Next Article in Journal
RNA-Seq Reveals the Underlying Molecular Mechanism of First Cleavage Time Affecting Porcine Embryo Development
Next Article in Special Issue
Exome Array Analysis of 9721 Ischemic Stroke Cases from the SiGN Consortium
Previous Article in Journal
Genome-Wide Analysis Identifies Candidate Genes Encoding Feather Color in Ducks
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Intracerebral Hemorrhage Genetics

by
Aleksandra Ekkert
1,*,
Aleksandra Šliachtenko
2,
Algirdas Utkus
3 and
Dalius Jatužis
1
1
Center of Neurology, Faculty of Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
2
Faculty of Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
3
Center for Medical Genetics, Faculty of Medicine, Vilnius University, LT-03101 Vilnius, Lithuania
*
Author to whom correspondence should be addressed.
Genes 2022, 13(7), 1250; https://doi.org/10.3390/genes13071250
Submission received: 11 June 2022 / Revised: 4 July 2022 / Accepted: 11 July 2022 / Published: 15 July 2022
(This article belongs to the Collection Feature Papers: Neurogenomics)

Abstract

:
Intracerebral hemorrhage (ICH) is a devastating type of stroke, frequently resulting in unfavorable functional outcomes. Up to 15% of stroke patients experience ICH and approximately half of those have a lethal outcome within a year. Considering the huge burden of ICH, timely prevention and optimized treatment strategies are particularly relevant. Nevertheless, ICH management options are quite limited, despite thorough research. More and more trials highlight the importance of the genetic component in the pathogenesis of ICH. Apart from distinct monogenic disorders of familial character, mostly occurring in younger subjects, there are numerous polygenic risk factors, such as hypertension, neurovascular inflammation, disorders of lipid metabolism and coagulation cascade, and small vessel disease. In this paper we describe gene-related ICH types and underlying mechanisms. We also briefly discuss the emerging treatment options and possible clinical relevance of the genetic findings in ICH management. Although existing data seems of more theoretical and scientific value so far, a growing body of evidence, combined with rapidly evolving experimental research, will probably serve clinicians in the future.

1. Introduction

Although intracerebral hemorrhage (ICH) is not the most common form of stroke (responsible for 10–15% of all strokes), it places a great burden on society [1]. It is a significant cause of mortality and disability worldwide with only 50% of sufferers having 1-year survivability [2]. Acute therapies for ICH are still under investigation and their effectiveness is limited, which makes predictive and preventive measures even more important. Unlike ischemic strokes, there are not so many modifiable risk factors in the case of ICH. In this case, hypertension is a major player and the management of high blood pressure was proved to be an effective preventive strategy [3,4]. However, the heritability of ICH is claimed to be up to 44%, and having a first-degree relative with ICH is associated with a six-fold higher risk of ICH, making the genetic component important in the pathogenesis of the disorder [5]. Although the number of potentially modifiable risk factors is modest, investigation of genetic factors and emerging data on gene therapy progress in other fields carry some hope for better individualized ICH prevention and treatment strategies.
ICH is classified according to etiopathogenesis. There are several classification systems based on anatomy or clinics, all of them have proved to be reliable if used properly [6]. The most common “anatomical” classification refers to the region of the brain as lobar (the junction of the cortical gray matter and subcortical white matter), and non-lobar or deep (the thalamus, basal ganglia, brainstem, and cerebellum). The “mechanical” systems use underlying pathological causes, such as hypertension, cerebral amyloid angiopathy (CAA), anticoagulation, and vessels’ structural defects to classify ICH. However, it is hard to use those classification systems separately, as hypertension is often associated with non-lobar ICH, but has little impact on lobar, whereas the most common histopathology found in lobar ICH is cerebral amyloid angiopathy (CAA) [7,8]. The overall heritability of lobar ICH is 48%, 30% for non-lobar ICH, and 29% for both types combined, according to International Stroke Genetics Consortium data [9].

2. Methods of Genetic Studies

Most of the information about ICH genetics comes from either the candidate gene or genome-wide association studies (GWAS). Candidate gene selection is based on known risk factors or association with disease. This approach helped to identify several genetic causes of ICH, however, as it relies on prior information, it has serious limitations, such as potential for bias and inability to find completely new genes [10]. On the other hand, GWAS is helpful when one intends to examine the entire genome for new genetic variants linked to a certain syndrome, which requires detecting more than a million single nucleotide polymorphisms (SNPs). This method empowers the researcher to identify not only new SNP in already known genes, but also completely novel genetic links.

3. Mendelian (Monogenic) Inheritance of ICH

Monogenic, also referred to as familial disorders, are rare genetic mutations following a clear Mendelian pattern of inheritance. They cause only a small number of ICH cases and tend to manifest at a younger age [11].

3.1. Familial Cerebral Amyloid Angiopathy (CAA)

Cerebral amyloid angiopathy is characterized by the depositions of amyloid-β peptide (Aβ), which is a cleavage product of the amyloid precursor protein (APP), in leptomeningeal arteries and cortical capillaries in the brain. Although numerous hypotheses have been proposed, the exact etiology of CAA remains unknown. It is unclear if the role of Aβ in the pathogenesis of the disease is causative, although the accumulation of Aβ fibrils in the vessels of the brain certainly correlates with the symptom occurrence and progression [12]. Typical manifestations include cognitive decline and intracranial hemorrhages [13]. Approximately 20% of all ICH are caused by CAA, affecting mostly the elderly population [13]. CAA-related hemorrhages are usually lobar and typically posteriorly localized [12]. The recurrence of the CAA-caused ICH is significantly higher, causing 7.4% annual recurrence risk in CAA-affected subjects, compared to 1.1% in the non-CAA group [14].
There are several genetic forms of CAA, sometimes classified as familial and sporadic. Familial types of CAA occur at a younger age and are characterized by a more severe course and a higher chance of an early lethal outcome [15]. Compared with the non-familial form of ICH, this CAA type is inherited in the Mendelian, autosomal dominant pattern [12]. The first detected gene related to familial CAA was the APP coding gene, located in the 21st chromosome [16]. SNPs in this gene cause early onset Alzheimer’s disease or CAA; whereas the variants affecting other Aβ regions manifest with prominent CAA. Polymorphisms in presenilin, cystatine C, and British precursor protein genes are associated with rare familial disorder forms characterized by ICH and the Alzheimer disease phenotype [12].
Sporadic CAA risk is caused by polygenic factors. This syndrome is described further in this article (part 4.3, “Lipid metabolism disorders”).

3.2. COL4A1 and COL4A2-Related ICH

Another frequent form of monogenic ICH is the COL4A1-related disorder. Similarly to CAA, COL4A1 has familial and sporadic forms. Familial cases represent the rare autosomal dominant mutation of the α-1 chain of the type IV collagen encoding gene. This subtype of collagen plays a crucial role in the foundation and strengthening of basement membranes of the blood vessels and many other tissues [17]. The exact pathological mechanism is the inhibition of the heterotrimer deposition in the vascular basement membrane, due to missense mutations of highly conserved hydrophobic glycine residues, which results in the alteration of vessels structural properties [18].
Clinical manifestation covers different age periods as well as intra- and extracranial symptoms. Perinatal ICH and porencephaly, ICH of various localizations in those of adult age, microbleeds, lacunar ischemic strokes due to small-vessel disease, and leukoaraiosis are typical findings [19]. COL4A1 mutations are also associated with cataracts, microcornea, Axenfeld–Rieger anomalies, and retinal hemorrhages [20]. Similarly, COL4A2 mutations have been detected in ICH and porencephaly, suggesting the same pathological mechanism [21,22,23].

3.3. CADASIL

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is caused by an autosomal dominant inherited mutation in the NOTCH3 gene, and is characterized by alterations in arterial development and compromised cerebral hemodynamics [24]. CADASIL is well known as the most common monogenic cause of ischemic stroke in adults. The most common clinical findings include migraine with aura, ischemic events, mood disturbances, and progressing dementia [25]. However, there are also several documented CADASIL-associated cases of ICH [26]. ICH is an uncommon, yet possible, manifestation of CADASIL, occurring mostly in deep white matter of the brain in male adults. Most of the described patients with CADASIL-associated ICH had concomitant hypertension [27].

4. Polygenic Inheritance of the ICH Risk Factors

4.1. Hypertension

The most important risk factor for ICH is undoubtedly hypertension, which is an especially significant pathogenetic mechanism of deep white matter ICH [28]. This opinion is supported by the data from the International Stroke Genetics Consortium that found that the association of certain genes was linked to hypertension and intracerebral hemorrhage [29]. Despite the fact that no single SNP was proven to be specifically associated with ICH, the authors had constructed a genetic risk score (GRS) including 38 single nucleotide polymorphisms (SNP) known to increase the risk of hypertension. The analysis highlighted the association between the aforementioned GRS and deep ICH. Interestingly, this association was even more prominent in non-hypertensive subjects, whereas incorporating clinical hypertension into a model did not affect the risk substantially.
The most studied gene related to hypertension is the angiotensin-converting enzyme gene (ACE) located in the 17 chromosome. This gene participates in the regulation of the renin–angiotensin–aldosterone system (RAAS), thus, it is crucial for blood pressure and water–electrolytes balance. The main risk variant rs1799752 has an insertion (I)/deletion (D) sequence in intron 16, which affects its activity and risk for stroke [30]. D allele was identified as a risk factor for ICH in Asian, but not Caucasian, populations [31,32,33].

4.2. Coagulation Disorders

Coagulation is a highly balanced system. Any dysregulation in it may lead to minor or major bleeding, including in the brain area. Some of the genetic polymorphisms associated with the deficits in the coagulation cascade are linked with ICH. Common variants of Factor XIII Subunit A (Tyr204/Phe204 and Leu564/Leu564) are associated with an increased risk of ICH in young white women (age < 45), especially in the presence of the plasminogen activator inhibitor polymorphism 5G/5G [34]. The -323Ins allele of Factor VII is associated with a 1.54-fold risk for ICH, whereas the -401G > T polymorphism of the same factor has no significant impact on hemorrhagic stroke [35,36]. As for factor V Leiden, the carriers seemed to be at lower risk of developing ICH [35]. Platelet-activating factor deficiency, caused by a missense V279F mutation in Exon 9, is also a genetic risk factor for ICH [37]. The β1-tubulin gene (TUBB1) Q43P polymorphism significantly increased both subarachnoid hemorrhage and ICH risk in men, and was associated with an earlier age of ICH occurrence [38].

4.3. Lipid Metabolism Disorders

The most studied and best-known ICH-causing genetic variant related to lipid metabolism is apolipoprotein E (APOE). APOE gene is located in the 19 chromosome and can be represented by three allele variants—ε2, ε3, and ε4. Ε3ε3 is the most common genotype that is not related to ICH, whereas both ε2 and ε4 are missense variants involved in the pathogenesis of sporadic CAA [39,40], increasing the risk of subsequent lobar ICH [41]. A meta-analysis of 11 case–control studies showed a significantly increased frequency of the APOE ε4 allele in ICH cases in Caucasians and Asians [42]. The association of the ε4 with CAA is dose-dependent, whereas the ε2 allele is associated with larger hemorrhage volume that results in more severe disability and higher mortality [41,43]. It has been shown that the presence of the APOE ε2 and ε4 alleles puts the subjects on warfarin at substantial risk of lobar ICH: each ε2 and ε4 allele is associated with a 2.5-fold and 3-fold increase in the risk, respectively [44]. Despite the fact that earlier data clearly showed APOE association with lobar ICH, an increasing number of studies has proven both the ε2 and ε4 link to deep ICH, as well [42,43,45,46]. One of the studies has identified both ε2 and ε4 as increasing the risk for lobar ICH and only ε4 for the deep one [46].
APOE is not the only lipoprotein-coding gene that influences ICH risk. Apolipoprotein H (APOH) participates in lipid metabolism, hemostasis, and synthesis of the antiphospholipid antibody [47]. Four polymorphisms of the APOH gene were examined in the Chinese population. One of them (A allele of G341A) was observed significantly more frequently in ICH patients, especially those with a history of hypertension or family strokes [48].
Apolipoprotein (a) forms lipoprotein Lp(a) and is controlled by LPA gene expression. Its main risk variant is the repetitive pentanucleotide sequence TTTTA in the upstream of the transcription start site. A low number of TTTTA is related to elevation in the levels of Lp(a) and an increased risk of ICH [49].
Both Endoplasmic Reticulum Lipid Raft Associated 1 (ERLIN1) and Low-Density Lipoprotein Receptor (LDLR), located in 10 and 19 chromosomes, respectively, are important in cholesterol homeostasis. The main variants of those genes (rs1324694 for ERLIN1 and rs688 for LDLR) are associated with a decreased risk of ICH [50,51]. Moreover, they are associated with a protective effect after the ICH.
Higher blood concentration of high-density lipoprotein cholesterol (HDL-C) has been described as a risk factor for ICH [52]. Another study found that genetic variants of the cholesteryl ester transfer protein gene (CETP) associated with increased HDL-C plasma levels also elevate risk of ICH with the strongest association with the rs173539 SNP [53]. CETP inhibitors leading to an increase in the HDL-C levels in blood plasma are thought to be one of the potential atherosclerosis treatment options [54,55]. Unfortunately, the data about intracerebral hemorrhage in CETP inhibitors’ clinical trials are quite scarce so far [55]. It seems reasonable to investigate cerebrovascular outcomes in these trials further, including not only ischemic cardiovascular events, but also intracerebral hemorrhage frequency.

4.4. Inflammation

Inflammation plays an important role in vascular damage and the severity of stroke outcome. The upregulation of genes influencing inflammation processes could be associated with the increased risk of ICH. The methylenetetrahydrofolate reductase (MTHFR) coding gene is located in the 1p36.22 chromosome. The function of MTHFR is to convert amino acids’ homocysteine to methionine. The rs1801133 polymorphism is the main risk variant, which reduces MTHFR activity and causes hyperhomocysteinemia, a well-known risk factor for inflammation, atherosclerosis, and endothelial dysfunction [56,57]. Large meta-analysis showed associations between ICH and MTHFR 677 T variant allele in both Asian and Caucasian populations [58].
Another essential cytokine in the inflammatory response to ICH is Interleukin 6 (IL6) [59]. It is responsible for endothelial cell activation and vascular dysfunction, as well as oxidative stress and an increased expression of pro-inflammatory cytokines. The rs1800796 polymorphism (C allele) is the functional promoter of IL6 and was noticed to be the major allele in the Asian population (79%) but the minor allele in the European population (<5%), according to the HapMap database. This genetic variant is related to an increased expression of IL6 and was significantly linked with an ICH risk in the Japanese population [60,61].
Another proinflammatory cytokine, initiating and regulating the inflammatory response, is tumor necrosis factor-a (TNF-a). The TNF-a concentration depends on different polymorphisms in the regulatory region. rs1799964, rs1800629, and rs1800630 SNP are associated with increased TNF-a expression [62,63]. A study of the Taiwanese population showed that rs1799964 and rs1800629 are risk factors for deep ICH in men, while rs1800630 has protective role [64].
The protein encoded by trafficking protein particle complex 9 (TRAPPC) is involved in the neuronal signaling of transcription factors and its overexpression enhances a cytokine-induced NF- κB signaling pathway [65]. An intronic polymorphism rs12679196 (C allele) has been proved to significantly elevate the risk of ICH in the Japanese population, while the T allele decreased it [50].
Interferon Epsilon (IFNE) is expressed in several tissues, including brain, and is responsible for controlling immune reactions in the central nervous system [66,67]. The rs2039381 polymorphism of the IFNE gene was an associated risk factor for ICH in Korean population [68].
Another study of the Korean population showed that the rs2228048 polymorphism of transforming growth factor β2 receptor 2 gene (TGFBR2) increased the risk of ICH [69]. TGFBR2 is mostly expressed in neurons and plays an important role in T cells’ development and homeostasis [70,71].
Tissue inhibitors of metalloproteinases (TIMPs) are inhibitors for matrix metalloproteinases (MMPs)—endopeptidases, involved in blood–brain barrier breakdown and inflammatory reactions [72]. MMPs, especially gelatin-binding, are able to degrade type IV and V collagen thus break down the vascular basement membrane and cause the rupture of the vessel walls [73,74]. TIMP-1 and TIMP-2 are located in the X and 13 chromosomes, respectively. The main TIMP-1 risk variant for ICH is rs2070584, whereas another variant (rs4898) provides a protective effect on deep ICH [75,76]. As for TIMP-2, the main risk variants are rs7503726 and s7503607; however, rs7503726 also was considered as protection against deep ICH in some populations [77,78].
Moreover, genes coding MMP themselves might also contribute to the ICH risk. SNPs in the MMP2 (rs2285053) and MMP9 (rs3787268, rs2250889) genes increase the risk of deep ICH in the older Asian population (>65 years old). The pathogenetic mechanism is thought to be extracellular matrix degradation promoted by MMP [79].

4.5. Small Vessel Disease

A recent study of genetic loci associated with stroke has found eight genes related to ICH. Four loci (EDNRA, 1q22, MMP12, SH3PXD2A) were the same in ICH and small vessel stroke, suggesting a mutual underlying mechanism [80]. An earlier study revealed 1q22 as a susceptibility locus for deep ICH with the main risk variant rs2984613. In the case of this SNP, each additional C allele increased the frequency of deep ICH by 24% [81].

4.6. Others

Meta-analysis by Woo et al. revealed new susceptibility loci for ICH. Polymorphisms in the 12q21.1 region (especially the rs11179580 variant) were associated with lobar ICH, but only in the European population. The exact mechanisms of the ICH caused by this gene polymorphism are quite unclear [81]. A large study by Traylor et al. has found eight genes related to ICH. Seven of them (1q22, COL4A2, EDNRA, LINC01492, MMP12, SH3PXD2A, CDK6) were influencing both ischemic stroke and ICH, while ANK2 (rs149538932) raised risk predominantly in ICH. As mentioned before, four of them (EDNRA, 1q22, MMP12, SH3PXD2A) are related to small vessel disease, whereas the exact mechanism of other polymorphisms is still unknown [80]. The summary of the most well-known genes and SNPs associated with the increase in polygenic ICH risk is provided in Table A1 (Appendix A).

5. Genetic Predictors of Outcome

Genetic factors could provide a hint regarding ICH prognosis. Newly discovered susceptibility risk locus in the 17p12 is associated with hematoma volume, clinical severity, and functional outcome in non-lobar ICH in patients of European ancestry [82]. The APOE ε2 allele is associated with a larger ICH volume, as well as an elevated risk of mortality and poor functional recovery in lobar ICH [83]. Inflammation also has an impact on ICH severity and outcome: high IL-6 plasma levels at admission could predict hematoma enlargement [84].

6. Gene Therapy Opportunities

So far there is no effective, specific treatment for ICH, although recent studies suggest promising results. The greatest interest is raised by strategies targeting secondary brain injury, in particular, mechanisms such as inflammation, oxidative stress, apoptosis, brain edema, and neuronal damage [85]. Therefore, genetic studies are crucial, as they provide information on mechanisms involved in the ICH pathogenesis and provide more possibilities for development of target-specific treatment.
Recent studies revealed some important interactions between the inflammatory gene expression in peripheral blood and ICH. One of them identified 122 genes with altered expression in ICH patients, mostly involved in neuroinflammation, apoptosis, interleukin, or transcriptional regulation. It also showed significant overlap between dysregulated genes in blood and perihematomal brain tissue, suggesting the importance of blood–brain interactions in ICH patients [86].
Another study performed an analysis of expressed genes in peripheral blood within 24 h of ICH onset and 72 h after the first sample. According to the results, the expression of 218 genes was significantly altered after ICH; the most-induced genes were related to the inflammation and the activation of the immune response [87]. Both of the studies open paths for novel target-specific treatment investigations, although they also require additional research on links between the study subject and ICH pathogenesis, and outcomes.

7. Conclusions

ICH is a severe disease resulting in high mortality and a large proportion of unfavorable functional outcomes, sometimes being recurrent and causing even more devastating consequences. Such a disorder requires timely risk factor evaluation and prevention. Genetic risk factors play an important role in the ICH pathogenesis, presenting by a large spectrum of pathologies, including hypertension, neurovascular inflammation, disorders of lipid metabolism, and coagulation cascade. Genetic data should be used for the analysis of potential ICH mechanisms and to optimize the prevention: e.g., patients who are genetically susceptible to deep ICH, which is frequently caused by hypertension, possibly should receive more strict blood pressure control. This information would enable clinicians to individualize ICH prevention and treatment strategies, and to plan the outcomes. So far, existing data seems largely of theoretical and scientific value; nevertheless, experimental research in genetics is rapidly evolving, giving us more hope regarding ICH patients’ management in future.

Author Contributions

A.Š. wrote Introduction, Methods of genetic studies, Mendelian (monogenic) inheritance of ICH and Gene therapy opportunities; A.E. wrote Polygenic inheritance of the ICH risk factors, Genetic predictors of outcome and Conclusions; A.U.—consulting on topic of genetics, final manuscript revision and corrections; D.J.—final manuscript revision and corrections. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Table A1. Genes contributing to polygenic risk of the ICH.
Table A1. Genes contributing to polygenic risk of the ICH.
SyndromeGeneChromosomePolymorphismsReference
HypertensionACE17q23.3rs1799752PMID: 24098401
CoagulationFactor XIII Subunit A6p25.1Tyr204/Phe204; Leu564/Leu564PMID: 11692020
PAI-1 (SERPINE1)7q21.3-q225G/5GPMID: 11692020
Factor VII13q34-323InsPMID: 11342420
TUBB120q13.32Q43PPMID: 17488662
Lipid metabolismAPOE19q13.32rs7412; rs429358PMID: 32891149
APOH17q24.2G341APMID: 14707422
LPA6q25-q26TTTTAPMID: 12791946
ERLIN110q24.31rs1324694PMID: 20198315
LDLR19p13.2rs688PMID: 24295502
CETP16q13rs173539PMID: 27717122
InflammationMTHFR1p36.22rs1801133PMID: 12560871
IL67p15.3rs1800796PMID: 16741147
TNF6p21.33rs1799964; rs1800629; rs1800630PMID: 20534169
PMID: 27809686
TRAPPC6p21.33rs12679196PMID: 20198315
IFNE9p21.3rs2039381PMID: 24055696
TGFBR23p24.1rs2228048PMID: 21609163
TIMP-1Xp11.3rs2070584; rs4898PMID: 23841813
PMID: 25932641
TIMP-213q34rs7503726; rs7503607PMID: 19844095
PMID: 26551785
MMP216q12.2rs2285053PMID: 26551785
MMP920q13.12rs3787268; rs2250889PMID: 25932641
Small vessel diseaseEDNRA4q31.22-q31.23rs5333PMID: 31150867
MMP1211q22.2rs660599PMID: 25078452
SH3PXD2A10q24.33N/APMID: 31306060
1q221q22rs2984613PMID: 24656865
COL4A213q34rs9521732; rs9521733; rs9515199PMID: 30518145
Other12q21.112q21.1rs11179580PMID: 24656865
LINC014929q31.1N/APMID: 31306060
CDK67q21.2N/APMID: 31306060
ANK24q25-q26rs149538932PMID: 31306060

References

  1. John, S.; Garg, R. Intracerebral hemorrhage. Handb. Stroke Neurocritical Care 2013, 1, 209–222. [Google Scholar]
  2. Poon, M.; Fonville, A.F.; Salman, R.A.-S. Long-term prognosis after intracerebral haemorrhage: Systematic review and meta-analysis. J. Neurol. Neurosurg. Psychiatry 2013, 85, 660–667. [Google Scholar] [CrossRef]
  3. Hemphill, J.C., 3rd; Greenberg, S.M.; Anderson, C.S.; Becker, K.; Bendok, B.R.; Cushman, M.; Fung, G.L.; Goldstein, J.N.; Macdonald, R.L.; Mitchell, P.H.; et al. Guidelines for the Management of Spontaneous Intracerebral Hemorrhage: A Guide-line for Healthcare Professionals from the American Heart Association/American Stroke Association. Stroke 2015, 46, 2032–2060. [Google Scholar] [CrossRef] [Green Version]
  4. Arima, H.; Tzourio, C.; Anderson, C.; Woodward, M.; Bousser, M.-G.; MacMahon, S.; Neal, B.; Chalmers, J. Effects of Perindopril-Based Lowering of Blood Pressure on Intracerebral Hemorrhage Related to Amyloid Angiopathy. Stroke 2010, 41, 394–396. [Google Scholar] [CrossRef] [Green Version]
  5. Hostettler, I.C.; Seiffge, D.J.; Werring, D.J. Intracerebral hemorrhage: An update on diagnosis and treatment. Expert Rev. Neurother. 2019, 19, 679–694. [Google Scholar] [CrossRef]
  6. Rannikmäe, K.; Woodfield, R.; Anderson, C.S.; Charidimou, A.; Chiewvit, P.; Greenberg, S.M.; Jeng, J.-S.; Meretoja, A.; Palm, F.; Putaala, J.; et al. Reliability of intracerebral hemorrhage classification systems: A systematic review. Int. J. Stroke 2016, 11, 626–636. [Google Scholar] [CrossRef]
  7. Woo, D.; Sauerbeck, L.R.; Kissela, B.M.; Khoury, J.C.; Szaflarski, J.P.; Gebel, J.; Shukla, R.; Pancioli, A.M.; Jauch, E.C.; Menon, A.G.; et al. Genetic and Environmental Risk Factors for Intracerebral Hemorrhage. Stroke 2002, 33, 1190–1196. [Google Scholar] [CrossRef] [Green Version]
  8. Charidimou, A.; Gang, Q.; Werring, D.J. Sporadic cerebral amyloid angiopathy revisited: Recent insights into pathophysiology and clinical spectrum. J. Neurol. Neurosurg. Psychiatry 2011, 83, 124–137. [Google Scholar] [CrossRef]
  9. Falcone, G.J.; Malik, R.; Dichgans, M.; Rosand, J. Current concepts and clinical applications of stroke genetics. Lancet Neurol. 2014, 13, 405–418. [Google Scholar] [CrossRef]
  10. Zhu, M.; Zhao, S. Candidate Gene Identification Approach: Progress and Challenges. Int. J. Biol. Sci. 2007, 3, 420–427. [Google Scholar] [CrossRef] [Green Version]
  11. Falcone, G.J.; Woo, D. Genetics of Spontaneous Intracerebral Hemorrhage. Stroke 2017, 48, 3420–3424. [Google Scholar] [CrossRef]
  12. Gatti, L.; Tinelli, F.; Scelzo, E.; Arioli, F.; Di Fede, G.; Obici, L.; Pantoni, L.; Giaccone, G.; Caroppo, P.; Parati, E.A.; et al. Understanding the Pathophysiology of Cerebral Amyloid Angiopathy. Int. J. Mol. Sci. 2020, 21, 3435. [Google Scholar] [CrossRef]
  13. Inoue, Y.; Ando, Y.; Misumi, Y.; Ueda, M. Current Management and Therapeutic Strategies for Cerebral Amyloid Angiopathy. Int. J. Mol. Sci. 2021, 22, 3869. [Google Scholar] [CrossRef]
  14. Charidimou, A.; Imaizumi, T.; Moulin, S.; Biffi, A.; Samarasekera, N.; Yakushiji, Y.; Peeters, A.; Vandermeeren, Y.; Laloux, P.; Baron, J.-C.; et al. Brain hemorrhage recurrence, small vessel disease type, and cerebral microbleeds. Neurology 2017, 89, 820–829. [Google Scholar] [CrossRef]
  15. Zhang-Nunes, S.X.; Maat‐Schieman, M.L.; van Duinen, S.G.; Roos, R.A.; Frosch, M.P.; Greenberg, S.M. The cerebral β-amyloid angiopathies: Hereditary and sporadic. Brain Pathol. 2006, 16, 30–39. [Google Scholar] [CrossRef]
  16. Karschnia, P.; Nishimura, S.; Louvi, A. Cerebrovascular disorders associated with genetic lesions. Cell. Mol. Life Sci. 2018, 76, 283–300. [Google Scholar] [CrossRef]
  17. Vahedi, K.; Kubis, N.; Boukobza, M.; Arnoult, M.; Massin, P.; Tournier-Lasserve, E.; Bousser, M.-G. COL4A1 Mutation in a Patient With Sporadic, Recurrent Intracerebral Hemorrhage. Stroke 2007, 38, 1461–1464. [Google Scholar] [CrossRef] [Green Version]
  18. 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] [Green Version]
  19. Van Der Knaap, M.S.; Smit, L.M.E.; Barkhof, F.; Pijnenburg, Y.A.L.; Zweegman, S.; Niessen, H.; Imhof, S.; Heutink, P. Neonatal porencephaly and adult stroke related to mutations in collagen IV A1. Ann. Neurol. 2006, 59, 504–511. [Google Scholar] [CrossRef]
  20. 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] [Green Version]
  21. 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 α2 Chain Cause Porencephaly. Am. J. Hum. Genet. 2012, 90, 86–90. [Google Scholar] [CrossRef] [Green Version]
  22. Verbeek, E.; Meuwissen, M.E.C.; 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]
  23. 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]
  24. Rutten, J.W.; Haan, J.; Terwindt, G.M.; van Duinen, S.G.; Boon, E.M.; Oberstein, S.A.L. Interpretation of NOTCH3 mutations in the diagnosis of CADASIL. Expert Rev. Mol. Diagn. 2014, 14, 593–603. [Google Scholar] [CrossRef]
  25. Chabriat, H.; Joutel, A.; Tournier-Lasserve, E.; Bousser, M.G. CADASIL: Yesterday, today, tomorrow. Eur. J. Neurol. 2020, 27, 1588–1595. [Google Scholar] [CrossRef]
  26. Chabriat, H.; Joutel, A.; Dichgans, M.; Tournier-Lasserve, E.; Bousser, M.-G. CADASIL. Lancet Neurol. 2009, 8, 643–653. [Google Scholar] [CrossRef]
  27. Palazzo, P.; Le Guyader, G.; Neau, J.-P. Intracerebral hemorrhage in CADASIL. Rev. Neurol. 2021, 177, 422–430. [Google Scholar] [CrossRef]
  28. Ikram, M.A.; Wieberdink, R.G.; Koudstaal, P.J. International Epidemiology of Intracerebral Hemorrhage. Curr. Atheroscler. Rep. 2012, 14, 300–306. [Google Scholar] [CrossRef] [Green Version]
  29. Falcone, G.J.; Biffi, A.; Devan, W.J.; Jagiella, J.M.; Schmidt, H.; Kissela, B.; Hansen, B.; Jimenez-Conde, J.; Steinhauer, E.G.; Elosua, R.; et al. Burden of Risk Alleles for Hypertension Increases Risk of Intracerebral Hemorrhage. Stroke 2012, 43, 2877–2883. [Google Scholar] [CrossRef]
  30. He, Q.; Fan, C.; Yu, M.; Wallar, G.; Zhang, Z.-F.; Wang, L.; Zhang, X.; Hu, R. Associations of ACE Gene Insertion/Deletion Polymorphism, ACE Activity, and ACE mRNA Expression with Hypertension in a Chinese Population. PLoS ONE 2013, 8, e75870. [Google Scholar] [CrossRef]
  31. Sun, Y.; Liu, Y.; Watts, L.T.; Sun, Q.; Zhong, Z.; Yang, G.-Y.; Bian, L. Genetic Associations of Angiotensin-Converting Enzyme with Primary Intracerebral Hemorrhage: A Meta-analysis. PLoS ONE 2013, 8, e67402. [Google Scholar] [CrossRef] [Green Version]
  32. Kalita, J.; Misra, U.; Bindu, I.; Kumar, B.; Mittal, B. Angiotensin-converting enzyme (rs4646994) and α ADDUCIN (rs4961) gene polymorphisms′ study in primary spontaneous intracerebral hemorrhage. Neurol. India 2011, 59, 41–46. [Google Scholar] [CrossRef]
  33. Kumar, A.; Prasad, K.N.; Vivekanandhan, S.; Srivastava, A.K.; Goswami, S.K.; Srivastava, M.V.P.; Tripathi, M. Association between angiotensin converting enzyme gene insertion/deletion polymorphism and intracerebral haemorrhage in North Indian population: A case control study and meta-analysis. Neurol. Sci. 2014, 35, 1983–1990. [Google Scholar] [CrossRef]
  34. Reiner, A.P.; Schwartz, S.M.; Frank, M.B.; Longstreth, J.W.; Hindorff, L.A.; Teramura, G.; Rosendaal, F.R.; Gaur, L.K.; Psaty, B.M.; Siscovick, D.S.; et al. Polymorphisms of Coagulation Factor XIII Subunit A and Risk of Nonfatal Hemorrhagic Stroke in Young White Women Editorial Comment. Stroke 2001, 32, 2580–2587. [Google Scholar] [CrossRef] [Green Version]
  35. Corral, J.; Iniesta, J.A.; González-Conejero, R.; Villalón, M.; Vicente, V. Polymorphisms of clotting factors modify the risk for primary intracranial hemorrhage. Blood 2001, 97, 2979–2982. [Google Scholar] [CrossRef] [Green Version]
  36. Greisenegger, S.; Weber, M.; Funk, M.; Endler, G.; Lang, W.; Ferrari, J.; Lalouschek, W.; Mannhalter, C. Polymorphisms in the coagulation factor VII gene and risk of primary intracerebral hemorrhage. Eur. J. Neurol. 2007, 14, 1098–1101. [Google Scholar] [CrossRef]
  37. Hiramoto, M.; Yoshida, H.; Imaizumi, T.; Yoshimizu, N.; Satoh, K. A Mutation in Plasma Platelet-Activating Factor Acetylhydrolase (Val 279→Phe) is a Genetic Risk Factor for Stroke. Stroke 1997, 28, 2417–2420. [Google Scholar] [CrossRef]
  38. Navarro-Nuñez, L.; Lozano, M.L.; Rivera, J.; Corral, J.; Roldan, V.; González-Conejero, R.; Iniesta, J.A.; Montaner, J.; Vicente, V.; Martínez, C. The association of the 1-tubulin Q43P polymorphism with intracerebral hemorrhage in men. Haematologica 2007, 92, 513–518. [Google Scholar] [CrossRef] [Green Version]
  39. Greenberg, S.M.; Vonsattel, J.-P.G.; Segal, A.Z.; Chiu, R.I.; Clatworthy, A.E.; Liao, A.; Hyman, B.T.; Rebeck, G.W. Association of apolipoprotein E 2 and vasculopathy in cerebral amyloid angiopathy. Neurology 1998, 50, 961–965. [Google Scholar] [CrossRef]
  40. Premkumar, D.R.; Cohen, D.L.; Hedera, P.; Friedland, R.P.; Kalaria, R.N. Apolipoprotein E-epsilon4 alleles in cerebral amyloid angiopathy and cerebrovascular pathology associated with Alzheimer’s disease. Am. J. Pathol. 1996, 148, 2083–2095. [Google Scholar]
  41. Biffi, A.; Anderson, C.D.; Jagiella, J.M.; Schmidt, H.; Kissela, B.; Hansen, B.M.; Jimenez-Conde, J.; Pires, C.R.; Ayres, A.M.; Schwab, K.; et al. APOE genotype and extent of bleeding and outcome in lobar intracerebral haemorrhage: A genetic association study. Lancet Neurol. 2011, 10, 702–709. [Google Scholar] [CrossRef] [Green Version]
  42. Zhang, R.; Wang, X.; Tang, Z.; Liu, J.; Yang, S.; Zhang, Y.; Wei, Y.; Luo, W.; Wang, J.; Li, J.; et al. Apolipoprotein E gene polymorphism and the risk of intracerebral hemorrhage: A meta-analysis of epidemiologic studies. Lipids Health Dis. 2014, 13, 47. [Google Scholar] [CrossRef] [Green Version]
  43. Rannikmäe, K.; Samarasekera, N.; Martínez-González, N.A.; Salman, R.A.-S.; Sudlow, C. Genetics of cerebral amyloid angiopathy: Systematic review and meta-analysis. J. Neurol. Neurosurg. Psychiatry 2013, 84, 901–908. [Google Scholar] [CrossRef]
  44. Falcone, G.J.; Radmanesh, F.; Brouwers, H.B.; Battey, T.W.; Devan, W.J.; Valant, V.; Raffeld, M.R.; Chitsike, L.P.; Ayres, A.M.; Schwab, K.; et al. APOE variants increase risk of warfarin-related intracerebral hemorrhage. Neurology 2014, 83, 1139–1146. [Google Scholar] [CrossRef] [Green Version]
  45. Tzourio, C.; Arima, H.; Harrap, S.; Anderson, C.; Godin, O.; Woodward, M.; Neal, B.; Bousser, M.-G.; Chalmers, J.; Cambien, F.; et al. APOE genotype, ethnicity, and the risk of cerebral hemorrhage. Neurology 2008, 70, 1322–1328. [Google Scholar] [CrossRef]
  46. Biffi, A.; Sonni, A.; Anderson, C.D.; Kissela, B.; Jagiella, J.M.; Schmidt, H.; Jimenez-Conde, J.; Bs, B.M.H.; Fernandez-Cadenas, I.; Msc, L.C.; et al. Variants at APOE influence risk of deep and lobar intracerebral hemorrhage. Ann. Neurol. 2010, 68, 934–943. [Google Scholar] [CrossRef] [Green Version]
  47. Schousboe, I. β 2-Glycoprotein I: A plasma inhibitor of the contact activation of the intrinsic blood coagulation pathway. Blood 1985, 66, 1086–1091. [Google Scholar] [CrossRef] [Green Version]
  48. Xia, J.; Yang, Q.-D.; Xu, H.-W.; Liu, Y.-H.; Zhang, L.; Zhou, Y.-H.; Wu, Z.-G.; Cao, G.-F. Apolipoprotein H Gene Polymorphisms and Risk of Primary Cerebral Hemorrhage in a Chinese Population. Cerebrovasc. Dis. 2003, 17, 197–203. [Google Scholar] [CrossRef]
  49. Sun, L.; Li, Z.; Zhang, H.; Ma, A.; Liao, Y.; Wang, D.; Zhao, B.; Zhu, Z.; Zhao, J.; Zhang, Z.; et al. Pentanucleotide TTTTA Repeat Polymorphism of Apolipoprotein(a) Gene and Plasma Lipoprotein(a) Are Associated with Ischemic and Hemorrhagic Stroke in Chinese. Stroke 2003, 34, 1617–1622. [Google Scholar] [CrossRef] [Green Version]
  50. Yamada, Y.; Yoshida, T.; Kato, K.; Yokoi, K.; Oguri, M.; Watanabe, S.; Metoki, N.; Yoshida, H.; Satoh, K.; Aoyagi, Y.; et al. Association of genetic variants with hemorrhagic stroke in Japanese individuals. Int. J. Mol. Med. 2010, 25, 649–656. [Google Scholar] [CrossRef] [Green Version]
  51. Lee, J.-D.; Hsiao, K.-M.; Lee, T.-H.; Kuo, Y.-W.; Huang, Y.-C.; Hsu, H.-L.; Lin, Y.-H.; Wu, C.-Y.; Huang, Y.-C.; Lee, M.; et al. Genetic polymorphism of LDLR (rs688) is associated with primary intracerebral hemorrhage. Curr. Neurovascular Res. 2014, 11, 10–15. [Google Scholar] [CrossRef]
  52. Wang, X.; Dong, Y.; Qi, X.; Huang, C.; Hou, L. Cholesterol Levels and Risk of Hemorrhagic Stroke. Stroke 2013, 44, 1833–1839. [Google Scholar] [CrossRef] [Green Version]
  53. Anderson, C.D.; Falcone, G.J.; Phuah, C.; Ma, R.A.S.; Brouwers, H.B.; Bs, T.W.K.B.; Biffi, A.; Peloso, G.M.; Liu, D.J.; Ba, A.M.A.; et al. Genetic variants in CETP increase risk of intracerebral hemorrhage. Ann. Neurol. 2016, 80, 730–740. [Google Scholar] [CrossRef]
  54. Tall, A.R.; Rader, D.J. Trials and Tribulations of CETP Inhibitors. Circ. Res. 2018, 122, 106–112. [Google Scholar] [CrossRef]
  55. Taheri, H.; Filion, K.B.; Windle, S.B.; Reynier, P.; Eisenberg, M.J. Cholesteryl Ester Transfer Protein Inhibitors and Cardiovascular Outcomes: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Cardiology 2020, 145, 236–250. [Google Scholar] [CrossRef]
  56. Ogino, S.; Wilson, R.B. Genotype and haplotype distributions of MTHFR 677C>T and 1298A>C single nucleotide polymorphisms: A meta-analysis. J. Hum. Genet. 2003, 48, 0001–0007. [Google Scholar] [CrossRef] [Green Version]
  57. Lai, W.K.C.; Kan, M.Y. Homocysteine-Induced Endothelial Dysfunction. Ann. Nutr. Metab. 2015, 67, 1–12. [Google Scholar] [CrossRef]
  58. Gao, S.; Li, H.; Xiao, H.; Yao, G.; Shi, Y.; Wang, Y.; Zhou, X.; Yu, H. Association of MTHFR 677T variant allele with risk of intracerebral haemorrhage: A meta-analysis. J. Neurol. Sci. 2012, 323, 40–45. [Google Scholar] [CrossRef]
  59. Sargurupremraj, M.; International Network against Thrombosis (INVENT) Consortium; Suzuki, H.; Jian, X.; Sarnowski, C.; Evans, T.E.; Bis, J.C.; Eiriksdottir, G.; Sakaue, S.; Terzikhan, N.; et al. Cerebral small vessel disease genomics and its implications across the lifespan. Nat. Commun. 2020, 11, 6285. [Google Scholar] [CrossRef]
  60. Yamada, Y.; Metoki, N.; Yoshida, H.; Satoh, K.; Ichihara, S.; Kato, K.; Kameyama, T.; Yokoi, K.; Matsuo, H.; Segawa, T.; et al. Genetic Risk for Ischemic and Hemorrhagic Stroke. Arter. Thromb. Vasc. Biol. 2006, 26, 1920–1925. [Google Scholar] [CrossRef] [Green Version]
  61. Fishman, D.; Faulds, G.; Jeffery, R.; Mohamed-Ali, V.; Yudkin, J.S.; Humphries, S.; Woo, P. The effect of novel polymorphisms in the interleukin-6 (IL-6) gene on IL-6 transcription and plasma IL-6 levels, and an association with systemic-onset juvenile chronic arthritis. J. Clin. Investig. 1998, 102, 1369–1376. [Google Scholar] [CrossRef] [Green Version]
  62. Wilson, A.G.; Symons, J.A.; McDowell, T.L.; McDevitt, H.O.; Duff, G.W. Effects of a polymorphism in the human tumor necrosis factor α promoter on transcriptional activation. Proc. Natl. Acad. Sci. USA 1997, 94, 3195–3199. [Google Scholar] [CrossRef] [Green Version]
  63. Higuchi, T.; Seki, N.; Kamizono, S.; Yamada, A.; Kimura, A.; Kato, H.; Itoh, K. Polymorphism of the 5′-flanking region of the human tumor necrosis factor (TNF)-α gene in Japanese. Tissue Antigens 1998, 51, 605–612. [Google Scholar] [CrossRef]
  64. Chen, Y.-C.; Hu, F.-J.; Chen, P.; Wu, Y.-R.; Wu, H.-C.; Chen, S.-T.; Lee-Chen, G.-J.; Chen, C.-M. Association of TNF-α gene with spontaneous deep intracerebral hemorrhage in the Taiwan population: A case control study. BMC Neurol. 2010, 10, 41–48. [Google Scholar] [CrossRef] [Green Version]
  65. Hu, W.-H.; Pendergast, J.S.; Mo, X.-M.; Brambilla, R.; Bracchi-Ricard, V.; Li, F.; Walters, W.M.; Blits, B.; He, L.; Schaal, S.M.; et al. NIBP, a Novel NIK and IKKβ-binding Protein That Enhances NF-κB Activation. J. Biol. Chem. 2005, 280, 29233–29241. [Google Scholar] [CrossRef] [Green Version]
  66. Pestka, S.; Krause, C.D.; Walter, M.R. Interferons, interferon-like cytokines, and their receptors. Immunol. Rev. 2004, 202, 8–32. [Google Scholar] [CrossRef]
  67. Nayak, D.; Johnson, K.R.; Heydari, S.; Roth, T.L.; Zinselmeyer, B.H.; McGAVERN, D.B. Type I Interferon Programs Innate Myeloid Dynamics and Gene Expression in the Virally Infected Nervous System. PLoS Pathog. 2013, 9, e1003395. [Google Scholar] [CrossRef] [Green Version]
  68. Kim, S.K.; Park, H.J.; Kim, J.W.; Chung, J.-H.; Yoo, S.D.; Kim, D.H.; Yun, D.H.; Kim, H.-S. T Allele of nonsense polymorphism (rs2039381, Gln71Stop) of interferon-ε is a risk factor for the development of intracerebral hemorrhage. Hum. Immunol. 2014, 75, 88–90. [Google Scholar] [CrossRef]
  69. Lim, Y.H.; Jeong, Y.S.; Kim, S.K.; Kim, N.H.; Yun, D.H.; Yoo, S.D.; Kim, H.-S.; Baik, H.H. Association between TGFBR2 Gene Polymorphism (rs2228048, Asn389Asn) and Intracerebral Hemorrhage in Korean Population. Immunol. Investig. 2011, 40, 569–580. [Google Scholar] [CrossRef]
  70. Travis, M.A.; Sheppard, D. TGF-β Activation and Function in Immunity. Annu. Rev. Immunol. 2014, 32, 51–82. [Google Scholar] [CrossRef] [Green Version]
  71. Yi, J.J.; Barnes, A.P.; Hand, R.; Polleux, F.; Ehlers, M.D. TGF-β Signaling Specifies Axons during Brain Development. Cell 2010, 142, 144–157. [Google Scholar] [CrossRef] [Green Version]
  72. Chang, J.J.; Emanuel, B.A.; Mack, W.J.; Tsivgoulis, G.; Alexandrov, A.V. Matrix Metalloproteinase-9: Dual Role and Temporal Profile in Intracerebral Hemorrhage. J. Stroke Cerebrovasc. Dis. 2014, 23, 2498–2505. [Google Scholar] [CrossRef]
  73. Newby, A.C. Dual Role of Matrix Metalloproteinases (Matrixins) in Intimal Thickening and Atherosclerotic Plaque Rupture. Physiol. Rev. 2005, 85, 1–31. [Google Scholar] [CrossRef] [Green Version]
  74. Florczak-Rzepka, M.; Grond-Ginsbach, C.; Montaner, J.; Steiner, T. Matrix Metalloproteinases in Human Spontaneous Intracerebral Hemorrhage: An Update. Cerebrovasc. Dis. 2012, 34, 249–262. [Google Scholar] [CrossRef] [Green Version]
  75. Wang, H.-X.; Yang, Q.-D.; Liu, B.-Q.; Zhang, L.; Ma, M.-M.; Hu, Z.-Y.; Xia, J.; Xu, H.-W.; Du, X.-P. TIMP-1 polymorphisms in a Chinese Han population with intracerebral hemorrhage. Int. J. Neurosci. 2013, 124, 61–67. [Google Scholar] [CrossRef]
  76. Ho, W.-M.; Chen, C.-M.; Lee, Y.-S.; Chang, K.-H.; Chen, H.-W.; Chen, S.-T.; Chen, Y.-C. Association of MMP-9 Haplotypes and TIMP-1 Polymorphism with Spontaneous Deep Intracerebral Hemorrhage in the Taiwan Population. PLoS ONE 2015, 10, e0125397. [Google Scholar] [CrossRef]
  77. Reuter, B.; Bugert, P.; Stroick, M.; Bukow, S.; Griebe, M.; Hennerici, M.G.; Fatar, M. TIMP-2 Gene Polymorphism Is Associated with Intracerebral Hemorrhage. Cerebrovasc. Dis. 2009, 28, 558–563. [Google Scholar] [CrossRef]
  78. Chen, Y.-C.; Ho, W.M.; Lee, Y.S.; Chen, H.W.; Chen, C.-M. Polymorphisms in the Promoters of the MMP-2 and TIMP-2 Genes Are Associated with Spontaneous Deep Intracerebral Hemorrhage in the Taiwan Population. PLoS ONE 2015, 10, e0142482. [Google Scholar] [CrossRef]
  79. Guo, H.; You, M.; Wu, J.; Chen, A.; Wan, Y.; Gu, X.; Tan, S.; Xu, Y.; He, Q.; Hu, B. Genetics of Spontaneous Intracerebral Hemorrhage: Risk and Outcome. Front. Neurosci. 2022, 16, 874962. [Google Scholar]
  80. Traylor, M.; Anderson, C.D.; Rutten-Jacobs, L.C.; Falcone, G.J.; Comeau, M.E.; Ay, H.; Sudlow, C.L.; Xu, H.; Mitchell, B.D.; Cole, J.W.; et al. Subtype Specificity of Genetic Loci Associated With Stroke in 16,664 Cases and 32,792 Controls. Circ. Genom. Precis. Med. 2019, 12, e002338. [Google Scholar] [CrossRef] [Green Version]
  81. Woo, D.; Falcone, G.J.; Devan, W.J.; Brown, W.M.; Biffi, A.; Howard, T.D.; Anderson, C.D.; Brouwers, H.B.; Valant, V.; Battey, T.W.; et al. Meta-analysis of Genome-wide Association Studies Identifies 1q22 as a Susceptibility Locus for Intracerebral Hemorrhage. Am. J. Hum. Genet. 2014, 94, 511–521. [Google Scholar] [CrossRef] [Green Version]
  82. Marini, S.; Devan, W.J.; Radmanesh, F.; Miyares, L.; Poterba, T.; Hansen, B.M.; Norrving, B.; Jimenez-Conde, J.; Giralt-Steinhauer, E.; Elosua, R.; et al. 17p12 Influences Hematoma Volume and Outcome in Spontaneous Intracerebral Hemorrhage. Stroke 2018, 49, 1618–1625. [Google Scholar] [CrossRef]
  83. Biffi, A.; Anderson, C.D.; Jagiella, J.M.; Schmidt, H.; Kissela, B.; Hansen, B.M.; Jimenez, J.-C.; Pires, C.R.; Ayres, A.M.; Schwab, K.; et al. APOE Genotype Predicts Extent of Bleeding and Outcome in Lobar Intracerebral Hemorrhage. Lancet Neurol. 2011, 10, 702–709. [Google Scholar] [CrossRef]
  84. Silva, Y.; Leira, R.; Tejada, J.; Lainez, J.M.; Castillo, J.; Dávalos, A. Molecular Signatures of Vascular Injury Are Associated With Early Growth of Intracerebral Hemorrhage. Stroke 2005, 36, 86–91. [Google Scholar] [CrossRef]
  85. Shao, A.; Zhu, Z.; Li, L.; Zhang, S.; Zhang, J. Emerging therapeutic targets associated with the immune system in patients with intracerebral haemorrhage (ICH): From mechanisms to translation. EBioMedicine 2019, 45, 615–623. [Google Scholar] [CrossRef] [Green Version]
  86. Durocher, M.; Ander, B.P.; Jickling, G.; Hamade, F.; Hull, H.; Knepp, B.; Liu, D.Z.; Zhan, X.; Tran, A.; Cheng, X.; et al. Inflammatory, regulatory, and autophagy co-expression modules and hub genes underlie the peripheral immune response to human intracerebral hemorrhage. J. Neuroinflamm. 2019, 16, 1–21. [Google Scholar] [CrossRef]
  87. Walsh, K.B.; Zhang, X.; Zhu, X.; Wohleb, E.; Woo, D.; Lu, L.; Adeoye, O. Intracerebral Hemorrhage Induces Inflammatory Gene Expression in Peripheral Blood: Global Transcriptional Profiling in Intracerebral Hemorrhage Patients. DNA Cell Biol. 2019, 38, 660–669. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ekkert, A.; Šliachtenko, A.; Utkus, A.; Jatužis, D. Intracerebral Hemorrhage Genetics. Genes 2022, 13, 1250. https://doi.org/10.3390/genes13071250

AMA Style

Ekkert A, Šliachtenko A, Utkus A, Jatužis D. Intracerebral Hemorrhage Genetics. Genes. 2022; 13(7):1250. https://doi.org/10.3390/genes13071250

Chicago/Turabian Style

Ekkert, Aleksandra, Aleksandra Šliachtenko, Algirdas Utkus, and Dalius Jatužis. 2022. "Intracerebral Hemorrhage Genetics" Genes 13, no. 7: 1250. https://doi.org/10.3390/genes13071250

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