Journal Home > Volume 7 , Issue 4

Cerebral small vessel disease (CSVD) refers to a type of syndrome caused by lesions in perforating arteries, small veins, small arteries, or capillaries, resulting in clinical, imaging, or pathological alterations. The occurrence and development of CSVD are related to various cerebrovascular risk factors, such as metabolism and genetic factors. CSVD is diagnosed based on brain imaging biomarkers; however, biomarkers capable of predicting and diagnosing CSVD early in its progression have not been found. Exploring biomarkers closely related to disease progression is of great significance for early diagnosis, prognosis, prevention, and treatment of CSVD. This article examines the research progress of CSVD biomarkers, from inflammatory biomarkers, coagulation and fibrinolysis markers, biomarkers of endothelial dysfunction, biomarkers related to cerebrospinal fluid, and gene markers.


menu
Abstract
Full text
Outline
About this article

Advances in biomarkers of cerebral small vessel disease

Show Author's information Xue Peng1,2,3Jianhua Zhao1,2,3( )Junli Liu1,2,3Shaomin Li4
First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453100, Henan, China
Henan Key Laboratory of Neurorestoratology, Xinxiang 453100, Henan, China
Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Xinxiang 453100, Henan, China
Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston 02115, Massachusetts, United States

Abstract

Cerebral small vessel disease (CSVD) refers to a type of syndrome caused by lesions in perforating arteries, small veins, small arteries, or capillaries, resulting in clinical, imaging, or pathological alterations. The occurrence and development of CSVD are related to various cerebrovascular risk factors, such as metabolism and genetic factors. CSVD is diagnosed based on brain imaging biomarkers; however, biomarkers capable of predicting and diagnosing CSVD early in its progression have not been found. Exploring biomarkers closely related to disease progression is of great significance for early diagnosis, prognosis, prevention, and treatment of CSVD. This article examines the research progress of CSVD biomarkers, from inflammatory biomarkers, coagulation and fibrinolysis markers, biomarkers of endothelial dysfunction, biomarkers related to cerebrospinal fluid, and gene markers.

Keywords: cerebral small vessel disease, biomarkers, risk factors

References(86)

[1]
SJ Catchlove, A Pipingas, ME Hughes, et al. Magnetic resonance imaging for assessment of cerebrovascular reactivity and its relationship to cognition: a systematic review. BMC Neurosci. 2018, 19: 21.
[2]
EE Smith, G Saposnik, GJ Biessels, et al. Prevention of stroke in patients with silent cerebrovascular disease: A scientific statement for healthcare professionals from the American heart association/American stroke association. Stroke. 2017, 48(2): e44-e71.
[3]
A Ter Telgte, EMC van Leijsen, K Wiegertjes, et al. Cerebral small vessel disease: from a focal to a global perspective. Nat Rev Neurol. 2018, 14(7): 387-398.
[4]
L Pantoni. Cerebral small vessel disease: from pathogenesis and clinical characteristics to therapeutic challenges. Lancet Neurol. 2010, 9(7): 689-701.
[5]
L Østergaard, TS Engedal, F Moreton, et al. Cerebral small vessel disease: Capillary pathways to stroke and cognitive decline. J Cereb Blood Flow Metab. 2016, 36(2): 302-325.
[7]
S Verma, SH Li, MV Badiwala, et al. Endothelin antagonism and interleukin-6 inhibition attenuate the proatherogenic effects of C-reactive protein. Circulation. 2002, 105(16): 1890-1896.
[8]
S Hilal, MA Ikram, MM Verbeek, et al. C-reactive protein, plasma amyloid-β levels, and their interaction with magnetic resonance imaging markers. Stroke. 2018, 49(11): 2692-2698.
[8]
MB Pepys, GM Hirschfield. C-reactive protein: a critical update. J Clin Invest. 2003, 111(12): 1805-1812.
[9]
S Mitaki, A Nagai, H Oguro, et al. C-reactive protein levels are associated with cerebral small vessel-related lesions. Acta Neurol Scand. 2016, 133(1): 68-74.
[10]
MS Elkind, JM Luna, LA McClure, et al. C-reactive protein as a prognostic marker after lacunar stroke: levels of inflammatory markers in the treatment of stroke study. Stroke. 2014, 45(3): 707-716.
[11]
AK Boehme, LA McClure, Y Zhang, et al. Inflammatory markers and outcomes after lacunar stroke: levels of inflammatory markers in treatment of stroke study. Stroke. 2016, 47(3): 659-667.
[12]
W Whiteley, C Jackson, S Lewis, et al. Inflammatory markers and poor outcome after stroke: a prospective cohort study and systematic review of interleukin-6. PLoS Med. 2009, 6(9): e1000145.
[13]
J Staszewski, R Piusińska-Macoch, B Brodacki, et al. IL-6, PF-4, sCD40 L, and homocysteine are associated with the radiological progression of cerebral small- vessel disease: a 2-year follow-up study. Clin Interv Aging. 2018, 13: 1135-1141.
[14]
J Staszewski, E Skrobowska, R Piusińska-Macoch, et al. IL-1α and IL-6 predict vascular events or death in patients with cerebral small vessel disease-Data from the SHEF-CSVD study. Adv Med Sci. 2019, 64(2): 258-266.
[15]
A Shoamanesh, CS Kwok, O Benavente. Cerebral microbleeds: histopathological correlation of neuroimaging. Cerebrovasc Dis. 2011, 32(6): 528-534.
[16]
D Faustman, M Davis. TNF receptor 2 pathway: drug target for autoimmune diseases. Nat Rev Drug Discov. 2010, 9(6): 482-493.
[17]
MO Breckwoldt, JW Chen, L Stangenberg, et al. Tracking the inflammatory response in stroke in vivo by sensing the enzyme myeloperoxidase. Proc Natl Acad Sci USA. 2008, 105(47): 18584-18589.
[18]
CM Ballantyne, RC Hoogeveen, H Bang, et al. Lipoprotein-associated phospholipase A2, high- sensitivity C-reactive protein, and risk for incident ischemic stroke in middle-aged men and women in the Atherosclerosis Risk in Communities (ARIC) study. Arch Intern Med. 2005, 165(21): 2479-2484.
[19]
SZ Zhu, XB Wei, XH Yang, et al. Plasma lipoprotein- associated phospholipase A2 and superoxide dismutase are independent predicators of cognitive impairment in cerebral small vessel disease patients: diagnosis and assessment. Aging Dis. 2019, 10(4): 834-846.
[20]
A Shoamanesh, SR Preis, AS Beiser, et al. Inflammatory biomarkers, cerebral microbleeds, and small vessel disease: Framingham heart study. Neurology. 2015, 84(8): 825-832.
[21]
HS Markus, B Hunt, K Palmer, et al. Markers of endothelial and hemostatic activation and progression of cerebral white matter hyperintensities: longitudinal results of the Austrian Stroke Prevention Study. Stroke. 2005, 36(7): 1410-1414.
[22]
IL Knottnerus, K Winckers, H Ten Cate, et al. Levels of heparin-releasable TFPI are increased in first-ever lacunar stroke patients. Neurology. 2012, 78(7): 493-498.
[23]
A Hassan, BJ Hunt, M O'Sullivan, et al. Markers of endothelial dysfunction in lacunar infarction and ischaemic leukoaraiosis. Brain. 2003, 126(Pt 2): 424-432.
[24]
SJ Wiseman, FN Doubal, FM Chappell, et al. Plasma biomarkers of inflammation, endothelial function and hemostasis in cerebral small vessel disease. Cerebrovasc Dis. 2015, 40(3/4): 157-164.
[25]
JC Chapin, KA Hajjar. Fibrinolysis and the control of blood coagulation. Blood Rev. 2015, 29(1): 17-24.
[26]
Y Suzuki, N Nagai, K Umemura. A review of the mechanisms of blood-brain barrier permeability by tissue-type plasminogen activator treatment for cerebral ischemia. Front Cell Neurosci. 2016, 10: 2.
[27]
F Correa, M Gauberti, J Parcq, et al. Tissue plasminogen activator prevents white matter damage following stroke. J Exp Med. 2011, 208(6): 1229-1242.
[28]
Y Aono, T Ohkubo, M Kikuya, et al. Plasma fibrinogen, ambulatory blood pressure, and silent cerebrovascular lesions: the Ohasama study. Arterioscler Thromb Vasc Biol. 2007, 27(4): 963-968.
[29]
Y Notsu, T Nabika, H Bokura, et al. Evaluation of asymmetric dimethylarginine and homocysteine in microangiopathy-related cerebral damage. Am J Hypertens. 2009, 22(3): 257-262.
[30]
K Tajiri, A Sato, T Harunari, et al. Impact of rivaroxaban compared with warfarin on the coagulation status in Japanese patients with non-valvular atrial fibrillation: a preliminary analysis of the prothrombin fragment 1+2 levels. J Cardiol. 2015, 65(3): 191-196.
[31]
J Staszewski, R Piusińska-Macoch, B Brodacki, et al. Association between hemostatic markers, serum lipid fractions and progression of cerebral small vessel disease: A 2-year follow-up study. Neurol Neurochir Pol. 2018, 52(1): 54-63.
[32]
K Kario, T Matsuo, H Kobayashi, et al. Hyperinsulinemia and hemostatic abnormalities are associated with silent lacunar cerebral infarcts in elderly hypertensive subjects. J Am Coll Cardiol. 2001, 37(3): 871-877.
[33]
JI Weitz, JC Fredenburgh, JW Eikelboom. A test in context: D-dimer. J Am Coll Cardiol. 2017, 70(19): 2411-2420.
[34]
S Schol-Gelok, F Morelli, LR Arends, et al. A revised systematic review and meta-analysis on the effect of statins on D-dimer levels. Eur J Clin Invest. 2019, 49(8): e13130.
[35]
H Tomimoto, I Akiguchi, H Wakita, et al. Coagulation activation in patients with binswanger disease. Arch Neurol. 1999, 56(9): 1104-1108.
[36]
JD van Buul, E Kanters, PL Hordijk. Endothelial signaling by ig-Like cell adhesion molecules. Arterioscler Thromb Vasc Biol. 2007, 27(9): 1870-1876.
[37]
ILH Knottnerus, JWP Govers-Riemslag, K Hamulyak, et al. Endothelial activation in lacunar stroke subtypes. Stroke. 2010, 41(8): 1617-1622.
[38]
M Fornage, YA Chiang, ES O'Meara, et al. Biomarkers of inflammation and MRI-defined small vessel disease of the brain: the cardiovascular health study. Stroke. 2008, 39(7): 1952-1959.
[39]
E Cuadrado-Godia, P Dwivedi, S Sharma, et al. Cerebral small vessel disease: A review focusing on pathophysiology, biomarkers, and machine learning strategies. J Stroke. 2018, 20(3): 302-320.
[40]
K Kozuka, T Kohriyama, E Nomura, et al. Endothelial markers and adhesion molecules in acute ischemic stroke—sequential change and differences in stroke subtype. Atherosclerosis. 2002, 161(1): 161-168.
[41]
MO Giwa, J Williams, K Elderfield, et al. Neuropathologic evidence of endothelial changes in cerebral small vessel disease. Neurology. 2012, 78(3): 167-174.
[42]
J Pegon, E Groot, P de Groot, et al. Regulation of von Willebrand factor-platelet interactions. Thromb Haemost. 2010, 104(9): 449-455.
[43]
X Wang, FM Chappell, M Valdes Hernandez, et al. Endothelial function, inflammation, thrombosis, and basal ganglia perivascular spaces in patients with stroke. J Stroke Cerebrovasc Dis. 2016, 25(12): 2925-2931.
[44]
S Wiseman, F Marlborough, F Doubal, et al. Blood markers of coagulation, fibrinolysis, endothelial dysfunction and inflammation in lacunar stroke versus non-lacunar stroke and non-stroke: systematic review and meta-analysis. Cerebrovasc Dis. 2014, 37(1): 64-75.
[45]
PC Lavallée, J Labreuche, D Faille, et al. Circulating markers of endothelial dysfunction and platelet activation in patients with severe symptomatic cerebral small vessel disease. Cerebrovasc Dis. 2013, 36(2): 131-138.
[46]
A Pikula, RH Böger, AS Beiser, et al. Association of plasma ADMA levels with MRI markers of vascular brain injury: Framingham offspring study. Stroke. 2009, 40(9): 2959-2964.
[47]
U Khan, A Hassan, P Vallance, et al. Asymmetric dimethylarginine in cerebral small vessel disease. Stroke. 2007, 38(2): 411-413.
[48]
JX Guan, CQ Yan, Q Gao, et al. Analysis of risk factors in patients with leukoaraiosis. Medicine. 2017, 96(8): e6153.
[49]
U Sen, PK Mishra, N Tyagi, et al. Homocysteine to hydrogen sulfide or hypertension. Cell Biochem Biophys. 2010, 57(2/3): 49-58.
[50]
C Feng, X Bai, Y Xu, et al. Hyperhomocysteinemia associates with small vessel disease more closely than large vessel disease. Int J Med Sci. 2013, 10(4): 408-412.
[51]
RP Kloppenborg, PJ Nederkoorn, Y van der Graaf, et al. Homocysteine and cerebral small vessel disease in patients with symptomatic atherosclerotic disease. The SMART-MR study. Atherosclerosis. 2011, 216(2): 461-466.
[52]
Homocysteine Studies Collaboration. Homocysteine and risk of ischemic heart disease and stroke: a meta- analysis. JAMA. 2002, 288(16): 2015-2022.
[53]
A Hassan, BJ Hunt, M O'Sullivan, et al. Homocysteine is a risk factor for cerebral small vessel disease, acting via endothelial dysfunction. Brain. 2004, 127(Pt 1): 212-219.
[54]
RS Beard Jr, JJ Reynolds, SE Bearden. Hyperhomocysteinemia increases permeability of the blood-brain barrier by NMDA receptor-dependent regulation of adherens and tight junctions. Blood. 2011, 118(7): 2007-2014.
[55]
AH Hainsworth, MJ Fisher. A dysfunctional blood- brain barrier and cerebral small vessel disease. Neurology. 2017, 88(5): 420-421.
[56]
LR Bridges, J Andoh, AJ Lawrence, et al. Blood-brain barrier dysfunction and cerebral small vessel disease (arteriolosclerosis) in brains of older people. J Neuropathol Exp Neurol. 2014, 73(11): 1026-1033.
[57]
JM Wardlaw, PAG Sandercock, MS Dennis, et al. Is breakdown of the blood-brain barrier responsible for lacunar stroke, leukoaraiosis, and dementia? Stroke. 2003, 34(3): 806-812.
[58]
R Uiterwijk, RJ van Oostenbrugge, M Huijts, et al. Total cerebral small vessel disease MRI score is associated with cognitive decline in executive function in patients with hypertension. Front Aging Neurosci. 2016, 8: 301.
[59]
T Gattringer, D Pinter, C Enzinger, et al. Serum neurofilament light is sensitive to active cerebral small vessel disease. Neurology. 2017, 89(20): 2108-2114.
[60]
CX Qiu, MF Cotch, S Sigurdsson, et al. Retinal and cerebral microvascular signs and diabetes: the age, gene/environment susceptibility-Reykjavik study. Diabetes. 2008, 57(6): 1645-1650.
[61]
T Sumii, EH Lo. Involvement of matrix metalloproteinase in thrombolysis-associated hemorrhagic transformation after embolic focal ischemia in rats. Stroke. 2002, 33(3): 831-836.
[62]
GC Jickling, DZ Liu, B Stamova, et al. Hemorrhagic transformation after ischemic stroke in animals and humans. J Cereb Blood Flow Metab. 2014, 34(2): 185-199.
[63]
W Walz, FS Cayabyab. Neutrophil infiltration and matrix metalloproteinase-9 in lacunar infarction. Neurochem Res. 2017, 42(9): 2560-2565.
[64]
ZA Corbin, N Rost, S Lorenzano, et al. White matter hyperintensity volume correlates with matrix metalloproteinase-2 in acute ischemic stroke. J Stroke Cerebrovasc Dis. 2014, 23(6): 1300-1306.
[65]
A Assareh, KA Mather, PR Schofield, et al. The genetics of white matter lesions. CNS Neurosci Ther. 2011, 17(5): 525-540.
[66]
MJ van Rijn, MJ Bos, A Isaacs, et al. Polymorphisms of the renin-angiotensin system are associated with blood pressure, atherosclerosis and cerebral white matter pathology. J Neurol Neurosurg Psychiatry. 2007, 78(10): 1083-1087.
[67]
A Shiga, H Nozaki, A Yokoseki, et al. Cerebral small-vessel disease protein HTRA1 controls the amount of TGF-β1 via cleavage of proTGF-β1. Hum Mol Genet. 2011, 20(9): 1800-1810.
[68]
I Di Donato, S Bianchi, N De Stefano, et al. 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. 2017, 15(1): 41.
[69]
H Chabriat, A Joutel, M Dichgans, et al. Cadasil. Lancet Neurol. 2009, 8(7): 643-653.
[70]
A Joutel, C Corpechot, A Ducros, et al. Notch3 mutations in CADASIL, a hereditary adult-onset condition causing stroke and dementia. Nature. 1996, 383(6602): 707-710.
[71]
S Tikka, M Baumann, M Siitonen, et al. CADASIL and CARASIL. Brain Pathol. 2014, 24(5): 525-544.
[72]
K Hara, A Shiga, T Fukutake, et al. Association of HTRA1 mutations and familial ischemic cerebral small-vessel disease. N Engl J Med. 2009, 360(17): 1729-1739.
[73]
E Verdura, D Hervé, E Scharrer, et al. Heterozygous HTRA1 mutations are associated with autosomal dominant cerebral small vessel disease. Brain. 2015, 138(Pt 8): 2347-2358.
[74]
DS Kuo, C Labelle-Dumais, DB Gould. COL4A1 and COL4A2 mutations and disease: insights into pathogenic mechanisms and potential therapeutic targets. Hum Mol Genet. 2012, 21(R1): R97-R110.
[75]
DB Gould, FC Phalan, SE van Mil, et al. Role of COL4A1 in small-vessel disease and hemorrhagic stroke. N Engl J Med. 2006, 354(14): 1489-1496.
[76]
S Lanfranconi, HS Markus. COL4A1 mutations as a monogenic cause of cerebral small vessel disease: a systematic review. Stroke. 2010, 41(8): e513-518.
[77]
LCA Rutten-Jacobs, NS Rost. Emerging insights from the genetics of cerebral small-vessel disease. Ann N Y Acad Sci. 2019, in press, .
[78]
S Miyatake, S Schneeberger, N Koyama, et al. Biallelic COLGALT1 variants are associated with cerebral small vessel disease. Ann Neurol. 2018, 84(6): 843-853.
[79]
A Charidimou. Defining retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations. Brain. 2016, 139(11): 2819-2821.
[80]
YG Yang, T Lindahl, DE Barnes. Trex1 exonuclease degrades ssDNA to prevent chronic checkpoint activation and autoimmune disease. Cell. 2007, 131(5): 873-886.
[81]
M Hasan, CS Fermaintt, NG Gao, et al. Cytosolic nuclease TREX1 regulates oligosaccharyltransferase activity independent of nuclease activity to suppress immune activation. Immunity. 2015, 43(3): 463-474.
[82]
MG Vervloet, S Sezer, ZA Massy, et al. The role of phosphate in kidney disease. Nat Rev Nephrol. 2017, 13(1): 27-38.
[83]
SR Li, R Liu, L Chen, et al. The correlation research of the lack of 25-(OH)-D and cerebral small vessel disease. Chin J Pract Nerv Dis. 2017, 20(5): 20-23.
[84]
PW Chung, KY Park, JM Kim, et al. 25-hydroxyvitamin D status is associated with chronic cerebral small vessel disease. Stroke. 2015, 46(1): 248-251.
[85]
CP Chung, LN Peng, KH Chou, et al. High circulatory phosphate level is associated with cerebral small-vessel diseases. Transl Stroke Res. 2019, 10(3): 265-272.
[86]
N Cheung, T Mosley, A Islam, et al. Retinal microvascular abnormalities and subclinical magnetic resonance imaging brain infarct: a prospective study. Brain. 2010, 133(Pt 7): 1987-1993.
Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 28 July 2019
Revised: 21 October 2019
Accepted: 28 October 2019
Published: 29 November 2019
Issue date: December 2019

Copyright

© The authors 2019

Acknowledgements

This work was supported by Henan Natural Science Foundation, China (No. 182300410389, grant to Jianhua Zhao); Scientific and Technological Project of Health and Family Planning Commission, Henan Province, China (No. 201303105, grant to Jianhua Zhao); Key Scientific Research Projects of Universities in Henan Province, China (No. 16B320019, grant to Jianhua Zhao), and the project for the disciplinary Group of Psychology and Neuroscience, Xinxiang Medical University, China (No. 2016PN-KFKT-16, grant to Shaomin Li).

Rights and permissions

This article is published with open access at http://jnr.tsinghuajournals.com

Return