Skip to main content
Erschienen in: Acta Neuropathologica 1/2018

03.05.2018 | Original Paper

CADASIL brain vessels show a HTRA1 loss-of-function profile

verfasst von: Andreas Zellner, Eva Scharrer, Thomas Arzberger, Chio Oka, Valérie Domenga-Denier, Anne Joutel, Stefan F. Lichtenthaler, Stephan A. Müller, Martin Dichgans, Christof Haffner

Erschienen in: Acta Neuropathologica | Ausgabe 1/2018

Einloggen, um Zugang zu erhalten

Abstract

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and a phenotypically similar recessive condition (CARASIL) have emerged as important genetic model diseases for studying the molecular pathomechanisms of cerebral small vessel disease (SVD). CADASIL, the most frequent and intensely explored monogenic SVD, is characterized by a severe pathology in the cerebral vasculature including the mutation-induced aggregation of the Notch3 extracellular domain (Notch3ECD) and the formation of protein deposits of insufficiently determined composition in vessel walls. To identify key molecules and pathways involved in this process, we quantitatively determined the brain vessel proteome from CADASIL patient and control autopsy samples (n = 6 for each group), obtaining 95 proteins with significantly increased abundance. Intriguingly, high-temperature requirement protein A1 (HTRA1), the extracellular protease mutated in CARASIL, was found to be strongly enriched (4.9-fold, p = 1.6 × 10−3) and to colocalize with Notch3ECD deposits in patient vessels suggesting a sequestration process. Furthermore, the presence of increased levels of several HTRA1 substrates in the CADASIL proteome was compatible with their reduced degradation as consequence of a loss of HTRA1 activity. Indeed, a comparison with the brain vessel proteome of HTRA1 knockout mice (n = 5) revealed a highly significant overlap of 18 enriched proteins (p = 2.2 × 10−16), primarily representing secreted and extracellular matrix factors. Several of them were shown to be processed by HTRA1 in an in vitro proteolysis assay identifying them as novel substrates. Our study provides evidence for a loss of HTRA1 function as a critical step in the development of CADASIL pathology linking the molecular mechanisms of two distinct SVD forms.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
2.
Zurück zum Zitat Arboleda-Velasquez JF, Manent J, Lee JH, Tikka S, Ospina C, Vanderburg CR, Frosch MP, Rodriguez-Falcon M, Villen J, Gygi S, Lopera F, Kalimo H, Moskowitz MA, Ayata C, Louvi A et al (2011) Hypomorphic Notch 3 alleles link Notch signaling to ischemic cerebral small-vessel disease. Proc Natl Acad Sci USA 108:E128–135. https://doi.org/10.1073/pnas.1101964108 CrossRefPubMed Arboleda-Velasquez JF, Manent J, Lee JH, Tikka S, Ospina C, Vanderburg CR, Frosch MP, Rodriguez-Falcon M, Villen J, Gygi S, Lopera F, Kalimo H, Moskowitz MA, Ayata C, Louvi A et al (2011) Hypomorphic Notch 3 alleles link Notch signaling to ischemic cerebral small-vessel disease. Proc Natl Acad Sci USA 108:E128–135. https://​doi.​org/​10.​1073/​pnas.​1101964108 CrossRefPubMed
4.
Zurück zum Zitat Beaufort N, Scharrer E, Kremmer E, Lux V, Ehrmann M, Huber R, Houlden H, Werring D, Haffner C, Dichgans M (2014) Cerebral small vessel disease-related protease HtrA1 processes latent TGF-beta binding protein 1 and facilitates TGF-beta signaling. Proc Natl Acad Sci USA 111:16496–16501. https://doi.org/10.1073/pnas.1418087111 CrossRefPubMed Beaufort N, Scharrer E, Kremmer E, Lux V, Ehrmann M, Huber R, Houlden H, Werring D, Haffner C, Dichgans M (2014) Cerebral small vessel disease-related protease HtrA1 processes latent TGF-beta binding protein 1 and facilitates TGF-beta signaling. Proc Natl Acad Sci USA 111:16496–16501. https://​doi.​org/​10.​1073/​pnas.​1418087111 CrossRefPubMed
7.
Zurück zum Zitat Capone C, Dabertrand F, Baron-Menguy C, Chalaris A, Ghezali L, Domenga-Denier V, Schmidt S, Huneau C, Rose-John S, Nelson MT, Joutel A (2016) Mechanistic insights into a TIMP3-sensitive pathway constitutively engaged in the regulation of cerebral hemodynamics. Elife 5. https://doi.org/10.7554/eLife.17536 CrossRef Capone C, Dabertrand F, Baron-Menguy C, Chalaris A, Ghezali L, Domenga-Denier V, Schmidt S, Huneau C, Rose-John S, Nelson MT, Joutel A (2016) Mechanistic insights into a TIMP3-sensitive pathway constitutively engaged in the regulation of cerebral hemodynamics. Elife 5. https://​doi.​org/​10.​7554/​eLife.​17536 CrossRef
8.
Zurück zum Zitat Carare RO, Hawkes CA, Jeffrey M, Kalaria RN, Weller RO (2013) Review: cerebral amyloid angiopathy, prion angiopathy, CADASIL and the spectrum of protein elimination failure angiopathies (PEFA) in neurodegenerative disease with a focus on therapy. Neuropathol Appl Neurobiol 39:593–611. https://doi.org/10.1111/nan.12042 CrossRefPubMed Carare RO, Hawkes CA, Jeffrey M, Kalaria RN, Weller RO (2013) Review: cerebral amyloid angiopathy, prion angiopathy, CADASIL and the spectrum of protein elimination failure angiopathies (PEFA) in neurodegenerative disease with a focus on therapy. Neuropathol Appl Neurobiol 39:593–611. https://​doi.​org/​10.​1111/​nan.​12042 CrossRefPubMed
11.
Zurück zum Zitat Cognat E, Baron-Menguy C, Domenga-Denier V, Cleophax S, Fouillade C, Monet-Lepretre M, Dewerchin M, Joutel A (2014) Archetypal Arg169Cys mutation in NOTCH3 does not drive the pathogenesis in cerebral autosomal dominant arteriopathy with subcortical infarcts and leucoencephalopathy via a loss-of-function mechanism. Stroke 45:842–849. https://doi.org/10.1161/STROKEAHA.113.003339 CrossRefPubMed Cognat E, Baron-Menguy C, Domenga-Denier V, Cleophax S, Fouillade C, Monet-Lepretre M, Dewerchin M, Joutel A (2014) Archetypal Arg169Cys mutation in NOTCH3 does not drive the pathogenesis in cerebral autosomal dominant arteriopathy with subcortical infarcts and leucoencephalopathy via a loss-of-function mechanism. Stroke 45:842–849. https://​doi.​org/​10.​1161/​STROKEAHA.​113.​003339 CrossRefPubMed
12.
Zurück zum Zitat Craggs L, Taylor J, Slade JY, Chen A, Hagel C, Kuhlenbaeumer G, Borjesson-Hanson A, Viitanen M, Kalimo H, Deramecourt V, Oakley AE, Kalaria RN (2016) Clusterin/Apolipoprotein J immunoreactivity is associated with white matter damage in cerebral small vessel diseases. Neuropathol Appl Neurobiol 42:194–209. https://doi.org/10.1111/nan.12248 CrossRefPubMed Craggs L, Taylor J, Slade JY, Chen A, Hagel C, Kuhlenbaeumer G, Borjesson-Hanson A, Viitanen M, Kalimo H, Deramecourt V, Oakley AE, Kalaria RN (2016) Clusterin/Apolipoprotein J immunoreactivity is associated with white matter damage in cerebral small vessel diseases. Neuropathol Appl Neurobiol 42:194–209. https://​doi.​org/​10.​1111/​nan.​12248 CrossRefPubMed
14.
Zurück zum Zitat de la Pena P, Bornstein B, del Hoyo P, Fernandez-Moreno MA, Martin MA, Campos Y, Gomez-Escalonilla C, Molina JA, Cabello A, Arenas J, Garesse R (2001) Mitochondrial dysfunction associated with a mutation in the Notch3 gene in a CADASIL family. Neurology 57:1235–1238CrossRefPubMed de la Pena P, Bornstein B, del Hoyo P, Fernandez-Moreno MA, Martin MA, Campos Y, Gomez-Escalonilla C, Molina JA, Cabello A, Arenas J, Garesse R (2001) Mitochondrial dysfunction associated with a mutation in the Notch3 gene in a CADASIL family. Neurology 57:1235–1238CrossRefPubMed
15.
Zurück zum Zitat Di Donato I, Bianchi S, De Stefano N, Dichgans M, Dotti MT, Duering M, Jouvent E, Korczyn AD, Lesnik-Oberstein SA, Malandrini A, Markus HS, Pantoni L, Penco S, Rufa A, Sinanovic O et al (2017) Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL) as a model of small vessel disease: update on clinical, diagnostic, and management aspects. BMC Med 15:41. https://doi.org/10.1186/s12916-017-0778-8 CrossRefPubMedPubMedCentral Di Donato I, Bianchi S, De Stefano N, Dichgans M, Dotti MT, Duering M, Jouvent E, Korczyn AD, Lesnik-Oberstein SA, Malandrini A, Markus HS, Pantoni L, Penco S, Rufa A, Sinanovic O et al (2017) Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL) as a model of small vessel disease: update on clinical, diagnostic, and management aspects. BMC Med 15:41. https://​doi.​org/​10.​1186/​s12916-017-0778-8 CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Hara K, Shiga A, Fukutake T, Nozaki H, Miyashita A, Yokoseki A, Kawata H, Koyama A, Arima K, Takahashi T, Ikeda M, Shiota H, Tamura M, Shimoe Y, Hirayama M et al (2009) Association of HTRA1 mutations and familial ischemic cerebral small-vessel disease. N Engl J Med 360:1729–1739. https://doi.org/10.1056/NEJMoa0801560 CrossRefPubMed Hara K, Shiga A, Fukutake T, Nozaki H, Miyashita A, Yokoseki A, Kawata H, Koyama A, Arima K, Takahashi T, Ikeda M, Shiota H, Tamura M, Shimoe Y, Hirayama M et al (2009) Association of HTRA1 mutations and familial ischemic cerebral small-vessel disease. N Engl J Med 360:1729–1739. https://​doi.​org/​10.​1056/​NEJMoa0801560 CrossRefPubMed
27.
Zurück zum Zitat Jones A, Kumar S, Zhang N, Tong Z, Yang JH, Watt C, Anderson J, Amrita Fillerup H, McCloskey M, Luo L, Yang Z, Ambati B, Marc R, Oka C et al (2011) Increased expression of multifunctional serine protease, HTRA1, in retinal pigment epithelium induces polypoidal choroidal vasculopathy in mice. Proc Natl Acad Sci USA 108:14578–14583. https://doi.org/10.1073/pnas.1102853108 CrossRefPubMed Jones A, Kumar S, Zhang N, Tong Z, Yang JH, Watt C, Anderson J, Amrita Fillerup H, McCloskey M, Luo L, Yang Z, Ambati B, Marc R, Oka C et al (2011) Increased expression of multifunctional serine protease, HTRA1, in retinal pigment epithelium induces polypoidal choroidal vasculopathy in mice. Proc Natl Acad Sci USA 108:14578–14583. https://​doi.​org/​10.​1073/​pnas.​1102853108 CrossRefPubMed
31.
Zurück zum Zitat Joutel A, Monet-Lepretre M, Gosele C, Baron-Menguy C, Hammes A, Schmidt S, Lemaire-Carrette B, Domenga V, Schedl A, Lacombe P, Hubner N (2010) Cerebrovascular dysfunction and microcirculation rarefaction precede white matter lesions in a mouse genetic model of cerebral ischemic small vessel disease. J Clin Invest 120:433–445. https://doi.org/10.1172/jci39733 CrossRefPubMedPubMedCentral Joutel A, Monet-Lepretre M, Gosele C, Baron-Menguy C, Hammes A, Schmidt S, Lemaire-Carrette B, Domenga V, Schedl A, Lacombe P, Hubner N (2010) Cerebrovascular dysfunction and microcirculation rarefaction precede white matter lesions in a mouse genetic model of cerebral ischemic small vessel disease. J Clin Invest 120:433–445. https://​doi.​org/​10.​1172/​jci39733 CrossRefPubMedPubMedCentral
38.
42.
Zurück zum Zitat Malandrini A, Albani F, Palmeri S, Fattapposta F, Gambelli S, Berti G, Bracco A, Tammaro A, Calzavara S, Villanova M, Ferrari M, Rossi A, Carrera P (2002) Asymptomatic cores and paracrystalline mitochondrial inclusions in CADASIL. Neurology 59:617–620CrossRefPubMed Malandrini A, Albani F, Palmeri S, Fattapposta F, Gambelli S, Berti G, Bracco A, Tammaro A, Calzavara S, Villanova M, Ferrari M, Rossi A, Carrera P (2002) Asymptomatic cores and paracrystalline mitochondrial inclusions in CADASIL. Neurology 59:617–620CrossRefPubMed
43.
Zurück zum Zitat Manousopoulou A, Gatherer M, Smith C, Nicoll JAR, Woelk CH, Johnson M, Kalaria R, Attems J, Garbis SD, Carare RO (2017) Systems proteomic analysis reveals that clusterin and tissue inhibitor of metalloproteinases 3 increase in leptomeningeal arteries affected by cerebral amyloid angiopathy. Neuropathol Appl Neurobiol 43:492–504. https://doi.org/10.1111/nan.12342 CrossRefPubMed Manousopoulou A, Gatherer M, Smith C, Nicoll JAR, Woelk CH, Johnson M, Kalaria R, Attems J, Garbis SD, Carare RO (2017) Systems proteomic analysis reveals that clusterin and tissue inhibitor of metalloproteinases 3 increase in leptomeningeal arteries affected by cerebral amyloid angiopathy. Neuropathol Appl Neurobiol 43:492–504. https://​doi.​org/​10.​1111/​nan.​12342 CrossRefPubMed
53.
Zurück zum Zitat Pippucci T, Maresca A, Magini P, Cenacchi G, Donadio V, Palombo F, Papa V, Incensi A, Gasparre G, Valentino ML, Preziuso C, Pisano A, Ragno M, Liguori R, Giordano C et al (2015) Homozygous NOTCH3 null mutation and impaired NOTCH3 signaling in recessive early-onset arteriopathy and cavitating leukoencephalopathy. EMBO Mol Med 7:848–858. https://doi.org/10.15252/emmm.201404399 CrossRefPubMedPubMedCentral Pippucci T, Maresca A, Magini P, Cenacchi G, Donadio V, Palombo F, Papa V, Incensi A, Gasparre G, Valentino ML, Preziuso C, Pisano A, Ragno M, Liguori R, Giordano C et al (2015) Homozygous NOTCH3 null mutation and impaired NOTCH3 signaling in recessive early-onset arteriopathy and cavitating leukoencephalopathy. EMBO Mol Med 7:848–858. https://​doi.​org/​10.​15252/​emmm.​201404399 CrossRefPubMedPubMedCentral
56.
58.
Zurück zum Zitat Shiga A, Nozaki H, Yokoseki A, Nihonmatsu M, Kawata H, Kato T, Koyama A, Arima K, Ikeda M, Katada S, Toyoshima Y, Takahashi H, Tanaka A, Nakano I, Ikeuchi T et al (2011) Cerebral small-vessel disease protein HTRA1 controls the amount of TGF-beta1 via cleavage of proTGF-beta1. Hum Mol Genet 20:1800–1810. https://doi.org/10.1093/hmg/ddr063 CrossRefPubMed Shiga A, Nozaki H, Yokoseki A, Nihonmatsu M, Kawata H, Kato T, Koyama A, Arima K, Ikeda M, Katada S, Toyoshima Y, Takahashi H, Tanaka A, Nakano I, Ikeuchi T et al (2011) Cerebral small-vessel disease protein HTRA1 controls the amount of TGF-beta1 via cleavage of proTGF-beta1. Hum Mol Genet 20:1800–1810. https://​doi.​org/​10.​1093/​hmg/​ddr063 CrossRefPubMed
63.
Zurück zum Zitat Verdura E, Herve D, Scharrer E, Amador Mdel M, Guyant-Marechal L, Philippi A, Corlobe A, Bergametti F, Gazal S, Prieto-Morin C, Beaufort N, Le Bail B, Viakhireva I, Dichgans M, Chabriat H et al (2015) Heterozygous HTRA1 mutations are associated with autosomal dominant cerebral small vessel disease. Brain 138:2347–2358. https://doi.org/10.1093/brain/awv155 CrossRefPubMed Verdura E, Herve D, Scharrer E, Amador Mdel M, Guyant-Marechal L, Philippi A, Corlobe A, Bergametti F, Gazal S, Prieto-Morin C, Beaufort N, Le Bail B, Viakhireva I, Dichgans M, Chabriat H et al (2015) Heterozygous HTRA1 mutations are associated with autosomal dominant cerebral small vessel disease. Brain 138:2347–2358. https://​doi.​org/​10.​1093/​brain/​awv155 CrossRefPubMed
67.
Zurück zum Zitat Wollenweber FA, Hanecker P, Bayer-Karpinska A, Malik M, Bäzner H, Moreton F, Muir KW, Müller S, Giese A, Opherk C, Dichgans M, Haffner C, Düring M (2015) Cysteine-sparing CADASIL mutations in NOTCH3 show pro-aggregatory properties in vitro. Stroke 46:786–792CrossRefPubMed Wollenweber FA, Hanecker P, Bayer-Karpinska A, Malik M, Bäzner H, Moreton F, Muir KW, Müller S, Giese A, Opherk C, Dichgans M, Haffner C, Düring M (2015) Cysteine-sparing CADASIL mutations in NOTCH3 show pro-aggregatory properties in vitro. Stroke 46:786–792CrossRefPubMed
69.
Zurück zum Zitat Yoshida H, Nagaoka A, Kusaka-Kikushima A, Tobiishi M, Kawabata K, Sayo T, Sakai S, Sugiyama Y, Enomoto H, Okada Y, Inoue S (2013) KIAA1199, a deafness gene of unknown function, is a new hyaluronan binding protein involved in hyaluronan depolymerization. Proc Natl Acad Sci USA 110:5612–5617. https://doi.org/10.1073/pnas.1215432110 CrossRefPubMed Yoshida H, Nagaoka A, Kusaka-Kikushima A, Tobiishi M, Kawabata K, Sayo T, Sakai S, Sugiyama Y, Enomoto H, Okada Y, Inoue S (2013) KIAA1199, a deafness gene of unknown function, is a new hyaluronan binding protein involved in hyaluronan depolymerization. Proc Natl Acad Sci USA 110:5612–5617. https://​doi.​org/​10.​1073/​pnas.​1215432110 CrossRefPubMed
Metadaten
Titel
CADASIL brain vessels show a HTRA1 loss-of-function profile
verfasst von
Andreas Zellner
Eva Scharrer
Thomas Arzberger
Chio Oka
Valérie Domenga-Denier
Anne Joutel
Stefan F. Lichtenthaler
Stephan A. Müller
Martin Dichgans
Christof Haffner
Publikationsdatum
03.05.2018
Verlag
Springer Berlin Heidelberg
Erschienen in
Acta Neuropathologica / Ausgabe 1/2018
Print ISSN: 0001-6322
Elektronische ISSN: 1432-0533
DOI
https://doi.org/10.1007/s00401-018-1853-8

Weitere Artikel der Ausgabe 1/2018

Acta Neuropathologica 1/2018 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.