ventral root axons of transgenic mice that express human SOD1 with a G93A mutation. J Cell Biol. 1997;139(5):1307–15.PubMedPubMedCentralCrossRef Zhang B, Tu P, Abtahian F, Trojanowski JQ, Lee VM. Neurofilaments and orthograde transport are reduced in ventral root axons of transgenic mice that express human SOD1 with a G93A mutation. J Cell Biol. 1997;139(5):1307–15.PubMedPubMedCentralCrossRef
40.
go back to reference Mackenzie IR, Bigio EH, Ince PG, Geser F, Neumann M, Cairns NJ, et al. Pathological TDP-43 distinguishes sporadic amyotrophic lateral sclerosis from amyotrophic lateral sclerosis with SOD1 mutations. Ann Neurol. 2007;61(5):427–34. doi:10.1002/ana.21147.PubMedCrossRef Mackenzie IR, Bigio EH, Ince PG, Geser F, Neumann M, Cairns NJ, et al. Pathological TDP-43 distinguishes sporadic amyotrophic lateral sclerosis from amyotrophic lateral sclerosis with SOD1 mutations. Ann Neurol. 2007;61(5):427–34. doi:10.​1002/​ana.​21147.PubMedCrossRef
41.
46.
go back to reference Brooks BR, Miller RG, Swash M, Munsat TL. World Federation of Neurology Research Group on Motor Neuron D. El Escorial revisited: revised criteria for the diagnosis of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Other Motor Neuron Disord. 2000;1(5):293–9.PubMedCrossRef Brooks BR, Miller RG, Swash M, Munsat TL. World Federation of Neurology Research Group on Motor Neuron D. El Escorial revisited: revised criteria for the diagnosis of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Other Motor Neuron Disord. 2000;1(5):293–9.PubMedCrossRef
47.
go back to reference Schrooten M, Smetcoren C, Robberecht W, Van Damme P. Benefit of the Awaji diagnostic algorithm for amyotrophic lateral sclerosis: a prospective study. Ann Neurol. 2011;70(1):79–83. doi:10.1002/ana.22380.PubMedCrossRef Schrooten M, Smetcoren C, Robberecht W, Van Damme P. Benefit of the Awaji diagnostic algorithm for amyotrophic lateral sclerosis: a prospective study. Ann Neurol. 2011;70(1):79–83. doi:10.​1002/​ana.​22380.PubMedCrossRef
50.
go back to reference Peretti-Viton P, Azulay JP, Trefouret S, Brunel H, Daniel C, Viton JM, et al. MRI of the intracranial corticospinal tracts in amyotrophic and primary lateral sclerosis. Neuroradiology. 1999;41(10):744–9.PubMedCrossRef Peretti-Viton P, Azulay JP, Trefouret S, Brunel H, Daniel C, Viton JM, et al. MRI of the intracranial corticospinal tracts in amyotrophic and primary lateral sclerosis. Neuroradiology. 1999;41(10):744–9.PubMedCrossRef
51.
go back to reference Waragai M. MRI and clinical features in amyotrophic lateral sclerosis. Neuroradiology. 1997;39(12):847–51.PubMedCrossRef Waragai M. MRI and clinical features in amyotrophic lateral sclerosis. Neuroradiology. 1997;39(12):847–51.PubMedCrossRef
53.
go back to reference Ciccarelli O, Behrens TE, Altmann DR, Orrell RW, Howard RS, Johansen-Berg H, et al. Probabilistic diffusion tractography: a potential tool to assess the rate of disease progression in amyotrophic lateral sclerosis. Brain. 2006;129(Pt 7):1859–71. doi:10.1093/brain/awl100.PubMedCrossRef Ciccarelli O, Behrens TE, Altmann DR, Orrell RW, Howard RS, Johansen-Berg H, et al. Probabilistic diffusion tractography: a potential tool to assess the rate of disease progression in amyotrophic lateral sclerosis. Brain. 2006;129(Pt 7):1859–71. doi:10.​1093/​brain/​awl100.PubMedCrossRef
60.
go back to reference Quartuccio N, Van Weehaeghe D, Cistaro A, Jonsson C, Van Laere K, Pagani M. Positron emission tomography neuroimaging in amyotrophic lateral sclerosis: what is new? Q J Nucl Med Mol Imaging. 2014;58(4):344–54.PubMed Quartuccio N, Van Weehaeghe D, Cistaro A, Jonsson C, Van Laere K, Pagani M. Positron emission tomography neuroimaging in amyotrophic lateral sclerosis: what is new? Q J Nucl Med Mol Imaging. 2014;58(4):344–54.PubMed
62.
go back to reference Ludolph AC, Langen KJ, Regard M, Herzog H, Kemper B, Kuwert T, et al. Frontal lobe function in amyotrophic lateral sclerosis: a neuropsychologic and positron emission tomography study. Acta Neurol Scand. 1992;85(2):81–9.PubMedCrossRef Ludolph AC, Langen KJ, Regard M, Herzog H, Kemper B, Kuwert T, et al. Frontal lobe function in amyotrophic lateral sclerosis: a neuropsychologic and positron emission tomography study. Acta Neurol Scand. 1992;85(2):81–9.PubMedCrossRef
63.
go back to reference Hoffman JM, Mazziotta JC, Hawk TC, Sumida R. Cerebral glucose utilization in motor neuron disease. Arch Neurol. 1992;49(8):849–54.PubMedCrossRef Hoffman JM, Mazziotta JC, Hawk TC, Sumida R. Cerebral glucose utilization in motor neuron disease. Arch Neurol. 1992;49(8):849–54.PubMedCrossRef
64.
go back to reference Renard D, Collombier L, Castelnovo G, Fourcade G, Kotzki PO, LaBauge P. Brain FDG-PET changes in ALS and ALS-FTD. Acta Neurol Belg. 2011;111(4):306–9.PubMed Renard D, Collombier L, Castelnovo G, Fourcade G, Kotzki PO, LaBauge P. Brain FDG-PET changes in ALS and ALS-FTD. Acta Neurol Belg. 2011;111(4):306–9.PubMed
65.
go back to reference Cistaro A, Valentini MC, Chio A, Nobili F, Calvo A, Moglia C, et al. Brain hypermetabolism in amyotrophic lateral sclerosis: a FDG PET study in ALS of spinal and bulbar onset. Eur J Nucl Med Mol Imaging. 2012;39(2):251–9. doi:10.1007/s00259-011-1979-6.PubMedCrossRef Cistaro A, Valentini MC, Chio A, Nobili F, Calvo A, Moglia C, et al. Brain hypermetabolism in amyotrophic lateral sclerosis: a FDG PET study in ALS of spinal and bulbar onset. Eur J Nucl Med Mol Imaging. 2012;39(2):251–9. doi:10.​1007/​s00259-011-1979-6.PubMedCrossRef
67.
go back to reference Van Laere K, Vanhee A, Verschueren J, De Coster L, Driesen A, Dupont P, et al. Value of 18fluorodeoxyglucose-positron-emission tomography in amyotrophic lateral sclerosis: a prospective study. JAMA Neurol. 2014;71(5):553–61. doi:10.1001/jamaneurol.2014.62.PubMedCrossRef Van Laere K, Vanhee A, Verschueren J, De Coster L, Driesen A, Dupont P, et al. Value of 18fluorodeoxyglucose-positron-emission tomography in amyotrophic lateral sclerosis: a prospective study. JAMA Neurol. 2014;71(5):553–61. doi:10.​1001/​jamaneurol.​2014.​62.PubMedCrossRef
68.
go back to reference Van Weehaeghe D, Ceccarini J, Delva A, Robberecht W, Van Damme P, Van Laere K. Prospective validation of 18F-FDG brain PET discriminant analysis methods in the diagnosis of amyotrophic lateral sclerosis. J Nucl Med. 2016. doi:10.2967/jnumed.115.166272.PubMed Van Weehaeghe D, Ceccarini J, Delva A, Robberecht W, Van Damme P, Van Laere K. Prospective validation of 18F-FDG brain PET discriminant analysis methods in the diagnosis of amyotrophic lateral sclerosis. J Nucl Med. 2016. doi:10.​2967/​jnumed.​115.​166272.PubMed
69.
go back to reference Pagani M, Oberg J, De Carli F, Calvo A, Moglia C, Canosa A, et al. Metabolic spatial connectivity in amyotrophic lateral sclerosis as revealed by independent component analysis. Hum Brain Mapp. 2016;37(3):942–53. doi:10.1002/hbm.23078.PubMedCrossRef Pagani M, Oberg J, De Carli F, Calvo A, Moglia C, Canosa A, et al. Metabolic spatial connectivity in amyotrophic lateral sclerosis as revealed by independent component analysis. Hum Brain Mapp. 2016;37(3):942–53. doi:10.​1002/​hbm.​23078.PubMedCrossRef
71.
go back to reference Cistaro A, Pagani M, Montuschi A, Calvo A, Moglia C, Canosa A, et al. The metabolic signature of C9ORF72-related ALS: FDG PET comparison with nonmutated patients. Eur J Nucl Med Mol Imaging. 2014;41(5):844–52. doi:10.1007/s00259-013-2667-5.PubMedCrossRef Cistaro A, Pagani M, Montuschi A, Calvo A, Moglia C, Canosa A, et al. The metabolic signature of C9ORF72-related ALS: FDG PET comparison with nonmutated patients. Eur J Nucl Med Mol Imaging. 2014;41(5):844–52. doi:10.​1007/​s00259-013-2667-5.PubMedCrossRef
74.
go back to reference Pellerin L, Magistretti PJ. Glutamate uptake into astrocytes stimulates aerobic glycolysis: a mechanism coupling neuronal activity to glucose utilization. Proc Natl Acad Sci U S A. 1994;91(22):10625–9.PubMedPubMedCentralCrossRef Pellerin L, Magistretti PJ. Glutamate uptake into astrocytes stimulates aerobic glycolysis: a mechanism coupling neuronal activity to glucose utilization. Proc Natl Acad Sci U S A. 1994;91(22):10625–9.PubMedPubMedCentralCrossRef
75.
go back to reference Abe K, Yorifuji S, Nishikawa Y. Reduced isotope uptake restricted to the motor area in patients with amyotrophic lateral sclerosis. Neuroradiology. 1993;35(6):410–1.PubMedCrossRef Abe K, Yorifuji S, Nishikawa Y. Reduced isotope uptake restricted to the motor area in patients with amyotrophic lateral sclerosis. Neuroradiology. 1993;35(6):410–1.PubMedCrossRef
76.
go back to reference Kew JJ, Leigh PN, Playford ED, Passingham RE, Goldstein LH, Frackowiak RS, et al. Cortical function in amyotrophic lateral sclerosis. a positron emission tomography study. Brain. 1993;116(Pt 3):655–80.PubMedCrossRef Kew JJ, Leigh PN, Playford ED, Passingham RE, Goldstein LH, Frackowiak RS, et al. Cortical function in amyotrophic lateral sclerosis. a positron emission tomography study. Brain. 1993;116(Pt 3):655–80.PubMedCrossRef
77.
go back to reference Waldemar G, Vorstrup S, Jensen TS, Johnsen A, Boysen G. Focal reductions of cerebral blood flow in amyotrophic lateral sclerosis: a [99mTc]-d, l-HMPAO SPECT study. J Neurol Sci. 1992;107(1):19–28.PubMedCrossRef Waldemar G, Vorstrup S, Jensen TS, Johnsen A, Boysen G. Focal reductions of cerebral blood flow in amyotrophic lateral sclerosis: a [99mTc]-d, l-HMPAO SPECT study. J Neurol Sci. 1992;107(1):19–28.PubMedCrossRef
78.
go back to reference Habert MO, Lacomblez L, Maksud P, El Fakhri G, Pradat JF, Meininger V. Brain perfusion imaging in amyotrophic lateral sclerosis: extent of cortical changes according to the severity and topography of motor impairment. Amyotroph Lateral Scler. 2007;8(1):9–15. doi:10.1080/14660820601048815.PubMedCrossRef Habert MO, Lacomblez L, Maksud P, El Fakhri G, Pradat JF, Meininger V. Brain perfusion imaging in amyotrophic lateral sclerosis: extent of cortical changes according to the severity and topography of motor impairment. Amyotroph Lateral Scler. 2007;8(1):9–15. doi:10.​1080/​1466082060104881​5.PubMedCrossRef
79.
go back to reference Borasio GD, Linke R, Schwarz J, Schlamp V, Abel A, Mozley PD, et al. Dopaminergic deficit in amyotrophic lateral sclerosis assessed with [I-123] IPT single photon emission computed tomography. J Neurol Neurosurg Psychiatry. 1998;65(2):263–5.PubMedPubMedCentralCrossRef Borasio GD, Linke R, Schwarz J, Schlamp V, Abel A, Mozley PD, et al. Dopaminergic deficit in amyotrophic lateral sclerosis assessed with [I-123] IPT single photon emission computed tomography. J Neurol Neurosurg Psychiatry. 1998;65(2):263–5.PubMedPubMedCentralCrossRef
80.
go back to reference Przedborski S, Dhawan V, Donaldson DM, Murphy PL, McKenna-Yasek D, Mandel FS, et al. Nigrostriatal dopaminergic function in familial amyotrophic lateral sclerosis patients with and without copper/zinc superoxide dismutase mutations. Neurology. 1996;47(6):1546–51.PubMedCrossRef Przedborski S, Dhawan V, Donaldson DM, Murphy PL, McKenna-Yasek D, Mandel FS, et al. Nigrostriatal dopaminergic function in familial amyotrophic lateral sclerosis patients with and without copper/zinc superoxide dismutase mutations. Neurology. 1996;47(6):1546–51.PubMedCrossRef
81.
go back to reference Takahashi H, Snow BJ, Bhatt MH, Peppard R, Eisen A, Calne DB. Evidence for a dopaminergic deficit in sporadic amyotrophic lateral sclerosis on positron emission scanning. Lancet. 1993;342(8878):1016–8.PubMedCrossRef Takahashi H, Snow BJ, Bhatt MH, Peppard R, Eisen A, Calne DB. Evidence for a dopaminergic deficit in sporadic amyotrophic lateral sclerosis on positron emission scanning. Lancet. 1993;342(8878):1016–8.PubMedCrossRef
82.
go back to reference Lloyd CM, Richardson MP, Brooks DJ, Al-Chalabi A, Leigh PN. Extramotor involvement in ALS: PET studies with the GABA(A) ligand [(11)C]flumazenil. Brain. 2000;123(Pt 11):2289–96.PubMedCrossRef Lloyd CM, Richardson MP, Brooks DJ, Al-Chalabi A, Leigh PN. Extramotor involvement in ALS: PET studies with the GABA(A) ligand [(11)C]flumazenil. Brain. 2000;123(Pt 11):2289–96.PubMedCrossRef
83.
go back to reference Turner MR, Hammers A, Al-Chalabi A, Shaw CE, Andersen PM, Brooks DJ, et al. Distinct cerebral lesions in sporadic and ‘D90A’ SOD1 ALS: studies with [11C]flumazenil PET. Brain. 2005;128(Pt 6):1323–9. doi:10.1093/brain/awh509.PubMedCrossRef Turner MR, Hammers A, Al-Chalabi A, Shaw CE, Andersen PM, Brooks DJ, et al. Distinct cerebral lesions in sporadic and ‘D90A’ SOD1 ALS: studies with [11C]flumazenil PET. Brain. 2005;128(Pt 6):1323–9. doi:10.​1093/​brain/​awh509.PubMedCrossRef
84.
go back to reference Wicks P, Turner MR, Abrahams S, Hammers A, Brooks DJ, Leigh PN, et al. Neuronal loss associated with cognitive performance in amyotrophic lateral sclerosis: an (11C)-flumazenil PET study. Amyotroph Lateral Scler. 2008;9(1):43–9. doi:10.1080/17482960701737716.PubMedCrossRef Wicks P, Turner MR, Abrahams S, Hammers A, Brooks DJ, Leigh PN, et al. Neuronal loss associated with cognitive performance in amyotrophic lateral sclerosis: an (11C)-flumazenil PET study. Amyotroph Lateral Scler. 2008;9(1):43–9. doi:10.​1080/​1748296070173771​6.PubMedCrossRef
85.
86.
89.
go back to reference Alexianu ME, Kozovska M, Appel SH. Immune reactivity in a mouse model of familial ALS correlates with disease progression. Neurology. 2001;57(7):1282–9.PubMedCrossRef Alexianu ME, Kozovska M, Appel SH. Immune reactivity in a mouse model of familial ALS correlates with disease progression. Neurology. 2001;57(7):1282–9.PubMedCrossRef
90.
go back to reference Hall ED, Oostveen JA, Gurney ME. Relationship of microglial and astrocytic activation to disease onset and progression in a transgenic model of familial ALS. Glia. 1998;23(3):249–56.PubMedCrossRef Hall ED, Oostveen JA, Gurney ME. Relationship of microglial and astrocytic activation to disease onset and progression in a transgenic model of familial ALS. Glia. 1998;23(3):249–56.PubMedCrossRef
91.
go back to reference McEnery MW, Snowman AM, Trifiletti RR, Snyder SH. Isolation of the mitochondrial benzodiazepine receptor: association with the voltage-dependent anion channel and the adenine nucleotide carrier. Proc Natl Acad Sci U S A. 1992;89(8):3170–4.PubMedPubMedCentralCrossRef McEnery MW, Snowman AM, Trifiletti RR, Snyder SH. Isolation of the mitochondrial benzodiazepine receptor: association with the voltage-dependent anion channel and the adenine nucleotide carrier. Proc Natl Acad Sci U S A. 1992;89(8):3170–4.PubMedPubMedCentralCrossRef
92.
go back to reference Papadopoulos V, Amri H, Boujrad N, Cascio C, Culty M, Garnier M, et al. Peripheral benzodiazepine receptor in cholesterol transport and steroidogenesis. Steroids. 1997;62(1):21–8.PubMedCrossRef Papadopoulos V, Amri H, Boujrad N, Cascio C, Culty M, Garnier M, et al. Peripheral benzodiazepine receptor in cholesterol transport and steroidogenesis. Steroids. 1997;62(1):21–8.PubMedCrossRef
93.
go back to reference Galiegue S, Tinel N, Casellas P. The peripheral benzodiazepine receptor: a promising therapeutic drug target. Curr Med Chem. 2003;10(16):1563–72.PubMedCrossRef Galiegue S, Tinel N, Casellas P. The peripheral benzodiazepine receptor: a promising therapeutic drug target. Curr Med Chem. 2003;10(16):1563–72.PubMedCrossRef
94.
go back to reference Lavisse S, Garcia-Lorenzo D, Peyronneau MA, Bodini B, Thiriez C, Kuhnast B, et al. Optimized quantification of translocator protein radioligand (1)(8)F-DPA-714 uptake in the brain of genotyped healthy volunteers. J Nucl Med. 2015;56(7):1048–54. doi:10.2967/jnumed.115.156083. Lavisse S, Garcia-Lorenzo D, Peyronneau MA, Bodini B, Thiriez C, Kuhnast B, et al. Optimized quantification of translocator protein radioligand (1)(8)F-DPA-714 uptake in the brain of genotyped healthy volunteers. J Nucl Med. 2015;56(7):1048–54. doi:10.​2967/​jnumed.​115.​156083.
96.
go back to reference Cagnin A, Gerhard A, Banati RB. In vivo imaging of neuroinflammation. Eur Neuropsychopharmacol. 2002;12(6):581–6.PubMedCrossRef Cagnin A, Gerhard A, Banati RB. In vivo imaging of neuroinflammation. Eur Neuropsychopharmacol. 2002;12(6):581–6.PubMedCrossRef
97.
go back to reference Benavides J, Fage D, Carter C, Scatton B. Peripheral type benzodiazepine binding sites are a sensitive indirect index of neuronal damage. Brain Res. 1987;421(1-2):167–72.PubMedCrossRef Benavides J, Fage D, Carter C, Scatton B. Peripheral type benzodiazepine binding sites are a sensitive indirect index of neuronal damage. Brain Res. 1987;421(1-2):167–72.PubMedCrossRef
98.
go back to reference Benavides J, Quarteronet D, Imbault F, Malgouris C, Uzan A, Renault C, et al. Labelling of “peripheral-type” benzodiazepine binding sites in the rat brain by using [3H]PK 11195, an isoquinoline carboxamide derivative: kinetic studies and autoradiographic localization. J Neurochem. 1983;41(6):1744–50.PubMedCrossRef Benavides J, Quarteronet D, Imbault F, Malgouris C, Uzan A, Renault C, et al. Labelling of “peripheral-type” benzodiazepine binding sites in the rat brain by using [3H]PK 11195, an isoquinoline carboxamide derivative: kinetic studies and autoradiographic localization. J Neurochem. 1983;41(6):1744–50.PubMedCrossRef
99.
go back to reference Le Fur G, Guilloux F, Rufat P, Benavides J, Uzan A, Renault C, et al. Peripheral benzodiazepine binding sites: effect of PK 11195, 1-(2-chlorophenyl)-N-methyl-(1-methylpropyl)-3 isoquinolinecarboxamide. II. In vivo studies. Life Sci. 1983;32(16):1849–56.PubMedCrossRef Le Fur G, Guilloux F, Rufat P, Benavides J, Uzan A, Renault C, et al. Peripheral benzodiazepine binding sites: effect of PK 11195, 1-(2-chlorophenyl)-N-methyl-(1-methylpropyl)-3 isoquinolinecarboxamide. II. In vivo studies. Life Sci. 1983;32(16):1849–56.PubMedCrossRef
100.
go back to reference Le Fur G, Perrier ML, Vaucher N, Imbault F, Flamier A, Benavides J, et al. Peripheral benzodiazepine binding sites: effect of PK 11195, 1-(2-chlorophenyl)-N-methyl-N-(1-methylpropyl)-3-isoquinolinecarboxamide. I. In vitro studies. Life Sci. 1983;32(16):1839–47.PubMedCrossRef Le Fur G, Perrier ML, Vaucher N, Imbault F, Flamier A, Benavides J, et al. Peripheral benzodiazepine binding sites: effect of PK 11195, 1-(2-chlorophenyl)-N-methyl-N-(1-methylpropyl)-3-isoquinolinecarboxamide. I. In vitro studies. Life Sci. 1983;32(16):1839–47.PubMedCrossRef
101.
go back to reference Rojas S, Martin A, Arranz MJ, Pareto D, Purroy J, Verdaguer E, et al. Imaging brain inflammation with [(11)C]PK11195 by PET and induction of the peripheral-type benzodiazepine receptor after transient focal ischemia in rats. J Cereb Blood Flow Metab. 2007;27(12):1975–86. doi:10.1038/sj.jcbfm.9600500.PubMedCrossRef Rojas S, Martin A, Arranz MJ, Pareto D, Purroy J, Verdaguer E, et al. Imaging brain inflammation with [(11)C]PK11195 by PET and induction of the peripheral-type benzodiazepine receptor after transient focal ischemia in rats. J Cereb Blood Flow Metab. 2007;27(12):1975–86. doi:10.​1038/​sj.​jcbfm.​9600500.PubMedCrossRef
104.
go back to reference Banati RB, Newcombe J, Gunn RN, Cagnin A, Turkheimer F, Heppner F, et al. The peripheral benzodiazepine binding site in the brain in multiple sclerosis: quantitative in vivo imaging of microglia as a measure of disease activity. Brain. 2000;123(Pt 11):2321–37.PubMedCrossRef Banati RB, Newcombe J, Gunn RN, Cagnin A, Turkheimer F, Heppner F, et al. The peripheral benzodiazepine binding site in the brain in multiple sclerosis: quantitative in vivo imaging of microglia as a measure of disease activity. Brain. 2000;123(Pt 11):2321–37.PubMedCrossRef
105.
go back to reference Sitte HH, Wanschitz J, Budka H, Berger ML. Autoradiography with [3H]PK11195 of spinal tract degeneration in amyotrophic lateral sclerosis. Acta Neuropathol. 2001;101(2):75–8.PubMed Sitte HH, Wanschitz J, Budka H, Berger ML. Autoradiography with [3H]PK11195 of spinal tract degeneration in amyotrophic lateral sclerosis. Acta Neuropathol. 2001;101(2):75–8.PubMed
106.
go back to reference Engelhardt JI, Tajti J, Appel SH. Lymphocytic infiltrates in the spinal cord in amyotrophic lateral sclerosis. Arch Neurol. 1993;50(1):30–6.PubMedCrossRef Engelhardt JI, Tajti J, Appel SH. Lymphocytic infiltrates in the spinal cord in amyotrophic lateral sclerosis. Arch Neurol. 1993;50(1):30–6.PubMedCrossRef
107.
go back to reference Troost D, Van den Oord JJ, de Jong Vianney JM. Immunohistochemical characterization of the inflammatory infiltrate in amyotrophic lateral sclerosis. Neuropathol Appl Neurobiol. 1990;16(5):401–10.PubMedCrossRef Troost D, Van den Oord JJ, de Jong Vianney JM. Immunohistochemical characterization of the inflammatory infiltrate in amyotrophic lateral sclerosis. Neuropathol Appl Neurobiol. 1990;16(5):401–10.PubMedCrossRef
108.
go back to reference Turner MR, Cagnin A, Turkheimer FE, Miller CC, Shaw CE, Brooks DJ, et al. Evidence of widespread cerebral microglial activation in amyotrophic lateral sclerosis: an [11C](R)-PK11195 positron emission tomography study. Neurobiol Dis. 2004;15(3):601–9. doi:10.1016/j.nbd.2003.12.012.PubMedCrossRef Turner MR, Cagnin A, Turkheimer FE, Miller CC, Shaw CE, Brooks DJ, et al. Evidence of widespread cerebral microglial activation in amyotrophic lateral sclerosis: an [11C](R)-PK11195 positron emission tomography study. Neurobiol Dis. 2004;15(3):601–9. doi:10.​1016/​j.​nbd.​2003.​12.​012.PubMedCrossRef
110.
go back to reference Chauveau F, Van Camp N, Dolle F, Kuhnast B, Hinnen F, Damont A, et al. Comparative evaluation of the translocator protein radioligands 11C-DPA-713, 18F-DPA-714, and 11C-PK11195 in a rat model of acute neuroinflammation. J Nucl Med. 2009;50(3):468–76. doi:10.2967/jnumed.108.058669.PubMedCrossRef Chauveau F, Van Camp N, Dolle F, Kuhnast B, Hinnen F, Damont A, et al. Comparative evaluation of the translocator protein radioligands 11C-DPA-713, 18F-DPA-714, and 11C-PK11195 in a rat model of acute neuroinflammation. J Nucl Med. 2009;50(3):468–76. doi:10.​2967/​jnumed.​108.​058669.PubMedCrossRef
111.
go back to reference James ML, Fulton RR, Vercoullie J, Henderson DJ, Garreau L, Chalon S, et al. DPA-714, a new translocator protein-specific ligand: synthesis, radiofluorination, and pharmacologic characterization. J Nucl Med. 2008;49(5):814–22. doi:10.2967/jnumed.107.046151.PubMedCrossRef James ML, Fulton RR, Vercoullie J, Henderson DJ, Garreau L, Chalon S, et al. DPA-714, a new translocator protein-specific ligand: synthesis, radiofluorination, and pharmacologic characterization. J Nucl Med. 2008;49(5):814–22. doi:10.​2967/​jnumed.​107.​046151.PubMedCrossRef
118.
go back to reference Yiangou Y, Facer P, Durrenberger P, Chessell IP, Naylor A, Bountra C, et al. COX-2, CB2 and P2X7-immunoreactivities are increased in activated microglial cells/macrophages of multiple sclerosis and amyotrophic lateral sclerosis spinal cord. BMC Neurol. 2006;6:12. doi:10.1186/1471-2377-6-12.PubMedPubMedCentralCrossRef Yiangou Y, Facer P, Durrenberger P, Chessell IP, Naylor A, Bountra C, et al. COX-2, CB2 and P2X7-immunoreactivities are increased in activated microglial cells/macrophages of multiple sclerosis and amyotrophic lateral sclerosis spinal cord. BMC Neurol. 2006;6:12. doi:10.​1186/​1471-2377-6-12.PubMedPubMedCentralCrossRef
119.
go back to reference Apolloni S, Amadio S, Montilli C, Volonte C, D’Ambrosi N. Ablation of P2X7 receptor exacerbates gliosis and motoneuron death in the SOD1-G93A mouse model of amyotrophic lateral sclerosis. Hum Mol Genet. 2013;22(20):4102–16. doi:10.1093/hmg/ddt259.PubMedCrossRef Apolloni S, Amadio S, Montilli C, Volonte C, D’Ambrosi N. Ablation of P2X7 receptor exacerbates gliosis and motoneuron death in the SOD1-G93A mouse model of amyotrophic lateral sclerosis. Hum Mol Genet. 2013;22(20):4102–16. doi:10.​1093/​hmg/​ddt259.PubMedCrossRef
120.
go back to reference Duan S, Anderson CM, Keung EC, Chen Y, Chen Y, Swanson RA. P2X7 receptor-mediated release of excitatory amino acids from astrocytes. J Neurosci. 2003;23(4):1320–8.PubMed Duan S, Anderson CM, Keung EC, Chen Y, Chen Y, Swanson RA. P2X7 receptor-mediated release of excitatory amino acids from astrocytes. J Neurosci. 2003;23(4):1320–8.PubMed
122.
go back to reference Guile SD, Alcaraz L, Birkinshaw TN, Bowers KC, Ebden MR, Furber M, et al. Antagonists of the P2X(7) receptor. from lead identification to drug development. J Med Chem. 2009;52(10):3123–41. doi:10.1021/jm801528x.PubMedCrossRef Guile SD, Alcaraz L, Birkinshaw TN, Bowers KC, Ebden MR, Furber M, et al. Antagonists of the P2X(7) receptor. from lead identification to drug development. J Med Chem. 2009;52(10):3123–41. doi:10.​1021/​jm801528x.PubMedCrossRef
124.
go back to reference Gunosewoyo H, Coster MJ, Kassiou M. Molecular probes for P2X7 receptor studies. Curr Med Chem. 2007;14(14):1505–23.PubMedCrossRef Gunosewoyo H, Coster MJ, Kassiou M. Molecular probes for P2X7 receptor studies. Curr Med Chem. 2007;14(14):1505–23.PubMedCrossRef
127.
go back to reference Michel AD, Chambers LJ, Clay WC, Condreay JP, Walter DS, Chessell IP. Direct labelling of the human P2X7 receptor and identification of positive and negative cooperativity of binding. Br J Pharmacol. 2007;151(1):103–14. doi:10.1038/sj.bjp.0707196.PubMedCrossRef Michel AD, Chambers LJ, Clay WC, Condreay JP, Walter DS, Chessell IP. Direct labelling of the human P2X7 receptor and identification of positive and negative cooperativity of binding. Br J Pharmacol. 2007;151(1):103–14. doi:10.​1038/​sj.​bjp.​0707196.PubMedCrossRef
128.
go back to reference Romagnoli R, Baraldi PG, Pavani MG, Tabrizi MA, Moorman AR, Di Virgilio F, et al. Synthesis, radiolabeling, and preliminary biological evaluation of [3H]-1-[(S)-N, O-bis-(isoquinolinesulfonyl)-N-methyl-tyrosyl]-4-(o-tolyl)-piperazi ne, a potent antagonist radioligand for the P2X7 receptor. Bioorg Med Chem Lett. 2004;14(22):5709–12. doi:10.1016/j.bmcl.2004.07.095.PubMedCrossRef Romagnoli R, Baraldi PG, Pavani MG, Tabrizi MA, Moorman AR, Di Virgilio F, et al. Synthesis, radiolabeling, and preliminary biological evaluation of [3H]-1-[(S)-N, O-bis-(isoquinolinesulfonyl)-N-methyl-tyrosyl]-4-(o-tolyl)-piperazi ne, a potent antagonist radioligand for the P2X7 receptor. Bioorg Med Chem Lett. 2004;14(22):5709–12. doi:10.​1016/​j.​bmcl.​2004.​07.​095.PubMedCrossRef
130.
go back to reference Ory D, Celen S, Gijsbers R, Van Den Haute C, Postnov A, Koole M, et al. Preclinical evaluation of a P2X7 receptor selective radiotracer: PET studies in a rat model with local overexpression of the human P2X7 receptor and in non-human primates. J Nucl Med. 2016. doi:10.2967/jnumed.115.169995.PubMed Ory D, Celen S, Gijsbers R, Van Den Haute C, Postnov A, Koole M, et al. Preclinical evaluation of a P2X7 receptor selective radiotracer: PET studies in a rat model with local overexpression of the human P2X7 receptor and in non-human primates. J Nucl Med. 2016. doi:10.​2967/​jnumed.​115.​169995.PubMed
131.
go back to reference Janssen B, Vugts DJ, Funke U, Spaans A, Schuit RC, Kooijman E, et al. Synthesis and initial preclinical evaluation of the P2X7 receptor antagonist [(1)(1)C]A-740003 as a novel tracer of neuroinflammation. J Labelled Comp Radiopharm. 2014;57(8):509–16. doi:10.1002/jlcr.3206.PubMedCrossRef Janssen B, Vugts DJ, Funke U, Spaans A, Schuit RC, Kooijman E, et al. Synthesis and initial preclinical evaluation of the P2X7 receptor antagonist [(1)(1)C]A-740003 as a novel tracer of neuroinflammation. J Labelled Comp Radiopharm. 2014;57(8):509–16. doi:10.​1002/​jlcr.​3206.PubMedCrossRef
132.
133.
134.
go back to reference Seibert K, Zhang Y, Leahy K, Hauser S, Masferrer J, Isakson P. Distribution of COX-1 and COX-2 in normal and inflamed tissues. Adv Exp Med Biol. 1997;400A:167–70.PubMedCrossRef Seibert K, Zhang Y, Leahy K, Hauser S, Masferrer J, Isakson P. Distribution of COX-1 and COX-2 in normal and inflamed tissues. Adv Exp Med Biol. 1997;400A:167–70.PubMedCrossRef
135.
go back to reference Yasojima K, Tourtellotte WW, McGeer EG, McGeer PL. Marked increase in cyclooxygenase-2 in ALS spinal cord: implications for therapy. Neurology. 2001;57(6):952–6.PubMedCrossRef Yasojima K, Tourtellotte WW, McGeer EG, McGeer PL. Marked increase in cyclooxygenase-2 in ALS spinal cord: implications for therapy. Neurology. 2001;57(6):952–6.PubMedCrossRef
136.
go back to reference Almer G, Guegan C, Teismann P, Naini A, Rosoklija G, Hays AP, et al. Increased expression of the pro-inflammatory enzyme cyclooxygenase-2 in amyotrophic lateral sclerosis. Ann Neurol. 2001;49(2):176–85.PubMedCrossRef Almer G, Guegan C, Teismann P, Naini A, Rosoklija G, Hays AP, et al. Increased expression of the pro-inflammatory enzyme cyclooxygenase-2 in amyotrophic lateral sclerosis. Ann Neurol. 2001;49(2):176–85.PubMedCrossRef
137.
go back to reference Pompl PN, Ho L, Bianchi M, McManus T, Qin W, Pasinetti GM. A therapeutic role for cyclooxygenase-2 inhibitors in a transgenic mouse model of amyotrophic lateral sclerosis. FASEB J. 2003;17(6):725–7. doi:10.1096/fj.02-0876fje.PubMed Pompl PN, Ho L, Bianchi M, McManus T, Qin W, Pasinetti GM. A therapeutic role for cyclooxygenase-2 inhibitors in a transgenic mouse model of amyotrophic lateral sclerosis. FASEB J. 2003;17(6):725–7. doi:10.​1096/​fj.​02-0876fje.PubMed
138.
go back to reference Kaur J, Tietz O, Bhardwaj A, Marshall A, Way J, Wuest M, et al. Design, synthesis, and evaluation of an (18)F-labeled radiotracer based on Celecoxib-NBD for positron emission tomography (PET) imaging of Cyclooxygenase-2 (COX-2). ChemMedChem. 2015;10(10):1635–40. doi:10.1002/cmdc.201500287.PubMedCrossRef Kaur J, Tietz O, Bhardwaj A, Marshall A, Way J, Wuest M, et al. Design, synthesis, and evaluation of an (18)F-labeled radiotracer based on Celecoxib-NBD for positron emission tomography (PET) imaging of Cyclooxygenase-2 (COX-2). ChemMedChem. 2015;10(10):1635–40. doi:10.​1002/​cmdc.​201500287.PubMedCrossRef
139.
go back to reference Tietz O, Marshall A, Wuest M, Wang M, Wuest F. Radiotracers for molecular imaging of cyclooxygenase-2 (COX-2) enzyme. Curr Med Chem. 2013;20(35):4350–69.PubMedCrossRef Tietz O, Marshall A, Wuest M, Wang M, Wuest F. Radiotracers for molecular imaging of cyclooxygenase-2 (COX-2) enzyme. Curr Med Chem. 2013;20(35):4350–69.PubMedCrossRef
140.
go back to reference Fowler JS, Wang GJ, Logan J, Xie S, Volkow ND, MacGregor RR, et al. Selective reduction of radiotracer trapping by deuterium substitution: comparison of carbon-11-l-deprenyl and carbon-11-deprenyl-D2 for MAO B mapping. J Nucl Med. 1995;36(7):1255–62.PubMed Fowler JS, Wang GJ, Logan J, Xie S, Volkow ND, MacGregor RR, et al. Selective reduction of radiotracer trapping by deuterium substitution: comparison of carbon-11-l-deprenyl and carbon-11-deprenyl-D2 for MAO B mapping. J Nucl Med. 1995;36(7):1255–62.PubMed
141.
go back to reference Aquilonius SM, Jossan SS, Ekblom JG, Askmark H, Gillberg PG. Increased binding of 3H-l-deprenyl in spinal cords from patients with amyotrophic lateral sclerosis as demonstrated by autoradiography. J Neural Transm Gen Sect. 1992;89(1-2):111–22.PubMedCrossRef Aquilonius SM, Jossan SS, Ekblom JG, Askmark H, Gillberg PG. Increased binding of 3H-l-deprenyl in spinal cords from patients with amyotrophic lateral sclerosis as demonstrated by autoradiography. J Neural Transm Gen Sect. 1992;89(1-2):111–22.PubMedCrossRef
142.
go back to reference Farid K, Carter SF, Rodriguez-Vieitez E, Almkvist O, Andersen P, Wall A, et al. Case report of complex amyotrophic lateral sclerosis with cognitive impairment and cortical amyloid deposition. J Alzheimers Dis. 2015;47(3):661–7. doi:10.3233/JAD-141965.PubMedCrossRef Farid K, Carter SF, Rodriguez-Vieitez E, Almkvist O, Andersen P, Wall A, et al. Case report of complex amyotrophic lateral sclerosis with cognitive impairment and cortical amyloid deposition. J Alzheimers Dis. 2015;47(3):661–7. doi:10.​3233/​JAD-141965.PubMedCrossRef
144.
go back to reference Coffey RG, Yamamoto Y, Snella E, Pross S. Tetrahydrocannabinol inhibition of macrophage nitric oxide production. Biochem Pharmacol. 1996;52(5):743–51.PubMedCrossRef Coffey RG, Yamamoto Y, Snella E, Pross S. Tetrahydrocannabinol inhibition of macrophage nitric oxide production. Biochem Pharmacol. 1996;52(5):743–51.PubMedCrossRef
146.
go back to reference Ahmad R, Koole M, Evens N, Serdons K, Verbruggen A, Bormans G, et al. Whole-body biodistribution and radiation dosimetry of the cannabinoid type 2 receptor ligand [11C]-NE40 in healthy subjects. Mol Imaging Biol. 2013;15(4):384–90. doi:10.1007/s11307-013-0626-y.PubMedCrossRef Ahmad R, Koole M, Evens N, Serdons K, Verbruggen A, Bormans G, et al. Whole-body biodistribution and radiation dosimetry of the cannabinoid type 2 receptor ligand [11C]-NE40 in healthy subjects. Mol Imaging Biol. 2013;15(4):384–90. doi:10.​1007/​s11307-013-0626-y.PubMedCrossRef
147.
go back to reference Mu L, Bieri D, Slavik R, Drandarov K, Muller A, Cermak S, et al. Radiolabeling and in vitro /in vivo evaluation of N-(1-adamantyl)-8-methoxy-4-oxo-1-phenyl-1,4-dihydroquinoline-3-carboxamide as a PET probe for imaging cannabinoid type 2 receptor. J Neurochem. 2013;126(5):616–24. doi:10.1111/jnc.12354.PubMedCrossRef Mu L, Bieri D, Slavik R, Drandarov K, Muller A, Cermak S, et al. Radiolabeling and in vitro /in vivo evaluation of N-(1-adamantyl)-8-methoxy-4-oxo-1-phenyl-1,4-dihydroquinoline-3-carboxamide as a PET probe for imaging cannabinoid type 2 receptor. J Neurochem. 2013;126(5):616–24. doi:10.​1111/​jnc.​12354.PubMedCrossRef
148.
149.
go back to reference Slavik R, Grether U, Muller Herde A, Gobbi L, Fingerle J, Ullmer C, et al. Discovery of a high affinity and selective pyridine analog as a potential positron emission tomography imaging agent for cannabinoid type 2 receptor. J Med Chem. 2015;58(10):4266–77. doi:10.1021/acs.jmedchem.5b00283.PubMedCrossRef Slavik R, Grether U, Muller Herde A, Gobbi L, Fingerle J, Ullmer C, et al. Discovery of a high affinity and selective pyridine analog as a potential positron emission tomography imaging agent for cannabinoid type 2 receptor. J Med Chem. 2015;58(10):4266–77. doi:10.​1021/​acs.​jmedchem.​5b00283.PubMedCrossRef
151.
go back to reference Aronica E, Catania MV, Geurts J, Yankaya B, Troost D. Immunohistochemical localization of group I and II metabotropic glutamate receptors in control and amyotrophic lateral sclerosis human spinal cord: upregulation in reactive astrocytes. Neuroscience. 2001;105(2):509–20.PubMedCrossRef Aronica E, Catania MV, Geurts J, Yankaya B, Troost D. Immunohistochemical localization of group I and II metabotropic glutamate receptors in control and amyotrophic lateral sclerosis human spinal cord: upregulation in reactive astrocytes. Neuroscience. 2001;105(2):509–20.PubMedCrossRef
152.
go back to reference Hamill TG, Krause S, Ryan C, Bonnefous C, Govek S, Seiders TJ, et al. Synthesis, characterization, and first successful monkey imaging studies of metabotropic glutamate receptor subtype 5 (mGluR5) PET radiotracers. Synapse. 2005;56(4):205–16. doi:10.1002/syn.20147.PubMedCrossRef Hamill TG, Krause S, Ryan C, Bonnefous C, Govek S, Seiders TJ, et al. Synthesis, characterization, and first successful monkey imaging studies of metabotropic glutamate receptor subtype 5 (mGluR5) PET radiotracers. Synapse. 2005;56(4):205–16. doi:10.​1002/​syn.​20147.PubMedCrossRef
153.
go back to reference Brownell AL, Kuruppu D, Kil KE, Jokivarsi K, Poutiainen P, Zhu A, et al. PET imaging studies show enhanced expression of mGluR5 and inflammatory response during progressive degeneration in ALS mouse model expressing SOD1-G93A gene. J Neuroinflammation. 2015;12(1):217. doi:10.1186/s12974-015-0439-9.PubMedPubMedCentralCrossRef Brownell AL, Kuruppu D, Kil KE, Jokivarsi K, Poutiainen P, Zhu A, et al. PET imaging studies show enhanced expression of mGluR5 and inflammatory response during progressive degeneration in ALS mouse model expressing SOD1-G93A gene. J Neuroinflammation. 2015;12(1):217. doi:10.​1186/​s12974-015-0439-9.PubMedPubMedCentralCrossRef
154.
155.
go back to reference Milanese M, Giribaldi F, Melone M, Bonifacino T, Musante I, Carminati E, et al. Knocking down metabotropic glutamate receptor 1 improves survival and disease progression in the SOD1(G93A) mouse model of amyotrophic lateral sclerosis. Neurobiol Dis. 2014;64:48–59. doi:10.1016/j.nbd.2013.11.006.PubMedCrossRef Milanese M, Giribaldi F, Melone M, Bonifacino T, Musante I, Carminati E, et al. Knocking down metabotropic glutamate receptor 1 improves survival and disease progression in the SOD1(G93A) mouse model of amyotrophic lateral sclerosis. Neurobiol Dis. 2014;64:48–59. doi:10.​1016/​j.​nbd.​2013.​11.​006.PubMedCrossRef
156.
go back to reference Zanotti-Fregonara P, Barth VN, Liow JS, Zoghbi SS, Clark DT, Rhoads E, et al. Evaluation in vitro and in animals of a new 11C-labeled PET radioligand for metabotropic glutamate receptors 1 in brain. Eur J Nucl Med Mol Imaging. 2013;40(2):245–53. doi:10.1007/s00259-012-2269-7.PubMedCrossRef Zanotti-Fregonara P, Barth VN, Liow JS, Zoghbi SS, Clark DT, Rhoads E, et al. Evaluation in vitro and in animals of a new 11C-labeled PET radioligand for metabotropic glutamate receptors 1 in brain. Eur J Nucl Med Mol Imaging. 2013;40(2):245–53. doi:10.​1007/​s00259-012-2269-7.PubMedCrossRef
157.
go back to reference Zanotti-Fregonara P, Barth VN, Zoghbi SS, Liow JS, Nisenbaum E, Siuda E, et al. 11C-LY2428703, a positron emission tomographic radioligand for the metabotropic glutamate receptor 1, is unsuitable for imaging in monkey and human brains. EJNMMI Res. 2013;3(1):47. doi:10.1186/2191-219X-3-47.PubMedPubMedCentralCrossRef Zanotti-Fregonara P, Barth VN, Zoghbi SS, Liow JS, Nisenbaum E, Siuda E, et al. 11C-LY2428703, a positron emission tomographic radioligand for the metabotropic glutamate receptor 1, is unsuitable for imaging in monkey and human brains. EJNMMI Res. 2013;3(1):47. doi:10.​1186/​2191-219X-3-47.PubMedPubMedCentralCrossRef
158.
go back to reference Zanotti-Fregonara P, Xu R, Zoghbi SS, Liow JS, Fujita M, Veronese M, et al. The PET radioligand 18F-FIMX images and quantifies metabotropic glutamate receptor 1 in proportion to the regional density of its gene transcript in human brain. J Nucl Med. 2016;57(2):242–7. doi:10.2967/jnumed.115.162461.PubMedCrossRef Zanotti-Fregonara P, Xu R, Zoghbi SS, Liow JS, Fujita M, Veronese M, et al. The PET radioligand 18F-FIMX images and quantifies metabotropic glutamate receptor 1 in proportion to the regional density of its gene transcript in human brain. J Nucl Med. 2016;57(2):242–7. doi:10.​2967/​jnumed.​115.​162461.PubMedCrossRef
159.
go back to reference Crevecoeur J, Kaminski RM, Rogister B, Foerch P, Vandenplas C, Neveux M, et al. Expression pattern of synaptic vesicle protein 2 (SV2) isoforms in patients with temporal lobe epilepsy and hippocampal sclerosis. Neuropathol Appl Neurobiol. 2014;40(2):191–204. doi:10.1111/nan.12054.PubMedCrossRef Crevecoeur J, Kaminski RM, Rogister B, Foerch P, Vandenplas C, Neveux M, et al. Expression pattern of synaptic vesicle protein 2 (SV2) isoforms in patients with temporal lobe epilepsy and hippocampal sclerosis. Neuropathol Appl Neurobiol. 2014;40(2):191–204. doi:10.​1111/​nan.​12054.PubMedCrossRef
160.
go back to reference Glantz LA, Lewis DA. Decreased dendritic spine density on prefrontal cortical pyramidal neurons in schizophrenia. Arch Gen Psychiatry. 2000;57(1):65–73.PubMedCrossRef Glantz LA, Lewis DA. Decreased dendritic spine density on prefrontal cortical pyramidal neurons in schizophrenia. Arch Gen Psychiatry. 2000;57(1):65–73.PubMedCrossRef
163.
165.
go back to reference Buckley K, Kelly RB. Identification of a transmembrane glycoprotein specific for secretory vesicles of neural and endocrine cells. J Cell Biol. 1985;100(4):1284–94.PubMedCrossRef Buckley K, Kelly RB. Identification of a transmembrane glycoprotein specific for secretory vesicles of neural and endocrine cells. J Cell Biol. 1985;100(4):1284–94.PubMedCrossRef
166.
go back to reference Mendoza-Torreblanca JG, Vanoye-Carlo A, Phillips-Farfan BV, Carmona-Aparicio L, Gomez-Lira G. Synaptic vesicle protein 2A: basic facts and role in synaptic function. Eur J Neurosci. 2013;38(11):3529–39. doi:10.1111/ejn.12360.PubMedCrossRef Mendoza-Torreblanca JG, Vanoye-Carlo A, Phillips-Farfan BV, Carmona-Aparicio L, Gomez-Lira G. Synaptic vesicle protein 2A: basic facts and role in synaptic function. Eur J Neurosci. 2013;38(11):3529–39. doi:10.​1111/​ejn.​12360.PubMedCrossRef
168.
go back to reference Nabulsi NB, Mercier J, Holden D, Carre S, Najafzadeh S, Vandergeten MC, et al. Synthesis and preclinical evaluation of 11C-UCB-J as a PET tracer for imaging the synaptic vesicle glycoprotein 2A in the brain. J Nucl Med. 2016;57(5):777–84. doi:10.2967/jnumed.115.168179.PubMedCrossRef Nabulsi NB, Mercier J, Holden D, Carre S, Najafzadeh S, Vandergeten MC, et al. Synthesis and preclinical evaluation of 11C-UCB-J as a PET tracer for imaging the synaptic vesicle glycoprotein 2A in the brain. J Nucl Med. 2016;57(5):777–84. doi:10.​2967/​jnumed.​115.​168179.PubMedCrossRef
171.
go back to reference Bruijn LI, Houseweart MK, Kato S, Anderson KL, Anderson SD, Ohama E, et al. Aggregation and motor neuron toxicity of an ALS-linked SOD1 mutant independent from wild-type SOD1. Science. 1998;281(5384):1851–4.PubMedCrossRef Bruijn LI, Houseweart MK, Kato S, Anderson KL, Anderson SD, Ohama E, et al. Aggregation and motor neuron toxicity of an ALS-linked SOD1 mutant independent from wild-type SOD1. Science. 1998;281(5384):1851–4.PubMedCrossRef
174.
go back to reference Zetterstrom P, Stewart HG, Bergemalm D, Jonsson PA, Graffmo KS, Andersen PM, et al. Soluble misfolded subfractions of mutant superoxide dismutase-1s are enriched in spinal cords throughout life in murine ALS models. Proc Natl Acad Sci U S A. 2007;104(35):14157–62. doi:10.1073/pnas.0700477104.PubMedPubMedCentralCrossRef Zetterstrom P, Stewart HG, Bergemalm D, Jonsson PA, Graffmo KS, Andersen PM, et al. Soluble misfolded subfractions of mutant superoxide dismutase-1s are enriched in spinal cords throughout life in murine ALS models. Proc Natl Acad Sci U S A. 2007;104(35):14157–62. doi:10.​1073/​pnas.​0700477104.PubMedPubMedCentralCrossRef
176.
go back to reference Bryson JB, Hobbs C, Parsons MJ, Bosch KD, Pandraud A, Walsh FS, et al. Amyloid precursor protein (APP) contributes to pathology in the SOD1(G93A) mouse model of amyotrophic lateral sclerosis. Hum Mol Genet. 2012;21(17):3871–82. doi:10.1093/hmg/dds215.PubMedCrossRef Bryson JB, Hobbs C, Parsons MJ, Bosch KD, Pandraud A, Walsh FS, et al. Amyloid precursor protein (APP) contributes to pathology in the SOD1(G93A) mouse model of amyotrophic lateral sclerosis. Hum Mol Genet. 2012;21(17):3871–82. doi:10.​1093/​hmg/​dds215.PubMedCrossRef
177.
go back to reference Yamakawa Y, Shimada H, Ataka S, Tamura A, Masaki H, Naka H, et al. Two cases of dementias with motor neuron disease evaluated by Pittsburgh compound B-positron emission tomography. Neurol Sci. 2012;33(1):87–92. doi:10.1007/s10072-011-0479-6.PubMedCrossRef Yamakawa Y, Shimada H, Ataka S, Tamura A, Masaki H, Naka H, et al. Two cases of dementias with motor neuron disease evaluated by Pittsburgh compound B-positron emission tomography. Neurol Sci. 2012;33(1):87–92. doi:10.​1007/​s10072-011-0479-6.PubMedCrossRef
178.
go back to reference Matias-Guiu JA, Pytel V, Cabrera-Martin MN, Galan L, Valles-Salgado M, Guerrero A, et al. Amyloid- and FDG-PET imaging in amyotrophic lateral sclerosis. Eur J Nucl Med Mol Imaging. 2016. doi:10.1007/s00259-016-3434-1.PubMed Matias-Guiu JA, Pytel V, Cabrera-Martin MN, Galan L, Valles-Salgado M, Guerrero A, et al. Amyloid- and FDG-PET imaging in amyotrophic lateral sclerosis. Eur J Nucl Med Mol Imaging. 2016. doi:10.​1007/​s00259-016-3434-1.PubMed
181.
go back to reference Fujibayashi Y, Taniuchi H, Yonekura Y, Ohtani H, Konishi J, Yokoyama A. Copper-62-ATSM: a new hypoxia imaging agent with high membrane permeability and low redox potential. J Nucl Med. 1997;38(7):1155–60.PubMed Fujibayashi Y, Taniuchi H, Yonekura Y, Ohtani H, Konishi J, Yokoyama A. Copper-62-ATSM: a new hypoxia imaging agent with high membrane permeability and low redox potential. J Nucl Med. 1997;38(7):1155–60.PubMed
183.
go back to reference Donnelly PS, Liddell JR, Lim S, Paterson BM, Cater MA, Savva MS, et al. An impaired mitochondrial electron transport chain increases retention of the hypoxia imaging agent diacetylbis(4-methylthiosemicarbazonato)copperII. Proc Natl Acad Sci U S A. 2012;109(1):47–52. doi:10.1073/pnas.1116227108.PubMedCrossRef Donnelly PS, Liddell JR, Lim S, Paterson BM, Cater MA, Savva MS, et al. An impaired mitochondrial electron transport chain increases retention of the hypoxia imaging agent diacetylbis(4-methylthiosemicarbazonato)copperII. Proc Natl Acad Sci U S A. 2012;109(1):47–52. doi:10.​1073/​pnas.​1116227108.PubMedCrossRef
184.
go back to reference Yoshii Y, Yoneda M, Ikawa M, Furukawa T, Kiyono Y, Mori T, et al. Radiolabeled Cu-ATSM as a novel indicator of overreduced intracellular state due to mitochondrial dysfunction: studies with mitochondrial DNA-less rho0 cells and cybrids carrying MELAS mitochondrial DNA mutation. Nucl Med Biol. 2012;39(2):177–85. doi:10.1016/j.nucmedbio.2011.08.008.PubMedCrossRef Yoshii Y, Yoneda M, Ikawa M, Furukawa T, Kiyono Y, Mori T, et al. Radiolabeled Cu-ATSM as a novel indicator of overreduced intracellular state due to mitochondrial dysfunction: studies with mitochondrial DNA-less rho0 cells and cybrids carrying MELAS mitochondrial DNA mutation. Nucl Med Biol. 2012;39(2):177–85. doi:10.​1016/​j.​nucmedbio.​2011.​08.​008.PubMedCrossRef
188.
go back to reference McAllum EJ, Lim NK, Hickey JL, Paterson BM, Donnelly PS, Li QX, et al. Therapeutic effects of CuII(atsm) in the SOD1-G37R mouse model of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener. 2013;14(7-8):586–90. doi:10.3109/21678421.2013.824000.PubMedCrossRef McAllum EJ, Lim NK, Hickey JL, Paterson BM, Donnelly PS, Li QX, et al. Therapeutic effects of CuII(atsm) in the SOD1-G37R mouse model of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener. 2013;14(7-8):586–90. doi:10.​3109/​21678421.​2013.​824000.PubMedCrossRef
189.
go back to reference Soon CP, Donnelly PS, Turner BJ, Hung LW, Crouch PJ, Sherratt NA, et al. Diacetylbis(N(4)-methylthiosemicarbazonato) copper(II) (CuII(atsm)) protects against peroxynitrite-induced nitrosative damage and prolongs survival in amyotrophic lateral sclerosis mouse model. J Biol Chem. 2011;286(51):44035–44. doi:10.1074/jbc.M111.274407.PubMedPubMedCentralCrossRef Soon CP, Donnelly PS, Turner BJ, Hung LW, Crouch PJ, Sherratt NA, et al. Diacetylbis(N(4)-methylthiosemicarbazonato) copper(II) (CuII(atsm)) protects against peroxynitrite-induced nitrosative damage and prolongs survival in amyotrophic lateral sclerosis mouse model. J Biol Chem. 2011;286(51):44035–44. doi:10.​1074/​jbc.​M111.​274407.PubMedPubMedCentralCrossRef
190.
go back to reference Williams JR, Trias E, Beilby PR, Lopez NI, Labut EM, Bradford CS, et al. Copper delivery to the CNS by CuATSM effectively treats motor neuron disease in SOD(G93A) mice co-expressing the Copper-Chaperone-for-SOD. Neurobiol Dis. 2016;89:1–9. doi:10.1016/j.nbd.2016.01.020.PubMedCrossRef Williams JR, Trias E, Beilby PR, Lopez NI, Labut EM, Bradford CS, et al. Copper delivery to the CNS by CuATSM effectively treats motor neuron disease in SOD(G93A) mice co-expressing the Copper-Chaperone-for-SOD. Neurobiol Dis. 2016;89:1–9. doi:10.​1016/​j.​nbd.​2016.​01.​020.PubMedCrossRef
Metadata
Title
Positron emission tomography in amyotrophic lateral sclerosis: Towards targeting of molecular pathological hallmarks
Authors
Stefanie M. A. Willekens
Donatienne Van Weehaeghe
Philip Van Damme
Koen Van Laere
Publication date
01-03-2017
Publisher
Springer Berlin Heidelberg
Published in
European Journal of Nuclear Medicine and Molecular Imaging / Issue 3/2017
Print ISSN: 1619-7070
Electronic ISSN: 1619-7089
DOI
https://doi.org/10.1007/s00259-016-3587-y

Other articles of this Issue 3/2017

European Journal of Nuclear Medicine and Molecular Imaging 3/2017 Go to the issue