L. Pantoni, Cerebral small vessel disease: from pathogenesis and clinical characteristics to therapeutic challenges, Lancet Neurol, vol.9, issue.7, pp.689-701, 2010.

J. M. Wardlaw, E. E. Smith, G. J. Biessels, C. Cordonnier, and F. Fazekas, , 2013.

, Neuroimaging standards for research into small vessel disease and its contribution to ageing and neurodegeneration, Lancet Neurol, vol.12, issue.8, pp.822-860

S. Love, K. Chalmers, P. Ince, M. Esiri, and J. Attems, Development, appraisal, validation and implementation of a consensus protocol for the assessment of cerebral amyloid angiopathy in post-mortem brain tissue, Am. J. Neurodegener. Dis, vol.3, issue.1, pp.19-32, 2014.

A. Charidimou, G. Boulouis, M. E. Gurol, C. Ayata, and B. J. Bacskai, Emerging concepts in sporadic cerebral amyloid angiopathy, Brain J. Neurol, vol.140, issue.7, pp.1829-50, 2017.

L. Craggs, Y. Yamamoto, V. Deramecourt, and R. N. Kalaria, Microvascular pathology and morphometrics of sporadic and hereditary small vessel diseases of the brain, Brain Pathol. Zurich Switz, vol.24, issue.5, pp.495-509, 2014.

S. Debette, S. Schilling, M. Duperron, S. C. Larsson, and H. S. Markus, Clinical Significance of Magnetic Resonance Imaging Markers of Vascular Brain Injury: A Systematic Review and Meta-analysis, JAMA Neurol, 2018.

J. M. Wardlaw, C. Smith, and M. Dichgans, Mechanisms of sporadic cerebral small vessel disease: insights from neuroimaging, Lancet Neurol, vol.12, issue.5, pp.483-97, 2013.

E. Rapsomaniki, A. Timmis, J. George, M. Pujades-rodriguez, and A. D. Shah, Blood pressure and incidence of twelve cardiovascular diseases: lifetime risks, healthy lifeyears lost, and age-specific associations in 1·25 million people, Lancet Lond. Engl, vol.383, issue.9932, pp.1899-1911, 2014.

P. Klarenbeek, R. J. Van-oostenbrugge, R. Rouhl, I. Knottnerus, and J. Staals, Ambulatory blood pressure in patients with lacunar stroke: association with total MRI burden of cerebral small vessel disease, Stroke, vol.44, issue.11, pp.2995-99, 2013.

G. Chauhan, H. Adams, C. L. Satizabal, J. C. Bis, and A. Teumer, Genetic and lifestyle risk factors for MRI-defined brain infarcts in a population-based setting, 2019.
URL : https://hal.archives-ouvertes.fr/inserm-02457571

D. C. Bezerra, A. R. Sharrett, K. Matsushita, R. F. Gottesman, and D. Shibata, Risk factors for lacune subtypes in the Atherosclerosis Risk in Communities (ARIC) Study, Neurology, vol.78, issue.2, pp.102-110, 2012.

E. J. Van-dijk, M. Breteler, R. Schmidt, K. Berger, and L. Nilsson, The association between blood pressure, hypertension, and cerebral white matter lesions: cardiovascular determinants of dementia study. Hypertens. Dallas Tex, vol.44, pp.625-655, 1979.

C. Cordonnier, A. Salman, R. Wardlaw, and J. , Spontaneous brain microbleeds: systematic review, subgroup analyses and standards for study design and reporting, Brain J. Neurol, vol.130, pp.1988-2003, 2007.

C. Dufouil, A. De-kersaint-gilly, V. Besançon, C. Levy, and E. Auffray, Longitudinal study of blood pressure and white matter hyperintensities: the EVA MRI Cohort, Neurology, vol.56, issue.7, pp.921-947, 2001.

K. K. Lau, L. Li, M. Simoni, Z. Mehta, and W. Küker, Long-Term Premorbid Blood Pressure and Cerebral Small Vessel Disease Burden on Imaging in Transient Ischemic Attack and Ischemic Stroke, Stroke, vol.49, issue.9, p.22, 2018.

. Variable-brain-consortium, The association between blood pressure variability (BPV) with dementia and cognitive function: a systematic review and metaanalysis protocol, Syst. Rev, vol.7, issue.1, p.163, 2018.

P. S. Koekkoek, L. J. Kappelle, E. Van-den-berg, G. Rutten, and G. J. Biessels, Cognitive function in patients with diabetes mellitus: guidance for daily care, Lancet Neurol, vol.14, issue.3, pp.329-369, 2015.

S. Schilling, C. Tzourio, C. Dufouil, Y. Zhu, and C. Berr, Plasma lipids and cerebral small vessel disease, Neurology, vol.83, issue.20, pp.1844-52, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01196098

X. Wang, Y. Dong, X. Qi, C. Huang, and L. Hou, Cholesterol levels and risk of hemorrhagic stroke: a systematic review and meta-analysis, Stroke, vol.44, issue.7, pp.1833-1872, 2013.

A. Hassan, B. J. Hunt, M. O'sullivan, R. Bell, D. Souza et al., Homocysteine is a risk factor for cerebral small vessel disease, acting via endothelial dysfunction, Brain J. Neurol, vol.127, pp.212-231, 2004.

S. C. Larsson, M. Traylor, and H. S. Markus, Homocysteine and Small Vessel Stroke: A Mendelian Randomization Analysis, Ann. Neurol, 2019.

J. M. Wardlaw, M. Allerhand, F. N. Doubal, V. Hernandez, M. Morris et al., Vascular risk factors, large-artery atheroma, and brain white matter hyperintensities, Neurology, vol.82, issue.15, pp.1331-1369, 2014.

T. S. Field, F. N. Doubal, W. Johnson, E. Backhouse, and C. Mchutchison, Early life characteristics and late life burden of cerebral small vessel disease in the Lothian Birth Cohort, Aging, vol.8, issue.9, p.23, 1936.

C. Haffner, R. Malik, and M. Dichgans, Genetic factors in cerebral small vessel disease and their impact on stroke and dementia, J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab, vol.36, issue.1, pp.158-71, 2016.

X. Ayrignac, C. Carra-dalliere, M. De-champfleur, N. Denier, C. Aubourg et al., Adult-onset genetic leukoencephalopathies: a MRI pattern-based approach in a comprehensive study of 154 patients, Brain J. Neurol, vol.138, issue.2, pp.284-92, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01138578

H. Chabriat, A. Joutel, M. Dichgans, E. Tournier-lasserve, and M. G. Bousser, Cadasil. Lancet Neurol, vol.8, issue.7, pp.643-53, 2009.

Y. Lee, C. Chung, N. Chao, J. Fuh, and F. Chang, Characterization of Heterozygous HTRA1 Mutations in Taiwanese Patients With Cerebral Small Vessel Disease, Stroke, vol.49, issue.7, pp.1593-1601, 2018.

A. Joutel, K. Vahedi, C. Corpechot, A. Troesch, and H. Chabriat, Strong clustering and stereotyped nature of Notch3 mutations in CADASIL patients, Lancet, vol.350, issue.9090, pp.1511-1526, 1997.

J. W. Rutten, B. J. Van-eijsden, M. Duering, E. Jouvent, and C. Opherk, The effect of NOTCH3 pathogenic variant position on CADASIL disease severity: NOTCH3 EGFr 1-6 pathogenic variant are associated with a more severe phenotype and lower survival compared with EGFr 7-34 pathogenic variant, Genet. Med. Off. J. Am. Coll. Med. Genet, vol.21, issue.3, pp.676-82, 2019.

R. Malik, G. Chauhan, M. Traylor, M. Sargurupremraj, and Y. Okada, Multiancestry genome-wide association study of 520,000 subjects identifies 32 loci associated with stroke and stroke subtypes, Nat. Genet, vol.50, issue.4, p.24, 2018.

R. Malik, K. Rannikmäe, M. Traylor, M. K. Georgakis, and M. Sargurupremraj, , 2018.

, Genome-wide meta-analysis identifies 3 novel loci associated with stroke, Ann. Neurol, vol.84, issue.6, pp.934-973

K. Rannikmäe, V. Sivakumaran, H. Millar, R. Malik, and C. D. Anderson, COL4A2 is associated with lacunar ischemic stroke and deep ICH: Meta-analyses among 21,500 cases and 40,600 controls, Neurology, vol.89, issue.17, pp.1829-1868, 2017.

A. Mishra, G. Chauhan, M. Violleau, D. Vojinovic, and X. Jian, Association of variants in HTRA1 and NOTCH3 with MRI-defined extremes of cerebral small vessel disease in older subjects, Brain J. Neurol, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02093539

C. Iadecola, The pathobiology of vascular dementia, Neuron, vol.80, issue.4, pp.844-66, 2013.

T. A. Longden, F. Dabertrand, M. Koide, A. L. Gonzales, and N. R. Tykocki, Capillary K+-sensing initiates retrograde hyperpolarization to increase local cerebral blood flow, Nat. Neurosci, vol.20, issue.5, pp.717-743, 2017.

M. J. Cipolla, The Cerebral Circulation, 2009.

M. D. Sweeney, Z. Zhao, A. Montagne, A. R. Nelson, and B. V. Zlokovic, Blood-Brain Barrier: From Physiology to Disease and Back, Physiol. Rev, vol.99, issue.1, pp.21-78, 2019.

M. K. Rasmussen, H. Mestre, and M. Nedergaard, The glymphatic pathway in neurological disorders, Lancet Neurol, vol.17, issue.11, pp.1016-1040, 2018.

A. Y. Shih, P. Blinder, P. S. Tsai, B. Friedman, and G. Stanley, The smallest stroke: occlusion of one penetrating vessel leads to infarction and a cognitive deficit, Nat. Neurosci, vol.16, issue.1, pp.55-63, 2013.

E. E. Smith and A. E. Beaudin, New insights into cerebral small vessel disease and vascular cognitive impairment from MRI, Curr. Opin. Neurol, vol.31, issue.1, pp.36-43, 2018.

A. Joutel and H. Chabriat, Pathogenesis of white matter changes in cerebral small vessel diseases: beyond vessel-intrinsic mechanisms, Clin. Sci. Lond. Engl, vol.131, issue.8, pp.635-51, 1979.
URL : https://hal.archives-ouvertes.fr/inserm-02067812

C. Huneau, M. Houot, A. Joutel, B. Béranger, and C. Giroux, Altered dynamics of neurovascular coupling in CADASIL, Ann. Clin. Transl. Neurol, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01812731

M. M. Santisteban and C. Iadecola, Hypertension, dietary salt and cognitive impairment, 2018.

J. Cereb, Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab, vol.38, issue.12, pp.2112-2140

P. Toth, S. Tarantini, A. Csiszar, and Z. Ungvari, Functional vascular contributions to cognitive impairment and dementia: mechanisms and consequences of cerebral autoregulatory dysfunction, endothelial impairment, and neurovascular uncoupling in aging, Am. J. Physiol. Heart Circ. Physiol, vol.312, issue.1, pp.1-20, 2017.

C. Iadecola and R. L. Davisson, Hypertension and cerebrovascular dysfunction, Cell Metab, vol.7, issue.6, pp.476-84, 2008.

G. L. Baumbach, C. D. Sigmund, T. Bottiglieri, and S. R. Lentz, Structure of cerebral arterioles in cystathionine beta-synthase-deficient mice, Circ. Res, vol.91, issue.10, pp.931-968, 2002.

S. Dayal, A. M. Devlin, R. B. Mccaw, M. Liu, and E. Arning, Cerebral vascular dysfunction in methionine synthase-deficient mice, Circulation, vol.112, issue.5, pp.737-781, 2005.

J. Duncombe, A. Kitamura, Y. Hase, M. Ihara, R. N. Kalaria et al., Chronic cerebral hypoperfusion: a key mechanism leading to vascular cognitive impairment and 26, 2017.

, Closing the translational gap between rodent models and human vascular cognitive impairment and dementia, Clin. Sci. Lond. Engl, vol.131, pp.2451-68, 1979.

D. Silva, T. M. Faraci, and F. M. , Microvascular Dysfunction and Cognitive Impairment, Cell. Mol. Neurobiol, vol.36, issue.2, pp.241-58, 2016.

C. Iadecola, The Neurovascular Unit Coming of Age: A Journey through Neurovascular Coupling in Health and Disease, Neuron, vol.96, issue.1, pp.17-42, 2017.

G. Faraco, Y. Sugiyama, D. Lane, L. Garcia-bonilla, and H. Chang, Perivascular macrophages mediate the neurovascular and cognitive dysfunction associated with hypertension, J. Clin. Invest, vol.126, issue.12, pp.4674-89, 2016.

C. Baron-menguy, V. Domenga-denier, L. Ghezali, F. M. Faraci, and A. Joutel, Increased Notch3 Activity Mediates Pathological Changes in Structure of Cerebral Arteries. Hypertens. Dallas Tex, vol.69, pp.60-70, 1979.

C. Capone, E. Cognat, L. Ghezali, C. Baron-menguy, and D. Aubin, Reducing Timp3 or vitronectin ameliorates disease manifestations in CADASIL mice, Ann. Neurol, vol.79, issue.3, pp.387-403, 2016.

A. Joutel, F. Andreux, S. Gaulis, V. Domenga, and M. Cecillon, The ectodomain of the Notch3 receptor accumulates within the cerebrovasculature of CADASIL patients, J Clin Invest, vol.105, issue.5, pp.597-605, 2000.

A. Joutel, M. Monet-lepretre, C. Gosele, C. Baron-menguy, and A. Hammes, Cerebrovascular dysfunction and microcirculation rarefaction precede white matter lesions in a mouse genetic model of cerebral ischemic small vessel disease, J Clin Invest, vol.120, issue.2, p.27, 2010.

M. Monet-leprêtre, I. Haddad, C. Baron-menguy, M. Fouillot-panchal, and M. Riani, Abnormal recruitment of extracellular matrix proteins by excess Notch3 ECD: a new pathomechanism in CADASIL, Brain J. Neurol, vol.136, pp.1830-1875, 2013.

F. Dabertrand, C. Krøigaard, A. D. Bonev, E. Cognat, and T. Dalsgaard, Potassium channelopathy-like defect underlies early-stage cerebrovascular dysfunction in a genetic model of small vessel disease, Proc. Natl. Acad. Sci. U. S. A, vol.112, issue.7, pp.796-805, 2015.

C. Capone, F. Dabertrand, C. Baron-menguy, A. Chalaris, and L. Ghezali, Mechanistic insights into a TIMP3-sensitive pathway constitutively engaged in the regulation of cerebral hemodynamics, vol.5, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01615584

F. Erd?, L. Denes, and E. De-lange, Age-associated physiological and pathological changes at the blood-brain barrier: A review, J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab, vol.37, issue.1, pp.4-24, 2017.

K. Kisler, A. R. Nelson, S. V. Rege, A. Ramanathan, and Y. Wang, Pericyte degeneration leads to neurovascular uncoupling and limits oxygen supply to brain, Nat. Neurosci, vol.20, issue.3, pp.406-422, 2017.

A. Montagne, A. M. Nikolakopoulou, Z. Zhao, A. P. Sagare, and G. Si, Pericyte degeneration causes white matter dysfunction in the mouse central nervous system, Nat. Med, vol.24, issue.3, pp.326-363, 2018.

M. H. Park, J. Y. Lee, K. H. Park, I. K. Jung, and K. Kim, Vascular and Neurogenic Rejuvenation in Aging Mice by Modulation of ASM, Neuron, vol.100, issue.1, pp.167-182, 2018.

R. Brown, H. Benveniste, S. E. Black, S. Charpak, and M. Dichgans, Understanding the role of the perivascular space in cerebral small vessel disease, Cardiovasc. Res, vol.114, issue.11, p.28, 2018.

H. Mestre, J. Tithof, T. Du, W. Song, and W. Peng, Flow of cerebrospinal fluid is driven by arterial pulsations and is reduced in hypertension, Nat. Commun, vol.9, issue.1, p.4878, 2018.

H. Mestre, L. M. Hablitz, A. L. Xavier, W. Feng, and W. Zou, Aquaporin-4-dependent glymphatic solute transport in the rodent brain, vol.7, 2018.

B. T. Kress, J. J. Iliff, M. Xia, M. Wang, and H. S. Wei, Impairment of paravascular clearance pathways in the aging brain, Ann. Neurol, vol.76, issue.6, pp.845-61, 2014.

B. Campbell, G. A. Donnan, K. R. Lees, W. Hacke, and P. Khatri, Endovascular stent thrombectomy: the new standard of care for large vessel ischaemic stroke, Lancet Neurol, vol.14, issue.8, pp.846-54, 2015.

L. Pantoni, F. Fierini, and A. Poggesi, Thrombolysis in acute stroke patients with cerebral small vessel disease, Cerebrovasc. Dis. Basel Switz, vol.37, issue.1, pp.5-13, 2014.

C. Eggers, C. Bocksrucker, and L. Seyfang, The efficacy of thrombolysis in lacunar stroke -evidence from the Austrian Stroke Unit Registry, Eur. J. Neurol, vol.24, issue.6, pp.780-87, 2017.

A. Charidimou, M. Pasi, M. Fiorelli, S. Shams, V. Kummer et al., Leukoaraiosis, Cerebral Hemorrhage, and Outcome After Intravenous Thrombolysis for Acute Ischemic Stroke: A Meta-Analysis (v1), Stroke, vol.47, issue.9, pp.2364-72, 2016.

C. Cordonnier, A. Demchuk, W. Ziai, and C. S. Anderson, Intracerebral haemorrhage: current approaches to acute management, Lancet Lond. Engl, vol.392, pp.1257-68, 2018.

J. Scaggiante, X. Zhang, J. Mocco, and C. P. Kellner, Minimally Invasive Surgery for Intracerebral Hemorrhage, Stroke, vol.49, issue.11, p.29, 2018.

H. M. Perry, B. R. Davis, T. R. Price, W. B. Applegate, and W. S. Fields, Effect of treating isolated systolic hypertension on the risk of developing various types and subtypes of stroke: the Systolic Hypertension in the Elderly Program (SHEP), JAMA, vol.284, issue.4, pp.465-71, 2000.

. Progress-collaborative-group, Randomised trial of a perindopril-based bloodpressure-lowering regimen among 6,105 individuals with previous stroke or transient ischaemic attack, Lancet Lond. Engl, vol.358, issue.9287, pp.1033-1074, 2001.

C. Dufouil, J. Chalmers, O. Coskun, V. Besançon, and M. Bousser, Effects of blood pressure lowering on cerebral white matter hyperintensities in patients with stroke: the PROGRESS (Perindopril Protection Against Recurrent Stroke Study) Magnetic Resonance Imaging Substudy, Circulation, vol.112, issue.11, pp.1644-50, 2005.
URL : https://hal.archives-ouvertes.fr/inserm-00175957

. Sprint-research-group, J. T. Wright, J. D. Williamson, P. K. Whelton, and J. K. Snyder, A Randomized Trial of Intensive versus Standard Blood-Pressure Control, N. Engl. J. Med, vol.373, issue.22, pp.2103-2119, 2015.

. Accord-study-group, W. C. Cushman, G. W. Evans, R. P. Byington, and D. C. Goff, Effects of intensive blood-pressure control in type 2 diabetes mellitus, N. Engl. J. Med, vol.362, issue.17, pp.1575-85, 2010.

S. Bangalore, B. Toklu, E. Gianos, A. Schwartzbard, and H. Weintraub, Optimal Systolic Blood Pressure Target After SPRINT: Insights from a Network Meta-Analysis of Randomized Trials, Am. J. Med, vol.130, issue.6, pp.707-719, 2017.

. Sps3-study-group, O. R. Benavente, C. S. Coffey, R. Conwit, and R. G. Hart, Bloodpressure targets in patients with recent lacunar stroke: the SPS3 randomised trial, Lancet Lond. Engl, vol.382, issue.9891, pp.507-522, 2013.

A. M. Murray, F. Hsu, J. D. Williamson, R. N. Bryan, and H. C. Gerstein, ACCORDION MIND: results of the observational extension of the ACCORD MIND randomised trial, Diabetologia, vol.60, issue.1, pp.69-80, 2017.

S. E. Kjeldsen, K. Narkiewicz, M. Burnier, and S. Oparil, Intensive blood pressure lowering prevents mild cognitive impairment and possible dementia and slows development of white matter lesions in brain: the SPRINT Memory and Cognition IN Decreased Hypertension (SPRINT MIND) study, Blood Press, vol.27, issue.5, pp.247-295, 2018.

C. A. Peralta, L. A. Mcclure, R. Scherzer, M. C. Odden, and C. L. White, Effect of Intensive Versus Usual Blood Pressure Control on Kidney Function Among Individuals With Prior Lacunar Stroke: A Post Hoc Analysis of the Secondary Prevention of Small Subcortical Strokes (SPS3) Randomized Trial, Circulation, vol.133, issue.6, pp.584-91, 2016.

J. D. Williamson, N. M. Pajewski, A. P. Auchus, and R. N. Bryan, Effect of Intensive vs Standard Blood Pressure Control on Probable Dementia: A Randomized Clinical Trial, 2019.

I. D. Croall, D. J. Tozer, B. Moynihan, U. Khan, and J. T. O'brien, Effect of Standard vs Intensive Blood Pressure Control on Cerebral Blood Flow in Small Vessel Disease: The PRESERVE Randomized Clinical Trial, JAMA Neurol, vol.75, issue.6, pp.720-747, 2018.

K. L. Furie, S. E. Kasner, R. J. Adams, G. W. Albers, and R. L. Bush, Guidelines for the prevention of stroke in patients with stroke or transient ischemic attack: a guideline for healthcare professionals from the american heart association/american stroke association, Stroke, vol.42, issue.1, pp.227-76, 2011.

, Collaborative overview of randomised trials of antiplatelet therapy--I: Prevention of death, myocardial infarction, and stroke by prolonged antiplatelet therapy in various 31 categories of patients. Antiplatelet Trialists' Collaboration, BMJ, vol.308, issue.6921, pp.81-106, 1994.

. Sps3-investigators, O. R. Benavente, R. G. Hart, L. A. Mcclure, and J. M. Szychowski, Effects of clopidogrel added to aspirin in patients with recent lacunar stroke, N. Engl. J. Med, vol.367, issue.9, pp.817-842, 2012.

A. Oesterle, U. Laufs, and J. K. Liao, Pleiotropic Effects of Statins on the Cardiovascular System, Circ. Res, vol.120, issue.1, pp.229-272, 2017.

. Isgcwtc-cc, Failure to validate association between 12p13 variants and ischemic stroke, N Engl J Med, vol.362, issue.16, pp.1547-50

P. Amarenco, J. Bogousslavsky, A. Callahan, L. B. Goldstein, and M. Hennerici, High-dose atorvastatin after stroke or transient ischemic attack, N. Engl. J. Med, vol.355, issue.6, pp.549-59, 2006.

P. Amarenco, J. Labreuche, P. Lavallée, and P. Touboul, Statins in stroke prevention and carotid atherosclerosis: systematic review and up-to-date meta-analysis, Stroke, vol.35, issue.12, pp.2902-2911, 2004.

P. Amarenco, O. Benavente, L. B. Goldstein, A. Callahan, and H. Sillesen, Results of the Stroke Prevention by Aggressive Reduction in Cholesterol Levels (SPARCL) trial by stroke subtypes, Stroke, vol.40, issue.4, pp.1405-1414, 2009.

D. G. Hackam, M. Woodward, L. K. Newby, D. L. Bhatt, and M. Shao, Statins and intracerebral hemorrhage: collaborative systematic review and meta-analysis, Circulation, vol.124, issue.20, pp.2233-2275, 2011.

M. Cavalieri, R. Schmidt, C. Chen, V. Mok, and G. R. De-freitas, B vitamins and magnetic resonance imaging-detected ischemic brain lesions in patients with recent transient ischemic attack or stroke: the VITAmins TO Prevent Stroke (VITATOPS), 2012.

. Mri-substudy, Stroke, vol.43, issue.12, pp.3266-70

P. M. Vanhoutte, Y. Zhao, A. Xu, and S. Leung, Thirty Years of Saying NO: Sources, Fate, Actions, and Misfortunes of the Endothelium-Derived Vasodilator Mediator, Circ. Res, vol.119, issue.2, pp.375-96, 2016.

G. Bagdy, P. Riba, V. Kecskeméti, C. D. Juhász, and G. , Headache-type adverse effects of NO donors: vasodilation and beyond, Br. J. Pharmacol, vol.160, issue.1, pp.20-35, 2010.

R. H. Ritchie, G. R. Drummond, C. G. Sobey, D. Silva, T. M. Kemp-harper et al., The opposing roles of NO and oxidative stress in cardiovascular disease, Pharmacol. Res, vol.116, pp.57-69, 2017.

P. Singh, S. Vijayakumar, A. Kalogeroupoulos, and J. Butler, Multiple Avenues of Modulating the Nitric Oxide Pathway in Heart Failure Clinical Trials, Curr. Heart Fail. Rep, vol.15, issue.2, pp.44-52, 2018.

A. A. Miller, G. R. Drummond, H. Schmidt, and C. G. Sobey, NADPH oxidase activity and function are profoundly greater in cerebral versus systemic arteries, Circ. Res, vol.97, issue.10, pp.1055-62, 2005.

M. H. Hoffmann and H. R. Griffiths, The dual role of Reactive Oxygen Species in autoimmune and inflammatory diseases: evidence from preclinical models. Free Radic, Biol. Med, vol.125, pp.62-71, 2018.

H. Girouard, L. Park, J. Anrather, P. Zhou, and C. Iadecola, Angiotensin II attenuates endothelium-dependent responses in the cerebral microcirculation through nox-2-derived radicals, Arterioscler. Thromb. Vasc. Biol, vol.26, issue.4, pp.826-858, 2006.

D. Silva, T. M. Modrick, M. L. Dabertrand, F. Faraci, and F. M. , Changes in Cerebral Arteries and Parenchymal Arterioles With Aging: Role of Rho Kinase 2 and Impact of Genetic Background. Hypertens. Dallas Tex, vol.71, pp.921-948, 1979.

A. Aartsma-rus and G. Van-ommen, Antisense-mediated exon skipping: a versatile tool with therapeutic and research applications, RNA N. Y. N, vol.13, issue.10, pp.1609-1633, 2007.

J. W. Rutten, H. G. Dauwerse, D. Peters, A. Goldfarb, and H. Venselaar, Therapeutic NOTCH3 cysteine correction in CADASIL using exon skipping: in vitro proof of concept, Brain J. Neurol, vol.139, pp.1123-1158, 2016.

A. Aartsma-rus and A. M. Krieg, FDA Approves Eteplirsen for Duchenne Muscular Dystrophy: The Next Chapter in the Eteplirsen Saga, Nucleic Acid Ther, vol.27, issue.1, pp.1-3, 2017.

L. Ghezali, C. Capone, C. Baron-menguy, J. Ratelade, and S. Christensen, Notch3ECD immunotherapy improves cerebrovascular responses in CADASIL mice, 2018.

, Ann. Neurol, vol.84, issue.2, pp.246-59

M. Monet-lepretre, B. Bardot, B. Lemaire, V. Domenga, and O. Godin, Distinct phenotypic and functional features of CADASIL mutations in the Notch3 ligand binding domain, Brain, vol.132, pp.1601-1613, 2009.
URL : https://hal.archives-ouvertes.fr/pasteur-02075498

A. I. Machuca-parra, A. A. Bigger-allen, A. V. Sanchez, A. Boutabla, and J. Cardona-vélez, Therapeutic antibody targeting of Notch3 signaling prevents mural cell loss in CADASIL, J. Exp. Med, vol.214, issue.8, pp.2271-82, 2017.

J. W. Rutten, E. Boon, M. K. Liem, J. G. Dauwerse, and M. J. Pont, Hypomorphic NOTCH3 alleles do not cause CADASIL in humans, Hum. Mutat, vol.34, issue.11, pp.1486-89, 2013.

E. Cognat, C. Baron-menguy, V. Domenga-denier, S. Cleophax, and C. Fouillade, , 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 J. Cereb. Circ, vol.45, issue.3, pp.842-891

P. R. Deshpande, S. Rajan, B. L. Sudeepthi, A. Nazir, and C. P. , Patient-reported outcomes: A new era in clinical research, Perspect. Clin. Res, vol.2, issue.4, pp.137-181, 2011.

P. Benjamin, E. Zeestraten, C. Lambert, I. C. Ster, and O. A. Williams, Progression of MRI markers in cerebral small vessel disease: Sample size considerations for clinical trials, J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab, vol.36, issue.1, pp.228-268, 2016.

B. Patel and H. S. Markus, Magnetic resonance imaging in cerebral small vessel disease and its use as a surrogate disease marker, Int. J. Stroke Off. J. Int. Stroke Soc, vol.6, issue.1, pp.47-59, 2011.

S. Benisty, S. Reyes, O. Godin, D. Hervé, and N. Zieren, White-matter lesions without lacunar infarcts in CADASIL, J. Alzheimers Dis. JAD, vol.29, issue.4, pp.903-914, 2012.

J. M. Biesbroek, N. A. Weaver, and G. J. Biessels, Lesion location and cognitive impact of cerebral small vessel disease, Clin. Sci. Lond. Engl, vol.131, issue.8, pp.715-743, 1979.

E. Baykara, B. Gesierich, R. Adam, A. M. Tuladhar, and J. M. Biesbroek, A Novel Imaging Marker for Small Vessel Disease Based on Skeletonization of White Matter Tracts and Diffusion Histograms, Ann. Neurol, vol.80, issue.4, pp.581-92, 2016.

I. D. Croall, V. Lohner, B. Moynihan, U. Khan, and A. Hassan, Using DTI to assess white matter microstructure in cerebral small vessel disease (SVD) in multicentre studies, Clin. Sci. Lond. Engl, vol.131, issue.12, pp.1361-73, 1979.

E. A. Zeestraten, A. J. Lawrence, C. Lambert, P. Benjamin, and R. L. Brookes, Change in multimodal MRI markers predicts dementia risk in cerebral small vessel disease, Neurology, vol.89, issue.18, pp.1869-76, 2017.

Y. Ling, D. Guio, F. Jouvent, E. Duering, M. Hervé et al., Clinical correlates of longitudinal MRI changes in CADASIL, J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab, pp.271678-18757875, 2018.

R. Uiterwijk, R. J. Van-oostenbrugge, M. Huijts, D. Leeuw, P. W. Kroon et al., Total Cerebral Small Vessel Disease MRI Score Is Associated with Cognitive Decline in Executive Function in Patients with, Hypertension. Front. Aging Neurosci, vol.8, p.301, 2016.

F. De-guio, E. Jouvent, G. J. Biessels, S. E. Black, and C. Brayne, Reproducibility and variability of quantitative magnetic resonance imaging markers in cerebral small vessel disease, J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab, vol.36, issue.8, pp.1319-1356, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01317759

S. Yusuf, H. Diener, R. L. Sacco, D. Cotton, and S. Ounpuu, Telmisartan to prevent recurrent stroke and cardiovascular events, N. Engl. J. Med, vol.359, issue.12, pp.1225-1262, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00626465

R. Weber, C. Weimar, J. Blatchford, K. Hermansson, and I. Wanke, Telmisartan on top of antihypertensive treatment does not prevent progression of cerebral white matter lesions in the prevention regimen for effectively avoiding second strokes (PRoFESS) MRI substudy, Stroke, vol.43, issue.9, pp.2336-2378, 2012.