Next Article in Journal
The ZT Biopolymer: A Self-Assembling Protein Scaffold for Stem Cell Applications
Next Article in Special Issue
Microbiome Influence in the Pathogenesis of Prion and Alzheimer’s Diseases
Previous Article in Journal
NOD2 Supports Crypt Survival and Epithelial Regeneration after Radiation-Induced Injury
Previous Article in Special Issue
Emerging Role of Genetic Alterations Affecting Exosome Biology in Neurodegenerative Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genetic Factors of Cerebral Small Vessel Disease and Their Potential Clinical Outcome

1
Department of Bionano Technology & Gachon Bionano Research Institute, Gachon University, Seongnam-si, Gyeonggi-do 461-701, Korea
2
Department of Neurology, Chung-Ang University College of Medicine, Seoul 06973, Korea
3
Department of Neurology, Seoul National University College of Medicine & Neurocognitive Behavior Center, Seoul National University Bundang Hospital, Seoul 06973, Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2019, 20(17), 4298; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20174298
Submission received: 7 August 2019 / Revised: 27 August 2019 / Accepted: 1 September 2019 / Published: 3 September 2019

Abstract

:
Cerebral small vessel diseases (SVD) have been causally correlated with ischemic strokes, leading to cognitive decline and vascular dementia. Neuroimaging and molecular genetic tests could improve diagnostic accuracy in patients with potential SVD. Several types of monogenic, hereditary cerebral SVD have been identified: cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL), cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), cathepsin A-related arteriopathy with strokes and leukoencephalopathy (CARASAL), hereditary diffuse leukoencephalopathy with spheroids (HDLS), COL4A1/2-related disorders, and Fabry disease. These disorders can be distinguished based on their genetics, pathological and imaging findings, clinical manifestation, and diagnosis. Genetic studies of sporadic cerebral SVD have demonstrated a high degree of heritability, particularly among patients with young-onset stroke. Common genetic variants in monogenic disease may contribute to pathological progress in several cerebral SVD subtypes, revealing distinct genetic mechanisms in different subtype of SVD. Hence, genetic molecular analysis should be used as the final gold standard of diagnosis. The purpose of this review was to summarize the recent discoveries made surrounding the genetics of cerebral SVD and their clinical significance, to provide new insights into the pathogenesis of cerebral SVD, and to highlight the possible convergence of disease mechanisms in monogenic and sporadic cerebral SVD.

Graphical Abstract

1. Introduction

Cerebral small vessel diseases (SVD) has been recognized as an important cause of cognitive impairment and dementia among the elderly [1,2,3]. The pathologic correlation of cerebral dysfunctions with stroke was stronger in the population in Asia than in the USA or Europe [1]. Cerebral SVD accounts for 15–26% of ischemic strokes in the USA and Europe [4,5,6,7], whereas this proportion in Asia ranges from 25% to 54% [8,9,10,11,12]. However, these observations were made only in hospital-based settings, and the studies were performed in non-Asian populations [13,14]. Cerebral SVD has an enormous social and economic impact. Currently, the causes of the majority of cerebral SVD are not well understood, which could limit effective treatment. Several traditional risk factors have been suggested to play important roles in the mechanisms and etiology of cerebral SVD [15]. In addition, a comparative investigation of vascular risk factors, as well as genetic and environmental susceptibility has suggested that ethnic differences could contribute to the prevalence of cerebral SVD. Generally, cerebral SVD subtypes may present similar pathogenesis, comprising lacunar stroke, hypertensive hemorrhage, leukoaraiosis, or cerebral microbleeds [16,17,18], which indicate a possible overlapping etiology with extracerebral small vessel vasculopathies [19,20,21,22]. The standard markers of SVD included lacunes, cerebral microbleeds, or white matter hyperintensities (WMHs). These markers, particularly in advanced structural neuroimaging magnetic resonance imaging (MRI) techniques, were used to assess the accumulated burden of cerebrovascular disease throughout life. However, the variations in the definitions and terms of descriptive features may cause difficulties with respect to the interpretation and comparison of results between studies. Until recently, the molecular, cellular, and pathophysiologic mechanisms underlying SVD were largely unknown. Mechanistic studies were hampered by a lack of animal models, difficulties in visualizing small blood vessels in vivo, particularly with technological challenges in making an animal model of brain microvessels for the physiologic and biochemical studies. Figure 1 summarized the pathogenesis of cerebral SVD manifestations. Although rare, these phenotypic extremes shared both clinical and radiological features with sporadic SVD, which could provide important insights into the mechanisms of the disease. Thus, diagnostic and therapeutic strategies are still limited [1,2,3].
Genetic disorders could be based on the combination of mutations in a single gene or on a cluster of genes with an autosomal dominant or recessive pattern [2,23,24]. Interestingly, particular traits suggested recessive patterns by receiving chromosomes from both parents, where different mutations of identical genes were responsible [21]. Similar to ischemic stroke, genetic factors revealed their significant impact on cerebral SVD, which could clarify the pathophysiology of sporadic cerebral SVD. The estimated heritability for cerebral white matter lesions as a surrogate marker of cerebral SVD ranged between 50% and 80% [25]. Since the human genome project in 2003, extraordinary progress has been made in genome sequencing technologies, genome-wide association studies (GWAS), and the meta-analysis of individual candidate gene studies. These advancements have contributed to a significant increase in the number of, and in-depth information available on the genetic loci for sporadic cerebral SVD [1]. Since the occurrences of cerebral SVD would increase in all age groups, in-depth investigations on the molecular mechanisms of the pathogenesis of cerebral SVD were needed [26]. Although the majority of cerebral SVD cases were sporadic, several reports of inherited forms of cerebral SVD pointed to single-gene disorders. Different monogenic cerebral SVD were discovered (Table 1), including cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) [27], cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL) [28,29], cathepsin A–related arteriopathy with strokes and leukoencephalopathy (CARASAL) [30], hereditary diffuse leukoencephalopathy with spheroids (HDLS) [31], COL4A1/2-related cerebral SVD [27,32,33], autosomal-dominant retinal vasculopathy with cerebral leukodystrophy, and Fabry disease. Thus, the recognition of the genetic aspect of cerebral SVD could contribute to the improved diagnosis and treatment of these rare single-gene disorders, as well as sporadic cerebral SVD.
The clinical features of autosomal-dominant SVD may share several similarities with sporadic SVD, such as arteriopathy, white matter dysfunctions, and infracts in subcortical regions [1,34,35]. These diseases could have wide a wide range of age of onset—some patients develop SVD at a younger age (under 20 years), while in other affected individuals the disease phenotypes could present after age 50 or 60 [26,35]. Genetics could play a significant role in the onset of SVD. Thus, genetic testing as part of routine diagnosis could identify the potential causative mutations in patients with a family history, suggesting possible connections between gene mutations and clinical symptoms of blood vessel diseases. The present work aimed to discuss the genetic background of the various forms of inherited cerebral SVD and the recent advances made in the genetic study of cerebral SVD pathogenesis.

2. Cerebral Autosomal-Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL)

CADASIL (OMIM 125310) is the most common hereditary stroke disorder, and it was also suggested as the most common inherited form of vascular dementia. Clinical symptoms appear at younger ages or during the adulthood. Common symptoms at the initial stage of disorder were stroke, migraine (with or without aura), or transient ischemic attack. Additional symptoms include cognitive impairment, dementia, mood disturbances or seizures [36]. CADASIL was associated with non-amyloid and/or non-atherosclerotic angiopathy in the brain vessels (small penetrating and lepto-meningeal arteries). The disease also affected the vessels in other organs, such as the muscles, skin, or heart [37]. One of the causes in ischemic brain injury was microangiopathy with granular osmiophilic deposits (GOM) in the basal membrane. The diagnosis of GOM could be confirmed with 100% accuracy using magnetic resonance imaging (MRI). Recently, skin biopsies were suggested [38]. MRI and computed tomography scan (CT) could reveal the hyperintensities and hypodensities in the white matter, respectively. Additionally, lacunar infarcts could be seen in different parts of the brain, such as the semioval center, thalamus, and basal ganglia. The third type of lesions were the cerebral microbleeds (CMBs) [39]. Affected brain areas were the thalamus, central semivovale, basal ganglia, temporal lobes or pons [37,40]. The imaging data of a typical CADASIL (A1), the atypical form of CADASIL with (A2) and without genetic mutations (A3), were summarized in Figure 2. Typical CADASIL manifests extensive white matter hyperintensities (WMHs) in different regions, which were less prominent in the atypical form. Patients with atypical CADASIL had stronger microstructural alterations in the bilateral frontal and temporal lobes and corpus callosum. The manifestation of similar symptoms and/or findings in relatives would strongly suggest familial CADASIL. Davous first suggested the diagnostic criteria for CADASIL in 1998 [41]. The clinical diagnosis was usually made on the basis of a combination of otherwise unexplained cerebral ischemic events or cognitive impairment, brain MRI abnormalities [42,43], and a family history of stroke or dementia [44].
Genetically, the missense mutations of cysteine-altering in NOTCH3 gene on chromosome 19 [27] with 33 exons were responsible for CADASIL. The NOTCH3 gene encoded for the 2321-amino-acid-long single-pass transmembrane receptor protein, one of the key moleculea of notch signaling, particularly during embryonal development [45]. In adults, NOTCH3 could be essential for the development, remodeling and differentiation of vascular system. A recent study revealed that patients with typical CADASIL with NOTCH3 variants showed distinct anatomic vulnerabilities in both grey and white matter structures [40] (Figure 2).
The extracellular domain of NOTCH3 (NOTCH3ECD) consisted of 34 epidermal growths factor-like repeats (EGFr). These EGFrs subunits of approximately 40 amino acids contained three disulfide bridges in their positions, stabilizing their domain structures. The unpaired cysteine residue from the mutation in NOTCH3ECD disrupted the disulfide bridge formation, and the shuffling and mismatched disulfide bridges would result in increased instability and multimerization properties in comparison to those in the wild-type NOTCH3ECD [46], destroying the NOTCH3 signaling cascade. Hence, these multimerized toxic aggregations of NOTCH3ECD were found in and around vascular smooth muscle cells (VSMCs) of small-to-medium-sized arteries, particularly in the brain. In addition, the multimeric depositions, which were also found in arteries of other organs and tissues [47,48]. The NOTCH3ECD aggregation could also reflect the pathological changes in the vessel wall, including the degeneration of VSMCs and the deposition of visualized granular osmiophilic materials by electron microscopy [49].
The majority of pathogenic mutations in NOTCH3 were missense mutations with rare deletions and insertions in one of 34 EGFr domains [50]. Although various NOTCH3 mutations have been reported in different populations, all CADASIL mutations occurred in exons 2–24, resulting in a gain or loss of cysteine residues in the extracellular N-terminal region [51], creating an uneven number of cysteine—either five or seven—which commonly induced CADASIL. However, several reports have described non-cysteine related mutations [52], referring to these as non-classical NOTCH3 mutations of CADASIL-like GOM-negative familial phenotypes with vascular dementia. Thus, clinical examination may raise suspicions for CADASIL, and the diagnosis should be confirmed by genetic testing, or by assessing for the detailed pathological characteristics of the disease (Table 2).
The recommendations for clinical diagnosis and interpretation of NOTCH3 (Notch homolog 3, Drosophila) mutations in CADASIL were suggested by Rutten et al. [44]. In general, a genetic test presenting an archetypal CADASIL mutation [27] of NOTCH3 gene would confirm the diagnosis [53]. If a mutation was found previously in an affected relative, the targeted molecular genetic testing could be performed for the particular loci. Otherwise, the whole gene or exons 2–24 (which harbor the majority of the mutations) should be sequenced [50]. The correct diagnostic interpretation of variants other than the stereotypical cysteine-altering missense mutations would require an expert opinion based on both the clinical features and distinguishing molecular aspects of CADASIL [44]. A recent study suggested that a single-particle in vitro aggregation assay might be a reliable tool to evaluate the clinical significance of the non-cysteine variants [54]. However, the study included only one family, and it was still debated whether the diagnosis was conclusive for selecting the method as diagnostic tool [44,54]. To date, more than 130 different mutations in NOTCH3 gene were reported in patients with CADASIL, 95% of which were missense point mutations [51]. CADASIL is most commonly a hereditary autosomal-dominant disease affecting all small cerebral arteries. In addition, recent studies of five patients with homozygous mutations (p.Arg133Cys, p.Arg578Cys, p.Gly528Cys, p.Arg544Cys, and p.Cys183Ser) supported their pathological effects [55,56,57,58,59]. Interestingly, several cases of de novo mutations were also found [47,60,61].
Five small deletions (four in non-frame [47,62,63] and one frameshift), one splice site mutation [64], and a small deletion of a non-cysteine related residue were reported [52]. Further, p.Ala1020Pro, p.Arg213Lys, p.Tyr1098Ser, and p.Arg75Pro variants were found in diagnosed patients with CADASIL (Table 2) [65,66,67,68,69,70,71,72,73,74,75,76,77,78], in whom exons 2–24 were sequenced from skin biopsy for confirmation [79,80,81,82]. Recently, a three-nucleotide insertion was reported as the first pathogenic insertion in the NOTCH3 gene [83]. However, the significance of such molecular variants still remains unclear. Therefore, complete screening of Notch3-coding exons should be performed in suspected cases for understanding the CADASIL phenotype and genotype spectra.

3. Cerebral Autosomal Recessive Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CARASIL)

A variety of rare genetic disorders may have symptoms similar to those found in CADASIL [67]. CARASIL was the second known form of ischemic, non-hypertensive, cerebral SVD with an identified gene, HTRA1 [28,86,87,88]. Patients would develop several phenotypes in the early lifetime (teenage years or 20–30 years old), such as lumbago, alopecia or even encephalopathy [72,73]. Additional symptoms may occur in patients older than 30 or 40 years of age, such as gait disturbances, premature scalp alopecia, ischemic stroke, acute mid- to lower-back pain, pseudobulbar palsy, pyramidal-or extrapyramidal symptoms, Babinski signs and progressive cognitive disturbances, leading to severe dementia [73,84]. Brain CT imaging revealed diffuse homogenous changes in white matter and dilatation in different areas, such as cerebral sulci. MRI imaging revealed WMH lesions, which could be the common characteristics of CARASIL [72,84,88]. Additional signs included multiple lacunar infarctions or cerebral angiography. Lesions could initially appear in the subcortical deep white matter, but later they appear in other brain areas such as the thalamus, cerebellum or brainstem [87,88]. Anterior temporal lobes and external capsules were affected, suggesting that CARASIL and CADASIL may share phenotypical similarities [87,89]. Unlike CADASIL, CARASIL may demonstrate no pathognomonic histological features; the histological findings most closely resemble those of nonhereditary ischemic cerebral SVD, or “earthen pipe phenomenon” [90]. A fibrous proliferation of the intima, hyaline degeneration of the media, loss of VSMCs, and thickening and fragmentation of the internal elastic lamina were observed in patients with CARASIL [88]. Granular appearance of GOMs were never observed in CARASIL [88,89]. These findings were limited to the cerebral small arteries—a skin biopsy was not helpful for the diagnosis [86,87].
CARASIL is a rare genetic disorder characterized by mutations in the HTRA1 gene located on chromosome 10q (10q25.3-q26.2). It is a very rare autosomal recessive form of familial SVD. CARASIL was identified to have an autosomal recessive inheritance pattern. Patients with mutations tend to produce protein products with a low protease activity; these proteins were unable to repress signaling by transforming growth factor-β (TGF-β) [28]. Approximately 56 cases have been reported: 48 from Japan, eight from China [91,92,93], and one case each from Spain [94], Romania [95], Turkey [96], and America [97]. These cases are summarized in Table 3. The mutation could result in the loss of HTRA1 protease activity, leading to cerebral small-vessel arteriopathy. Recently, heterozygous HTRA1 mutations have been described in patients with late-onset familial SVD [29,85,93,98,99,100]. It has also been speculated that the upregulation of TGF-β family signaling by the mutant HTRA1 may be responsible for the non-neurological manifestations of CARASIL, for example, alopecia and degenerative spine disease [28].
To date, at least 16 mutations in the HTRA1 gene have been identified in 25 families. These mutations were mostly located in exons 3–6 and may result in a decreased level of protease activity, leading to an increase in TGF-β signaling [101], which in turn could cause degeneration of smooth muscle cells in the cerebral small vessels and angiopathy. The majority of missense mutations have been reported among Japanese patients; in particular, four pathogenic homozygous mutations including two missense (p.Ala252Thr; p.Val297Met) and two nonsense (p.Arg302X; p.Arg370X) mutations were discovered in this cohort. These mutations resulted in reduction of the protease activity by 21–50%, which was not enough to repress by the TGF-β family. Conversely, the other nonsense mutation (p.Arg370X) caused the loss of HTRA1 protease activity by the nonsense-mediated decay of mRNA [28]. The diagnosis of CARASIL was confirmed through molecular genetic testing, which identifies characteristic mutations in the HTRA1 gene. In CARASIL patients, mutations in the HTRA1 gene may cause disturbances in the regulation of TGF-β signaling. This abnormally increased TGF-β signaling altered the small blood vessel structure in the brain [28]. These blood vessel abnormalities (described as arteriopathy) increased the risk of stroke highly, leading to neuronal loss in several areas of the brain. The dysregulation of TGF-β signaling may also underlie the hair loss and back pain in CARASIL patients, although the relationship between abnormal TGF-β signaling and these features have not yet been fully clarified. Abnormally increased TGF-β signaling seemed to cause the degeneration of VSMCs, because TGF-β played an important role in the differentiation of these cells.

4. Cathepsin A–Related Arteriopathy with Strokes and Leukoencephalopathy (CARASAL)

CARASAL is a novel hereditary adult-onset cerebral SVD. The disease was characterized by therapy-resistant hypertension, strokes, and slow and late cognitive deteriorations. Impairment in the lower cranial nerve function was also prominent, resulting in several symptoms, such as vertigo, motor dysfunctions (including facial nerves), refractory hypertension, as well as a dry mouth and eyes [30]. In CARASAL, the MRI pattern showed a diffuse, progressive leukoencephalopathy that preceded the onset of strokes and was disproportionate to the degree of clinical severity. There were also multifocal signal changes in the cerebral white matter and basal nuclei, thalami, and brainstem, a pattern suggestive of SVD [15]. These changes caused the leukoencephalopathy to become virtually diffuse. Hervé et al. reported a French family with autosomal-dominant vascular leukoencephalopathy with an MRI pattern similar to that in CARASAL [101,102]. Neuropathological findings included a diffuse white matter-and myelin pallor, asymmetric fibrous thickening in small arterioles or astroglyosis. Lacunar changes may also be possible, such as perivascular tissue refraction or axonal loss [30].
The French family showed linkage with an 11.2-Mb interval on chromosome 20q13, encompassing the 1,145-kb region of the CTSA variant, presenting a strong argument that it was the same disease [102]. The CTSA gene encodes protective protein cathepsin A (PPCA, genomic coordinates (GRCh37): 20:44 519 590–44 527 458) protein and is located on chromosome 20q13. Structurally, the CTSA gene overlaps at its 5′ and 3′ ends, and two other genes were both transcribed from the antisense strand relative to CTSA. The gene at the 3′ end (PLTP) encoded a phospholipid transfer protein, whereas that at the 5′ end (Neuralized E3 Ubiquitin Protein Ligase 2, NEURL2) encodes OZZ, a striated muscle-specific E3-ubiquitin ligase. Thus, mutation analysis of the patient’s CTSA gene and biochemical assessment of the cathepsin A, β-galactosidase, and neuraminidase-1 activities in cultured fibroblasts should be performed to support the diagnosis of early infantile galactosialidosis. Recessive CTSA mutations may cause galactosialidosis. One of the numerous functions of cathepsin A was to degrade endothelin-1. Thus far, 19 disease-causing mutations have been identified for galactosialidosis in the CTSA gene [103], the majority of which were missense mutations resulting in single amino acid substitutions. An incomplete PPCA functional protein was resulted from the absence of CTSA mRNA, leading to either a premature termination of translated protein or a truncated protein product [30]. This defective protein was caused by small deletions/insertions, missense mutations, splicing variants, or a nonsense mutation. In addition to cerebral vascular abnormalities, endothelin-1 may have a role in the pathogenesis of CARASAL [30,102].
While homozygous CTSA mutations may play a role in galactosialidosis because of a deficiency in β-galactosidase and neuraminidase-1 [103,104], heterozygous CTSA mutations were not associated with the disease previously. Although the possible functional role of the CTSA mutation in CARASAL remained unexplained, the Arg325Cys missense could have a dominant inheritance and possibly some toxic effect [30]. The extra cysteine could affect the stabilization, folding, and structure of the protein, probably through the extra disulfide bond [74]. Notably, CADASIL was the most common monogenic SVD and is primarily caused by NOTCH3 mutations that affected cysteines, as described above. On the other hand, a recent report demonstrated that endothelin-1 mediated the inhibition of oligodendrocyte maturation and remyelination by reactive astrocytes [105]. A higher abundance of astrocytic endothelin-1 was detected in the brains of patients with CARASAL compared to controls, potentially leading to reduced cathepsin an activity [30].
CARASAL should be classified as a very rare disease, since currently, limited diagnostic tools were available [30,102]. CARASAL may represent a new phenotype to the spectrum of SVD. More genetic studies on CTSA mutations should be performed to assess the prevalence of the disease [30]. Nevertheless, for SVD patients with a positive family history, an unusual extensive leukoencephalopathy, and an absence of NOTCH3, HTRA1, and COL4A1/A2 mutations, a molecular analysis of the CTSA gene should be considered [30]. It was worthwhile to investigate whether heterozygous CTSA mutations were associated with an increased risk of SVD, particularly in patients with galactosialidosis [30,102]. Further studies should elucidate whether the variant in CTSA was associated with CARASAL and whether the risk of an SVD was also seen with other mutations.

5. Hereditary Diffuse Leukoencephalopathy with Spheroids (HDLS)

Hereditary diffuse leukoencephalopathy with spheroids (HDLS) is an autosomal-dominant white-matter disease with a high age-dependent penetrance that differs between genders [106]. The disease is characterized by different clinical symptoms, including dementia and personality changes (depression, schizophrenia, anxiety or irritability) [106,107]. Motor dysfunctions also appear, such as gait instability, pyramidal signs, and an instable posture [31,107,108,109]. Urinary incontinence could also be prominent among the patients [106]. HDLS causes adult-onset cognitive impairment when patients were in their 40s and 50s [31]. Cognitive impairment was the most frequent initial symptom in women whose disease began when they were 20–30 years old [107]. Since the clinical presentation of HDLS has not been clearly elucidated yet, patients may be clinically misdiagnosed as having frontotemporal dementia, multiple sclerosis (MS), cerebral autosomal-dominant arteriopathy with subcortical infarcts, leukoencephalopathy [25], AD [80], or corticobasal degeneration [106]. The disease is characterized by the degeneration of white matter, loss of the myelin sheath, and destruction of the spheroids in axons [108]. MRI findings on patients with HDLS often revealed cerebral white matter lesions, which were asymmetrical in the early disease stage, but become symmetrical with disease progression. Several brain areas were affected, particularly the frontal and parietal white matter [31]. Microbleeds may be missing in cases of HDLS, and grey matter could be spared from disease pathology [109].
Colony-stimulating factor 1 receptor [CSF1R, MIM*164770] mutations cause HDLS, which was first identified by Axelsson et al. in 1984 [31,110]. As a transmembrane protein, CSF1R served as a tyrosine kinase and was involved in the activation of mononuclear phagocytic cells, such as microglia. Microglia cells participated as immune effector cells and played a role homeostasis and surveillance in the brain [111]. Dysfunctions in microglia due to CSF1R mutations was assumed to be the primary disease-causing mechanism in HDLS [112]. On the basis of these observations, the name of adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) was proposed to encompass both of these CSF1R-related diseases [113]. Most of the mutations were located in the tyrosine kinase domain of CSF1R and were thought to cause CSF1R loss-of-function [31,107].
Recently, mutations in CSF1R have also been identified in families with pigmented orthochromatic leukodystrophy [113], which was another disease that affects the white matter and was clinically and pathologically similar to HDLS. In addition, functional studies have suggested that the mutations affect the kinase activity of the protein, probably by altering the phosphorylation of downstream targets [31]. Although mutations in the CSF1R gene may cause HDLS, mutations in this gene were detected in cases with different clinical manifestations, further demonstrating the difficulties in the clinical diagnosis of HDLS. It was found that 79% of the mutations were located in the distal part of the TKD of CSF1R (102 cases) protein [103]. TKD is a type III receptor tyrosine kinase belonging to the platelet-derived growth factor (PDGF) receptor family, whose members included PDGF-α and PDGF-β, the FMS-like tyrosine kinase 3 (FLT3), and the receptor for stem cell factor (c-KIT). The protein kinase domains, which were a key regulator of most of the cellular pathways that might associate with disease, were structurally conserved and often oncogenic.
The number of HDLS cases confirmed by genetic analysis is increasing in various populations, suggesting that the prevalence of HDLS is higher than previously thought. CSF1R mutations were reported in families from the USA, Europe, and Asia—however, there was no obvious relationship between the specific mutations and country or race [107]. Although ALSP was usually inherited in an autosomal-dominant manner, sporadic form of diseases could comprise 40% of the cases in all families [107]. Figure 3 summarized all mutations discovered in the CSF1R gene from worldwide. The majority of the described variants were missense mutations. To date, approximately 60 pathogenic variants have been reported in patients with HDLS, including 51 missense-, nine splice-site, two frameshift-, one nonsense mutation, and one deletion mutation. All of these mutations were located within the TKD, however, the p.Thr567fsX44 mutation was found outside of the TKD. Interestingly, the p.Ser688-GlufsX13 mutation was within the kinase insertion domain and could interrupt the TKD [114]. Mutations were found more frequently in the distal kinase domain encoded by exons 17–21 (46 mutations) than in the proximal kinase domain encoded by exons 12–15 (13 mutations). The most common mutation was p.Ile794Thr mutation, which was found in 14 families around the world—a majority of them were found in Japan (8), but additional families were also positive for this mutation from USA (two), Germany (two), Netherlands (two), and Taiwan (two) [107].
A recent study analyzed 122 cases from 90 families with CSF1R mutations and demonstrated that the phenotype of HDLS caused by CSF1R mutations was influenced by gender. The mean age of onset was 43 years (range, 18–78 years), the mean age at death was 53 years (range, 23–84 years), and the mean disease duration was 6.8 years (range, 1–29 years) [107]. The CSF1R mutations were effectively gain-of-function mutations, producing dominant negative repressors [31]. HDLS is an autosomal-dominant disease, and its diagnosis should be essential even in the absence of a positive family history in which a specific CSF1R mutation showed a reduced penetrance. Since pharmacological targeting of CSF1R with tyrosine kinase inhibitors prevented the disease progression in mouse models of neurodegenerative disorders [2,3,24], a potential pharmacological benefit of CSF1R inhibition remains to be elucidated for patients with HDLS [115]. Since mutations in the CSF1R gene could cause HDLS, further genetic studies should be undertaken to address the difficulties in the clinical diagnosis of HDLS. Probably, these aforementioned facts about the role of CSF1R in monocyte/macrophages, as well as in microglia biology, suggested a fundamental impact and therapeutic potential for HDLS.

6. COL4A1/2-Related Brain Small-Vessel Disease

Collagen type IV alpha 1 chain (COL4A1) related disorders were characterized as cerebral SVD with diverse disease phenotypes that include porencephaly, seizures, dementia, intellectual disability, migraine, stroke, visual impairment (visual loss, cataract, glaucoma), muscle dysfunction or hereditary angiopathy with nephropathy, aneurysms, and muscle cramps (HANAC) syndrome [32,116]. The disease phenotypes appear in early lifetime, even in childhood, but also in the adult life. Imaging of COL4A1-related disease revealed changes in white matter, porencephalic cyst, haemmorrhage-or microhaemorrhage, possible involvement in basal ganglia [117,118]. Collagen type IV alpha 2 chain (COL4A2) related disease was also suggested as an adult onset SVD, where similar phenotypes were present, such as porencephaly, scattered white matter lesions, carotid aneurysm, myopia, amblyopia, cerebellar- and visual abnormalities [119]. COL4A1 and COL4A2 encoded the collagen chains α1 (IV) and α2(IV), respectively, which constituted a major component of the vascular basement membrane [120]. COL4A1 and COL4A2 mutations have been reported in a broad spectrum of disorders, including myopathy, glaucoma, cerebrovascular disease, and renal, ophthalmological, cardiac, and muscular abnormalities, which were collectively termed “COL4A1 and COL4A2 mutation-related disorders” [32,121,122,123,124]. Furthermore, missense mutations in COL4A1/COL4A2 caused rare familial forms of cerebral SVD, manifesting as deep intracerebral hemorrhages, lacunar ischemic strokes, and WMHs [35,118]. These mutations were associated with porencephaly and infantile hemiparesis and have been recently recognized as a monogenic cause of SVD that could present in adulthood.
Collagen type IV alpha 1 chain (COL4A1; NM_001845) and Collagen type IV alpha 2 chain (COL4A2, NM_001846) genes comprised of 52 and 48 exons, respectively, and were arranged head-to-head on opposite strands of human chromosome 13 (13q34). COL4A1 and COL4A2 genes, located on chromosome 13q34 (in tandem), shared a unique, bidirectional promoter. Their transcripts were strongly controlled by epigenetic mechanisms, including regulation by a microRNA family, which could be involved in the down-regulation of their expression [83,84]. Additional microRNAs may also indirectly regulate collagen synthesis [125,126]. Three main domains of COL4A1 and COL4A2 proteins could be distinguished: an N-termimal 7S domain, a central triple-helix-forming (collagenous) domain, and a C-terminal non-collagenous (NC1) domain. Human and mouse COL4A1 were similar, since they both had 21 conservative repeats, which divided the collagenous domain into 22 sub-domains. In addition, they both had 23 conserved repeat interruptions which align with those in COL4A1. All cysteine residues in the collagenous domain of COL4A1 and COL4A2 were present inside repeat interruptions, which suggested that these cysteine repeats may also be important sites for cross-linking between different molecules [123]. There were multisystem disorders that result from COL4A1 and COL4A2 mutations. Emerging studies were available on COL4A1 mutations, and the first COL4A2 mutations were recently reported [127,128]. In total, data from 67 families with COL4A1 and COL4A2 mutations were reported [124]. In these families, 33 mutations (50%) were found associated with familial form of disease—a de novo mutation was identified in 17 cases (25%), and in 17 cases (25%), there was a lack of data on parental sequencing [124]. These data indicated that the de novo mutation be COL4A1 and COL4A2 was high. In addition, with a high de novo mutation rate of 40%, 21 COL4A1 (12 of them novel) and 3 COL4A2 pathogenic mutations were identified, mostly in children with porencephaly or other patterns of parenchymal hemorrhage [124].
Together, these findings suggested that patients with COL4A1 and COL4A2 mutations may be at a higher risk of retinal hemorrhages, and retinal examinations may be useful for identifying patients with COL4A1 and COL4A2 mutations who may also be at a higher risk of hemorrhagic strokes [129]. In addition to prenatal and perinatal hemorrhages, COL4A1 and COL4A2 mutations also caused the sporadic and recurrent intracerebral hemorrhages (ICH) in young and old patients [123]. Most of the reported COL4A1 mutations were heterozygous missense mutations that affected a glycine residue within the collagenous region of the protein. However, many studies have shown that dominant missense mutations in COL4A1/COL4A2 caused rare familial forms of cerebral SVD, manifesting as deep ICHs, lacunar ischemic strokes, and WMHs [35,118]. Genes causing rare familial forms of cerebral SVD may also contain variants conferring risk for common forms of cerebral SVD [130,131].
The COL4A1 and COL4A2 mutations were important causes of cerebrovascular disease with a high mutation detection rate in porencephaly and childhood cerebral hemorrhage, and a relatively high rate of de novo mutations. This mutation was less prominent in (sporadic) adult-onset intracerebral hemorrhage, with an incidence of 6% [127,132]. In addition, the precise role of COL4A1 and COL4A2 mutations in cortical malformations needed to be elucidated, but it appeared to contribute to the malformations that resulted from vascular insults during fetal development [133]. Follow-up data on COL4A1 and COL4A2 mutation carriers would be important for developing appropriate surveillance protocols and adapting treatment.

7. Fabry Disease

Fabry disease, an X-linked lysosomal storage disorder, is caused by the deficiency or absence of alpha-galactosidase A (α-Gal-A), which leads to an accumulation of globotriaosylceramide/ceramide trihexoside (Gb3) with a terminal a-D-galactosyl residue, particularly globotriaosylceramide (GL-3, Gb3, CTH) and globotriaosylsphingosine (Lyso-GL-3, lyso-Gb3) [134,135]. This enzyme deficiency results in the accumulation of globotriaosylceramide within the lysosomes of various organs, such as the blood vessels, kidneys, heart, and dorsal root ganglia. The main disease symptom is acroparaesthesia, which appears in childhood or early adulthood. Many different clinical phenotypes were observed in patients with Fabry disease such as corneal dystrophy, angiokeratomas, visual impairment, reduced cognitive function, hearing loss, dizziness, problems with digestion, vertigo, depression, or abnormalities in cardiovascular system [135]. Patients with classical Fabry disease could represent the neuropathic pain, verticillata, and angiokeratoma, followed by stroke, heart disturbances, and kidney failure. The atypical form of the disorder has a later disease onset. The disease progression may affect a single organ or have a milder phenotype [136]. The most prominent MRI finding in Fabry disease was multifocal leukoencephalopathy, but additional abnormalities may be also observed, such as lacunar ischaemic lesions (basal ganglia, brainstem), cerebral atrophy or pulvinar sign [137]. The disease affected both myelinated and unmyelinated neurons, resulting in symmetrical small fiber neuropathy. It started from the feet and spreads proximally. Both males and females are affected, but the disease phenotype is more severe and has an earlier onset in affected males than in females [138].
Fabry disease is caused by the genetic mutations in the alpha-galactosidase-A gene (GLA-gene), located on the long arm of the X-chromosome (Xq22.1). Thus far, more than 790 different mutations have been described, including missense and nonsense mutations; however, small deletions, insertions, and splicing defects have also been reported in “The Human Gene Mutation Database” (http://www.hgmd.cf.ac.uk/ac/gene.php?gene=GLA, accessed on 1 July 2019). Most of the pathogenic GLA mutations were restricted to single families [139]. Interestingly, the disease manifestations may vary within families carrying the same mutation [140], however, the genotype-phenotype correlation was confined to individual families. In general, however, mutations that have been associated with more attenuated late-onset disease were frequently missense mutations, e.g., the N215S genotype. One missense variant, the D313Y genotype, was considered to be a non-damaging polymorphism [141]. The incidence of Fabry disease has been estimated to be 4.5% in men and 3.4% in women among patients with cryptogenic stroke [142], whereas the incidence was one per 17,000 to 117,000 in the general population [143,144]. The prevalence ranges from 0.6% to 11.1%. In addition, the disease seems to be rare, with an estimated prevalence of one in 40,000 to 60,000 in males of all ethnicities. As an X-linked disease, the genetic defect that causes Fabry disease could be transmitted by both males and females, but it seemed to be transmitted less frequently in females. Remarkably, the only coding variant (c.937G > T, p.Asp313Tyr) was detected approximately ten times more frequently (approximately 5%) in our cohort than in the previous study, which reported a frequency of 0.45% [145].
Many missense mutations affecting cysteine residues were associated with the classic Fabry disease phenotype due to the formation of disulfide bonds in the wild-type α-Gal structure, resulting in negligible or reduced residual enzyme activity [146]. This observation attests to the importance of these genes for normal enzyme function. Several missense mutations that introduced an extra cysteine into the amino acid sequence of α-Gal have been identified in Fabry disease patients. The Cys174Arg [115,147] mutation was expected to be a pathogenic mutation. GLA Cys174Gly were recently identified in a patient presenting with an unusual late-onset renal variant of Fabry disease [148], suggesting that this mutation was not clinically benign [149]. In addition, the GLA Arg118Cys variant was recently reported in large case-finding studies in different European populations [150,151,152,153] and Brazil [154] among patients with stroke, left ventricular hypertrophy, or on chronic dialysis. However, the GLA variants that cause Fabry disease phenotypes were still inconsistent. More recently, Ferreira et al. (2015) reported that the GLA Arg118Cys caused the Fabry disease phenotype; the findings were based on the analysis of a series of Iberian (Portuguese and Spanish) individuals and families [146]. Surprisingly, carriers of the Cys118 allele may present only with a typical eruption of angiokeratomas, which was usually considered a manifestation of classic Fabry disease. These data may explain the absence of a familial history of Fabry disease in an Italian patient identified by newborn screening [155,156]. The identification of a GLA mutation associated with a clinically relevant phenotype was crucial for patient management and risk stratification. However, the unresolved issue of how to appropriately match complementary genotypes and phenotypes remains a major impediment to the clinical application of such diagnostic information.

8. Possible Mechanisms of Cerebral Small Vessel Diseases Association Genetics

Several genes and mutations were identified in familial SVD and described above. These variants in phenotype share both clinical and radiological features with sporadic SVD and provide important insights into the mechanisms of the disease. SVD were known as complex disorders [1,22,42]. SVD disorders have several subtypes (such as CARASIL, CADASIL, HDLS, Fabry disease), and the exact molecular mechanisms of these diseases remain incompletely understood [1,22,40,42]. Several risk factors that could play a role in SVD have been identified, such as aging, head injury or stroke. Possible mechanisms of dysfunctions could be associated with dysfunctions in vessels, reduced defensive mechanism, and disturbances in vascular reactivity [42]. Post-mortem and animal studies were performed to find out the exact pathways, associated with SVD mechanisms [22,157]. The classical hypothesis of disease suggests that it was caused by cerebral reduced blood flow and the impairment of cerebral autoregulation. Elevated permeability and disruption of the blood-brain barrier (BBB) may also play a significant role in disease [20]. Reactive oxygen species (ROS) were also suggested to be important contributor in SVD. NADPH oxidases serve as ROS producing enzymes and could play a role in vascular diseases by producing superoxides [34]. In addition, genetic mutations in different genes could also play a role in the onset of different forms of SVD. Dysfunction in the extracellular matrix pathways, were associated with different diseases, such disturbances in potassium channels by NOTCH3 aggregation (CADASIL). NOTCH3 is a single transmembrane receptor involved in the Notch signaling. Mutations resulted in dysfunctions in the Notch signaling pathways. Mutant NOTCH3 accumulated in the vasculature and formed granular osmiophilic material (GOM). The exact role of GOMs in CADASIL remained unclear, but it may be possible that they result in impairment in glymphatic system. NOTCH3 also played a role in the development and homeostasis vascular system, and its dysfunctions could result in abnormal small vessel development, breakdown of BBB [47]. In addition, the genetic pathogenesis similarities existed between CADASIL and Alzheimer’s disease, affecting the small vessels of the brain. Depending on the mutation, plausible dual molecular mechanisms could be involved in vascular damage and their impact on brain function, meaning one gene could influence both diseases [2,23,24,158,159,160].
HTRA1 gene encodes a serine protease and is involved in TGFβ signaling and angiogenesis. Mutations in HTRA1 cause CARASIL by the attenuation of TGFβ function. HTRA1 cleaves the latent TGF-β binding protein 1 (LTBP-1), which is an important regulator of TGF-β [161]. COL4A1/COL4A2 related angiopathy was associated with collagen dysfunctions and ER stress, but the exact mechanism remains unclear. Apoptosis could play an important role in the onset of microcephaly [22]. CSF1R is a tyrosine kinase involved in HDLS which plays a key role in development and survival of microglia. Mutations in the receptor domain of CSF1R eliminated the kinase activity, ability of dimer formation, and CSF1-CSF1R interaction. CSF1R deficiency resulted in a reduction of microglia density and neuronal loss [31]. CTSA mutations (CARASAL) caused in endothelin-1 degradation and reduced oligodendrocyte regulation [30]. Additional pathways could also be associated with SVD causing mutations, for example, they could impair the blood-brain barrier integrity or the protection against DNA damage [22]. CSF1R is a tyrosine kinase transmembrane protein involved in activation of mononuclear phagocytic cells, for example, microglia that function as immune effector cells with homeostatic and surveillance tasks in the brain [111]. As microglial dysfunction due to CSF1R mutations is assumed to be the primary disease-causing mechanism, HDLS is classified as microgliopathy [111].
In several cases, genetics of SVD remained unexplained. Genome-wide association studies (GWAS) discovered several possible genetic factors that could contribute to sporadic form of SVD, such as NEURL1 (E3 ubiquitin protein ligase 1), PDCD11 (programmed cell death 11) and SH3PXD2A (SH3 and PX Domains 2A), which could be related to WMHs. Currently, it remains unclear whether the disease associated pathways were similar in all individuals, or whether there was additional different disease mechanism. GWAS revealed SVD-related pathways, but in some cases, these pathways were shared [22]. Heterozygous frameshift mutations in TREX1 (three prime repair exonuclease 1) were also be involved in vascular dysfunctions, and caused cerebroretinal vasculopathy or hereditary endotheliopathy, retinopathy, nephropathy and stroke. These mutations cause premature STOP codon and could possibly reduce the DNA repair mechanisms in case of oxidative stress. This process could result in premature aging in the vascular system [162]. FOXC1 (forkhead box transcription factor) duplications or deletions could cause abnormal development of cerebellum, impaired neovascularization in cornea. Missense mutations may also affect the FOXC1 protein function. Brain MRI of patients with FOXC1 mutations (duplication and deletion) revealed WMHs, lacunar infractions and dilated perivascular spaces. FOXC1 dysfunctions may impair the vascular cell stability by inducing dysfunctions in development of neural crest and mural cell recruitment. PITX2 (paired like homeodomain 2) could interact with FOXC1, and associated with several dysfunctions, such as atrial fibrillation, cardioembolic stroke, and white matter hyperintensities in brain [163].
Although the roles of most of the common genetic variants (e.g., NOTCH3, HTRA1, CTSA, CSF1R COL4A1, COL4A2, and GLA) emerging in association with sporadic forms of cerebral SVD are yet to be fully elucidated, it became clear that each of these variants could potentially disrupt specific components of the neurovascular unit’s structure and function. A detailed characterization of these pathways would be needed for the discovery of novel molecular targets for future therapies. Shared pathways affecting the integrity and function of the extracellular matrix appeared to play an integral role in these disease’s pathways. It was likely that there were multiple shared pathways, each being involved to different degrees in different manifestations or subtypes of SVD. These genetic mechanisms, as well as their interactions with environmental factors, may provide explanations as to why different patients in the sporadic disease population exhibit each feature of SVD to a different extent.

9. Conclusions and Future Perspective

Studies in both monogenic forms of SVD and the genetics of sporadic SVD were able to fill in the blank edges in the map of the disease processes in SVD. Blood circulation in the brain may be impaired by conditions that affect the large vessels, causing ischemic and hemorrhagic infarcts, and the small vessels, causing cerebral SVD. The pathological processes of cerebral SVD are complicated and incompletely understood. Hereditary SVDs were associated with cognitive and motor impairments, as well as stroke. These diseases affected not only the brain, but other organs as well, such as the kidney or visual system. Currently, no effective therapy is available for SVD [164]. In the last few years, GWAS and next generation sequencing techniques have facilitated the discovery of genes which could be involved in SVDs [1]. Genetic studies on the various forms of SVD could reveal a shared pathogenesis, re-conceptualizing the development and progression of cerebral SVD and closing the gaps in our understanding of the disease processes. These genetic mechanisms, as well as their interactions with environmental factors, may provide explanations and support for diagnosing the various forms of cerebral SVD. Since a correct diagnosis may be difficult to achieve based solely upon the clinical symptoms, molecular testing is becoming more widely available [165]. Patients should be more frequently diagnosed, and a clearer picture will emerge of both the prevalence and clinical spectrum of the disease.
Hence, the genetic factors of cerebral SVD play a pivotal role in terms of unraveling their molecular mechanism, and further studies could be considered a powerful tool for understanding the disease. Further genetic studies in cerebral SVD will likely provide further insight into the mechanisms involved in both the monogenic and sporadic forms of the disease. If large enough patient cohorts were available, a whole-genome association study could be an effective way to study the genetic factors underlying the phenotypes. Thus, further studies of autosomal-dominant diseases could use linkage analysis paired with targeted re-sequencing or whole-genome sequencing. Together, these studies could aid in the development of novel diagnostic strategies and treatments for the various forms of cerebral SVD. A challenge in genetic studies could be the lack of cell and animal models, which may be helpful in understanding the disease mechanism in terms of mutations. Studies on epigenetics and gene environmental interactions may also open more avenues in the prediction of risk for SVD [21]. In addition, some important issues with cerebral SVD were clinically heterogeneous and share several common features with other neurodegenerative diseases [43], such as a possible genetic background, assembly of misfolded proteins, or neuronal loss rather than a focal brain disease [166,167,168]. Thus, 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. Once disease mechanisms underlying cerebral SVD are established, individual genetic risk profiles may suggest particularly effective medical or dietary therapies to halt cerebral SVD’s common and devastating manifestations. In addition, despite intensive research efforts, the molecular mechanisms underlying cerebral SVD remain poorly understood, which has hampered the development of cerebral SVD-specific therapies. Important steps forward in understanding the disease mechanisms underlying cerebral SVD have been made using genetic approaches in studies of both monogenic and sporadic SVD. Further genetic studies in SVD will likely provide more conclusive evidence of an overlap of disease pathways involved in both monogenic and sporadic disease. Although understanding the processes of each disease, whether a monogenic form of SVD or sporadic disease, may aid the development of treatment options for the specific disease, it is possible that the distinction between each of the diseases is blurred, and that the same few convergent processes will eventually serve as therapeutic targets.

Author Contributions

Writing—Original Draft Preparation, V.V.G.; Literature Search, V.V.G. and E.B.; Writing—Review & Editing, V.V.G. and E.B.; Supervision, S.S.A.A., Y.C.Y. and S.Y.K.

Funding

Manuscript was supported by a National Research Foundation of Korea (NRF) Grants, awarded by the Korean government (NRF-2017R1A2B4012636 & NRF-2019R1G1A109740011).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Choi, J.C. Genetics of Cerebral Small Vessel Disease. J. Stroke 2015, 17, 7–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Giau, V.V.; Bagyinszky, E.; An, S.S.A.; Kim, S. Clinical genetic strategies for early onset neurodegenerative diseases. Mol. Cell. Toxicol. 2018, 14, 123–142. [Google Scholar] [CrossRef]
  3. van Giau, V.; An, S.S.A.; Bagyinszky, E.; Kim, S. Gene panels and primers for next generation sequencing studies on neurodegenerative disorders. Mol. Cell. Toxicol. 2015, 11, 89–143. [Google Scholar] [CrossRef]
  4. White, H.; Boden-Albala, B.; Wang, C.; Elkind, M.S.; Rundek, T.; Wright, C.B.; Sacco, R.L. Ischemic stroke subtype incidence among whites, blacks, and Hispanics: The Northern Manhattan Study. Circulation 2005, 111, 1327–1331. [Google Scholar] [CrossRef] [PubMed]
  5. Sacco, S.; Marini, C.; Totaro, R.; Russo, T.; Cerone, D.; Carolei, A. A population-based study of the incidence and prognosis of lacunar stroke. Neurology 2006, 66, 1335–1338. [Google Scholar] [CrossRef] [PubMed]
  6. Chamorro, A.; Sacco, R.L.; Mohr, J.P.; Foulkes, M.A.; Kase, C.S.; Tatemichi, T.K.; Wolf, P.A.; Price, T.R.; Hier, D.B. Clinical-computed tomographic correlations of lacunar infarction in the Stroke Data Bank. Stroke 1991, 22, 175–181. [Google Scholar] [CrossRef] [PubMed]
  7. Petty, G.W.; Brown, R.D., Jr.; Whisnant, J.P.; Sicks, J.D.; O’Fallon, W.M.; Wiebers, D.O. Ischemic stroke subtypes: A population-based study of incidence and risk factors. Stroke 1999, 30, 2513–2516. [Google Scholar] [CrossRef]
  8. Jung, K.H.; Lee, S.H.; Kim, B.J.; Yu, K.H.; Hong, K.S.; Lee, B.C.; Roh, J.K. Secular trends in ischemic stroke characteristics in a rapidly developed country: Results from the Korean Stroke Registry Study (secular trends in Korean stroke). Circ. Cardiovasc. Qual. Outcomes 2012, 5, 327–334. [Google Scholar] [CrossRef]
  9. Syed, N.A.; Khealani, B.A.; Ali, S.; Hasan, A.; Akhtar, N.; Brohi, H.; Mozaffar, T.; Ahmed, N.; Hameed, A.; Baig, S.M.; et al. Ischemic stroke subtypes in Pakistan: The Aga Khan University Stroke Data Bank. JPMA 2003, 53, 584–588. [Google Scholar]
  10. Tsai, C.F.; Thomas, B.; Sudlow, C.L. Epidemiology of stroke and its subtypes in Chinese vs. white populations: A systematic review. Neurology 2013, 81, 264–272. [Google Scholar] [CrossRef]
  11. Turin, T.C.; Kita, Y.; Rumana, N.; Nakamura, Y.; Takashima, N.; Ichikawa, M.; Sugihara, H.; Morita, Y.; Hirose, K.; Okayama, A.; et al. Ischemic stroke subtypes in a Japanese population: Takashima Stroke Registry, 1988–2004. Stroke 2010, 41, 1871–1876. [Google Scholar] [CrossRef] [PubMed]
  12. Kubo, M.; Hata, J.; Doi, Y.; Tanizaki, Y.; Iida, M.; Kiyohara, Y. Secular trends in the incidence of and risk factors for ischemic stroke and its subtypes in Japanese population. Circulation 2008, 118, 2672–2678. [Google Scholar] [CrossRef] [PubMed]
  13. Wolma, J.; Nederkoorn, P.J.; Goossens, A.; Vergouwen, M.D.; van Schaik, I.N.; Vermeulen, M. Ethnicity a risk factor? The relation between ethnicity and large- and small-vessel disease in White people, Black people, and Asians within a hospital-based population. Eur. J. Neurol. 2009, 16, 522–527. [Google Scholar] [CrossRef] [PubMed]
  14. Hilal, S.; Mok, V.; Youn, Y.C.; Wong, A.; Ikram, M.K.; Chen, C.L. Prevalence, risk factors and consequences of cerebral small vessel diseases: Data from three Asian countries. J. Neurol. Neurosurg. Psychiatry 2017, 88, 669–674. [Google Scholar] [CrossRef] [PubMed]
  15. Wardlaw, J.M.; Smith, E.E.; Biessels, G.J.; Cordonnier, C.; Fazekas, F.; Frayne, R.; Lindley, R.I.; O’Brien, J.T.; Barkhof, F.; Benavente, O.R.; et al. Neuroimaging standards for research into small vessel disease and its contribution to ageing and neurodegeneration. Lancet Neurol. 2013, 12, 822–838. [Google Scholar] [CrossRef] [Green Version]
  16. Moran, C.; Phan, T.G.; Srikanth, V.K. Cerebral small vessel disease: A review of clinical, radiological, and histopathological phenotypes. Int. J. Stroke 2012, 7, 36–46. [Google Scholar] [CrossRef] [PubMed]
  17. Rost, N.S.; Fitzpatrick, K.; Biffi, A.; Kanakis, A.; Devan, W.; Anderson, C.D.; Cortellini, L.; Furie, K.L.; Rosand, J. White matter hyperintensity burden and susceptibility to cerebral ischemia. Stroke 2010, 41, 2807–2811. [Google Scholar] [CrossRef]
  18. Rost, N.S.; Rahman, R.M.; Biffi, A.; Smith, E.E.; Kanakis, A.; Fitzpatrick, K.; Lima, F.; Worrall, B.B.; Meschia, J.F.; Brown, R.D.; et al. White matter hyperintensity volume is increased in small vessel stroke subtypes. Neurology 2010, 75, 1670–1677. [Google Scholar] [CrossRef]
  19. Cheung, N.; Liew, G.; Lindley, R.I.; Liu, E.Y.; Wang, J.J.; Hand, P.; Baker, M.; Mitchell, P.; Wong, T.Y. Retinal fractals and acute lacunar stroke. Ann. Neurol. 2010, 68, 107–111. [Google Scholar] [CrossRef]
  20. Toyoda, K. Cerebral Small Vessel Disease and Chronic Kidney Disease. J. Stroke 2015, 17, 31–37. [Google Scholar] [CrossRef] [Green Version]
  21. Martini, S.; Williams, S.; Moretti, P.; Woo, D.; Worrall, B. A molecular/genetic approach to cerebral small-vessel disease: Beyond aging and hypertension. Brain Circ. 2015, 1, 79–87. [Google Scholar] [CrossRef]
  22. Tan, R.; Traylor, M.; Rutten-Jacobs, L.; Markus, H. New insights into mechanisms of small vessel disease stroke from genetics. Clin. Sci. (London, England 1979) 2017, 131, 515–531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Giau, V.V.; Bagyinszky, E.; Yang, Y.S.; Youn, Y.C.; An, S.S.A.; Kim, S.Y. Genetic analyses of early-onset Alzheimer’s disease using next generation sequencing. Sci. Rep. 2019, 9, 8368. [Google Scholar] [CrossRef] [PubMed]
  24. Giau, V.V.; Senanarong, V.; Bagyinszky, E.; An, S.S.A.; Kim, S. Analysis of 50 Neurodegenerative Genes in Clinically Diagnosed Early-Onset Alzheimer’s Disease. Int. J. Mol. Sci. 2019, 20, 1514. [Google Scholar] [CrossRef] [PubMed]
  25. Dichgans, M. Genetics of ischaemic stroke. Lancet Neurol. 2007, 6, 149–161. [Google Scholar] [CrossRef]
  26. Craggs, L.J.; Yamamoto, Y.; Deramecourt, V.; Kalaria, R.N. Microvascular pathology and morphometrics of sporadic and hereditary small vessel diseases of the brain. Brain Pathol. (Zur. Switz.) 2014, 24, 495–509. [Google Scholar] [CrossRef] [PubMed]
  27. Joutel, A.; Corpechot, C.; Ducros, A.; Vahedi, K.; Chabriat, H.; Mouton, P.; Alamowitch, S.; Domenga, V.; Cecillion, M.; Marechal, E.; et al. Notch3 mutations in CADASIL, a hereditary adult-onset condition causing stroke and dementia. Nature 1996, 383, 707–710. [Google Scholar] [CrossRef] [PubMed]
  28. Hara, K.; Shiga, A.; Fukutake, T.; Nozaki, H.; Miyashita, A.; Yokoseki, A.; Kawata, H.; Koyama, A.; Arima, K.; Takahashi, T.; et al. Association of HTRA1 mutations and familial ischemic cerebral small-vessel disease. N. Engl. J. Med. 2009, 360, 1729–1739. [Google Scholar] [CrossRef] [PubMed]
  29. Verdura, E.; Herve, D.; Scharrer, E.; Amador Mdel, M.; Guyant-Marechal, L.; Philippi, A.; Corlobe, A.; Bergametti, F.; Gazal, S.; Prieto-Morin, C.; et al. Heterozygous HTRA1 mutations are associated with autosomal dominant cerebral small vessel disease. Brain 2015, 138, 2347–2358. [Google Scholar] [CrossRef]
  30. Bugiani, M.; Kevelam, S.H.; Bakels, H.S.; Waisfisz, Q.; Ceuterick-de Groote, C.; Niessen, H.W.; Abbink, T.E.; Lesnik Oberstein, S.A.; van der Knaap, M.S. Cathepsin A-related arteriopathy with strokes and leukoencephalopathy (CARASAL). Neurology 2016, 87, 1777–1786. [Google Scholar] [CrossRef]
  31. Rademakers, R.; Baker, M.; Nicholson, A.M.; Rutherford, N.J.; Finch, N.; Soto-Ortolaza, A.; Lash, J.; Wider, C.; Wojtas, A.; DeJesus-Hernandez, M.; et al. Mutations in the colony stimulating factor 1 receptor (CSF1R) gene cause hereditary diffuse leukoencephalopathy with spheroids. Nat. Genet. 2011, 44, 200–205. [Google Scholar] [CrossRef] [PubMed]
  32. Gould, D.B.; Phalan, F.C.; van Mil, S.E.; Sundberg, J.P.; Vahedi, K.; Massin, P.; Bousser, M.G.; Heutink, P.; Miner, J.H.; Tournier-Lasserve, E.; et al. Role of COL4A1 in small-vessel disease and hemorrhagic stroke. N. Engl. J. Med. 2006, 354, 1489–1496. [Google Scholar] [CrossRef] [PubMed]
  33. Revesz, T.; Holton, J.L.; Lashley, T.; Plant, G.; Frangione, B.; Rostagno, A.; Ghiso, J. Genetics and molecular pathogenesis of sporadic and hereditary cerebral amyloid angiopathies. Acta Neuropathol. 2009, 118, 115–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. De Silva, T.M.; Miller, A.A. Cerebral Small Vessel Disease: Targeting Oxidative Stress as a Novel Therapeutic Strategy? Front. Pharmacol. 2016, 7, 61. [Google Scholar] [CrossRef] [PubMed]
  35. Yamamoto, Y.; Craggs, L.; Baumann, M.; Kalimo, H.; Kalaria, R.N. Review: Molecular genetics and pathology of hereditary small vessel diseases of the brain. Neuropathol. Appl. Neurobiol. 2011, 37, 94–113. [Google Scholar] [CrossRef] [PubMed]
  36. Desmond, D.W.; Moroney, J.T.; Lynch, T.; Chan, S.; Chin, S.S.; Mohr, J.P. The natural history of CADASIL: A pooled analysis of previously published cases. Stroke 1999, 30, 1230–1233. [Google Scholar] [CrossRef] [PubMed]
  37. Fattapposta, F.; Restuccia, R.; Pirro, C.; Malandrini, A.; Locuratolo, N.; Amabile, G.; Bianco, F. Early diagnosis in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL): The role of MRI. Funct. Neurol. 2004, 19, 239–242. [Google Scholar] [PubMed]
  38. Malandrini, A.; Gaudiano, C.; Gambelli, S.; Berti, G.; Serni, G.; Bianchi, S.; Federico, A.; Dotti, M.T. Diagnostic value of ultrastructural skin biopsy studies in CADASIL. Neurology 2007, 68, 1430–1432. [Google Scholar] [CrossRef] [PubMed]
  39. Stojanov, D.; Aracki-Trenkic, A.; Vojinovic, S.; Ljubisavljevic, S.; Benedeto-Stojanov, D.; Tasic, A.; Vujnovic, S. Imaging characteristics of cerebral autosomal dominant arteriopathy with subcortical infarcts and leucoencephalopathy (CADASIL). Bosn. J. Basic Med. Sci. 2015, 15, 1–8. [Google Scholar] [CrossRef]
  40. Kim, K.W.; Kwon, H.; Kim, Y.-E.; Yoon, C.W.; Kim, Y.J.; Kim, Y.B.; Lee, J.M.; Yoon, W.T.; Kim, H.J.; Lee, J.S.; et al. Multimodal imaging analyses in patients with genetic and sporadic forms of small vessel disease. Sci. Rep. 2019, 9, 787. [Google Scholar] [CrossRef]
  41. Davous, P. CADASIL: A review with proposed diagnostic criteria. Eur. J. Neurol. 1998, 5, 219–233. [Google Scholar] [CrossRef] [PubMed]
  42. Smith, E.E.; Beaudin, A.E. New insights into cerebral small vessel disease and vascular cognitive impairment from MRI. Curr. Opin. Neurol. 2018, 31, 36–43. [Google Scholar] [CrossRef] [PubMed]
  43. ter Telgte, A.; van Leijsen, E.M.C.; Wiegertjes, K.; Klijn, C.J.M.; Tuladhar, A.M.; de Leeuw, F.-E. Cerebral small vessel disease: From a focal to a global perspective. Nat. Rev. Neurol. 2018, 14, 387–398. [Google Scholar] [CrossRef] [PubMed]
  44. Rutten, J.W.; Haan, J.; Terwindt, G.M.; van Duinen, S.G.; Boon, E.M.; Lesnik Oberstein, S.A. Interpretation of NOTCH3 mutations in the diagnosis of CADASIL. Expert Rev. Mol. Diagn. 2014, 14, 593–603. [Google Scholar] [CrossRef] [PubMed]
  45. Bianchi, S.; Dotti, M.T.; Federico, A. Physiology and pathology of notch signalling system. J. Cell. Physiol. 2006, 207, 300–308. [Google Scholar] [CrossRef] [PubMed]
  46. Duering, M.; Karpinska, A.; Rosner, S.; Hopfner, F.; Zechmeister, M.; Peters, N.; Kremmer, E.; Haffner, C.; Giese, A.; Dichgans, M.; et al. Co-aggregate formation of CADASIL-mutant NOTCH3: A single-particle analysis. Hum. Mol. Genet. 2011, 20, 3256–3265. [Google Scholar] [CrossRef]
  47. Joutel, A.; Andreux, F.; Gaulis, S.; Domenga, V.; Cecillon, M.; Battail, N.; Piga, N.; Chapon, F.; Godfrain, C.; Tournier-Lasserve, E. The ectodomain of the Notch3 receptor accumulates within the cerebrovasculature of CADASIL patients. J. Clin. Investig. 2000, 105, 597–605. [Google Scholar] [CrossRef] [PubMed]
  48. Monet-Lepretre, M.; Haddad, I.; Baron-Menguy, C.; Fouillot-Panchal, M.; Riani, M.; Domenga-Denier, V.; Dussaule, C.; Cognat, E.; Vinh, J.; Joutel, A. Abnormal recruitment of extracellular matrix proteins by excess Notch3 ECD: A new pathomechanism in CADASIL. Brain 2013, 136, 1830–1845. [Google Scholar] [CrossRef] [PubMed]
  49. Ruchoux, M.M.; Guerouaou, D.; Vandenhaute, B.; Pruvo, J.P.; Vermersch, P.; Leys, D. Systemic vascular smooth muscle cell impairment in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Acta Neuropathol. 1995, 89, 500–512. [Google Scholar] [CrossRef] [PubMed]
  50. Tikka, S.; Mykkanen, K.; Ruchoux, M.M.; Bergholm, R.; Junna, M.; Poyhonen, M.; Yki-Jarvinen, H.; Joutel, A.; Viitanen, M.; Baumann, M.; et al. Congruence between NOTCH3 mutations and GOM in 131 CADASIL patients. Brain 2009, 132, 933–939. [Google Scholar] [CrossRef] [Green Version]
  51. Federico, A.; Bianchi, S.; Dotti, M.T. The spectrum of mutations for CADASIL diagnosis. Neurol. Sci. 2005, 26, 117–124. [Google Scholar] [CrossRef] [PubMed]
  52. Mazzei, R.; Conforti, F.L.; Lanza, P.L.; Sprovieri, T.; Lupo, M.R.; Gallo, O.; Patitucci, A.; Magariello, A.; Caracciolo, M.; Gabriele, A.L.; et al. A novel Notch3 gene mutation not involving a cysteine residue in an Italian family with CADASIL. Neurology 2004, 63, 561–564. [Google Scholar] [CrossRef] [PubMed]
  53. Coupland, K.; Lendahl, U.; Karlström, H. Role of NOTCH3 Mutations in the Cerebral Small Vessel Disease Cerebral Autosomal Dominant Arteriopathy With Subcortical Infarcts and Leukoencephalopathy. Stroke 2018, 49, 2793–2800. [Google Scholar] [CrossRef] [PubMed]
  54. Wollenweber, F.A.; Hanecker, P.; Bayer-Karpinska, A.; Malik, R.; Bazner, H.; Moreton, F.; Muir, K.W.; Muller, S.; Giese, A.; Opherk, C.; et al. Cysteine-sparing CADASIL mutations in NOTCH3 show proaggregatory properties in vitro. Stroke 2015, 46, 786–792. [Google Scholar] [CrossRef] [PubMed]
  55. Tuominen, S.; Juvonen, V.; Amberla, K.; Jolma, T.; Rinne, J.O.; Tuisku, S.; Kurki, T.; Marttila, R.; Poyhonen, M.; Savontaus, M.L.; et al. Phenotype of a homozygous CADASIL patient in comparison to 9 age-matched heterozygous patients with the same R133C Notch3 mutation. Stroke 2001, 32, 1767–1774. [Google Scholar] [CrossRef] [PubMed]
  56. Liem, M.K.; Lesnik Oberstein, S.A.; Vollebregt, M.J.; Middelkoop, H.A.; van der Grond, J.; van den Helderman Enden, A.T. Homozygosity for a NOTCH3 mutation in a 65-year-old CADASIL patient with mild symptoms: A family report. J. Neurol. 2008, 255, 1978–1980. [Google Scholar] [CrossRef]
  57. Ragno, M.; Pianese, L.; Morroni, M.; Cacchio, G.; Manca, A.; Di Marzio, F.; Silvestri, S.; Miceli, C.; Scarcella, M.; Onofrj, M.; et al. “CADASIL coma” in an Italian homozygous CADASIL patient: Comparison with clinical and MRI findings in age-matched heterozygous patients with the same G528C NOTCH3 mutation. Neurol. Sci. 2013, 34, 1947–1953. [Google Scholar] [CrossRef]
  58. Soong, B.W.; Liao, Y.C.; Tu, P.H.; Tsai, P.C.; Lee, I.H.; Chung, C.P.; Lee, Y.C. A homozygous NOTCH3 mutation p.R544C and a heterozygous TREX1 variant p.C99MfsX3 in a family with hereditary small vessel disease of the brain. J. Chin. Med. Assoc. 2013, 76, 319–324. [Google Scholar] [CrossRef] [Green Version]
  59. Vinciguerra, C.; Rufa, A.; Bianchi, S.; Sperduto, A.; De Santis, M.; Malandrini, A.; Dotti, M.T.; Federico, A. Homozygosity and severity of phenotypic presentation in a CADASIL family. Neurol. Sci. 2014, 35, 91–93. [Google Scholar] [CrossRef]
  60. Coto, E.; Menendez, M.; Navarro, R.; Garcia-Castro, M.; Alvarez, V. A new de novo Notch3 mutation causing CADASIL. Eur. J. Neurol. 2006, 13, 628–631. [Google Scholar] [CrossRef]
  61. Stojanov, D.; Grozdanovic, D.; Petrovic, S.; Benedeto-Stojanov, D.; Stefanovic, I.; Stojanovic, N.; Ilic, D.N. De novo mutation in the NOTCH3 gene causing CADASIL. Bosn. J. Basic Med. Sci. 2014, 14, 48–50. [Google Scholar] [CrossRef] [PubMed]
  62. Dichgans, M.; Ludwig, H.; Muller-Hocker, J.; Messerschmidt, A.; Gasser, T. Small in-frame deletions and missense mutations in CADASIL: 3D models predict misfolding of Notch3 EGF-like repeat domains. Eur. J. Hum. Genet. 2000, 8, 280–285. [Google Scholar] [CrossRef] [PubMed]
  63. Dotti, M.T.; De Stefano, N.; Bianchi, S.; Malandrini, A.; Battisti, C.; Cardaioli, E.; Federico, A. A novel NOTCH3 frameshift deletion and mitochondrial abnormalities in a patient with CADASIL. Arch. Neurol. 2004, 61, 942–945. [Google Scholar] [CrossRef] [PubMed]
  64. Dichgans, M.; Herzog, J.; Gasser, T. NOTCH3 mutation involving three cysteine residues in a family with typical CADASIL. Neurology 2001, 57, 1714–1717. [Google Scholar] [CrossRef] [PubMed]
  65. Muiño, E.; Gallego-Fabrega, C.; Cullell, N.; Carrera, C.; Torres, N.; Krupinski, J.; Roquer, J.; Montaner, J.; Fernández-Cadenas, I. Systematic Review of Cysteine-Sparing NOTCH3 Missense Mutations in Patients with Clinical Suspicion of CADASIL. Int. J. Mol. Sci. 2017, 18, 1964. [Google Scholar] [CrossRef] [PubMed]
  66. Brass, S.D.; Smith, E.E.; Arboleda-Velasquez, J.F.; Copen, W.A.; Frosch, M.P. Case records of the Massachusetts General Hospital. Case 12-2009. A 46-year-old man with migraine, aphasia, and hemiparesis and similarly affected family members. N. Engl. J. Med. 2009, 360, 1656–1665. [Google Scholar] [CrossRef] [PubMed]
  67. Kim, Y.; Choi, E.J.; Choi, C.G.; Kim, G.; Choi, J.H.; Yoo, H.W.; Kim, J.S. Characteristics of CADASIL in Korea: A novel cysteine-sparing Notch3 mutation. Neurology 2006, 66, 1511–1516. [Google Scholar] [CrossRef]
  68. Ungaro, C.; Mazzei, R.; Conforti, F.L.; Sprovieri, T.; Servillo, P.; Liguori, M.; Citrigno, L.; Gabriele, A.L.; Magariello, A.; Patitucci, A.; et al. CADASIL: Extended polymorphisms and mutational analysis of the NOTCH3 gene. J. Neurosci. Res. 2009, 87, 1162–1167. [Google Scholar] [CrossRef]
  69. Ge, W.; Kuang, H.; Wei, B.; Bo, L.; Xu, Z.; Xu, X.; Geng, D.; Sun, M. A novel cysteine-sparing NOTCH3 mutation in a Chinese family with CADASIL. PLoS ONE 2014, 9, e104533. [Google Scholar] [CrossRef]
  70. Ampuero, I.; Alegre-Abarrategui, J.; Rodal, I.; Espana, A.; Ros, R.; Sendon, J.L.; Galloway, E.G.; Cervello, A.; Caminero, A.B.; Zabala, A.; et al. On the diagnosis of CADASIL. J. Alzheimer’s Dis. 2009, 17, 787–794. [Google Scholar] [CrossRef]
  71. Roy, B.; Maksemous, N.; Smith, R.A.; Menon, S.; Davies, G.; Griffiths, L.R. Two novel mutations and a previously unreported intronic polymorphism in the NOTCH3 gene. Mutat. Res. 2012, 732, 3–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Santa, Y.; Uyama, E.; Chui, D.H.; Arima, M.; Kotorii, S.; Takahashi, K.; Tabira, T. Genetic, clinical and pathological studies of CADASIL in Japan: A partial contribution of Notch3 mutations and implications of smooth muscle cell degeneration for the pathogenesis. J. Neurol. Sci. 2003, 212, 79–84. [Google Scholar] [CrossRef]
  73. Abramycheva, N.; Stepanova, M.; Kalashnikova, L.; Zakharova, M.; Maximova, M.; Tanashyan, M.; Lagoda, O.; Fedotova, E.; Klyushnikov, S.; Konovalov, R.; et al. New mutations in the Notch3 gene in patients with cerebral autosomal dominant arteriopathy with subcortical infarcts and leucoencephalopathy (CADASIL). J. Neurol. Sci. 2015, 349, 196–201. [Google Scholar] [CrossRef] [PubMed]
  74. Homer, V.; George, P.M.; du Toit, S.; Davidson, J.S.; Wilson, C.J. Novel human pathological mutations. Gene symbol: APOB. Disease: Normotriglyceridemic hypobetalipoproteinemia. Hum. Genet. 2007, 121, 645–646. [Google Scholar] [PubMed]
  75. Fouillade, C.; Chabriat, H.; Riant, F.; Mine, M.; Arnoud, M.; Magy, L.; Bousser, M.G.; Tournier-Lasserve, E.; Joutel, A. Activating NOTCH3 mutation in a patient with small-vessel-disease of the brain. Hum. Mutat. 2008, 29, 452. [Google Scholar] [CrossRef] [PubMed]
  76. Nishimoto, Y.; Shibata, M.; Nihonmatsu, M.; Nozaki, H.; Shiga, A.; Shirata, A.; Yamane, K.; Kosakai, A.; Takahashi, K.; Nishizawa, M.; et al. A novel mutation in the HTRA1 gene causes CARASIL without alopecia. Neurology 2011, 76, 1353–1355. [Google Scholar] [CrossRef]
  77. Bersano, A.; Ranieri, M.; Ciammola, A.; Cinnante, C.; Lanfranconi, S.; Dotti, M.T.; Candelise, L.; Baschirotto, C.; Ghione, I.; Ballabio, E.; et al. Considerations on a mutation in the NOTCH3 gene sparing a cysteine residue: A rare polymorphism rather than a CADASIL variant. Funct. Neurol. 2012, 27, 247–252. [Google Scholar] [PubMed]
  78. Chavoshi Tarzjani, S.P.; Shahzadeh Fazeli, S.A.; Sanati, M.H.; Mirzayee, Z. Genetic study of the NOTCH3 gene in CADASIL patients. Egypt. J. Med. Hum. Genet. 2018, 19, 425–427. [Google Scholar] [CrossRef]
  79. Wang, Z.; Yuan, Y.; Zhang, W.; Lv, H.; Hong, D.; Chen, B.; Liu, Y.; Luan, X.; Xie, S.; Wu, S. NOTCH3 mutations and clinical features in 33 mainland Chinese families with CADASIL. J. Neurol. Neurosurg. Psychiatry 2011, 82, 534–539. [Google Scholar] [CrossRef]
  80. Mizuno, T.; Muranishi, M.; Torugun, T.; Tango, H.; Nagakane, Y.; Kudeken, T.; Kawase, Y.; Kawabe, K.; Oshima, F.; Yaoi, T.; et al. Two Japanese CADASIL families exhibiting Notch3 mutation R75P not involving cysteine residue. Intern. Med. (Tokyo Jpn.) 2008, 47, 2067–2072. [Google Scholar] [CrossRef]
  81. Kotorii, S.; Takahashi, K.; Kamimura, K.; Nishio, T.; Arima, K.; Yamada, H.; Uyama, E.; Uchino, M.; Suenaga, A.; Matsumoto, M.; et al. Mutations of the notch3 gene in non-caucasian patients with suspected CADASIL syndrome. Dement. Geriatr. Cogn. Disord. 2001, 12, 185–193. [Google Scholar] [CrossRef] [PubMed]
  82. Scheid, R.; Heinritz, W.; Leyhe, T.; Thal, D.R.; Schober, R.; Strenge, S.; von Cramon, D.Y.; Froster, U.G. Cysteine-sparing notch3 mutations: Cadasil or cadasil variants? Neurology 2008, 71, 774–776. [Google Scholar] [CrossRef] [PubMed]
  83. Mazzei, R.; Guidetti, D.; Ungaro, C.; Conforti, F.L.; Muglia, M.; Cenacchi, G.; Lanza, P.L.; Patitucci, A.; Sprovieri, T.; Riguzzi, P.; et al. First evidence of a pathogenic insertion in the NOTCH3 gene causing CADASIL. J. Neurol. Neurosurg. Psychiatry 2008, 79, 108–110. [Google Scholar] [CrossRef] [PubMed]
  84. Kilarski, L.L.; Rutten-Jacobs, L.C.; Bevan, S.; Baker, R.; Hassan, A.; Hughes, D.A.; Markus, H.S. Prevalence of CADASIL and Fabry Disease in a Cohort of MRI Defined Younger Onset Lacunar Stroke. PLoS ONE 2015, 10, e0136352. [Google Scholar] [CrossRef] [PubMed]
  85. Cai, B.; Zeng, J.; Lin, Y.; Lin, Y.; Lin, W.; Lin, W.; Li, Z.; Wang, N. A frameshift mutation in HTRA1 expands CARASIL syndrome and peripheral small arterial disease to the Chinese population. Neurol. Sci. 2015, 36, 1387–1391. [Google Scholar] [CrossRef] [PubMed]
  86. Fukutake, T.; Hirayama, K. Familial young-adult-onset arteriosclerotic leukoencephalopathy with alopecia and lumbago without arterial hypertension. Eur. Neurol. 1995, 35, 69–79. [Google Scholar] [CrossRef]
  87. Oide, T.; Nakayama, H.; Yanagawa, S.; Ito, N.; Ikeda, S.; Arima, K. Extensive loss of arterial medial smooth muscle cells and mural extracellular matrix in cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL). Neuropathology 2008, 28, 132–142. [Google Scholar] [CrossRef]
  88. Arima, K.; Yanagawa, S.; Ito, N.; Ikeda, S. Cerebral arterial pathology of CADASIL and CARASIL (Maeda syndrome). Neuropathology 2003, 23, 327–334. [Google Scholar] [CrossRef]
  89. Fukutake, T. Cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL): From discovery to gene identification. J. Stroke Cerebrovasc. Dis. 2011, 20, 85–93. [Google Scholar] [CrossRef]
  90. Okeda, R.; Murayama, S.; Sawabe, M.; Kuroiwa, T. Pathology of the cerebral artery in Binswanger’s disease in the aged: Observation by serial sections and morphometry of the cerebral arteries. Neuropathology 2004, 24, 21–29. [Google Scholar] [CrossRef]
  91. Chen, Y.; He, Z.; Meng, S.; Li, L.; Yang, H.; Zhang, X. A novel mutation of the high-temperature requirement A serine peptidase 1 (HTRA1) gene in a Chinese family with cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL). J. Int. Med. Res. 2013, 41, 1445–1455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Wang, X.L.; Li, C.F.; Guo, H.W.; Cao, B.Z. A novel mutation in the HTRA1 gene identified in Chinese CARASIL pedigree. CNS Neurosci. Ther. 2012, 18, 867–869. [Google Scholar] [CrossRef] [PubMed]
  93. Di Donato, I.; Bianchi, S.; Gallus, G.N.; Cerase, A.; Taglia, I.; Pescini, F.; Nannucci, S.; Battisti, C.; Inzitari, D.; Pantoni, L.; et al. Heterozygous mutations of HTRA1 gene in patients with familial cerebral small vessel disease. CNS Neurosci. Ther. 2017, 23, 759–765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Mendioroz, M.; Fernandez-Cadenas, I.; Del Rio-Espinola, A.; Rovira, A.; Sole, E.; Fernandez-Figueras, M.T.; Garcia-Patos, V.; Sastre-Garriga, J.; Domingues-Montanari, S.; Alvarez-Sabin, J.; et al. A missense HTRA1 mutation expands CARASIL syndrome to the Caucasian population. Neurology 2010, 75, 2033–2035. [Google Scholar] [CrossRef] [PubMed]
  95. Bianchi, S.; Di Palma, C.; Gallus, G.N.; Taglia, I.; Poggiani, A.; Rosini, F.; Rufa, A.; Muresanu, D.F.; Cerase, A.; Dotti, M.T.; et al. Two novel HTRA1 mutations in a European CARASIL patient. Neurology 2014, 82, 898–900. [Google Scholar] [CrossRef] [PubMed]
  96. Bayrakli, F.; Balaban, H.; Gurelik, M.; Hizmetli, S.; Topaktas, S. Mutation in the HTRA1 gene in a patient with degenerated spine as a component of CARASIL syndrome. Turk. Neurosurg. 2014, 24, 67–69. [Google Scholar] [CrossRef] [PubMed]
  97. Ibrahimi, M.; Nozaki, H.; Lee, A.; Onodera, O.; Reichwein, R.; Wicklund, M.; El-Ghanem, M. A CARASIL Patient from Americas with Novel Mutation and Atypical Features: Case Presentation and Literature Review. Cerebrovasc. Dis. (Basel Switz.) 2017, 44, 135–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Bougea, A.; Velonakis, G.; Spantideas, N.; Anagnostou, E.; Paraskevas, G.; Kapaki, E.; Kararizou, E. The first Greek case of heterozygous cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy: An atypical clinico-radiological presentation. Neuroradiol. J. 2017, 30, 583–585. [Google Scholar] [CrossRef]
  99. Nozaki, H.; Kato, T.; Nihonmatsu, M.; Saito, Y.; Mizuta, I.; Noda, T.; Koike, R.; Miyazaki, K.; Kaito, M.; Ito, S.; et al. Distinct molecular mechanisms of HTRA1 mutants in manifesting heterozygotes with CARASIL. Neurology 2016, 86, 1964–1974. [Google Scholar] [CrossRef]
  100. Menezes Cordeiro, I.; Nzwalo, H.; Sa, F.; Ferreira, R.B.; Alonso, I.; Afonso, L.; Basilio, C. Shifting the CARASIL paradigm: Report of a non-Asian family and literature review. Stroke 2015, 46, 1110–1112. [Google Scholar] [CrossRef]
  101. Nozaki, H.; Nishizawa, M.; Onodera, O. Features of cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy. Stroke 2014, 45, 3447–3453. [Google Scholar] [CrossRef] [PubMed]
  102. Herve, D.; Chabriat, H.; Rigal, M.; Dalloz, M.A.; Kawkabani Marchini, A.; De Lepeleire, J.; Fontaine, B.; Ceuterick-de Groote, C.; Alili, N.; Mine, M.; et al. A novel hereditary extensive vascular leukoencephalopathy mapping to chromosome 20q13. Neurology 2012, 79, 2283–2287. [Google Scholar] [CrossRef] [PubMed]
  103. d’Azzo, A.; Bonten, E. Molecular Mechanisms of Pathogenesis in a Glycosphigolipid and a Glycoprotein Storage Disease. Biochem. Soc. Trans. 2010, 38, 1453–1457. [Google Scholar] [CrossRef] [PubMed]
  104. Strisciuglio, P.; Parenti, G.; Giudice, C.; Lijoi, S.; Hoogeveen, A.T.; d’Azzo, A. The presence of a reduced amount of 32-kd “protective” protein is a distinct biochemical finding in late infantile galactosialidosis. Hum. Genet. 1988, 80, 304–306. [Google Scholar] [CrossRef] [PubMed]
  105. Hammond, T.R.; Gadea, A.; Dupree, J.; Kerninon, C.; Nait-Oumesmar, B.; Aguirre, A.; Gallo, V. Astrocyte-derived endothelin-1 inhibits remyelination through notch activation. Neuron 2014, 81, 588–602. [Google Scholar] [CrossRef] [PubMed]
  106. Baba, Y.; Ghetti, B.; Baker, M.C.; Uitti, R.J.; Hutton, M.L.; Yamaguchi, K.; Bird, T.; Lin, W.; DeLucia, M.W.; Dickson, D.W.; et al. Hereditary diffuse leukoencephalopathy with spheroids: Clinical, pathologic and genetic studies of a new kindred. Acta Neuropathol. 2006, 111, 300–311. [Google Scholar] [CrossRef]
  107. Konno, T.; Yoshida, K.; Mizuno, T.; Kawarai, T.; Tada, M.; Nozaki, H.; Ikeda, S.I.; Nishizawa, M.; Onodera, O.; Wszolek, Z.K.; et al. Clinical and genetic characterization of adult-onset leukoencephalopathy with axonal spheroids and pigmented glia associated with CSF1R mutation. Eur. J. Neurol. 2017, 24, 37–45. [Google Scholar] [CrossRef]
  108. Sundal, C.; Ekholm, S.; Andersen, O. White matter disorders with autosomal dominant heredity: A review with personal clinical case studies and their MRI findings. Acta Neurol. Scand. 2010, 121, 328–337. [Google Scholar] [CrossRef]
  109. Sundal, C.; Jonsson, L.; Ljungberg, M.; Zhong, J.; Tian, W.; Zhu, T.; Linden, T.; Borjesson-Hanson, A.; Andersen, O.; Ekholm, S. Different stages of white matter changes in the original HDLS family revealed by advanced MRI techniques. J. Neuroimaging 2014, 24, 444–452. [Google Scholar] [CrossRef]
  110. Axelsson, R.; Roytta, M.; Sourander, P.; Akesson, H.O.; Andersen, O. Hereditary diffuse leucoencephalopathy with spheroids. Acta Psychiatr. Scand. Suppl. 1984, 314, 1–65. [Google Scholar]
  111. Prinz, M.; Priller, J. Microglia and brain macrophages in the molecular age: From origin to neuropsychiatric disease. Nat. Rev. Neurosci. 2014, 15, 300–312. [Google Scholar] [CrossRef] [PubMed]
  112. Sasaki, A. Microglia and brain macrophages: An update. Neuropathology 2017, 37, 452–464. [Google Scholar] [CrossRef] [PubMed]
  113. Nicholson, A.M.; Baker, M.C.; Finch, N.A.; Rutherford, N.J.; Wider, C.; Graff-Radford, N.R.; Nelson, P.T.; Clark, H.B.; Wszolek, Z.K.; Dickson, D.W.; et al. CSF1R mutations link POLD and HDLS as a single disease entity. Neurology 2013, 80, 1033–1040. [Google Scholar] [CrossRef] [PubMed]
  114. Locascio, L.E.; Donoghue, D.J. KIDs rule: Regulatory phosphorylation of RTKs. Trends Biochem. Sci. 2013, 38, 75–84. [Google Scholar] [CrossRef]
  115. Kraya, T.; Quandt, D.; Pfirrmann, T.; Kindermann, A.; Lampe, L.; Schroeter, M.L.; Kohlhase, J.; Stoevesandt, D.; Hoffmann, K.; Villavicencio-Lorini, P. Functional characterization of a novel CSF1R mutation causing hereditary diffuse leukoencephalopathy with spheroids. Mol. Genet. Genom. Med. 2019, 7, e00595. [Google Scholar] [CrossRef] [PubMed]
  116. Vahedi, K.; Mascart, F.; Mary, J.Y.; Laberenne, J.E.; Bouhnik, Y.; Morin, M.C.; Ocmant, A.; Velly, C.; Colombel, J.F.; Matuchansky, C. Reliability of antitransglutaminase antibodies as predictors of gluten-free diet compliance in adult celiac disease. Am. J. Gastroenterol. 2003, 98, 1079–1087. [Google Scholar] [CrossRef]
  117. Shah, S.; Ellard, S.; Kneen, R.; Lim, M.; Osborne, N.; Rankin, J.; Stoodley, N.; van der Knaap, M.; Whitney, A.; Jardine, P. Childhood presentation of COL4A1 mutations. Dev. Med. Child Neurol. 2012, 54, 569–574. [Google Scholar] [CrossRef] [Green Version]
  118. Lanfranconi, S.; Markus, H.S. COL4A1 mutations as a monogenic cause of cerebral small vessel disease: A systematic review. Stroke 2010, 41, e513–e518. [Google Scholar] [CrossRef]
  119. Verbeek, E.; Meuwissen, M.E.; Verheijen, F.W.; Govaert, P.P.; Licht, D.J.; Kuo, D.S.; Poulton, C.J.; Schot, R.; Lequin, M.H.; Dudink, J.; et al. COL4A2 mutation associated with familial porencephaly and small-vessel disease. Eur. J. Hum. Genet. 2012, 20, 844–851. [Google Scholar] [CrossRef] [Green Version]
  120. Khoshnoodi, J.; Pedchenko, V.; Hudson, B.G. Mammalian collagen IV. Microsc. Res. Tech. 2008, 71, 357–370. [Google Scholar] [CrossRef] [Green Version]
  121. Gould, D.B.; Phalan, F.C.; Breedveld, G.J.; van Mil, S.E.; Smith, R.S.; Schimenti, J.C.; Aguglia, U.; van der Knaap, M.S.; Heutink, P.; John, S.W. Mutations in Col4a1 cause perinatal cerebral hemorrhage and porencephaly. Science 2005, 308, 1167–1171. [Google Scholar] [CrossRef] [PubMed]
  122. Plaisier, E.; Gribouval, O.; Alamowitch, S.; Mougenot, B.; Prost, C.; Verpont, M.C.; Marro, B.; Desmettre, T.; Cohen, S.Y.; Roullet, E.; et al. COL4A1 mutations and hereditary angiopathy, nephropathy, aneurysms, and muscle cramps. N. Engl. J. Med. 2007, 357, 2687–2695. [Google Scholar] [CrossRef] [PubMed]
  123. Kuo, D.S.; Labelle-Dumais, C.; Gould, D.B. COL4A1 and COL4A2 mutations and disease: Insights into pathogenic mechanisms and potential therapeutic targets. Hum. Mol. Genet. 2012, 21, R97–R110. [Google Scholar] [CrossRef] [PubMed]
  124. Meuwissen, M.E.; Halley, D.J.; Smit, L.S.; Lequin, M.H.; Cobben, J.M.; de Coo, R.; van Harssel, J.; Sallevelt, S.; Woldringh, G.; van der Knaap, M.S.; et al. The expanding phenotype of COL4A1 and COL4A2 mutations: Clinical data on 13 newly identified families and a review of the literature. Genet. Med. 2015, 17, 843–853. [Google Scholar] [CrossRef] [PubMed]
  125. Kato, M.; Wang, L.; Putta, S.; Wang, M.; Yuan, H.; Sun, G.; Lanting, L.; Todorov, I.; Rossi, J.J.; Natarajan, R. Post-transcriptional up-regulation of Tsc-22 by Ybx1, a target of miR-216a, mediates TGF-{beta}-induced collagen expression in kidney cells. J. Biol. Chem. 2010, 285, 34004–34015. [Google Scholar] [CrossRef] [PubMed]
  126. Chung, A.C.; Huang, X.R.; Meng, X.; Lan, H.Y. miR-192 mediates TGF-beta/Smad3-driven renal fibrosis. J. Am. Soc. Nephrol. 2010, 21, 1317–1325. [Google Scholar] [CrossRef] [PubMed]
  127. Jeanne, M.; Labelle-Dumais, C.; Jorgensen, J.; Kauffman, W.B.; Mancini, G.M.; Favor, J.; Valant, V.; Greenberg, S.M.; Rosand, J.; Gould, D.B. COL4A2 Mutations Impair COL4A1 and COL4A2 Secretion and Cause Hemorrhagic Stroke. Am. J. Hum. Genet. 2012, 90, 91–101. [Google Scholar] [CrossRef] [Green Version]
  128. Yoneda, Y.; Haginoya, K.; Arai, H.; Yamaoka, S.; Tsurusaki, Y.; Doi, H.; Miyake, N.; Yokochi, K.; Osaka, H.; Kato, M.; et al. De novo and inherited mutations in COL4A2, encoding the type IV collagen alpha2 chain cause porencephaly. Am. J. Hum. Genet. 2012, 90, 86–90. [Google Scholar] [CrossRef]
  129. Alavi, M.V.; Mao, M.; Pawlikowski, B.T.; Kvezereli, M.; Duncan, J.L.; Libby, R.T.; John, S.W.M.; Gould, D.B. Col4a1 mutations cause progressive retinal neovascular defects and retinopathy. Sci. Rep. 2016, 6, 18602. [Google Scholar] [CrossRef]
  130. Schmidt, H.; Zeginigg, M.; Wiltgen, M.; Freudenberger, P.; Petrovic, K.; Cavalieri, M.; Gider, P.; Enzinger, C.; Fornage, M.; Debette, S.; et al. Genetic variants of the NOTCH3 gene in the elderly and magnetic resonance imaging correlates of age-related cerebral small vessel disease. Brain 2011, 134, 3384–3397. [Google Scholar] [CrossRef] [Green Version]
  131. Weng, Y.C.; Sonni, A.; Labelle-Dumais, C.; de Leau, M.; Kauffman, W.B.; Jeanne, M.; Biffi, A.; Greenberg, S.M.; Rosand, J.; Gould, D.B. COL4A1 mutations in patients with sporadic late-onset intracerebral hemorrhage. Ann. Neurol. 2012, 71, 470–477. [Google Scholar] [CrossRef] [PubMed]
  132. Weng, J.; Li, Y.; Xu, W.; Shi, L.; Zhang, Q.; Zhu, D.; Hu, Y.; Zhou, Z.; Yan, X.; Tian, H.; et al. Effect of intensive insulin therapy on beta-cell function and glycaemic control in patients with newly diagnosed type 2 diabetes: A multicentre randomised parallel-group trial. Lancet (Lond. Engl.) 2008, 371, 1753–1760. [Google Scholar] [CrossRef]
  133. Robin, N.H.; Taylor, C.J.; McDonald-McGinn, D.M.; Zackai, E.H.; Bingham, P.; Collins, K.J.; Earl, D.; Gill, D.; Granata, T.; Guerrini, R.; et al. Polymicrogyria and deletion 22q11.2 syndrome: Window to the etiology of a common cortical malformation. Am. J. Med. Genet. A 2006, 140, 2416–2425. [Google Scholar] [CrossRef] [PubMed]
  134. Brady, R.O.; Gal, A.E.; Bradley, R.M.; Martensson, E.; Warshaw, A.L.; Laster, L. Enzymatic defect in Fabry’s disease. Ceramidetrihexosidase deficiency. N. Engl. J. Med. 1967, 276, 1163–1167. [Google Scholar] [CrossRef] [PubMed]
  135. Guffon, N. Clinical presentation in female patients with Fabry disease. J. Med. Genet. 2003, 40, e38. [Google Scholar] [CrossRef] [PubMed]
  136. Arends, M.; Wanner, C.; Hughes, D.; Mehta, A.; Oder, D.; Watkinson, O.T.; Elliott, P.M.; Linthorst, G.E.; Wijburg, F.A.; Biegstraaten, M.; et al. Characterization of Classical and Nonclassical Fabry Disease: A Multicenter Study. J. Am. Soc. Nephrol. 2017, 28, 1631–1641. [Google Scholar] [CrossRef] [PubMed]
  137. Buechner, S.; Moretti, M.; Burlina, A.P.; Cei, G.; Manara, R.; Ricci, R.; Mignani, R.; Parini, R.; Di Vito, R.; Giordano, G.P.; et al. Central nervous system involvement in Anderson-Fabry disease: A clinical and MRI retrospective study. J. Neurol. Neurosurg. Psychiatry 2008, 79, 1249–1254. [Google Scholar] [CrossRef] [PubMed]
  138. Hopkin, R.J.; Jefferies, J.L.; Laney, D.A.; Lawson, V.H.; Mauer, M.; Taylor, M.R.; Wilcox, W.R. The management and treatment of children with Fabry disease: A United States-based perspective. Mol. Genet. Metab. 2016, 117, 104–113. [Google Scholar] [CrossRef] [PubMed]
  139. Gal, A. Molecular Genetics of Fabry Disease and Genotype—Phenotype Correlation. In Fabry Disease; Elstein, D., Altarescu, G., Beck, M., Eds.; Springer: Dordrecht, The Netherlands, 2010; pp. 3–19. [Google Scholar]
  140. Verovnik, F.; Benko, D.; Vujkovac, B.; Linthorst, G.E. Remarkable variability in renal disease in a large Slovenian family with Fabry disease. Eur. J. Hum. Genet. 2004, 12, 678–681. [Google Scholar] [CrossRef] [PubMed]
  141. Froissart, R.; Guffon, N.; Vanier, M.T.; Desnick, R.J.; Maire, I. Fabry disease: D313Y is an alpha-galactosidase A sequence variant that causes pseudodeficient activity in plasma. Mol. Genet. Metab. 2003, 80, 307–314. [Google Scholar] [CrossRef]
  142. Shi, Q.; Chen, J.; Pongmoragot, J.; Lanthier, S.; Saposnik, G. Prevalence of Fabry disease in stroke patients—A systematic review and meta-analysis. J. Stroke Cerebrovasc. Dis. 2014, 23, 985–992. [Google Scholar] [CrossRef] [PubMed]
  143. Meikle, P.J.; Hopwood, J.J.; Clague, A.E.; Carey, W.F. Prevalence of lysosomal storage disorders. JAMA 1999, 281, 249–254. [Google Scholar] [CrossRef] [PubMed]
  144. Houge, G.; Skarbovik, A.J. Fabry disease—A diagnostic and therapeutic challenge. Tidsskr. Nor. Laegeforen. 2005, 125, 1004–1006. [Google Scholar] [PubMed]
  145. Yasuda, M.; Shabbeer, J.; Benson, S.D.; Maire, I.; Burnett, R.M.; Desnick, R.J. Fabry disease: Characterization of alpha-galactosidase A double mutations and the D313Y plasma enzyme pseudodeficiency allele. Hum. Mutat. 2003, 22, 486–492. [Google Scholar] [CrossRef] [PubMed]
  146. Ferreira, S.; Ortiz, A.; Germain, D.P.; Viana-Baptista, M.; Caldeira-Gomes, A.; Camprecios, M.; Fenollar-Cortes, M.; Gallegos-Villalobos, A.; Garcia, D.; Garcia-Robles, J.A.; et al. The alpha-galactosidase A p.Arg118Cys variant does not cause a Fabry disease phenotype: Data from individual patients and family studies. Mol. Genet. Metab. 2015, 114, 248–258. [Google Scholar] [CrossRef]
  147. Meng, Y.; Zhang, W.M.; Shi, H.P.; Wei, M.; Huang, S.Z. Clinical manifestations and mutation study in 16 Chinese patients with Fabry disease. Zhonghua Yi Xue Za Zhi 2010, 90, 551–554. [Google Scholar] [PubMed]
  148. Mukdsi, J.H.; Gutiérrez, S.; Barrón, B.; Novoa, P.; Fernández, S.; de Diller, A.B.; Torres, A.I.; Formica, R.N., Jr.; Orías, M. A renal variant of Fabry disease: A case with a novel Gal A hemizygote mutation. J. Nephropathol. 2012, 1, 194–197. [Google Scholar] [CrossRef] [Green Version]
  149. Sanchez-Niño, M.D.; Ortiz, A. Is it or is it not a pathogenic mutation? Is it or is it not the podocyte? J. Nephropathol. 2012, 1, 152–154. [Google Scholar] [CrossRef] [Green Version]
  150. Baptista, M.V.; Ferreira, S.; Pinho, E.M.T.; Carvalho, M.; Cruz, V.T.; Carmona, C.; Silva, F.A.; Tuna, A.; Rodrigues, M.; Ferreira, 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, 431–436. [Google Scholar] [CrossRef]
  151. Gaspar, P.; Herrera, J.; Rodrigues, D.; Cerezo, S.; Delgado, R.; Andrade, C.F.; Forascepi, R.; Macias, J.; del Pino, M.D.; Prados, M.D.; et al. Frequency of Fabry disease in male and female haemodialysis patients in Spain. BMC Med. Genet. 2010, 11, 19. [Google Scholar] [CrossRef]
  152. Elliott, P.; Baker, R.; Pasquale, F.; Quarta, G.; Ebrahim, H.; Mehta, A.B.; Hughes, D.A. Prevalence of Anderson-Fabry disease in patients with hypertrophic cardiomyopathy: The European Anderson-Fabry Disease survey. Heart (Br. Card. Soc.) 2011, 97, 1957–1960. [Google Scholar] [CrossRef] [PubMed]
  153. Rolfs, A.; Fazekas, F.; Grittner, U.; Dichgans, M.; Martus, P.; Holzhausen, M.; Bottcher, T.; Heuschmann, P.U.; Tatlisumak, T.; Tanislav, C.; et al. Acute cerebrovascular disease in the young: The Stroke in Young Fabry Patients study. Stroke 2013, 44, 340–349. [Google Scholar] [CrossRef] [PubMed]
  154. Turaca, L.T.; Pessoa, J.G.; Motta, F.L.; Munoz Rojas, M.V.; Muller, K.B.; Lourenco, C.M.; Junior Marques, W.; D’Almeida, V.; Martins, A.M.; Pesquero, J.B. New mutations in the GLA gene in Brazilian families with Fabry disease. J. Hum. Genet. 2012, 57, 347–351. [Google Scholar] [CrossRef] [PubMed]
  155. Spada, M.; Pagliardini, S.; Yasuda, M.; Tukel, T.; Thiagarajan, G.; Sakuraba, H.; Ponzone, A.; Desnick, R.J. High Incidence of Later-Onset Fabry Disease Revealed by Newborn Screening. Am. J. Hum. Genet. 2006, 79, 31–40. [Google Scholar] [CrossRef] [PubMed]
  156. Hsu, T.R.; Niu, D.M. Fabry disease: Review and experience during newborn screening. Trends Cardiovasc. Med. 2018, 28, 274–281. [Google Scholar] [CrossRef] [PubMed]
  157. Parfenov, V.A.; Ostroumova, O.D.; Ostroumova, T.M.; Kochetkov, A.I.; Fateeva, V.V.; Khacheva, K.K.; Khakimova, G.R.; Epstein, O.I. Vascular cognitive impairment: Pathophysiological mechanisms, insights into structural basis, and perspectives in specific treatments. Neuropsychiatr. Dis. Treat. 2019, 15, 1381–1402. [Google Scholar] [CrossRef] [PubMed]
  158. Giau, V.V.; Bagyinszky, E.; An, S.S.A.; Kim, S.Y. Role of apolipoprotein E in neurodegenerative diseases. Neuropsychiatr. Dis. Treat. 2015, 11, 1723–1737. [Google Scholar] [CrossRef] [PubMed]
  159. Giau, V.V.; Lee, H.; Shim, K.H.; Bagyinszky, E.; An, S.S.A. Genome-editing applications of CRISPR-Cas9 to promote in vitro studies of Alzheimer’s disease. Clin. Interv. Aging 2018, 13, 221–233. [Google Scholar] [CrossRef]
  160. Shen, L.; An, S.S.A.; Bagyinszky, E.; Van Giau, V.; Choi, S.H.; Kim, S.Y. Novel GRN mutations in Koreans with Alzheimer’s disease. Mol. Cell. Toxicol. 2019, 15, 345–352. [Google Scholar] [CrossRef]
  161. Beaufort, N.; Scharrer, E.; Kremmer, E.; Lux, V.; Ehrmann, M.; Huber, R.; Houlden, H.; Werring, D.; Haffner, C.; Dichgans, M. Cerebral small vessel disease-related protease HtrA1 processes latent TGF-beta binding protein 1 and facilitates TGF-beta signaling. Proc. Natl. Acad. Sci. USA 2014, 111, 16496–16501. [Google Scholar] [CrossRef]
  162. Richards, A.; van den Maagdenberg, A.M.; Jen, J.C.; Kavanagh, D.; Bertram, P.; Spitzer, D.; Liszewski, M.K.; Barilla-Labarca, M.L.; Terwindt, G.M.; Kasai, Y.; et al. C-terminal truncations in human 3’–5’ DNA exonuclease TREX1 cause autosomal dominant retinal vasculopathy with cerebral leukodystrophy. Nat. Genet. 2007, 39, 1068–1070. [Google Scholar] [CrossRef] [PubMed]
  163. French, C.R.; Seshadri, S.; Destefano, A.L.; Fornage, M.; Arnold, C.R.; Gage, P.J.; Skarie, J.M.; Dobyns, W.B.; Millen, K.J.; Liu, T.; et al. Mutation of FOXC1 and PITX2 induces cerebral small-vessel disease. J. Clin. Investig. 2014, 124, 4877–4881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Sondergaard, C.B.; Nielsen, J.E.; Hansen, C.K.; Christensen, H. Hereditary cerebral small vessel disease and stroke. Clin. Neurol. Neurosurg. 2017, 155, 45–57. [Google Scholar] [CrossRef] [PubMed]
  165. Giau, V.V.; Bagyinszky, E.; An, S.S.A. Potential Fluid Biomarkers for the Diagnosis of Mild Cognitive Impairment. Int. J. Mol. Sci. 2019, 20, 4149. [Google Scholar] [CrossRef] [PubMed]
  166. Fleischer, V.; Radetz, A.; Ciolac, D.; Muthuraman, M.; Gonzalez-Escamilla, G.; Zipp, F.; Groppa, S. Graph Theoretical Framework of Brain Networks in Multiple Sclerosis: A Review of Concepts. Neuroscience 2017, 403, 35–53. [Google Scholar] [CrossRef] [PubMed]
  167. Giau, V.V.; An, S.S.A.; Hulme, J.J. Mitochondrial therapeutic interventions in Alzheimer’s disease. Neurol. Sci. 2018, 395, 62–70. [Google Scholar] [CrossRef] [PubMed]
  168. Van Giau, V.; An, S.S. Emergence of exosomal miRNAs as a diagnostic biomarker for Alzheimer’s disease. J. Neurol. Sci. 2016, 360, 141–152. [Google Scholar] [CrossRef]
Figure 1. Pathogenesis of cerebral SVD manifestations in development and its progression.
Figure 1. Pathogenesis of cerebral SVD manifestations in development and its progression.
Ijms 20 04298 g001
Figure 2. Multimodal imaging analyses in CADASIL and SVCI patients with and without NOTCH3 variants. WMH frequency maps of forms of CADASIL (A) and comparison of frequency maps between the typical CADASIL and SVCI groups (B). (A-1) Typical CADASIL patients show extensive WMH distributed throughout the periventricle, posterior temporal white matter, and anterior temporal white matter. (A-2) SVCI patients with NOTCH3 variant and (A-3) SVCI patients without NOTCH3 variants showed similar WMH frequency maps. (B-1,B-2) Typical CADASIL cases reveal significantly prevalent WMH distribution in the bilateral posterior temporal region compared with SVCI patients with and without NOTCH3 variants. Reprinted with permission from ref [40]. Copyright under a CC BY license (Creative Commons Attribution 4.0 International License).
Figure 2. Multimodal imaging analyses in CADASIL and SVCI patients with and without NOTCH3 variants. WMH frequency maps of forms of CADASIL (A) and comparison of frequency maps between the typical CADASIL and SVCI groups (B). (A-1) Typical CADASIL patients show extensive WMH distributed throughout the periventricle, posterior temporal white matter, and anterior temporal white matter. (A-2) SVCI patients with NOTCH3 variant and (A-3) SVCI patients without NOTCH3 variants showed similar WMH frequency maps. (B-1,B-2) Typical CADASIL cases reveal significantly prevalent WMH distribution in the bilateral posterior temporal region compared with SVCI patients with and without NOTCH3 variants. Reprinted with permission from ref [40]. Copyright under a CC BY license (Creative Commons Attribution 4.0 International License).
Ijms 20 04298 g002
Figure 3. Worldwide distribution of CSF1R mutations. The countries in which the mutation was reported were shown in purple. This world map was created using mapchart (http://mapchart.net/).
Figure 3. Worldwide distribution of CSF1R mutations. The countries in which the mutation was reported were shown in purple. This world map was created using mapchart (http://mapchart.net/).
Ijms 20 04298 g003
Table 1. Characteristics of single-gene disorders causing cerebral SVD.
Table 1. Characteristics of single-gene disorders causing cerebral SVD.
DiseasesInheritanceChromosome Locus GeneMutation FindingsProteinCerebral FeaturesNeuroimaging FindingPathological FindingsReferences
CADASILAutosomal dominant19q12NOTCH3Over 256 missense mutations or rare deletions and insertions were reportedTransmembrane receptorMigraine with & aura, recurrent was chemic strokes, mood disturbance, cognitive decline, disability > death 65–70 years WMH, lacunar infarcts, dilated PVS, micro bleeds, brain atrophyTypical of cerebral SVD plus in anterior temporal lobe and external capsularGranular osmiophilic material found in the walls of affected arterioles[27]
CARASILAutosomal recessive10q25HTRA1At least 17 mutations were identified in 25 families worldwideHtrA serine peptidase/protease 1Recurrent is chemic strokes, Cognitive decline, disability > deathSimilar to CADASIL, diffuse WM lesions and small infarcts in basal gangliaArteriosclerotic changes, WM changes. No GOM deposition. Hyaline degeneration and thickening and splitting of internal lamina[28,29]
CARASALAutosomal dominant20q13.12CTSAMany galactosialidosis patients related to CTSA gene point mutations were reportedCathepsin A (CathA)Therapy-resistant hypertension, strokes, and slow and late cognitive deteriorationA diffuse, progressive leukoencephalopathy preceding the onset of strokes and disproportionate to the degree of clinical severityEndothelin-1 overexpression coincides with increased numbers of premyelinating OPCs, decreased MBP amounts, abundance of axons without myelin, and features of remyelination failure[30]
HDLSAutosomal dominant5q32CSF1RApproximately 60 pathogenic variants have been reported in patients with HDLSCSF-1 receptorStroke episodes with pyramidal, bulbar andDiffuse WM lesions, lacunar strokes and atrophyDiffuse gliosis, moderate loss of axons and many axonal spheroids[31]
COL4-related disordersAutosomal dominant13q34COL4A, COL4A2Over 50 types of mutations have been reported to datCollagen type IV, alpha chainsInfantileemiparesis, intracerebralhemorrhage (perinatal, youngoradult) porencephaliccysts, microbleeds, WMH, intracranial aneurysms (HANAC)Typical of cerebral SVDDefects in the basement membrane[32]
FDX-linkedXq22GLAAround 585 pathogenic mutations have been reported in the GLA gene to dateLysosomal α-galactosidase ATypically, stroke is considered a manifestation of end stageMultifocal WMH lesions, intracranial arterial dolichectasiaLysomal storage materials in vascular endothelial cells and smooth muscle cells[33,34]
CADASIL: Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy; CARASIL: Cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy; AD: autosomal dominant; AR, autosomal recessive; SVD: small vessel disease; WMH: white matter hyperintensities; HDLS: Hereditary Diffuse Leukoencephalopathy with Spheroids; RVCL: retinal vasculopathy with cerebral leukodystrophy; FD: Fabry disease; OPC: Oligodendrocyteprogenitor cells; MBP: myelin basic protein.
Table 2. Various typical clinical CADASIL-causing variants in NOTCH3 that do not affect cysteine amino acids.
Table 2. Various typical clinical CADASIL-causing variants in NOTCH3 that do not affect cysteine amino acids.
MutationExonAge of OnsetGenderPopulationClinical PhenotypesReferences
p.Arg61Trp246NAAmericanMigraine, aphasia, hemiparesis, probable familial[66]
p.Arg75Pro353MKoreanTypical CADASIL symptoms, granular osmiophilic granules on skin biopsy in a patient. Probable positive family history[67]
p.Arg75Pro47F
p.Arg75Pro65M
p.Arg75Pro40–50sFJapaneseProband: depression, akinetic mutism, pseudobalbar palsy,
and tetraplegia; MRI: mild frontal and temporal lobe atrophy multiple cerebral infarctions at the bilateral basal ganglia
other family members: stiffness, headache, vertigo, dizziness; MRI: e infarctions at the bilateral basal ganglia
[80]
p.Arg75Pro64Fright thalamic hemorrhage, akinetic mutism, pseudobalbar palsy, repeated cerebral infarctions MRI: diffuse cerebral atrophy
Family members were affected with dementia
p.Arg75Pro34MChineseCerebral infarction, transient ischaemic attack with tinnitus, irritable; MRI: small infract in different brain areas (deep white matter, basal ganglia)[79]
p.Asp80Gly50sFGermanImaging and clinical symptoms: typical CADASIL phenotype. Mutation may be segregated with diseases[54]
p.Arg107TrpNANAGermanCADASIL, no clinical phenotype[68]
63MUKHypertension, Mother was affected with stroke[84]
p.Pro109Thr57FIranianCADASIL, no detailed clinical phenotype. Mutation co-existed with NOTCH3 Pro203His mutation.[78]
p.Gly149Val439FChineseInitial symptoms: progressive dizziness, memory dysfunctions. MRI: leukoencephalopathy and confluent signal abnormalities; Family history probable positive[69]
p.Gln151GluNANAItalianCADASIL, white matter hyperintensities, migraine, no detailed clinical phenotype[68]
p.Gln151GluNANASpanishCADASIL, white matter hyperintensities, stroke, no detailed clinical phenotype[70]
p.His170ArgNANASpanishCADASIL, white matter hyperintensities not specified, no detailed clinical phenotype[70]
p.His170ArgNANAOceanianCADASIL, not specified white matter hyperintensities, stroke, no clinical phenotype[71]
p.Ala198ThrNANAItalianCADASIL, white matter hyperintensities, migraine, no detailed clinical phenotype[68]
p.Ala202ValNAFOceanianCADASIL, not specified white matter hyperintensities, stroke, no clinical phenotype[70]
p.Pro203His57FIranianCADASIL, no detailed clinical phenotype. Mutation co-existed with NOTCH3 Pro109Thr mutation.[78]
p.Arg207HisNANAItalianCADASIL, white matter hyperintensities, migraine no detailed clinical phenotype[68]
p.Arg213Lys36MJapaneseCADASIL, white matter hyperintensities, migraine, dementia, stroke, positive family history[71]
p.Arg213Lys10MJapaneseCADASIL, white matter hyperintensities, dementia migraine, stroke, positive family history[72]
p.Val237Met571FJapaneseCADASIL, white matter hyperintensities, stroke, gait disturbances, dementia, positive family history[81]
p.Val252Met63MRussianstrokes and/or transient ischemic attacks, pseudobulbar palsy, pyramidal signs. MRI: hyperintensities of the external capsules, temporal lobes, lacunar infarcts in the cerebellum and/or brainstem, cerebral hemispheres[73]
p.Glu309Lys6NANAItalianCADASIL, white matter hyperintensities, migraine, dementia, family history positive[68]
p.Ser497Lys9NANARussianCADASIL, non-specified white matter hyperintensities, appeared in unaffected individuals[73]
p.Thr577Ala11NANAPortugueseUnclear phenotype, non-specified white matter hyperintensities[74]
p.Arg592Ser11NANAItalianCADASIL, white matter hyperintensities, migraine, dementia, probable positive family history[68]
p.Val644Asp12NANAItalianCADASIL, white matter hyperintensities, migraine, dementia, probable positive family history[68]
p.Ser978Arg18NAFPortugueseCADASIL, white matter hyperintensities, stroke seizure, psychiatric symptoms [74]
p.Ala1020Pro19AdolescenceFGermanCADASIL, white matter hyperintensities, hypertension, migraine, dementia, positive family history[82]
p.Ala1020Pro19NAFGermanCADASIL, white matter hyperintensities, hypertension, psychiatric disturbance, positive family history
p.Thr1098Ser2039MChineseCADASIL, white matter hyperintensities, stroke, psychiatric disturbances, dementia, positive family history[79]
p.His1133Gln21UnknownUnknownRussianCADASIL, non-specified white matter hyperintensities,[85]
p.His1235Lys22UnknownUnknownItalianCADASIL, white matter hyperintensities[68]
p.Lys1515Pro2535FFrenchCADASIL, white matter hyperintensities, migraine, dementia, positive family history[75]
p.Val1762Met29ChildhoodFItalianCADASIL, white matter hyperintensities, psychiatric dysfunctions, positive family history[77]
Table 3. HTRA1 mutations in patients with CARASIL.
Table 3. HTRA1 mutations in patients with CARASIL.
MutationExonGenderAOOClinical PhenotypesMutation TypeEthnicityReferences
Asian patients
p.Ala252Thr3FTeenageLumbago in her teens, leukoaraiosis in brain, stroke, gait disturbance, pseudobulbar palsy, pyramidal signsHomozygous missenseJapanese[28]
p.Val297Met4M/F14–16Alopecia in teens, leukoaraiosis in brain, spondylosis, dementia, gait disturbance, pseudobulbar palsy, pyramidal signs
p.Arg302X4F/M14/16Alopecia in teens, spondylosis, dementia, gait disturbance, possible stroke, pseudobulbar palsy, pyramidal signs
p.Arg370X6F18Alopecia in teens, spondylosis, dementia, gait disturbance, possible stroke, pseudobulbar palsy, pyramidal signs
p.Arg274Gln4F14Lumbago in her teens, lumbar and cervical spondylosis in her 30s, later subcortical ischemic lesions and spastic paraparesis and intellectual dysfunctions[76]
p.Gly283Glu4M49Cognitive impairment, gait disturbance, spondylosis, pseudobulbar palsy, hyperreflexia in limbsHeterozygous missense[99]
p.Pro285Leu4M20Alopecia in his 20s, gait distubances in his 30s, cognitive dysfunction in his 50s. Spondylosis, hyperreflexia in limbs, Babinski reflexes
M51Stroke, cognitive impairment and gait disturbance in his 50s. Spondylosis, pseudobulbar palsy, hyperreflexia in limbs
p.Arg302Gln4M20-63Alopecia possible in their 20–40s. Spondylosis, pseudobulbar palsy (not all patients), hyperreflexia in limbs, Babinski reflex
p.Thr319Ile4M53Spondylosis, pseudobulbar palsy, hyperreflexia in limbs, Babinski reflex
p.Pro285Leu4F24Progressive alopecia from birth, right limb disability, lumbar pain, lethargy, memory dysfunctionsHomozygous missenseChinese[91]
p.Leu364Pro6F25right foot and lumbar pain, prolapse of lumbar intervertebral disk, mild alopecia, intellectual dysfunction, spastic gait Babinski signsHomozygous missense[92]
p.Gly56Alafs*1601M28recurrent stroke, hair loss and low back pain, lower limb weakness, alopecia, pyramidal signsHomozygous/Frameshift[85]
Caucasian patients
p.Gly295Arg4M34Alopecia, dysarthria, dysphagia, emotional instability, and spastic gait, Babinski signLater, cognitive impairment and upper limb weakness, pseudobulbar syndromeHomozygous missenseSpanish[94]
p.Arg370X6F29Alopecia, back and neck pain, right-sided weakness, difficulty in walkingHomozygous nonsenseTurkish[96]
p.Glu42fs1F29chronic lumbar and cervical pain from the age of 14, ischemic strokes with left hemiparesis and dysarthria, without alopecia and cognitive dysfunctionscompound heterozygous deletion & missenseRomanian[95]
p.Ala321Thr4
p.Arg166Cys3M33Alopecia, transient ischemic attacks, lacunar strokes, cervical, and lumbar pain, acute psychosis, cognitive impairment, dysarthria, spastic paraparesis, hyperreflexia, bilateral Babinski’s sign.Homozygous missensePortuguese[100]
p.Gly206Arg3M24Alopecia, chronic back pain presented with recurrent ischemic strokes, hearing impairmenthomozygous missenseAmerican[97]

Share and Cite

MDPI and ACS Style

Giau, V.V.; Bagyinszky, E.; Youn, Y.C.; An, S.S.A.; Kim, S.Y. Genetic Factors of Cerebral Small Vessel Disease and Their Potential Clinical Outcome. Int. J. Mol. Sci. 2019, 20, 4298. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20174298

AMA Style

Giau VV, Bagyinszky E, Youn YC, An SSA, Kim SY. Genetic Factors of Cerebral Small Vessel Disease and Their Potential Clinical Outcome. International Journal of Molecular Sciences. 2019; 20(17):4298. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20174298

Chicago/Turabian Style

Giau, Vo Van, Eva Bagyinszky, Young Chul Youn, Seong Soo A. An, and Sang Yun Kim. 2019. "Genetic Factors of Cerebral Small Vessel Disease and Their Potential Clinical Outcome" International Journal of Molecular Sciences 20, no. 17: 4298. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms20174298

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop