Genetics in Ischemic Stroke: Current Perspectives and Future Directions
Abstract
:1. Introduction
2. Mechanisms of Ischemic Stroke and Stroke Genetics
2.1. Cerebral Small Vessel Disease
2.2. Large Vessel Disease
2.3. Cardioembolic Stroke
2.4. Embolic Stroke of Undetermined Source
2.5. Mitochondrial Dysfunction
2.6. Haematological Disorders
3. Approaches to the Study of Genetics in Ischemic Stroke
3.1. Linkage Analysis
3.2. Genome-Wide Association Studies (GWASs)
- GWASs predominantly focus on identifying common genetic variants, often those with minor allele frequencies exceeding 1–5% [47]. While rare-variant GWASs are also performed to identify associations between higher-impact rare variants with disease risk, studies are generally underpowered for such analysis due to the large sample size and case control ratios required for discovery [48]. Novel statistical models and methods have been developed for more robust rare-variant GWAS analysis to address this [49].
- GWASs can establish associations between genomic loci and ischemic stroke. With large numbers of loci being identified through GWASs, key challenges remain in interpreting the biological significance of associated loci, in spite of the large repertoire of available tools and methods [50]. For example, a significant locus may be in a non-coding region [51] or exist in linkage disequilibrium with the true causal variant that was captured during sequencing [52], resulting in challenges with biological interpretation.
- Ischemic stroke GWASs have mostly been carried out in populations of European descent. This limits the applicability of various genomic findings, such as PRS and genomic underpinnings of ischemic stroke. This key limitation is further coupled with the heterogenous nature of ischemic stroke, in which population-specific differences could contribute to limited replicability across ethnicities.
4. Genes Related to Ischemic Stroke Occurrence
4.1. Monogenic Ischemic Stroke
4.2. Polygenic Ischemic Stroke
4.3. Polygenic Risk Score
Trial Name | Author and Publication Year | Study Type | Number of Subjects | Ethnicity | Genes Found | Remarks |
---|---|---|---|---|---|---|
STROMICS [57] | Yongjun Wang et al., 2023 | GWAS of all ischemic stroke, LAS, CES, SVS | 10,241 cases, control number was not specified | Chinese | 77 loci (>42% novel) | Largest Chinese GWAS |
GWAS of Intracranial Artery Stenosis (ICAS) [65] | Shogo Dofuku et al., 2023 | GWAS of LAS | 757 (408 cases and 349 controls) | Japanese | rs112735431 in RNF213 | First GWAS of ICAS |
MEGAStroke [55] | Aniket Mishra et al., 2022 | Meta-analysis | >520,000 (67,162 cases and 454,450 controls) | Multi-ancestry (largely European, East Asia and African) | 89 loci (61 novel) (SH3PXD2A, FURIN GRK5 and NOS3) | The largest published GWAS meta-analysis |
COMPASS [58] | Keith L. Keene et al., 2020 | Meta-analysis of any ischemic stroke | >22,000 (3734 cases and 18,317 controls) | African | 1 locus confirmed (rs55931441 near HNF1A gene) 29 other potential variants including those mapped to SFXN4 and TMEM108 genes | Consortium of Minority Population Genome-Wide Association Studies of Stroke |
SiGN Multi-ancestry GWAS [56] | Rainer Malik et al., 2018 | Meta-analysis Of any ischemic stroke, LAS, CES | 521,612 (67,162 cases and 454,450 controls) | Multi-ancestry (Largely European, also East Asian, African, South Asian, mixed Asian and Latin American) | 32 loci(22 novel) CASZ1, WNT2B, KCNK3 for any stroke. CDK6, PDE3A, PRPF8, ILF3-SLC44A2 for any ischemic stroke EDNRA, LINC01492 for LAS RGS7, NKX2-5 for CES | NINDS Stroke Genetics Network (SiGN), MEGASTROKE Consortium |
GWAS on SVO stroke [66] | Tsong-Hai Lee et al., 2017 | GWAS of SVS | 2073 (342 cases and 1731 controls) | East Asian (Han Chinese) | rs2594966, rs2594973, rs4684776 in ATG7 | |
CHARGE [67] | Audrey Y Chu et al., 2016 | Meta-analysis of any type of stroke | 84,961 (4348 cases and 80,613 controls) | European ancestry | rs12204590 near FOXF2 associated with risk of all-stroke | Neurology Working Group of the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium |
MetaStroke [68] | Traylor M et al., 2012 | GWAS of LAS, CES | 74,393 (12,389 cases and 62,004 controls) | European ancestry | Cardioembolic stroke near PITX2 and ZFHX3 Large-vessel stroke at a 9p21 locus and HDAC9 | Goal to validate associations and identify novel genetic associations for ischemic stroke and its subtypes |
GWAS for large vessel stroke [69] | Hugh S Markus et al., 2012 | GWAS of LAS, CES | 12,140 (5859 cases and 6281 controls) | European ancestry | HDAC9 gene for large vessel stroke PITX2 and ZFHX3 for cardioembolic stroke |
4.4. SARS-CoV-2 Infection and Ischemic Stroke Genetics
5. Genes Related to Stroke Treatment Response: Pharmacogenetics and Pharmacogenomics
5.1. Recombinant Tissue Plasminogen Activator (rtPA):
5.2. Antiplatelet Therapy
5.2.1. Aspirin
5.2.2. Platelet P2Y12 Receptor Antagonist, P2Y12 Inhibitor (Clopidogrel and Ticagrelor)
5.3. Oral Anticoagulants (Warfarin and Direct-Acting Oral Anticoagulants)
6. Genetics of Stroke Recovery
7. Clinical and Translational Applications of Genetics in Ischemic Stroke
- Molecular diagnosis, prognosis, and counselling of patients with typical clinical or radiological phenotypes suggestive of a monogenic disorder, e.g., anterior temporal lobe white matter hyperintensities seen in CADASIL.
- Molecular diagnosis of monogenic disorders in early-onset stroke, stroke with systemic manifestations, or patients with a significant family history.
- Tailoring antiplatelet therapy using pharmacogenetics approaches, e.g., CYP2C19 genotyping for clopidogrel.
- Risk prediction based on common genetic variants using polygenic risk scores.
8. Future Directions
- (1)
- Next-generation sequencing on a large scale
- (2)
- Integration of multi-omics data to understand stroke pathophysiology
- (3)
- Gene therapy
- (4)
- Precision medicine
- (5)
- New drug development
- (6)
- Expanding genetic studies to underrepresented populations
- (7)
- Enhancement of global collaboration
9. Conclusions
Author Contributions
Funding
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- GBD 2019 Stroke Collaborators. Global, regional, and national burden of stroke and its risk factors, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet Neurol. 2021, 20, 795–820. [Google Scholar] [CrossRef] [PubMed]
- Liao, D.; Myers, R.; Hunt, S.; Shahar, E.; Paton, C.; Burke, G.; Province, M.; Heiss, G. Familial history of stroke and stroke risk. The Family Heart Study. Stroke 1997, 28, 1908–1912. [Google Scholar] [CrossRef]
- Osler, M.; Villumsen, M.D.; Jorgensen, M.B.; Hjelmborg, J.V.B.; Christensen, K.; Wium-Andersen, M.K. Familial risk and heritability of ischemic heart disease and stroke in Danish twins. Scand. J. Public Health 2022, 50, 199–204. [Google Scholar] [CrossRef]
- Bevan, S.; Traylor, M.; Adib-Samii, P.; Malik, R.; Paul, N.L.; Jackson, C.; Farrall, M.; Rothwell, P.M.; Sudlow, C.; Dichgans, M.; et al. Genetic heritability of ischemic stroke and the contribution of previously reported candidate gene and genomewide associations. Stroke 2012, 43, 3161–3167. [Google Scholar] [CrossRef]
- Chojdak-Lukasiewicz, J.; Dziadkowiak, E.; Budrewicz, S. Monogenic Causes of Strokes. Genes 2021, 12, 1855. [Google Scholar] [CrossRef]
- Fox, C.S.; Polak, J.F.; Chazaro, I.; Cupples, A.; Wolf, P.A.; D’Agostino, R.A.; O’Donnell, C.J.; Framingham Heart, S. Genetic and environmental contributions to atherosclerosis phenotypes in men and women: Heritability of carotid intima-media thickness in the Framingham Heart Study. Stroke 2003, 34, 397–401. [Google Scholar] [CrossRef]
- Jiang, Y.; Liu, Z.; Liao, Y.; Sun, S.; Dai, Y.; Tang, Y. Ischemic stroke: From pathological mechanisms to neuroprotective strategies. Front. Neurol. 2022, 13, 1013083. [Google Scholar] [CrossRef]
- Doyle, K.P.; Simon, R.P.; Stenzel-Poore, M.P. Mechanisms of ischemic brain damage. Neuropharmacology 2008, 55, 310–318. [Google Scholar] [CrossRef]
- Pantoni, L. Cerebral small vessel disease: From pathogenesis and clinical characteristics to therapeutic challenges. Lancet Neurol. 2010, 9, 689–701. [Google Scholar] [CrossRef] [PubMed]
- Cuadrado-Godia, E.; Dwivedi, P.; Sharma, S.; Ois Santiago, A.; Roquer Gonzalez, J.; Balcells, M.; Laird, J.; Turk, M.; Suri, H.S.; Nicolaides, A.; et al. Cerebral Small Vessel Disease: A Review Focusing on Pathophysiology, Biomarkers, and Machine Learning Strategies. J. Stroke 2018, 20, 302–320. [Google Scholar] [CrossRef] [PubMed]
- Chojdak-Lukasiewicz, J.; Dziadkowiak, E.; Zimny, A.; Paradowski, B. Cerebral small vessel disease: A review. Adv. Clin. Exp. Med. 2021, 30, 349–356. [Google Scholar] [CrossRef]
- Wardlaw, J.M.; Smith, C.; Dichgans, M. Small vessel disease: Mechanisms and clinical implications. Lancet Neurol. 2019, 18, 684–696. [Google Scholar] [CrossRef] [PubMed]
- Li, Q.; Yang, Y.; Reis, C.; Tao, T.; Li, W.; Li, X.; Zhang, J.H. Cerebral Small Vessel Disease. Cell Transplant. 2018, 27, 1711–1722. [Google Scholar] [CrossRef] [PubMed]
- Wang, Z.; Chen, Q.; Chen, J.; Yang, N.; Zheng, K. Risk factors of cerebral small vessel disease: A systematic review and meta-analysis. Medicine 2021, 100, e28229. [Google Scholar] [CrossRef] [PubMed]
- Bhagat, R.; Marini, S.; Romero, J.R. Genetic considerations in cerebral small vessel diseases. Front. Neurol. 2023, 14, 1080168. [Google Scholar] [CrossRef] [PubMed]
- Locatelli, M.; Padovani, A.; Pezzini, A. Pathophysiological Mechanisms and Potential Therapeutic Targets in Cerebral Autosomal Dominant Arteriopathy With Subcortical Infarcts and Leukoencephalopathy (CADASIL). Front. Pharmacol. 2020, 11, 321. [Google Scholar] [CrossRef] [PubMed]
- Rutten, J.W.; Hack, R.J.; Duering, M.; Gravesteijn, G.; Dauwerse, J.G.; Overzier, M.; van den Akker, E.B.; Slagboom, E.; Holstege, H.; Nho, K.; et al. Broad phenotype of cysteine-altering NOTCH3 variants in UK Biobank: CADASIL to nonpenetrance. Neurology 2020, 95, e1835–e1843. [Google Scholar] [CrossRef] [PubMed]
- Chabriat, H.; Joutel, A.; Dichgans, M.; Tournier-Lasserve, E.; Bousser, M.G. Cadasil. Lancet Neurol. 2009, 8, 643–653. [Google Scholar] [CrossRef]
- Haffner, C.; Malik, R.; Dichgans, M. Genetic factors in cerebral small vessel disease and their impact on stroke and dementia. J. Cereb. Blood Flow Metab. 2016, 36, 158–171. [Google Scholar] [CrossRef]
- Cole, J.W. Large Artery Atherosclerotic Occlusive Disease. Continuum 2017, 23, 133–157. [Google Scholar] [CrossRef]
- Marulanda-Londono, E.; Chaturvedi, S. Stroke due to large vessel atherosclerosis: Five new things. Neurol. Clin. Pract. 2016, 6, 252–258. [Google Scholar] [CrossRef] [PubMed]
- Arida, A.; Protogerou, A.D.; Kitas, G.D.; Sfikakis, P.P. Systemic Inflammatory Response and Atherosclerosis: The Paradigm of Chronic Inflammatory Rheumatic Diseases. Int. J. Mol. Sci. 2018, 19, 1890. [Google Scholar] [CrossRef] [PubMed]
- Alfaddagh, A.; Martin, S.S.; Leucker, T.M.; Michos, E.D.; Blaha, M.J.; Lowenstein, C.J.; Jones, S.R.; Toth, P.P. Inflammation and cardiovascular disease: From mechanisms to therapeutics. Am. J. Prev. Cardiol. 2020, 4, 100130. [Google Scholar] [CrossRef] [PubMed]
- Biros, E.; Karan, M.; Golledge, J. Genetic variation and atherosclerosis. Curr. Genom. 2008, 9, 29–42. [Google Scholar] [CrossRef] [PubMed]
- Kamada, F.; Aoki, Y.; Narisawa, A.; Abe, Y.; Komatsuzaki, S.; Kikuchi, A.; Kanno, J.; Niihori, T.; Ono, M.; Ishii, N.; et al. A genome-wide association study identifies RNF213 as the first Moyamoya disease gene. J. Hum. Genet. 2011, 56, 34–40. [Google Scholar] [CrossRef]
- Duan, L.; Wei, L.; Tian, Y.; Zhang, Z.; Hu, P.; Wei, Q.; Liu, S.; Zhang, J.; Wang, Y.; Li, D.; et al. Novel Susceptibility Loci for Moyamoya Disease Revealed by a Genome-Wide Association Study. Stroke 2018, 49, 11–18. [Google Scholar] [CrossRef]
- Mukawa, M.; Nariai, T.; Onda, H.; Yoneyama, T.; Aihara, Y.; Hirota, K.; Kudo, T.; Sumita, K.; Maehara, T.; Kawamata, T.; et al. Exome Sequencing Identified CCER2 as a Novel Candidate Gene for Moyamoya Disease. J. Stroke Cerebrovasc. Dis. 2017, 26, 150–161. [Google Scholar] [CrossRef]
- Yang, Y.; Wang, J.; Liang, Q.; Wang, Y.; Chen, X.; Zhang, Q.; Na, S.; Liu, Y.; Yan, T.; Hang, C.; et al. PHACTR1 is associated with disease progression in Chinese Moyamoya disease. PeerJ 2020, 8, e8841. [Google Scholar] [CrossRef]
- Feng, X.; Yu, F.; Zhou, X.; Liu, Z.; Liao, D.; Huang, Q.; Li, X.; Jin, X.; Xia, J. MMP9 rs17576 Is Simultaneously Correlated with Symptomatic Intracranial Atherosclerotic Stenosis and White Matter Hyperintensities in Chinese Population. Cerebrovasc. Dis. 2021, 50, 4–11. [Google Scholar] [CrossRef]
- Liu, M.; Gutierrez, J. Genetic Risk Factors of Intracranial Atherosclerosis. Curr. Atheroscler. Rep. 2020, 22, 13. [Google Scholar] [CrossRef]
- Pillai, A.A.; Tadi, P.; Kanmanthareddy, A. Cardioembolic Stroke. In StatPearls; StatPearls Publishing LLC.: Treasure Island, FL, USA, 2023. [Google Scholar]
- Mahida, S.; Lubitz, S.A.; Rienstra, M.; Milan, D.J.; Ellinor, P.T. Monogenic atrial fibrillation as pathophysiological paradigms. Cardiovasc. Res. 2011, 89, 692–700. [Google Scholar] [CrossRef] [PubMed]
- Gudbjartsson, D.F.; Holm, H.; Gretarsdottir, S.; Thorleifsson, G.; Walters, G.B.; Thorgeirsson, G.; Gulcher, J.; Mathiesen, E.B.; Njolstad, I.; Nyrnes, A.; et al. A sequence variant in ZFHX3 on 16q22 associates with atrial fibrillation and ischemic stroke. Nat. Genet. 2009, 41, 876–878. [Google Scholar] [CrossRef] [PubMed]
- Gretarsdottir, S.; Thorleifsson, G.; Manolescu, A.; Styrkarsdottir, U.; Helgadottir, A.; Gschwendtner, A.; Kostulas, K.; Kuhlenbaumer, G.; Bevan, S.; Jonsdottir, T.; et al. Risk variants for atrial fibrillation on chromosome 4q25 associate with ischemic stroke. Ann. Neurol. 2008, 64, 402–409. [Google Scholar] [CrossRef] [PubMed]
- Zou, R.; Zhang, D.; Lv, L.; Shi, W.; Song, Z.; Yi, B.; Lai, B.; Chen, Q.; Yang, S.; Hua, P. Bioinformatic gene analysis for potential biomarkers and therapeutic targets of atrial fibrillation-related stroke. J. Transl. Med. 2019, 17, 45. [Google Scholar] [CrossRef]
- Kamel, H.; Merkler, A.E.; Iadecola, C.; Gupta, A.; Navi, B.B. Tailoring the Approach to Embolic Stroke of Undetermined Source: A Review. JAMA Neurol. 2019, 76, 855–861. [Google Scholar] [CrossRef] [PubMed]
- Hart, R.G.; Catanese, L.; Perera, K.S.; Ntaios, G.; Connolly, S.J. Embolic Stroke of Undetermined Source: A Systematic Review and Clinical Update. Stroke 2017, 48, 867–872. [Google Scholar] [CrossRef]
- Georgakis, M.K.; Parodi, L.; Frerich, S.; Mayerhofer, E.; Tsivgoulis, G.; Pirruccello, J.P.; Slowik, A.; Rundek, T.; Malik, R.; Dichgans, M.; et al. Genetic Architecture of Stroke of Undetermined Source: Overlap with Known Stroke Etiologies and Associations with Modifiable Risk Factors. Ann. Neurol. 2022, 91, 640–651. [Google Scholar] [CrossRef]
- Weng, L.C.; Khurshid, S.; Gunn, S.; Trinquart, L.; Lunetta, K.L.; Xu, H.; NINDS Stroke Genetics Network; Benjamin, E.J.; Ellinor, P.T.; Anderson, C.D.; et al. Clinical and Genetic Atrial Fibrillation Risk and Discrimination of Cardioembolic From Noncardioembolic Stroke. Stroke 2023, 54, 1777–1785. [Google Scholar] [CrossRef]
- Lorenzoni, P.J.; Scola, R.H.; Kay, C.S.; Arndt, R.C.; Freund, A.A.; Bruck, I.; Santos, M.L.; Werneck, L.C. MELAS: Clinical features, muscle biopsy and molecular genetics. Arq. Neuropsiquiatr. 2009, 67, 668–676. [Google Scholar] [CrossRef]
- Dichgans, M.; Markus, H.S. Genetic association studies in stroke: Methodological issues and proposed standard criteria. Stroke 2005, 36, 2027–2031. [Google Scholar] [CrossRef]
- Hegele, R.A. Genetic association studies of stroke: Hope, signal, and noise. Stroke 2002, 33, 2769. [Google Scholar]
- Tournier-Lasserve, E.; Joutel, A.; Melki, J.; Weissenbach, J.; Lathrop, G.M.; Chabriat, H.; Mas, J.L.; Cabanis, E.A.; Baudrimont, M.; Maciazek, J.; et al. Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy maps to chromosome 19q12. Nat. Genet. 1993, 3, 256–259. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Hassan, A.; Sham, P.C.; Markus, H.S. Planning genetic studies in human stroke: Sample size estimates based on family history data. Neurology 2002, 58, 1483–1488. [Google Scholar] [CrossRef] [PubMed]
- Szolnoki, Z.; Melegh, B. Gene-gene and gene-environment interplay represent specific susceptibility for different types of ischaemic stroke and leukoaraiosis. Curr. Med. Chem. 2006, 13, 1627–1634. [Google Scholar] [CrossRef] [PubMed]
- Marees, A.T.; de Kluiver, H.; Stringer, S.; Vorspan, F.; Curis, E.; Marie-Claire, C.; Derks, E.M. A tutorial on conducting genome-wide association studies: Quality control and statistical analysis. Int. J. Methods Psychiatr. Res. 2018, 27, e1608. [Google Scholar] [CrossRef]
- Lee, S.; Abecasis, G.R.; Boehnke, M.; Lin, X. Rare-variant association analysis: Study designs and statistical tests. Am. J. Hum. Genet. 2014, 95, 5–23. [Google Scholar] [CrossRef] [PubMed]
- Mbatchou, J.; Barnard, L.; Backman, J.; Marcketta, A.; Kosmicki, J.A.; Ziyatdinov, A.; Benner, C.; O’Dushlaine, C.; Barber, M.; Boutkov, B.; et al. Computationally efficient whole-genome regression for quantitative and binary traits. Nat. Genet. 2021, 53, 1097–1103. [Google Scholar] [CrossRef]
- Edwards, S.L.; Beesley, J.; French, J.D.; Dunning, A.M. Beyond GWASs: Illuminating the dark road from association to function. Am. J. Hum. Genet. 2013, 93, 779–797. [Google Scholar] [CrossRef]
- Hindorff, L.A.; Sethupathy, P.; Junkins, H.A.; Ramos, E.M.; Mehta, J.P.; Collins, F.S.; Manolio, T.A. Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc. Natl. Acad. Sci. USA 2009, 106, 9362–9367. [Google Scholar] [CrossRef]
- Rodney, A.; Karanjeet, K.; Benzow, K.; Koob, M.D. A common Alu element insertion in the 3′UTR of TMEM106B is associated with an increased risk of dementia. bioRxiv 2023. [Google Scholar] [CrossRef]
- Li, J.; Abedi, V.; Zand, R. Dissecting Polygenic Etiology of Ischemic Stroke in the Era of Precision Medicine. J. Clin. Med. 2022, 11, 5980. [Google Scholar] [CrossRef] [PubMed]
- Ekkert, A.; Sliachtenko, A.; Grigaite, J.; Burnyte, B.; Utkus, A.; Jatuzis, D. Ischemic Stroke Genetics: What Is New and How to Apply It in Clinical Practice? Genes 2021, 13, 48. [Google Scholar] [CrossRef] [PubMed]
- Mishra, A.; Malik, R.; Hachiya, T.; Jurgenson, T.; Namba, S.; Posner, D.C.; Kamanu, F.K.; Koido, M.; Le Grand, Q.; Shi, M.; et al. Stroke genetics informs drug discovery and risk prediction across ancestries. Nature 2022, 611, 115–123. [Google Scholar] [CrossRef] [PubMed]
- Malik, R.; Chauhan, G.; Traylor, M.; Sargurupremraj, M.; Okada, Y.; Mishra, A.; Rutten-Jacobs, L.; Giese, A.K.; van der Laan, S.W.; Gretarsdottir, S.; et al. Multiancestry genome-wide association study of 520,000 subjects identifies 32 loci associated with stroke and stroke subtypes. Nat. Genet. 2018, 50, 524–537. [Google Scholar] [CrossRef]
- Cheng, S.; Xu, Z.; Bian, S.; Chen, X.; Shi, Y.; Li, Y.; Duan, Y.; Liu, Y.; Lin, J.; Jiang, Y.; et al. The STROMICS genome study: Deep whole-genome sequencing and analysis of 10K Chinese patients with ischemic stroke reveal complex genetic and phenotypic interplay. Cell Discov. 2023, 9, 75. [Google Scholar] [CrossRef] [PubMed]
- Keene, K.L.; Hyacinth, H.I.; Bis, J.C.; Kittner, S.J.; Mitchell, B.D.; Cheng, Y.C.; Pare, G.; Chong, M.; O’Donnell, M.; Meschia, J.F.; et al. Genome-Wide Association Study Meta-Analysis of Stroke in 22 000 Individuals of African Descent Identifies Novel Associations With Stroke. Stroke 2020, 51, 2454–2463. [Google Scholar] [CrossRef]
- Li, J.; Chaudhary, D.P.; Khan, A.; Griessenauer, C.; Carey, D.J.; Zand, R.; Abedi, V. Polygenic Risk Scores Augment Stroke Subtyping. Neurol. Genet. 2021, 7, e560. [Google Scholar] [CrossRef] [PubMed]
- Neumann, J.T.; Riaz, M.; Bakshi, A.; Polekhina, G.; Thao, L.T.P.; Nelson, M.R.; Woods, R.L.; Abraham, G.; Inouye, M.; Reid, C.M.; et al. Predictive Performance of a Polygenic Risk Score for Incident Ischemic Stroke in a Healthy Older Population. Stroke 2021, 52, 2882–2891. [Google Scholar] [CrossRef]
- Abraham, G.; Malik, R.; Yonova-Doing, E.; Salim, A.; Wang, T.; Danesh, J.; Butterworth, A.S.; Howson, J.M.M.; Inouye, M.; Dichgans, M. Genomic risk score offers predictive performance comparable to clinical risk factors for ischaemic stroke. Nat. Commun. 2019, 10, 5819, Erratum in Nat. Commun. 2020, 11, 1036. [Google Scholar] [CrossRef]
- O’Sullivan, J.W.; Shcherbina, A.; Justesen, J.M.; Turakhia, M.; Perez, M.; Wand, H.; Tcheandjieu, C.; Clarke, S.L.; Rivas, M.A.; Ashley, E.A. Combining Clinical and Polygenic Risk Improves Stroke Prediction Among Individuals with Atrial Fibrillation. Circ. Genom. Precis. Med. 2021, 14, e003168. [Google Scholar] [CrossRef] [PubMed]
- Cui, Q.; Liu, F.; Li, J.; Niu, X.; Liu, Z.; Shen, C.; Hu, D.; Huang, K.; Chen, S.; Cao, J.; et al. Integrating polygenic and clinical risks to improve stroke risk stratification in prospective Chinese cohorts. Sci. China Life Sci. 2023, 66, 1626–1635. [Google Scholar] [CrossRef] [PubMed]
- Duncan, L.; Shen, H.; Gelaye, B.; Meijsen, J.; Ressler, K.; Feldman, M.; Peterson, R.; Domingue, B. Analysis of polygenic risk score usage and performance in diverse human populations. Nat. Commun. 2019, 10, 3328. [Google Scholar] [CrossRef] [PubMed]
- Dofuku, S.; Sonehara, K.; Miyawaki, S.; Sakaue, S.; Imai, H.; Shimizu, M.; Hongo, H.; Shinya, Y.; Ohara, K.; Teranishi, Y.; et al. Genome-Wide Association Study of Intracranial Artery Stenosis Followed by Phenome-Wide Association Study. Transl. Stroke Res. 2023, 14, 322–333. [Google Scholar] [CrossRef]
- Lee, T.H.; Ko, T.M.; Chen, C.H.; Chang, Y.J.; Lu, L.S.; Chang, C.H.; Huang, K.L.; Chang, T.Y.; Lee, J.D.; Chang, K.C.; et al. A genome-wide association study links small-vessel ischemic stroke to autophagy. Sci. Rep. 2017, 7, 15229. [Google Scholar] [CrossRef]
- Neurology Working Group of the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium; The Stroke Genetics Network (SiGN); The International Stroke Genetics Consortium (ISGC). Identification of additional risk loci for stroke and small vessel disease: A meta-analysis of genome-wide association studies. Lancet Neurol. 2016, 15, 695–707. [Google Scholar] [CrossRef]
- Traylor, M.; Farrall, M.; Holliday, E.G.; Sudlow, C.; Hopewell, J.C.; Cheng, Y.C.; Fornage, M.; Ikram, M.A.; Malik, R.; Bevan, S.; et al. Genetic risk factors for ischaemic stroke and its subtypes (the METASTROKE collaboration): A meta-analysis of genome-wide association studies. Lancet Neurol. 2012, 11, 951–962. [Google Scholar] [CrossRef]
- International Stroke Genetics, C.; Wellcome Trust Case Control, C.; Bellenguez, C.; Bevan, S.; Gschwendtner, A.; Spencer, C.C.; Burgess, A.I.; Pirinen, M.; Jackson, C.A.; Traylor, M.; et al. Genome-wide association study identifies a variant in HDAC9 associated with large vessel ischemic stroke. Nat. Genet. 2012, 44, 328–333. [Google Scholar] [CrossRef]
- Belani, P.; Schefflein, J.; Kihira, S.; Rigney, B.; Delman, B.N.; Mahmoudi, K.; Mocco, J.; Majidi, S.; Yeckley, J.; Aggarwal, A.; et al. COVID-19 Is an Independent Risk Factor for Acute Ischemic Stroke. AJNR Am. J. Neuroradiol. 2020, 41, 1361–1364. [Google Scholar] [CrossRef]
- Kihira, S.; Schefflein, J.; Mahmoudi, K.; Rigney, B.; Delman, B.N.; Mocco, J.; Doshi, A.; Belani, P. Association of Coronavirus Disease (COVID-19) With Large Vessel Occlusion Strokes: A Case-Control Study. AJR Am. J. Roentgenol. 2021, 216, 150–156. [Google Scholar] [CrossRef]
- Tu, T.M.; Seet, C.Y.H.; Koh, J.S.; Tham, C.H.; Chiew, H.J.; De Leon, J.A.; Chua, C.Y.K.; Hui, A.C.; Tan, S.S.Y.; Vasoo, S.S.; et al. Acute Ischemic Stroke During the Convalescent Phase of Asymptomatic COVID-2019 Infection in Men. JAMA Netw. Open 2021, 4, e217498. [Google Scholar] [CrossRef]
- Dmytriw, A.A.; Dibas, M.; Phan, K.; Efendizade, A.; Ospel, J.; Schirmer, C.; Settecase, F.; Heran, M.K.S.; Kuhn, A.L.; Puri, A.S.; et al. Acute ischaemic stroke associated with SARS-CoV-2 infection in North America. J. Neurol. Neurosurg. Psychiatry 2022, 93, 360–368. [Google Scholar] [CrossRef]
- Luo, W.; Liu, X.; Bao, K.; Huang, C. Ischemic stroke associated with COVID-19: A systematic review and meta-analysis. J. Neurol. 2022, 269, 1731–1740. [Google Scholar] [CrossRef]
- Zhang, S.; Zhang, J.; Wang, C.; Chen, X.; Zhao, X.; Jing, H.; Liu, H.; Li, Z.; Wang, L.; Shi, J. COVID-19 and ischemic stroke: Mechanisms of hypercoagulability (Review). Int. J. Mol. Med. 2021, 47, 21. [Google Scholar] [CrossRef]
- Sagris, D.; Papanikolaou, A.; Kvernland, A.; Korompoki, E.; Frontera, J.A.; Troxel, A.B.; Gavriatopoulou, M.; Milionis, H.; Lip, G.Y.H.; Michel, P.; et al. COVID-19 and ischemic stroke. Eur. J. Neurol. 2021, 28, 3826–3836. [Google Scholar] [CrossRef] [PubMed]
- Bereda, G. How is ischemic stroke linked to a COVID-19-infected patient different from other cardiovascular risk factors? a case report. Ann. Med. Surg. 2023, 85, 2995–2998. [Google Scholar] [CrossRef]
- Wijeratne, T.; Gillard Crewther, S.; Sales, C.; Karimi, L. COVID-19 Pathophysiology Predicts That Ischemic Stroke Occurrence Is an Expectation, Not an Exception-A Systematic Review. Front. Neurol. 2020, 11, 607221. [Google Scholar] [CrossRef] [PubMed]
- Parodi, L.; Myserlis, E.P.; Chung, J.; Georgakis, M.K.; Mayerhofer, E.; Henry, J.; Montgomery, B.E.; Moy, M.; Xu, H.; Malik, R.; et al. Shared genetic background between SARS-CoV-2 infection and large artery stroke. Int. J. Stroke 2022, 8, 873–880. [Google Scholar] [CrossRef]
- Llucia-Carol, L.; Muino, E.; Cullell, N.; Carcel-Marquez, J.; Lledos, M.; Gallego-Fabrega, C.; Martin-Campos, J.; Marti-Fabregas, J.; Aguilera-Simon, A.; Planas, A.M.; et al. Genetic Architecture of Ischaemic Strokes after COVID-19 Shows Similarities with Large Vessel Strokes. Int. J. Mol. Sci. 2023, 24, 13452. [Google Scholar] [CrossRef] [PubMed]
- Llucia-Carol, L.; Muino, E.; Gallego-Fabrega, C.; Carcel-Marquez, J.; Martin-Campos, J.; Lledos, M.; Cullell, N.; Fernandez-Cadenas, I. Pharmacogenetics studies in stroke patients treated with rtPA: A review of the most interesting findings. Pharmacogenomics 2021, 22, 1091–1097. [Google Scholar] [CrossRef]
- Asiimwe, I.G.; Pirmohamed, M. Drug-Drug-Gene Interactions in Cardiovascular Medicine. Pharmgenom. Pers. Med. 2022, 15, 879–911. [Google Scholar] [CrossRef] [PubMed]
- Amici, S.; Paciaroni, M.; Agnelli, G.; Caso, V. Gene-drug interaction in stroke. Stroke Res. Treat. 2011, 2011, 212485. [Google Scholar] [CrossRef] [PubMed]
- Yaghi, S.; Willey, J.Z.; Cucchiara, B.; Goldstein, J.N.; Gonzales, N.R.; Khatri, P.; Kim, L.J.; Mayer, S.A.; Sheth, K.N.; Schwamm, L.H.; et al. Treatment and Outcome of Hemorrhagic Transformation After Intravenous Alteplase in Acute Ischemic Stroke: A Scientific Statement for Healthcare Professionals From the American Heart Association/American Stroke Association. Stroke 2017, 48, e343–e361. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez-Conejero, R.; Fernandez-Cadenas, I.; Iniesta, J.A.; Marti-Fabregas, J.; Obach, V.; Alvarez-Sabin, J.; Vicente, V.; Corral, J.; Montaner, J.; Proyecto Ictus Research, G. Role of fibrinogen levels and factor XIII V34L polymorphism in thrombolytic therapy in stroke patients. Stroke 2006, 37, 2288–2293. [Google Scholar] [CrossRef]
- Muino, E.; Carcel-Marquez, J.; Carrera, C.; Llucia-Carol, L.; Gallego-Fabrega, C.; Cullell, N.; Lledos, M.; Castillo, J.; Sobrino, T.; Campos, F.; et al. RP11-362K2.2:RP11-767I20.1 Genetic Variation Is Associated with Post-Reperfusion Therapy Parenchymal Hematoma. A GWAS Meta-Analysis. J. Clin. Med. 2021, 10, 3137. [Google Scholar] [CrossRef]
- Kernan, W.N.; Ovbiagele, B.; Black, H.R.; Bravata, D.M.; Chimowitz, M.I.; Ezekowitz, M.D.; Fang, M.C.; Fisher, M.; Furie, K.L.; Heck, D.V.; et al. Guidelines for the prevention of stroke in patients with stroke and transient ischemic attack: A guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke 2014, 45, 2160–2236. [Google Scholar] [CrossRef]
- Hankey, G.J.; Eikelboom, J.W. Aspirin resistance. Lancet 2006, 367, 606–617. [Google Scholar] [CrossRef]
- Parsa-Kondelaji, M.; Mansouritorghabeh, H. Aspirin and clopidogrel resistance; a neglected gap in stroke and cardiovascular practice in Iran: A systematic review and meta-analysis. Thromb. J. 2023, 21, 79. [Google Scholar] [CrossRef]
- Collet, J.P.; Montalescot, G. Platelet function testing and implications for clinical practice. J. Cardiovasc. Pharmacol. Ther. 2009, 14, 157–169. [Google Scholar] [CrossRef]
- Gorelick, P.B.; Farooq, M.U. Advances in our understanding of “resistance” to antiplatelet agents for prevention of ischemic stroke. Stroke Res. Treat. 2013, 2013, 727842. [Google Scholar] [CrossRef]
- Collette, S.L.; Bokkers, R.P.H.; Dierckx, R.; van der Laan, M.J.; Zeebregts, C.J.; Uyttenboogaart, M. Clinical importance of testing for clopidogrel resistance in patients undergoing carotid artery stenting-a systematic review. Ann. Transl. Med. 2021, 9, 1211. [Google Scholar] [CrossRef] [PubMed]
- Sibbing, D.; Koch, W.; Gebhard, D.; Schuster, T.; Braun, S.; Stegherr, J.; Morath, T.; Schomig, A.; von Beckerath, N.; Kastrati, A. Cytochrome 2C19*17 allelic variant, platelet aggregation, bleeding events, and stent thrombosis in clopidogrel-treated patients with coronary stent placement. Circulation 2010, 121, 512–518. [Google Scholar] [CrossRef] [PubMed]
- Mega, J.L.; Simon, T.; Collet, J.P.; Anderson, J.L.; Antman, E.M.; Bliden, K.; Cannon, C.P.; Danchin, N.; Giusti, B.; Gurbel, P.; et al. Reduced-function CYP2C19 genotype and risk of adverse clinical outcomes among patients treated with clopidogrel predominantly for PCI: A meta-analysis. JAMA 2010, 304, 1821–1830. [Google Scholar] [CrossRef] [PubMed]
- Xu, J.; Wang, A.; Wangqin, R.; Mo, J.; Chen, Z.; Dai, L.; Meng, X.; Zhao, X.; Wang, Y.; Li, H.; et al. Efficacy of clopidogrel for stroke depends on CYP2C19 genotype and risk profile. Ann. Neurol. 2019, 86, 419–426. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Zhao, X.; Lin, J.; Li, H.; Johnston, S.C.; Lin, Y.; Pan, Y.; Liu, L.; Wang, D.; Wang, C.; et al. Association Between CYP2C19 Loss-of-Function Allele Status and Efficacy of Clopidogrel for Risk Reduction Among Patients With Minor Stroke or Transient Ischemic Attack. JAMA 2016, 316, 70–78. [Google Scholar] [CrossRef] [PubMed]
- Bonney, P.A.; Yim, B.; Brinjikji, W.; Walcott, B.P. Pharmacogenomic considerations for antiplatelet agents: The era of precision medicine in stroke prevention and neurointerventional practice. Cold Spring Harb. Mol. Case Stud. 2019, 5, a003731. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Meng, X.; Wang, A.; Xie, X.; Pan, Y.; Johnston, S.C.; Li, H.; Bath, P.M.; Dong, Q.; Xu, A.; et al. Ticagrelor versus Clopidogrel in CYP2C19 Loss-of-Function Carriers with Stroke or TIA. N. Engl. J. Med. 2021, 385, 2520–2530. [Google Scholar] [CrossRef]
- Johnson, J.A.; Gong, L.; Whirl-Carrillo, M.; Gage, B.F.; Scott, S.A.; Stein, C.M.; Anderson, J.L.; Kimmel, S.E.; Lee, M.T.; Pirmohamed, M.; et al. Clinical Pharmacogenetics Implementation Consortium Guidelines for CYP2C9 and VKORC1 genotypes and warfarin dosing. Clin. Pharmacol. Ther. 2011, 90, 625–629. [Google Scholar] [CrossRef]
- Rieder, M.J.; Reiner, A.P.; Gage, B.F.; Nickerson, D.A.; Eby, C.S.; McLeod, H.L.; Blough, D.K.; Thummel, K.E.; Veenstra, D.L.; Rettie, A.E. Effect of VKORC1 haplotypes on transcriptional regulation and warfarin dose. N. Engl. J. Med. 2005, 352, 2285–2293. [Google Scholar] [CrossRef]
- Manolopoulos, V.G.; Ragia, G.; Tavridou, A. Pharmacogenetics of coumarinic oral anticoagulants. Pharmacogenomics 2010, 11, 493–496. [Google Scholar] [CrossRef]
- Limdi, N.A.; Arnett, D.K.; Goldstein, J.A.; Beasley, T.M.; McGwin, G.; Adler, B.K.; Acton, R.T. Influence of CYP2C9 and VKORC1 on warfarin dose, anticoagulation attainment and maintenance among European-Americans and African-Americans. Pharmacogenomics 2008, 9, 511–526. [Google Scholar] [CrossRef] [PubMed]
- McDonald, M.G.; Rieder, M.J.; Nakano, M.; Hsia, C.K.; Rettie, A.E. CYP4F2 is a vitamin K1 oxidase: An explanation for altered warfarin dose in carriers of the V433M variant. Mol. Pharmacol. 2009, 75, 1337–1346. [Google Scholar] [CrossRef] [PubMed]
- Kampouraki, E.; Kamali, F. Pharmacogenetics of anticoagulants used for stroke prevention in patients with atrial fibrillation. Expert Opin. Drug. Metab. Toxicol. 2019, 15, 449–458. [Google Scholar] [CrossRef] [PubMed]
- Duarte, J.D.; Cavallari, L.H. Pharmacogenetics to guide cardiovascular drug therapy. Nat. Rev. Cardiol. 2021, 18, 649–665. [Google Scholar] [CrossRef] [PubMed]
- Mega, J.L.; Walker, J.R.; Ruff, C.T.; Vandell, A.G.; Nordio, F.; Deenadayalu, N.; Murphy, S.A.; Lee, J.; Mercuri, M.F.; Giugliano, R.P.; et al. Genetics and the clinical response to warfarin and edoxaban: Findings from the randomised, double-blind ENGAGE AF-TIMI 48 trial. Lancet 2015, 385, 2280–2287. [Google Scholar] [CrossRef]
- Mola-Caminal, M.; Carrera, C.; Soriano-Tarraga, C.; Giralt-Steinhauer, E.; Diaz-Navarro, R.M.; Tur, S.; Jimenez, C.; Medina-Dols, A.; Cullell, N.; Torres-Aguila, N.P.; et al. PATJ Low Frequency Variants Are Associated With Worse Ischemic Stroke Functional Outcome. Circ. Res. 2019, 124, 114–120. [Google Scholar] [CrossRef] [PubMed]
- Soderholm, M.; Pedersen, A.; Lorentzen, E.; Stanne, T.M.; Bevan, S.; Olsson, M.; Cole, J.W.; Fernandez-Cadenas, I.; Hankey, G.J.; Jimenez-Conde, J.; et al. Genome-wide association meta-analysis of functional outcome after ischemic stroke. Neurology 2019, 92, e1271–e1283. [Google Scholar] [CrossRef]
- Zhu, R.; Zhao, Y.; Tian, D.; Guo, N.; Zhang, C.; Liu, X. GWAS-linked hot loci predict short-term functional outcome and recurrence of ischemic stroke in Chinese population. Am. J. Transl. Res. 2021, 13, 4521–4534. [Google Scholar]
- Aldridge, C.M.; Robynne, B.; Keene, K.L.; Hsu, F.C.; Sale, M.M.; Worrall, B.B. Post Stroke Motor Recovery Genome Wide Association Study: A Domain-Specific Approach. medRxiv 2023. [Google Scholar] [CrossRef]
- Dichgans, M.; Beaufort, N.; Debette, S.; Anderson, C.D. Stroke Genetics: Turning Discoveries into Clinical Applications. Stroke 2021, 52, 2974–2982. [Google Scholar] [CrossRef]
- Bycroft, C.; Freeman, C.; Petkova, D.; Band, G.; Elliott, L.T.; Sharp, K.; Motyer, A.; Vukcevic, D.; Delaneau, O.; O’Connell, J.; et al. The UK Biobank resource with deep phenotyping and genomic data. Nature 2018, 562, 203–209. [Google Scholar] [CrossRef]
- Leitsalu, L.; Haller, T.; Esko, T.; Tammesoo, M.L.; Alavere, H.; Snieder, H.; Perola, M.; Ng, P.C.; Magi, R.; Milani, L.; et al. Cohort Profile: Estonian Biobank of the Estonian Genome Center, University of Tartu. Int. J. Epidemiol. 2015, 44, 1137–1147. [Google Scholar] [CrossRef] [PubMed]
- Kurki, M.I.; Karjalainen, J.; Palta, P.; Sipila, T.P.; Kristiansson, K.; Donner, K.M.; Reeve, M.P.; Laivuori, H.; Aavikko, M.; Kaunisto, M.A.; et al. FinnGen provides genetic insights from a well-phenotyped isolated population. Nature 2023, 613, 508–518, Erratum in Nature 2023, 615, E19. [Google Scholar] [CrossRef] [PubMed]
- Kubo, M.; Guest, E. BioBank Japan project: Epidemiological study. J. Epidemiol. 2017, 27, S1. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Shao, C.; Zhou, H.; Du, H.; Chen, H.; Wan, H.; He, Y. Multi-omics research strategies in ischemic stroke: A multidimensional perspective. Ageing Res. Rev. 2022, 81, 101730. [Google Scholar] [CrossRef] [PubMed]
- Skukan, L.; Brezak, M.; Ister, R.; Klimaschewski, L.; Vojta, A.; Zoldos, V.; Gajovic, S. Lentivirus- or AAV-mediated gene therapy interventions in ischemic stroke: A systematic review of preclinical in vivo studies. J. Cereb. Blood Flow Metab. 2022, 42, 219–236. [Google Scholar] [CrossRef]
- Chen, Y.C.; Ma, N.X.; Pei, Z.F.; Wu, Z.; Do-Monte, F.H.; Keefe, S.; Yellin, E.; Chen, M.S.; Yin, J.C.; Lee, G.; et al. A NeuroD1 AAV-Based Gene Therapy for Functional Brain Repair after Ischemic Injury through In Vivo Astrocyte-to-Neuron Conversion. Mol. Ther. 2020, 28, 217–234. [Google Scholar] [CrossRef]
- Ansah, E.O. Ethical Challenges and Controversies in the Practice and Advancement of Gene Therapy. Adv. Cell Gene Ther. 2022, 2022, 1015996. [Google Scholar] [CrossRef]
- Lewis, A.C.F.; Green, R.C. Polygenic risk scores in the clinic: New perspectives needed on familiar ethical issues. Genome Med. 2021, 13, 14. [Google Scholar] [CrossRef]
- Dergunova, L.V.; Filippenkov, I.B.; Limborska, S.A.; Myasoedov, N.F. Neuroprotective Peptides and New Strategies for Ischemic Stroke Drug Discoveries. Genes 2023, 14, 953. [Google Scholar] [CrossRef]
- Nozohouri, S.; Sifat, A.E.; Vaidya, B.; Abbruscato, T.J. Novel approaches for the delivery of therapeutics in ischemic stroke. Drug Discov. Today 2020, 25, 535–551. [Google Scholar] [CrossRef] [PubMed]
- Akinyemi, R.O.; Ovbiagele, B.; Adeniji, O.A.; Sarfo, F.S.; Abd-Allah, F.; Adoukonou, T.; Ogah, O.S.; Naidoo, P.; Damasceno, A.; Walker, R.W.; et al. Stroke in Africa: Profile, progress, prospects and priorities. Nat. Rev. Neurol. 2021, 17, 634–656. [Google Scholar] [CrossRef] [PubMed]
- Huang, Q.; Lan, X.; Chen, H.; Li, H.; Sun, Y.; Ren, C.; Xing, C.; Bo, X.; Wang, J.; Jin, X.; et al. Association between genetic predisposition and disease burden of stroke in China: A genetic epidemiological study. Lancet Reg. Health West. Pac. 2023, 36, 100779. [Google Scholar] [CrossRef] [PubMed]
- The International Stroke Genetics Consortium (ISGC). Available online: https://www.strokegenetics.org/ (accessed on 15 November 2023).
- The MEGASTROKE Consortium. Available online: https://www.megastroke.org/ (accessed on 15 November 2023).
- Maguire, J.M.; Bevan, S.; Stanne, T.M.; Lorenzen, E.; Fernandez-Cadenas, I.; Hankey, G.J.; Jimenez-Conde, J.; Jood, K.; Lee, J.M.; Lemmens, R.; et al. GISCOME—Genetics of Ischaemic Stroke Functional Outcome network: A protocol for an international multicentre genetic association study. Eur. Stroke J. 2017, 2, 229–237. [Google Scholar] [CrossRef]
Mechanism of Ischemic Stroke | Disease | Related Genes | Inheritance Pattern | Key Clinical Features |
---|---|---|---|---|
Small vessel disease | CADASIL | NOTCH3 | AD | Migraine, recurrent transient ischemic attacks and lacunar infarcts, vascular cognitive impairment, anterior temporal lobe white matter hyperintensities |
CARASIL | HTRA1 | AR | Similar to CADASIL, but of autosomal recessive inheritance pattern | |
Cathepsin A-related arteriopathy with strokes and leukoencephalopathy (CARASAL) | CTSA | AD | Migraine, transient ischemic attack, recurrent strokes, vertigo, dysphagia, cognitive impairment, REM-sleep behavioral disorder | |
Retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations (RVCL-S) | TREX1 | AD | Visual loss, vascular retinopathy, migraine, cognitive impairment microvascular renal disease, Raynaud phenomenon | |
Fabry disease | GAL | X-linked recessive | Transient ischemic attack, stroke. May also be associated with other stroke phenotypes including large vessel disease Acroparasthesias, corneal and lenticular opacities, cardiomyopathy | |
FOXC1-deletion related cerebral small vessel disease | FOXC1 | AD | Hearing impairment, cerebellar malformations, white matter hyperintensities on MRI Brain | |
Type IV collagenopathy | COL4A1 and A2 | AD | Recurrent strokes, seizures, migraine, visual loss, nephropathy, myopathy, arrhythmias, intracranial aneurysms or dolichoectasia | |
Pontine autosomal-dominant microangiopathy with leukoencephalopathy (PADMAL) | COL4A1 | AD | Recurrent lacunar infarcts with predilection to the pons, progressive cognitive impairment | |
Large vessel disease | Ehlers–Danlos Syndrome Type IV | COL3A1 | AD | Facial acrogeria, skin fragility and bruising, large and medium vessel artery dissections, including extracranial and intracranial vertebral and carotid arteries |
Pseudoxanthoma elasticum | ABCC6 | AR | Increased skin elasticity, skin discoloration, ocular angioid streaks | |
Marfan syndrome | FBN1 | AD | Marfanoid features, ascending aorta dissection | |
Moyamoya disease | RNF213 | AD or AR | Steno-occlusive disease of the terminal internal carotid artery with “puff of smoke” collaterals | |
Haematological disorders | Sickle cell disease | HBB | AR | Pain crises, seizures, myelopathy, anemia, thrombosis Can be associated with both large vessel and small vessel infarcts |
Polycythemia rubra vera (PV) | JAK2 | AD or AR | Ischemic stroke due to hyperviscosity state | |
Essential thrombocythemia (ET) | JAK2 MPL | AD or AR | Ischemic stroke due to hyperviscosity state | |
Homocystinuria Hyperhomocysteinemia | CBS italic>MTHFR | AR | Raised plasma homocysteine level, atherosclerosis, thrombosis Classic homocystinuria: ectopia lentis, tall stature, pectus excavatum, developmental delay, thromboembolism | |
Hereditary thrombophilias | Factor V Leiden mutation (FVL) | F5 | AD | Can be associated with small vessel, large vessel, and embolic infarcts |
Prothrombin gene mutation (Factor II mutation) | F2 | AD | ||
Protein C deficiency | PROC | AD or AR | ||
Protein S deficiency | PROS1 | AD or AR | ||
Antithrombin III deficiency | SERPINC1 | AD | ||
Paroxysmal nocturnal hemoglobinuria (PNH) | PIGA | X linked | Complement-mediated hemolysis. Can be associated with small vessel, large vessel and embolic infarcts |
Variant | Gene | Remarks |
---|---|---|
m.3243A>G m.3271T>C m.3252A>G | MT-TL1 | Encodes the transfer RNA for leucine. This mutation results in the disruption of the function of mitochondrial tRNA, affecting protein synthesis within the mitochondria. |
m.13513G>A | MT-ND5 | A part of complex I of the mitochondrial respiratory chain. |
m.8344A>G | MT-TK | Encodes the transfer RNA for lysine. This mutation is more commonly associated with another mitochondrial disorder known as MERRF (Myoclonic Epilepsy with Ragged Red Fibers), but it can sometimes cause MELAS or a MELAS/MERRF overlap syndrome. |
m.14453G>A | MT-ND6 | Associated with MELAS phenotype with dystonia. |
m.13042G>A | MT-ND5 | Associated with MELAS syndrome with a cardiomyopathy phenotype. |
m.8993T>G/C | MT-ATP6 | Associated with NARP (Neuropathy, Ataxia, and Retinitis Pigmentosa) syndrome, but can sometimes produce MELAS symptoms. |
Drug | Related Genes | |
---|---|---|
Recombinant tissue plasminogen activator (rtPA) or Alteplase | Increased recanalization: PAI-1, TAFI, IL1B, vWF, ACE Increased hemorrhagic conversion: PAI-1, MMP9, FXIII and FXII, A2M, ZBTB46, ACE | |
Aspirin | COX-1, COX-2, P1A1/A2COL1A1, COL1A2, vWF, ITGA2B, UGTIA6*2, ADRA2A, TXBA2R, PLA2G7 | |
Clopidogrel | Hepatic metabolism: CYP3A4, CYP1A2, CYP2C19 Intestinal absorption: ABCB1 Glycoprotein: ABCB1 Platelet surface receptors: P2Y1, P2Y12 | |
Ticagrelor | SLCO1B1, UGT2B7, CYP3A4 | |
Warfarin | VKORC1, and CYP2C9 | |
DOAC | Dabigatran | Activation: CES1, CES2 Transport: ABCB1 Metabolism: UGT1A9, UGT2B7, UGT2B15 |
Rivaroxaban | Transport: ABCB1, ABCG2 Metabolism: CYP3A4/5,CYP2J2 | |
Apixaban | Transport: ABCB1, ABCG2 Metabolism: CYP3A4/5,CYP2J2,CYP1A2 | |
Edoxaban | Transport: ABCB1, SLCO1B1 Metabolism: CES1, CYP3A4/5 | |
Betrixaban | Transport: ABCB1 Metabolism: CYP450-independent hydrolysis |
Trial Name | Author and Publication Year | Study Type | Number of Subjects | Ethnicity | Genes Found |
---|---|---|---|---|---|
Post Stroke Motor Recovery GWAS: A Domain-Specific Approach [110] | Chad M Aldridge et al., 2023 | GWAS | 2100 cases | Not mentioned | No genome-wide significant loci found. 115 SNPs’ subthreshold associations were identified. CLDN23 gene had the most convincing association, which affects blood–brain barrier integrity, neurodevelopment, and immune cell transmigration |
Six GWAS-linked hot loci on stroke outcome [109] | Ruixia Zhu et al., 2021 | GWAS | 982 cases | Northern Chinese | ALDH2 rs10744777, HDAC9 rs2107595, ABO rs532436 (associated with increased stroke recurrence), PATJ rs76221407, LOC105372028 rs1842681, PTCH1 rs2236406 (associated with poor stroke outcome) |
GWAS of functional outcome (GISCOME) [108] | Martin Söderholm et al., 2019 | Meta-analysis | 6165 cases | Europe, US, and Australia | LOC105372028 rs1842681 was associated with brain plasticity and good stroke outcome |
PATJ Variants Are Associated with Worse Ischemic Stroke Functional Outcome [107] | Marina Mola-Caminal et al., 2019 | Meta-analysis | >5000 cases | European ancestry | PATJ (Pals1-associated tight junction) gene (associated with worse functional outcome at 3 months after stroke) |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Zhang, K.; Loong, S.S.E.; Yuen, L.Z.H.; Venketasubramanian, N.; Chin, H.-L.; Lai, P.S.; Tan, B.Y.Q. Genetics in Ischemic Stroke: Current Perspectives and Future Directions. J. Cardiovasc. Dev. Dis. 2023, 10, 495. https://doi.org/10.3390/jcdd10120495
Zhang K, Loong SSE, Yuen LZH, Venketasubramanian N, Chin H-L, Lai PS, Tan BYQ. Genetics in Ischemic Stroke: Current Perspectives and Future Directions. Journal of Cardiovascular Development and Disease. 2023; 10(12):495. https://doi.org/10.3390/jcdd10120495
Chicago/Turabian StyleZhang, Ka, Shaun S. E. Loong, Linus Z. H. Yuen, Narayanaswamy Venketasubramanian, Hui-Lin Chin, Poh San Lai, and Benjamin Y. Q. Tan. 2023. "Genetics in Ischemic Stroke: Current Perspectives and Future Directions" Journal of Cardiovascular Development and Disease 10, no. 12: 495. https://doi.org/10.3390/jcdd10120495
APA StyleZhang, K., Loong, S. S. E., Yuen, L. Z. H., Venketasubramanian, N., Chin, H. -L., Lai, P. S., & Tan, B. Y. Q. (2023). Genetics in Ischemic Stroke: Current Perspectives and Future Directions. Journal of Cardiovascular Development and Disease, 10(12), 495. https://doi.org/10.3390/jcdd10120495