• Users Online: 1653
  • Home
  • Print this page
  • Email this page
Home About us Editorial board Ahead of print Current issue Search Archives Submit article Instructions Subscribe Contacts Login 

   Table of Contents      
REVIEW ARTICLE
Year : 2015  |  Volume : 1  |  Issue : 1  |  Page : 79-87

A molecular/genetic approach to cerebral small-vessel disease: Beyond aging and hypertension


1 Neurology Care Line; Center for Translational Research in Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center; Department of Neurology, Baylor College of Medicine, Houston, Texas, USA
2 Department of Neurology; Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
3 Neurology Care Line, Michael E. DeBakey Veterans Affairs Medical Center; Department of Neurology; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
4 Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, Ohio, USA

Date of Submission14-Apr-2015
Date of Acceptance23-Jun-2015
Date of Web Publication30-Sep-2015

Correspondence Address:
Sharyl R Martini
Department of Veterans Affairs, Michael E. DeBakey VA Medical Center, Neurology Care Line, 2002 Holcombe Boulevard, MS 127 Houston, Texas - 77030-4211
USA
Login to access the Email id

Source of Support: None, Conflict of Interest: None


DOI: 10.4103/2394-8108.166376

Rights and Permissions
  Abstract 

Lacunar infarction, white matter hyperintensities (WMH), deep cerebral microbleeds (dCMB), and deep intracerebral hemorrhage (ICH) are increasingly recognized as manifestations of a common underlying vasculopathy, encompassed by the term "cerebral small-vessel disease" (CSVD). Epidemiologic studies have found robust associations of the individual aspects of CSVD with aging and hypertension; however, heritability estimates and the disproportionate burden of CSVD in underrepresented minorities suggest that genetic factors contribute substantially to CSVD risk. Here we present the rationale for studying these phenotypes as part of a spectrum of CSVD, review aspects of genetic study design, summarize current knowledge of genetic contribution to CSVD, and discuss the next steps required to translate these genetic discoveries into therapies for this devastating disease. Genetic studies were identified using PubMed. Regions achieving genome-wide significance in association studies, meta-analyses of candidate gene studies, and studies of genes associated with Mendelian conditions exhibiting CSVD phenotypes have been summarized.

Keywords: Cerebral microbleeds, cerebral small-vessel disease (CSVD), genetics, intracerebral hemorrhage (ICH), lacunar stroke, white matter hyperintensities (WMH)


How to cite this article:
Martini SR, Williams SR, Moretti P, Woo D, Worrall BB. A molecular/genetic approach to cerebral small-vessel disease: Beyond aging and hypertension. Brain Circ 2015;1:79-87

How to cite this URL:
Martini SR, Williams SR, Moretti P, Woo D, Worrall BB. A molecular/genetic approach to cerebral small-vessel disease: Beyond aging and hypertension. Brain Circ [serial online] 2015 [cited 2023 Jun 3];1:79-87. Available from: http://www.braincirculation.org/text.asp?2015/1/1/79/166376

Sharyl R Martini, Stephen R Williams and Bradford B Worrall
These authors contributed equally to this work.



  Introduction Top


The term cerebral small-vessel disease (CSVD) encompasses a spectrum of pathologies affecting brain vessels 50-500 μm in diameter. Age- and hypertension-related pathologies causing intrinsic disease of the small penetrating arterioles is the most common type of CSVD. CSVDs such as cerebral amyloid angiopathy involve other small brain arteries [Figure 1]. [1] Age- and hypertension-related CSVD pathologies manifest as small subcortical "lacunar" ischemic strokes, deep intracerebral hemorrhage (ICH), magnetic resonance imaging (MRI) white matter hyperintensities (WMH), or deep cerebral microbleeds (dCMB). These manifestations frequently coexist, often silently, and are associated with progressive cognitive impairment and vascular dementia. [2],[3] Our understanding of the molecular underpinnings of CSVD has lagged significantly behind our pathological and epidemiologic understanding, impeding development of targeted therapies for this common disease. Recognizing the interrelatedness of these clinical and radiographic manifestations has allowed us to start uncovering their shared and distinct pathophysiologic mechanisms. The purpose of this review is to frame CSVD as a spectrum of interrelated conditions with shared etiologies, review underlying genetic mechanisms (many of which are shared), and discuss the steps following successful identification of genes contributing to CSVD.
Figure 1: CSVD. (a) Regions affected by CSVD (b-d) Axial FLAIR image demonstrating lacunar infarcts (B, arrows), deep ICH (C, asterisk), and diffuse WMH (e) Axial gradient echo image demonstrating dCMB (arrow) and deep ICH (asterisk).
CSVD = Cerebral small-vessel disease, FLAIR = fluid-attenuated inversion recovery, ICH = Intracerebral hemorrhage, WMH = White matter hyperintensities, dCMB = Deep cerebral microbleeds


Click here to view


To accomplish this purpose, we took the following approach: First we discuss reconceptualization of the CSVD spectrum, allowing for more accurate phenotyping. We next describe CSVD clinical presentations and provide examples of "phenocopies" that reduce the power of genetic studies. Finally, we describe the advantages and disadvantages of genetic approaches including family-based studies, candidate gene studies, and genome-wide association studies (GWAS), detailing methods for molecular characterization of the identified variants. For this review, genetic studies were identified using PubMed. Regions achieving genome-wide significance and meta-analyses of candidate gene studies were identified using search terms for each phenotype ("lacunar" or "small vessel" and stroke, "white matter hyperintensities," "cerebral microbleeds" or "intracerebral hemorrhage") plus ("gwas," "gene," or "genetic") to identify cohort-based association studies. For polymorphisms with multiple conflicting independent studies, only meta-analyses were summarized. Genes associated with Mendelian conditions exhibiting CSVD phenotypes were searched individually by combining the CSVD phenotype search terms mentioned above with the gene name.


  Reconceptualizing the CSVD Phenotype Top


Reliance on phenomenology to classify cerebrovascular diseases has led to the collapsing of distinct diseases into broad categories or the failure to recognize related processes due to distinct clinical, pathological, or radiographic features. Advances in neuroimaging, vascular imaging, and various "-omics" strategies have created the opportunity to reclassify brain and vascular disorders based on shared associations and advances in our understanding of underlying pathophysiology. This strategy allowed reconceptualization of CVSD through a shared pathogenesis encompassing lacunar stroke, hypertensive hemorrhage, leukoaraiosis, and cerebral microbleeds, [Figure 2] [4],[5],[6] and recognition of the overlap with extracerebral small-vessel vasculopathies. [7],[8] Precise definitions of the clinical phenotypes are crucial for the design of successful genetic and mechanistic studies.
Figure 2: Reconceptualizing manifestations of CSVD.
CSVD = Cerebral small-vessel disease, dCMB = Deep cerebral microbleeds, ICH = Intracerebral hemorrhage


Click here to view


The important roles that aging and hypertension play as risk factors for CSVD [9],[10],[11],[12],[13],[14],[15],[16],[17],[18],[19],[20] led to theories that such disease is inevitable "wear and tear" on the brain. However, heritability studies have found that 34% of ICH risk is conferred by genetic makeup, [21] and studies of WMH estimate heritability at 55-80%. [22],[23],[24] This evidence for heritability suggests a substantial molecular component, with potential to yield new targets for prevention or intervention.

Phenotypic presentation

CSVD has a range of phenotypes that can be studied individually, although each has advantages and disadvantages [Table 1]. Lacunar infarcts represent 20% of ischemic strokes and occur with high frequency in the population, [13] providing ample power. However, phenocopies (infarcts of similar appearance due to alternate etiologies) reduce the homogeneity of the clinical subtype, making it more difficult to identify true associations. [11],[25],[26],[27],[28] Examples of such phenocopies include occlusion of a small penetrating artery from parent vessel atherosclerosis or cardioembolism. This may be one reason that the heritability estimate for lacunar stroke was only 16.1%, much lower than for other manifestations of CSVD. [29] Another disadvantage of lacunar infarctions is that up to 89% are clinically silent, [12],[13],[30] requiring cohort study design to avoid selection bias. Other issues include variable progression pattern and radiographic permanence. [31] Deep ICH refers to the rupture of small cerebral vessels. Deep ICH is readily detected by noncontrast head computed tomography (CT), >90% after age 45 are due to intrinsic CSVD, [32] and most come to clinical attention, allowing for case-based ascertainment. [18] A significant disadvantage in studying deep ICH is that the incidence rate is approximately 20 per 100,000 per year, [33] limiting recruitment and thereby power. The high observed early mortality associated with ICH may result in selection bias, [34] although "hot-pursuit" methodology [18],[35] or validation using cohort studies that recruit cases prior to stroke occurrence can attenuate this bias.
Table 1: CSVD phenotypes

Click here to view


Studying intermediate phenotypes in progression toward clinical disease may allow for more robust association with causative genetic variants. [36] WMH and dCMB are two such intermediate CSVD phenotypes detected by MRI. WMH increase with age and hypertension, and are typically located around the lateral ventricles or in the subcortical or brainstem white matter. The advantages of studying WMH include their high prevalence in the population [14],[37],[38] as well their quantifiable nature, allowing volumetric analysis. [39],[40] Disadvantages include the need for MRI to sensitively define lesion burden, variable appearance depending on acquisition parameters, and the possibility of regression over time. [3],[41],[42] Like lacunes, WMHs have significant phenocopies: Demyelinating disease, remote injuries, migraine headaches, and metabolic disorders/genetic leukodystrophies. [43] Like WMH, dCMBs are highly associated with other manifestations of CSVD, quantifiable, and require MRI for detection. Both dCMBs and WMHs tend to be clinically silent. [44],[45],[46],[47] Although there is a lower incidence of dCMB compared to WMH, no significant dCMB phenocopies exist. [4],[48] Given the clinically silent nature of most WMH and dCMB, [20],[49] cohort design studies are required to minimize selection bias.

Regardless of the specific manifestation of CSVD studied, its increasing occurrence with age is an important consideration to prevent presymptomatic individuals from being misclassified as controls. Age matching for case-control studies and selecting older-aged individuals for cohort studies reduces this concern.

Designing studies

Family-based studies

Once the phenotype is precisely defined, the presumed mode of inheritance should be considered when designing a genetic study [Figure 3]. Families with multiple affected individuals, particularly with a Mendelian inheritance pattern, can help identify genetic variants with large effect sizes. Although such monogenic disorders are usually responsible for a small fraction of disease, identification of the genes responsible may reveal the association of less severe variants with sporadic disease and shed light on the underlying molecular pathogenesis. Several monogenic disorders cause CSVD, resulting in lacunar infarcts, deep ICH, or WMH [Table 2]. These disorders include cerebral autosomal dominant arteriopathy with silent infarcts and leukoencephalopathy (CADASIL), cerebral autosomal recessive arteriopathy with silent infarcts and leukoencephalopathy (CARASIL), Fabry disease, COL41A-related disorders, and retinal vasculopathy with cerebral leukodystrophy (RVCL). [1] The recently described autosomal recessive syndrome "deficiency of ADA2" (DADA2) includes small-vessel, lacunar-type stroke, deep hemorrhage, microbleeds, and systemic inflammation in a largely pediatric population. [50] The putative genetic variants were loss-of-function mutations in CECR1 (cat eye syndrome chromosome region, candidate 1) encoding the adenosine deaminase 2 (ADA2) protein: the same gene implicated in Sneddon's syndrome.
Figure 3: Strategies for identifying genetic variants. Allele frequency and magnitude of effect determine the most suitable genetic study design. Increasing allele frequency (x axis) is indicated by increasing yellow intensity; increasing genetic variant effect size (y axis) is indicated by increasing blue intensity

Click here to view
Table 2: Monogenic disorders exhibiting CSVD

Click here to view


Variants in genes for these Mendelian disorders have been associated with sporadic disease. Variants in the CADASIL gene Notch3 have been associated with sporadic WMH. [51] Variants in the Fabry disease gene alpha galactosidase A (or reductions in enzymatic activity) have been found in 1-4% of Portuguese [52] and Belgian [53] young stroke populations, and are associated with multiple lacunar infarctions in the Japanese elderly. [54] Recently, variants in the COL4A1/COL4A2 genomic region were found to be associated with sporadic deep ICH, with nonsignificant trends toward association with lacunar stroke and WMHs. [55] This result confirms a prior report of COL4A1 mutations in spontaneous late-onset ICH. [56] Point mutations in TREX1, the gene responsible for RVCL, were found in two apparently sporadic young stroke patients with severe white matter disease and recurrent strokes. [57] Two brothers heterozygous for a disease-associated mutation of CECR1 presented with typical, albeit aggressively recurrent, lacunar disease in their seventh and eighth decades. [50] The identification of variants for single-gene disease in sporadic cases (presumably lacking the classic clinical picture) underscores the utility of studying Mendelian disorders to understand sporadic disease.

Candidate gene studies

Candidate gene approaches select genes for study by their potential pathophysiologic mechanism, association with related phenotypes or pathway analyses. Unfortunately, these studies tend to limit candidate selection to pathways that are already presumed important for the phenotype, and have frequently yielded results not confirmed by subsequent studies. Meta-analyses have found the angiotensin-converting enzyme [ACE; Online Mendelian Inheritance in Man (OMIM) 106180] insertion/deletion polymorphism to be associated with ischemic stroke (particularly small-vessel), ICH and WMH with the largest effect in Asian populations. [58],[59],[60] Similarly, the CC polymorphism of methylenetetrahydrofolate reductase (MTHFR; OMIM 607093) has been associated in meta-analyses with large-vessel ischemic stroke and ICH, but not significantly with WMH. [58],[61],[62] Polymorphism of protein kinase C-eta (PRKCH; OMIM 605437) has been associated with lacunar stroke and ICH in a Chinese population. [63] Recent data associate apolipoprotein E alleles APOE2 and APOE4 (OMIM: 107741) with deep ICH, albeit with smaller effect sizes than lobar ICH due to cerebral amyloid angiopathy, for which these variants are best known. [64]

Genome-wide association studies

Unlike Mendelian diseases in which a single gene with high penetrance is responsible for disease, in a polygenic model multiple genetic variants, each with a small effect size, contribute to disease risk. This is the likely model for most complex traits and chronic diseases such as CSVD. Genetic and environmental variables such as aging interact with each genetic variant to modify its contribution to disease pathogenesis [Figure 4].
Figure 4: Genetic and environmental factors interact to influence development and progression of CSVD. CSVD = Cerebral smallvessel disease

Click here to view


Complete sequencing of the human genome allows unbiased approaches to evaluate the polygenic model of disease. Such studies simultaneously examine hundreds of thousands to millions of variants with high degrees of genome coverage. Although a given variant may not be causative, a nearby causative variant may be captured, as it is inherited along with the marker: a phenomenon known as linkage disequilibrium.

The genome-wide association study (GWAS) is predicated on the polygenic model. Most GWASs examine polymorphisms present in >5% of the population, since these are the single-nucleotide polymorphisms (SNPs) on most commercially available platforms. The simultaneous testing of hundreds of thousands of associations requires rigorous correction for multiple testing. A P value threshold for significance of <5 × 10 -8 is commonly applied (correcting for 1 million comparisons, as opposed to <0.05 for a single hypothesis or association). [65]

The consistent association of a SNP with disease (typically through replication) indicates that either that change itself or a tightly linked variant in a nearby position of the genome predisposes to disease. This approach allows identification of genes or molecular pathways not previously suspected to play a role in the pathophysiology of disease. Current GWAS technologies do not evaluate all regions of the genome with equal depth. For example, current commercial platforms cover the APOE region poorly as a biallelic variant. One limitation of GWAS is that the specific functional variants responsible for disease susceptibility are rarely identified, requiring additional effort to confirm the role of that genomic region using fine mapping and functional studies. [65],[66] Another limitation is that common SNP platforms represent genetic variation well in whites of European descent, but may not capture genetic variation as effectively in other racial/ethnic groups.

GWAS studies have identified a number of loci associated with CSVD phenotypes [Table 3]. Variants at 1q22 have achieved genome-wide significance for deep ICH as well as WMH. [55],[67],[68] Additional loci achieving genome-wide significance for WMH include 17q25, 10q24, 2p21, and 2p16. [67],[69],[70],[71] Focused GWA analysis at 6p25 identified SNPs associated with WMH strongly influencing expression of the forkhead box C1 transcription factor (FOXC1). [72] Interestingly, this study also found that human subjects with alterations in FOXC1 and paired-like homeodomain transcription factor 2 (PITX2) (OMIM: 601542) had CSVD, although PITX2 SNPs have only achieved genome-wide significance for cardioembolic stroke. [73] Despite the high prevalence of lacunar stroke and large-scale meta-analyses of stroke by subtype no SNPs have yet achieved genome-wide significance for lacunar stroke. The greater number of SNPs associated with WMH relative to ICH underscores the power of evaluating intermediate phenotypes while searching for causative variants of complex diseases.
Table 3: Genome-wide associations for CSVD

Click here to view



  Sequencing and Rare Variant Analysis Top


Unbiased approaches can also investigate the rare variant hypothesis that common diseases are due to a multitude of rare variants or private mutations, each with a large effect size. In this model, rare variants from different individuals will cluster preferentially in genes playing a role in the pathophysiology of the disease and cumulatively account for the population burden of disease.

Detection of rare variants is currently feasible using whole-genome or -exome sequencing [Table 4]. Burden and other analysis methods then identify those genes with a preponderance of rare variants. [74] For example, in Parkinson's disease, rare variants would be expected to cluster in alpha-synuclein (SNCA; OMIM 163890), Parkinson's disease 1 (PARK1; OMIM 168601) and leucine-rich repeat kinase (LRRK2; OMIM 609007) in cases but not controls, implicating these genes in disease pathogenesis. This approach has been piloted in ischemic stroke, including small-vessel (lacunar) disease, with promising results. [75],[76] Small sample sizes for rare diseases may lack sufficient power to detect association with rare variants. As deep sequencing of the entire genome becomes more common, variants will not be limited to coding regions, requiring a more comprehensive investigation (similar to that currently required for GWAS-identified SNPs) in order to understand their biological effects. [66] The analytic approaches for whole-exome sequencing have proved computationally intensive, requiring innovation and creativity. Whole-genome sequencing will undoubtedly require further analytic ingenuity. [77]
Table 4: Comparison of whole-genome versus whole-exome sequencing

Click here to view



  Population-Specific Considerations Top


Initial commercially available platforms ("SNP chips") were developed based on sequencing information largely derived from individuals of European descent. Genetic variation, and thus genetic risk for disease, may differ among populations. There are several examples of population-specific differences in the genetic architecture of CSVD. An ancestral deletion involving the gene TM4SF20 (OMIM 615404) found exclusively in individuals of Southeast Asian descent predisposes to WMH. [78] In another example, we found no association of 1q22 SNPs with ICH in Hispanic individuals, despite their robust association in other race/ethnic groups. [68] Hypertension is a significant risk factor for CSVD, and the frequency of related polymorphisms varies among populations. For example, specific polymorphisms in the alpha-1- and alpha-2-adrenergic receptors are much more common in African Americans. [79],[80] The relationship between specific genetic polymorphisms and hypertension is complex, but polymorphisms in alpha adrenergic receptors are associated with lacunar infarcts, [81] and hypertension risk allele burden has been shown to increase ICH risk. [82] These examples underscore the importance of evaluating genetic differences within specific populations.


  Following Up Regions of Interest Top


After studies identify regions of interest, the next task is to determine precisely which variant(s) within that particular stretch of DNA predispose to disease. On occasion, the identified SNP is itself the causal variant. In most cases however, the disease-associated SNP is merely "a signpost around which one must do a finer search." [65] Initial steps in that finer search might include haplotype analysis and deep sequencing of the region. Identifying causal variants can be challenging due to the extensive sequence variation in human DNA and the difficulty in interpreting functional effects of that variation.

Interpretation of DNA variation within the coding sequence of a gene is more straightforward than interpretation of noncoding variants. The altered sequence may cause absent or reduced levels of the protein impeding its normal function, or may cause the protein to gain an additional, abnormal function (often called a dominant negative). Other times, the protein or its RNA message cannot be degraded and becomes toxic. Noncoding variants may affect splicing or stability of the RNA message, or may affect gene expression through alteration in promoter and enhancer regions, DNA methylation, or chromatin structure. Analysis of expression quantitative trait loci (eQTL) determines the effect of a SNP on expression of nearby genes (cis), or genes on different chromosomes (trans). Effects on gene expression are often cell-type-specific [66] and brain or cerebral vessels are not generally accessible, making gene expression studies in these tissues difficult. Large consortia such as the Genotype-Tissue Expression project (GTEx) are beginning to address this limitation in a systematic fashion. [83]

Once the genetic variant is identified, defining the context within which it predisposes to a complex disease may still prove challenging. The variant itself is likely not sufficient to cause disease, and will predispose to disease through its interaction with other genetic or environmental factors. Mathematical models are one solution for deciphering this complexity. Another is to identify a molecular or biochemical phenotype associated with the genetic variant, which can then be linked with the disease. [84] Such intermediate molecular phenotypes may exhibit stronger association with disease, as they are more proximal to the disease state than the genetic variant. [85]

Finally, experimentation in model systems is required to elucidate the functional role of these genetic and biochemical effectors in development of CSVD. Such experimentation may be carried out in cell culture. The use of induced pluripotent stem cells that can be reprogrammed into disease-specific cell types allows investigation into functional effects of genetic variation in a model that is more biologically relevant than cell lines used previously. In fact, such cells may be induced to form "organoids" reminiscent of particular tissues. [86],[87] Culture models of the neurovascular unit have yet to be developed, making animal studies particularly useful. Although no animal model perfectly recapitulates human CSVD, the spontaneously hypertensive stroke-prone rat model has a number of pathophysiologic similarities to human CSVD. [88] With the advent of the clustered regularly interspaced short pallindromic repeats (CRISPR)/Cas9 system, we have the ability to introduce specific genetic variants into cell culture and model systems with great accuracy and speed. [89] Both cell culture and animal work will be required to elucidate the molecular basis for CSVD and assess the effects of manipulating identified pathways in order to slow or reverse CSVD.


  Future Directions Top


Genetic studies are a powerful tool for understanding the molecular underpinnings of CSVD. As classification of CSVD phenotypic variation becomes more accurate, additional genes that directly affect clinical risk factors and outcomes will be identified. Maximizing information gained from genetic studies requires collaborative study utilizing thousands of human samples as well as cell culture and animal models to clarify specific disease mechanisms. Additionally, understanding how age and environmental exposures interact with genetic background will greatly enhance our ability to predict CSVD risk. Studying underrepresented minority populations is critical, as CSVD occurs earlier and often with greater severity than in individuals of European descent. Indeed, genetic differences in CSVD susceptibility likely contribute to racial/ethnic disparities in stroke. Once disease mechanisms underlying CSVD are established, this iterative bedside-to-bench-to-bedside approach will be imperative for translating that knowledge into targeted therapies. Perhaps, as we gain a deeper understanding of these complex mechanisms of disease, individual genetic risk profiles may suggest particularly effective medical or dietary therapies to halt CSVD's common and devastating manifestations.

Financial support and sponsorship

Nil

Conflicts of interest

There are no conflicts of interest.

 
  References Top

1.
Pantoni L, Gorelick PB. Cerebral Small Vessel Disease. Cambridge, United Kingdom: Cambridge University Press; 2014.  Back to cited text no. 1
    
2.
Pantoni L. Cerebral small vessel disease: From pathogenesis and clinical characteristics to therapeutic challenges. Lancet Neurol 2010;9:689-701.  Back to cited text no. 2
    
3.
Ankolekar S, Renton C, Bereczki D, Sprigg N, Payne T, Gommans J, et al. ENOS Trial Investigators. Effect of the neutral CLOTS 1 trial on the use of graduated compression stockings in the Efficacy of Nitric Oxide Stroke (ENOS) trial. J Neurol Neurosurg Psychiatry 2013;84:342-7.  Back to cited text no. 3
    
4.
Moran C, Phan TG, Srikanth VK. Cerebral small vessel disease: A review of clinical, radiological, and histopathological phenotypes. Int J Stroke 2012;7:36-46.  Back to cited text no. 4
    
5.
Rost NS, Fitzpatrick K, Biffi A, Kanakis A, Devan W, Anderson CD, et al. White matter hyperintensity burden and susceptibility to cerebral ischemia. Stroke 2010;41:2807-11.  Back to cited text no. 5
    
6.
Rost NS, Rahman RM, Biffi A, Smith EE, Kanakis A, Fitzpatrick K, et al. White matter hyperintensity volume is increased in small vessel stroke subtypes. Neurology 2010;75:1670-7.  Back to cited text no. 6
    
7.
Cheung N, Liew G, Lindley RI, Liu EY, Wang JJ, Hand P, et al. Retinal fractals and acute lacunar stroke. Ann Neurol 2010;68:107-11.  Back to cited text no. 7
    
8.
Toyoda K. Cerebral small vessel disease and chronic kidney disease. J Stroke 2015;17:31-7.  Back to cited text no. 8
    
9.
Dozono K, Ishii N, Nishihara Y, Horie A. An autopsy study of the incidence of lacunes in relation to age, hypertension, and arteriosclerosis. Stroke 1991;22:993-6.  Back to cited text no. 9
    
10.
Tuszynski MH, Petito CK, Levy DE. Risk factors and clinical manifestations of pathologically verified lacunar infarctions. Stroke 1989;20:990-9.  Back to cited text no. 10
    
11.
Khan U, Porteous L, Hassan A, Markus HS. Risk factor profile of cerebral small vessel disease and its subtypes. J Neurol Neurosurg Psychiatry 2007;78:702-6.  Back to cited text no. 11
    
12.
Das RR, Seshadri S, Beiser AS, Kelly-Hayes M, Au R, Himali JJ, et al. Prevalence and correlates of silent cerebral infarcts in the Framingham offspring study. Stroke 2008;39:2929-35.  Back to cited text no. 12
    
13.
Longstreth WT Jr, Bernick C, Manolio TA, Bryan N, Jungreis CA, Price TR. Lacunar infarcts defined by magnetic resonance imaging of 3660 elderly people: The Cardiovascular Health Study. Arch Neurol 1998;55:1217-25.  Back to cited text no. 13
    
14.
Liao D, Cooper L, Cai J, Toole J, Bryan N, Burke G, et al. The prevalence and severity of white matter lesions, their relationship with age, ethnicity, gender, and cardiovascular disease risk factors: The ARIC Study. Neuroepidemiology 1997;16:149-62.  Back to cited text no. 14
    
15.
Longstreth WT Jr, Manolio TA, Arnold A, Burke GL, Bryan N, Jungreis CA, et al. Clinical correlates of white matter findings on cranial magnetic resonance imaging of 3301 elderly people. The Cardiovascular Health Study. Stroke 1996;27:1274-82.  Back to cited text no. 15
    
16.
Jeerakathil T, Wolf PA, Beiser A, Massaro J, Seshadri S, D'Agostino RB, et al. Stroke risk profile predicts white matter hyperintensity volume: The Framingham Study. Stroke 2004;35:1857-61.  Back to cited text no. 16
    
17.
Dufouil C, de Kersaint-Gilly A, Besançon V, Levy C, Auffray E, Brunnereau L, et al. Longitudinal study of blood pressure and white matter hyperintensities: The EVA MRI Cohort. Neurology 2001;56:921-6.  Back to cited text no. 17
    
18.
Martini SR, Flaherty ML, Brown WM, Haverbusch M, Comeau ME, Sauerbeck LR, et al. Risk factors for intracerebral hemorrhage differ according to hemorrhage location. Neurology 2012;79:2275-82.  Back to cited text no. 18
    
19.
Howard G, Cushman M, Howard VJ, Kissela BM, Kleindorfer DO, Moy CS, et al. Risk factors for intracerebral hemorrhage: The REasons for geographic and racial differences in stroke (REGARDS) study. Stroke 2013;44:1282-7.  Back to cited text no. 19
    
20.
Ylikoski A, Erkinjuntti T, Raininko R, Sarna S, Sulkava R, Tilvis R. White matter hyperintensities on MRI in the neurologically nondiseased elderly. Analysis of cohorts of consecutive subjects aged 55 to 85 years living at home. Stroke 1995;26:1171-7.  Back to cited text no. 20
    
21.
Devan WJ, Falcone GJ, Anderson CD, Jagiella JM, Schmidt H, Hansen BM, et al. Heritability estimates identify a substantial genetic contribution to risk and outcome of intracerebral hemorrhage. Stroke 2013;44:1578-83.  Back to cited text no. 21
    
22.
Atwood LD, Wolf PA, Heard-Costa NL, Massaro JM, Beiser A, D'Agostino RB, et al. Genetic variation in white matter hyperintensity volume in the Framingham Study. Stroke 2004;35:1609-13.  Back to cited text no. 22
    
23.
Carmelli D, DeCarli C, Swan GE, Jack LM, Reed T, Wolf PA, et al. Evidence for genetic variance in white matter hyperintensity volume in normal elderly male twins. Stroke 1998;29:1177-81.  Back to cited text no. 23
    
24.
Turner ST, Jack CR, Fornage M, Mosley TH, Boerwinkle E, de Andrade M. Heritability of leukoaraiosis in hypertensive sibships. Hypertension 2004;43:483-7.  Back to cited text no. 24
    
25.
Arauz A, Murillo L, Cantu C, Barinagarrementeria F, Higuera J. Prospective study of single and multiple lacunar infarcts using magnetic resonance imaging: Risk factors, recurrence, and outcome in 175 consecutive cases. Stroke 2003;34:2453-8.  Back to cited text no. 25
    
26.
Nah HW, Kang DW, Kwon SU, Kim JS. Diversity of single small subcortical infarctions according to infarct location and parent artery disease: Analysis of indicators for small vessel disease and atherosclerosis. Stroke 2010;41:2822-7.  Back to cited text no. 26
    
27.
Caplan LR. Lacunar infarction and small vessel disease: Pathology and pathophysiology. J Stroke 2015;17:2-6.  Back to cited text no. 27
    
28.
Park YS, Chung PW, Kim YB, Moon HS, Suh BC, Yoon WT, et al. Small deep infarction in patients with atrial fibrillation: Evidence of lacunar pathogenesis. Cerebrovasc Dis 2013;36:205-10.  Back to cited text no. 28
    
29.
Bevan S, Traylor M, Adib-Samii P, Malik R, Paul NL, Jackson C, et al. Genetic heritability of ischemic stroke and the contribution of previously reported candidate gene and Genome-Wide Associations. Stroke 2012;43:3161-7.  Back to cited text no. 29
    
30.
Prabhakaran S, Wright CB, Yoshita M, Delapaz R, Brown T, DeCarli C, et al. Prevalence and determinants of subclinical brain infarction: The Northern Manhattan Study. Neurology 2008;70:425-30.  Back to cited text no. 30
    
31.
Loos CM, Staals J, Wardlaw JM, van Oostenbrugge RJ. Cavitation of deep lacunar infarcts in patients with first-ever lacunar stroke: A 2-year follow-up study with MR. Stroke 2012;43:2245-7.  Back to cited text no. 31
    
32.
Zhu XL, Chan MS, Poon WS. Spontaneous intracranial hemorrhage: Which patients need diagnostic cerebral angiography? A prospective study of 206 cases and review of the literature. Stroke 1997;28:1406-9.  Back to cited text no. 32
    
33.
Woo D, Broderick JP. Spontaneous intracerebral hemorrhage: Epidemiology and clinical presentation. Neurosurg Clin N Am 2002;13:265-79, v.  Back to cited text no. 33
    
34.
Zahuranec DB, Lisabeth LD, Sanchez BN, Smith MA, Brown DL, Garcia NM, et al. Intracerebral hemorrhage mortality is not changing despite declining incidence. Neurology 2014;82:2180-6.  Back to cited text no. 34
    
35.
Woo D, Rosand J, Kidwell C, McCauley JL, Osborne J, Brown MW, et al. The ethnic/racial variations of intracerebral hemorrhage (ERICH) study protocol. Stroke 2013;44:e120-5.  Back to cited text no. 35
    
36.
Dichgans M, Markus HS. Genetic association studies in stroke: Methodological issues and proposed standard criteria. Stroke 2005;36:2027-31.  Back to cited text no. 36
    
37.
Manolio TA, Kronmal RA, Burke GL, Poirier V, O'Leary DH, Gardin JM, et al. Magnetic resonance abnormalities and cardiovascular disease in older adults. The Cardiovascular Health Study. Stroke 1994;25:318-27.  Back to cited text no. 37
    
38.
Lindgren A, Roijer A, Rudling O, Norrving B, Larsson EM, Eskilsson J, et al. Cerebral lesions on magnetic resonance imaging, heart disease, and vascular risk factors in subjects without stroke. A population-based study. Stroke 1994;25:929-34.  Back to cited text no. 38
    
39.
Rost NS, Sadaghiani S, Biffi A, Fitzpatrick KM, Cloonan L, Rosand J, et al. Setting a gold standard for quantification of leukoaraiosis burden in patients with ischemic stroke: The Atherosclerosis Risk in Communities Study. J Neurosci Methods 2014;221:196-201.  Back to cited text no. 39
    
40.
Tiehuis AM, Vincken KL, Mali WP, Kappelle LJ, Anbeek P, Algra A, et al. Automated and visual scoring methods of cerebral white matter hyperintensities: Relation with age and cognitive function. Cerebrovasc Dis 2008;25:59-66.  Back to cited text no. 40
    
41.
Wardlaw JM, Brindle W, Casado AM, Shuler K, Henderson M, Thomas B, et al. SINAPSE Collaborative Group. A systematic review of the utility of 1.5 versus 3 Tesla magnetic resonance brain imaging in clinical practice and research. Eur Radiol 2012;22:2295-303.  Back to cited text no. 41
    
42.
Cho AH, Kim HR, Kim W, Yang DW. White matter hyperintensity in ischemic stroke patients: It may regress over time. J Stroke 2015;17:60-6.  Back to cited text no. 42
    
43.
Kanekar S, Devgun P. A pattern approach to focal white matter hyperintensities on magnetic resonance imaging. Radiol Clin North Am 2014;52:241-61.  Back to cited text no. 43
    
44.
Wiegman AF, Meier IB, Schupf N, Manly JJ, Guzman VA, Narkhede A, et al. Cerebral microbleeds in a multiethnic elderly community: Demographic and clinical correlates. J Neurol Sci 2014;345:125-30.  Back to cited text no. 44
    
45.
Kato H, Izumiyama M, Izumiyama K, Takahashi A, Itoyama Y. Silent cerebral microbleeds on T2*-weighted MRI: Correlation with stroke subtype, stroke recurrence, and leukoaraiosis. Stroke 2002;33:1536-40.  Back to cited text no. 45
    
46.
Vernooij MW, van der Lugt A, Ikram MA, Wielopolski PA, Niessen WJ, Hofman A, et al. Prevalence and risk factors of cerebral microbleeds: The Rotterdam Scan Study. Neurology 2008;70:1208-14.  Back to cited text no. 46
    
47.
Smith EE, Nandigam KR, Chen YW, Jeng J, Salat D, Halpin A, et al. MRI markers of small vessel disease in lobar and deep hemispheric intracerebral hemorrhage. Stroke 2010;41:1933-8.  Back to cited text no. 47
    
48.
Poels MM, Zaccai K, Verwoert GC, Vernooij MW, Hofman A, van der Lugt A, et al. Arterial stiffness and cerebral small vessel disease: The Rotterdam Scan Study. Stroke 2012;43:2637-42.  Back to cited text no. 48
    
49.
Sandeman EM, Hernandez Mdel C, Morris Z, Bastin ME, Murray C, Gow AJ, et al. Incidental findings on brain MR imaging in older community-dwelling subjects are common but serious medical consequences are rare: A cohort study. PloS One 2013;8:e71467.  Back to cited text no. 49
    
50.
Zhou Q, Yang D, Ombrello AK, Zavialov AV, Toro C, Zavialov AV, et al. Early-onset stroke and vasculopathy associated with mutations in ADA2. N Engl J Med 2014;370:911-20.  Back to cited text no. 50
    
51.
Schmidt H, Zeginigg M, Wiltgen M, Freudenberger P, Petrovic K, Cavalieri M, et al. CHARGE consortium Neurology working group. Genetic variants of the NOTCH3 gene in the elderly and magnetic resonance imaging correlates of age-related cerebral small vessel disease. Brain 2011;134:3384-97.  Back to cited text no. 51
    
52.
Baptista MV, Ferreira S, Pinho-E-Melo T, Carvalho M, Cruz VT, Carmona C, et al. Mutations of the GLA gene in young patients with stroke: The PORTYSTROKE study--screening genetic conditions in Portuguese young stroke patients. Stroke 2010;41:431-6.  Back to cited text no. 52
    
53.
Brouns R, Thijs V, Eyskens F, Van den Broeck M, Belachew S, Van Broeckhoven C, et al. BeFaS Investigators. Belgian Fabry study: Prevalence of Fabry disease in a cohort of 1000 young patients with cerebrovascular disease. Stroke 2010;41:863-8.  Back to cited text no. 53
    
54.
Nakamura K, Sekijima Y, Nakamura K, Hattori K, Nagamatsu K, Shimizu Y, et al. p.E66Q mutation in the GLA gene is associated with a high risk of cerebral small-vessel occlusion in elderly Japanese males. Eur J Neurol 2014;21:49-56.  Back to cited text no. 54
    
55.
Rannikmäe K, Davies G, Thomson PA, Bevan S, Devan WJ, Falcone GJ, et al. METASTROKE Consortium; CHARGE WMH Group; ISGC ICH GWAS Study Collaboration; WMH in Ischemic Stroke GWAS Study Collaboration; International Stroke Genetics Consortium. Common variation in COL4A1/COL4A2 is associated with sporadic cerebral small vessel disease. Neurology 2015;84:918-26.  Back to cited text no. 55
    
56.
Weng YC, Sonni A, Labelle-Dumais C, de Leau M, Kauffman WB, Jeanne M, et al. COL4A1 mutations in patients with sporadic late-onset intracerebral hemorrhage. Ann Neurol 2012;71:470-7.  Back to cited text no. 56
    
57.
Pelzer N, de Vries B, Boon EM, Kruit MC, Haan J, Ferrari MD, et al. Heterozygous TREX1 mutations in early-onset cerebrovascular disease. J Neurol 2013;260:2188-90.  Back to cited text no. 57
    
58.
Paternoster L, Chen W, Sudlow CL. Genetic determinants of white matter hyperintensities on brain scans: A systematic assessment of 19 candidate gene polymorphisms in 46 studies in 19,000 subjects. Stroke 2009;40:2020-6.  Back to cited text no. 58
    
59.
Zhang Z, Xu G, Liu D, Fan X, Zhu W, Liu X. Angiotensin-converting enzyme insertion/deletion polymorphism contributes to ischemic stroke risk: A meta-analysis of 50 case-control studies. PloS One 2012;7:e46495.  Back to cited text no. 59
    
60.
Sun Y, Liu Y, Watts LT, Sun Q, Zhong Z, Yang GY, et al. Genetic Associations of Angiotensin-converting enzyme with primary intracerebral hemorrhage: A meta-analysis. PloS One 2013;8:e67402.  Back to cited text no. 60
    
61.
Cotlarciuc I, Malik R, Holliday EG, Ahmadi KR, Paré G, Psaty BM, et al. METASTROKE and the InternationalStroke Genetics Consortium. Effect of genetic variants associated with plasma homocysteine levels on stroke risk. Stroke 2014;45:1920-4.  Back to cited text no. 61
    
62.
Gao S, Li H, Xiao H, Yao G, Shi Y, Wang Y, et al. Association of MTHFR 677T variant allele with risk of intracerebral haemorrhage: A meta-analysis. J Neurol Sci 2012;323:40-5.  Back to cited text no. 62
    
63.
Wu L, Shen Y, Liu X, Ma X, Xi B, Mi J, et al. The 1425G/A SNP in PRKCH is associated with ischemic stroke and cerebral hemorrhage in a Chinese population. Stroke 2009;40:2973-6.  Back to cited text no. 63
    
64.
Biffi A, Sonni A, Anderson CD, Kissela B, Jagiella JM, Schmidt H, et al. International Stroke Genetics Consortium. Variants at APOE influence risk of deep and lobar intracerebral hemorrhage. Ann Neurol 2010;68:934-43.  Back to cited text no. 64
    
65.
Musunuru K, Hickey KT, Al-Khatib SM, Delles C, Fornage M, Fox CS, et al. American Heart Association Council on Functional Genomics and Translational Biology, Council on Clinical Cardiology, Council on Cardiovascular Disease in the Young, Council on Cardiovascular and Stroke Nursing, Council on Epidemiology and Prevention, Council on Hypertension, Council on Lifestyle and Cardiometabolic Health, Council on Quality of Care and Outcomes Research, and Stroke Council. Basic concepts and potential applications of genetics and genomics for cardiovascular and stroke clinicians: A scientific statement from the American Heart Association. Circ Cardiovasc Genet 2015;8:216-42.  Back to cited text no. 65
    
66.
Ward LD, Kellis M. Interpreting noncoding genetic variation in complex traits and human disease. Nat Biotechnol 2012;30:1095-106.  Back to cited text no. 66
    
67.
Verhaaren BF, Debette S, Bis JC, Smith JA, Ikram MK, Adams HH, et al. Multiethnic Genome-Wide Association Study of cerebral white matter hyperintensities on MRI. Circ Cardiovasc Genet 2015;8:398-409.  Back to cited text no. 67
    
68.
Woo D, Falcone GJ, Devan WJ, Brown WM, Biffi A, Howard TD, et al. International Stroke Genetics Consortium. Meta-analysis of Genome-Wide Association Studies identifies 1q22 as a susceptibility locus for intracerebral hemorrhage. Am J Hum Genet 2014;94:511-21.  Back to cited text no. 68
    
69.
Fornage M, Debette S, Bis JC, Schmidt H, Ikram MA, Dufouil C, et al. Genome-Wide Association Studies of cerebral white matter lesion burden: The CHARGE consortium. Ann Neurol 2011;69:928-39.  Back to cited text no. 69
    
70.
Adib-Samii P, Rost N, Traylor M, Devan W, Biffi A, Lanfranconi S, et al. International Stroke Genetics Consortium. 17q25 Locus is associated with white matter hyperintensity volume in ischemic stroke, but not with lacunar stroke status. Stroke 2013;44:1609-15.  Back to cited text no. 70
    
71.
Verhaaren BF, de Boer R, Vernooij MW, Rivadeneira F, Uitterlinden AG, Hofman A, et al. Replication study of chr17q25 with cerebral white matter lesion volume. Stroke 2011;42:3297-9.  Back to cited text no. 71
    
72.
French CR, Seshadri S, Destefano AL, Fornage M, Arnold CR, Gage PJ, et al. Mutation of FOXC1 and PITX2 induces cerebral small-vessel disease. J Clin Invest 2014;124:4877-81.  Back to cited text no. 72
    
73.
Traylor M, Farrall M, Holliday EG, Sudlow C, Hopewell JC, Cheng YC, et al. International Stroke Genetics Consortium. Genetic risk factors for ischaemic stroke and its subtypes (the METASTROKE collaboration): A meta-analysis of Genome-Wide Association Studies. Lancet Neurol 2012;11:951-62.  Back to cited text no. 73
    
74.
Lee S, Abecasis GR, Boehnke M, Lin X. Rare-variant association analysis: Study designs and statistical tests. Am J Hum Genet 2014;95:5-23.  Back to cited text no. 74
    
75.
Cole JW, Stine OC, Liu X, Pratap A, Cheng Y, Tallon LJ, et al. Rare variants in ischemic stroke: An exome pilot study. PloS One 2012;7:e35591.  Back to cited text no. 75
    
76.
Kochunov P, Jahanshad N, Marcus D, Winkler A, Sprooten E, Nichols TE, et al. Heritability of fractional anisotropy in human white matter: A comparison of Human Connectome Project and ENIGMA-DTI data. Neuroimage 2015;111:300-11.  Back to cited text no. 76
    
77.
Xu C, Ciampi A, Greenwood CM; UK10K Consortium. Exploring the potential benefits of stratified false discovery rates for region-based testing of association with rare genetic variation. Front Genet 2014;5:11.  Back to cited text no. 77
    
78.
Wiszniewski W, Hunter JV, Hanchard NA, Willer JR, Shaw C, Tian Q, et al. TM4SF20 ancestral deletion and susceptibility to a pediatric disorder of early language delay and cerebral white matter hyperintensities. Am J Hum Genet 2013;93:197-210.  Back to cited text no. 78
    
79.
Svetkey LP, Timmons PZ, Emovon O, Anderson NB, Preis L, Chen YT. Association of hypertension with beta2- and alpha2c10-adrenergic receptor genotype. Hypertension 1996;27:1210-5.  Back to cited text no. 79
    
80.
Lockette W, Ghosh S, Farrow S, MacKenzie S, Baker S, Miles P, et al. Alpha 2-adrenergic receptor gene polymorphism and hypertension in blacks. Am J Hypertens 1995;8:390-4.  Back to cited text no. 80
    
81.
Kim JO, Jeon YJ, Kim OJ, Oh SH, Kim HS, Shin BS, et al. Association between common genetic variants of α2A-, α2B- and α2C-adrenoceptors and the risk of silent brain infarction. Mol Med Rep 2014;9:2459-66.  Back to cited text no. 81
    
82.
Falcone GJ, Biffi A, Devan WJ, Jagiella JM, Schmidt H, Kissela B, et al. International Stroke Genetics Consortium. Burden of risk alleles for hypertension increases risk of intracerebral hemorrhage. Stroke 2012;43:2877-83.  Back to cited text no. 82
    
83.
GTEx Consortium. The Genotype-Tissue Expression (GTEx) project. Nat Genet 2013;45:580-5.  Back to cited text no. 83
    
84.
Williams SR, Yang Q, Chen F, Liu X, Keene KL, Jacques P, et al. Genomics and Randomized Trials Network;Framingham Heart Study. Genome-wide meta-analysis of homocysteine and methionine metabolism identifies five one carbon metabolism loci and a novel association of ALDH1L1 with ischemic stroke. PLoS Genet 2014;10:e1004214.  Back to cited text no. 84
    
85.
Shah SH, Kraus WE, Newgard CB. Metabolomic profiling for the identification of novel biomarkers and mechanisms related to common cardiovascular diseases: Form and function. Circulation 2012;126:1110-20.  Back to cited text no. 85
    
86.
Dye BR, Hill DR, Ferguson MA, Tsai YH, Nagy MS, Dyal R, et al. In vitro generation of human pluripotent stem cell derived lung organoids. Elife 2015;4.  Back to cited text no. 86
    
87.
Spence JR, Mayhew CN, Rankin SA, Kuhar MF, Vallance JE, Tolle K, et al. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature 2011;470:105-9.  Back to cited text no. 87
    
88.
Bailey EL, Smith C, Sudlow CL, Wardlaw JM. Is the spontaneously hypertensive stroke prone rat a pertinent model of sub cortical ischemic stroke? A systematic review. Int J Stroke 2011;6:434-44.  Back to cited text no. 88
    
89.
Hsu PD, Lander ES, Zhang F. Development and applications of CRISPR-Cas9 for genome engineering. Cell 2014;157:1262-78.  Back to cited text no. 89
    


    Figures

  [Figure 1], [Figure 2], [Figure 3], [Figure 4]
 
 
    Tables

  [Table 1], [Table 2], [Table 3], [Table 4]


This article has been cited by
1 Heavy Metal-Induced Cerebral Small Vessel Disease: Insights into Molecular Mechanisms and Possible Reversal Strategies
Jayant Patwa,Swaran Jeet Singh Flora
International Journal of Molecular Sciences. 2020; 21(11): 3862
[Pubmed] | [DOI]
2 Genetic Factors of Cerebral Small Vessel Disease and Their Potential Clinical Outcome
Vo Van Giau,Eva Bagyinszky,Young Chul Youn,Seong Soo A. An,Sang Yun Kim
International Journal of Molecular Sciences. 2019; 20(17): 4298
[Pubmed] | [DOI]
3 Risk of hemorrhagic transformation after ischemic stroke in patients with antiphospholipid antibody syndrome
Tapan Mehta,Mohammed Hussain,Khushboo Sheth,Yuchuan Ding,Louise D. McCullough
Neurological Research. 2017; 39(6): 477
[Pubmed] | [DOI]
4 Targeting G protein-coupled receptor for pain management
Hongyan Li, Rong Wang, Yinying Lu, Xuehua Xu, Jiaxiang Ni
Brain Circulation. 2017; 3(2): 109
[Pubmed] | [DOI]



 

Top
 
 
  Search
 
    Similar in PUBMED
   Search Pubmed for
   Search in Google Scholar for
 Related articles
    Access Statistics
    Email Alert *
    Add to My List *
* Registration required (free)  

 
  In this article
Abstract
Introduction
Reconceptualizin...
Sequencing and R...
Population-Speci...
Following Up Reg...
Future Directions
References
Article Figures
Article Tables

 Article Access Statistics
    Viewed6005    
    Printed255    
    Emailed0    
    PDF Downloaded656    
    Comments [Add]    
    Cited by others 4    

Recommend this journal