1
|
Lindgren AG. Genomics of stroke recovery and outcome. J Cereb Blood Flow Metab 2025:271678X251332528. [PMID: 40215404 PMCID: PMC11993556 DOI: 10.1177/0271678x251332528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/10/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
The understanding of genomics has improved tremendously during the last decades. The concept of recovery and outcome after stroke has also progressed during this time. However, the connection between genomics and stroke recovery has only begun to emerge in a more structural and comprehensive way. Different types of outcomes and recovery occur after stroke. This depends on domain of neurological deficit, severity, resilience, receptivity for rehabilitation measures, and concomitant morbidity. Methods for assessing stroke patients' prognosis depend on these factors. Different genetic approaches are possible and there is an increasing need for linking genetic findings to other omics as well as to clinically meaningful results. This review addresses recent advances and views in clinical genomics of stroke recovery and outcome in humans with focus on current and previous studies, concepts, and future perspectives.
Collapse
Affiliation(s)
- Arne G Lindgren
- Department of Neurology, Skåne University Hospital; Department of Clinical Sciences Lund, Neurology, Lund University, Lund, Sweden
| |
Collapse
|
2
|
Awuah WA, Shah MH, Sanker V, Mannan KM, Ranganathan S, Nkrumah-Boateng PA, Frimpong M, Darko K, Tan JK, Abdul-Rahman T, Atallah O. Advances in chromosomal microarray analysis: Transforming neurology and neurosurgery. BRAIN & SPINE 2025; 5:104197. [PMID: 39990116 PMCID: PMC11847126 DOI: 10.1016/j.bas.2025.104197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/25/2025]
Abstract
Over the past two decades, genomics has transformed our understanding of various clinical conditions, with Chromosomal Microarray Analysis (CMA) standing out as a key technique. Offering unparalleled sensitivity, CMA detects submicroscopic chromosomal imbalances, enabling the examination of DNA for copy number variations, deletions, duplications, and other structural differences. In neurology, CMA has revolutionised diagnoses, personalised treatment plans, and patient outcomes. By identifying genetic anomalies linked to neurological conditions, CMA allows clinicians to tailor treatments based on individual genetic profiles, enhancing precision medicine. CMA's clinical utility spans numerous neurological conditions, providing crucial insights into neurodevelopmental disorders, CNS tumours, neurodegenerative diseases, cerebrovascular diseases, and epilepsy. In neurodevelopmental disorders, CMA aids in diagnosing autism and intellectual disabilities, facilitating early interventions that improve long-term outcomes. In epilepsy, CMA helps identify genetic causes of drug-resistant seizures, enabling more targeted therapies and reducing adverse reactions. CMA also aids in stratifying risk for cerebrovascular diseases, enabling preventive interventions that improve patient prognosis. Despite its potential, challenges remain, such as interpreting variants of uncertain significance (VOUS), the lack of standardised testing guidelines, and issues of cost and accessibility. Addressing these challenges will optimise CMA's impact, advancing personalised medicine and reshaping neurology. This review discusses CMA's pivotal role in bridging the gap between genomics and clinical practice, underscoring its potential to transform neurogenetics and ultimately improve patient care.
Collapse
Affiliation(s)
| | | | - Vivek Sanker
- Department of Neurosurgery, Trivandrum Medical College, India
- Department of Neurosurgery, Stanford University, CA, USA
| | | | - Sruthi Ranganathan
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | - Mabel Frimpong
- Bryn Mawr College, 101 N Merion Avenue, Bryn Mawr, PA, USA
| | - Kwadwo Darko
- Department of Neurosurgery, Korle Bu Teaching Hospital, Accra, Ghana
| | - Joecelyn Kirani Tan
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, United Kingdom
| | | | - Oday Atallah
- Department of Neurosurgery, Carl Von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
3
|
Zhang L, Chu Y, You Y, Dong M, Pang X, Chen L, Zhu L, Yang S, Zhou L, Shang K, Deng G, Xiao J, Wang W, Qin C, Tian D. Systematic Druggable Genome-Wide Mendelian Randomization Identifies Therapeutic Targets for Functional Outcome After Ischemic Stroke. J Am Heart Assoc 2024; 13:e034749. [PMID: 39119979 PMCID: PMC11963950 DOI: 10.1161/jaha.124.034749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Stroke is a leading cause of death worldwide, with a lack of effective treatments for improving the prognosis. The aim of the present study was to identify novel therapeutic targets for functional outcome after ischemic stroke . METHODS AND RESULTS Cis-expression quantitative trait loci data for druggable genes were used as instrumental variables. The primary outcome was the modified Rankin Scale score at 3 months after ischemic stroke, evaluated as a dichotomous variable (3-6 versus 0-2) and also as an ordinal variable. Drug target Mendelian randomization, Steiger filtering analysis, and colocalization analysis were performed. Additionally, phenome-wide Mendelian randomization analysis was performed to identify the safety of the drug target genes at the genetic level. Among >2600 druggable genes, genetically predicted expression of 16 genes (ABCC2, ATRAID, BLK, CD93, CHST13, NR1H3, NRBP1, PI3, RIPK4, SEMG1, SLC22A4, SLC22A5, SLCO3A1, TEK, TLR4, and WNT10B) demonstrated the causal associations with ordinal modified Rankin Scale (P<1.892×10-5) or poor functional outcome (modified Rankin Scale 3-6 versus 0-2, P<1.893×10-5). Steiger filtering analysis suggested potential directional stability (P<0.05). Colocalization analysis provided further support for the associations between genetically predicted expression of ABCC2, NRBP1, PI3, and SEMG1 with functional outcome after ischemic stroke. Furthermore, phenome-wide Mendelian randomization revealed additional beneficial indications and few potential safety concerns of therapeutics targeting ABCC2, NRBP1, PI3, and SEMG1, but the robustness of these results was limited by low power. CONCLUSIONS The present study revealed 4 candidate therapeutic targets for improving functional outcome after ischemic stroke, while the underlying mechanisms need further investigation.
Collapse
Affiliation(s)
- Lu‐Yang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Neural Injury and Functional ReconstructionHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yun‐Hui Chu
- Department of Neurology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Neural Injury and Functional ReconstructionHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yun‐Fan You
- Department of Neurology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Neural Injury and Functional ReconstructionHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ming‐Hao Dong
- Department of Neurology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Neural Injury and Functional ReconstructionHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiao‐Wei Pang
- Department of Neurology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Neural Injury and Functional ReconstructionHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Lian Chen
- Department of Neurology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Neural Injury and Functional ReconstructionHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Li‐Fang Zhu
- Department of Neurology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Neural Injury and Functional ReconstructionHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Neural Injury and Functional ReconstructionHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Luo‐Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Neural Injury and Functional ReconstructionHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ke Shang
- Department of Neurology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Neural Injury and Functional ReconstructionHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Gang Deng
- Department of Neurology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Neural Injury and Functional ReconstructionHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jun Xiao
- Department of Neurology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Neural Injury and Functional ReconstructionHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Neural Injury and Functional ReconstructionHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Neural Injury and Functional ReconstructionHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Dai‐Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Neural Injury and Functional ReconstructionHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
4
|
Debette S, Paré G. Stroke Genetics, Genomics, and Precision Medicine. Stroke 2024; 55:2163-2168. [PMID: 38511336 DOI: 10.1161/strokeaha.123.044212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Affiliation(s)
- Stéphanie Debette
- University of Bordeaux, INSERM, Bordeaux Population Health, France (S.D.)
- Department of Neurology, Institute for Neurodegenerative Diseases, Bordeaux University Hospital, France (S.D.)
| | - Guillaume Paré
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada (G.P.)
| |
Collapse
|
5
|
Zhang S, Zhu D, Wu Z, Yang S, Liu Y, Kang X, Chen X, Zhu Z, Dong Q, Suo C, Han X. GWAS-based polygenic risk scoring for predicting cerebral artery dissection in the Chinese population. BMC Neurol 2024; 24:258. [PMID: 39054468 PMCID: PMC11271197 DOI: 10.1186/s12883-024-03759-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
OBJECTIVE Cerebral artery dissection (CeAD) is a rare but serious disease. Genetic risk assessment for CeAD is lacking in Chinese population. We performed genome-wide association study (GWAS) and computed polygenic risk score (PRS) to explore genetic susceptibility factors and prediction model of CeAD based on patients in Huashan Hospital. METHODS A total of 210 CeAD patients and 280 controls were enrolled from June 2017 to September 2022 in Department of Neurology, Huashan Hospital, Fudan University. We performed GWAS to identify genetic variants associated with CeAD in 140 CeAD patients and 210 control individuals according to a case and control 1:1.5 design rule in the training dataset, while the other 70 patients with CeAD and 70 controls were used as validation. Then Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and Gene Ontology (GO) enrichment analyses were utilized to identify the significant pathways. We constructed a PRS by capturing all independent GWAS SNPs in the analysis and explored the predictivity of PRS, age, and sex for CeAD. RESULTS Through GWAS analysis of the 140 cases and 210 controls in the training dataset, we identified 13 leading SNPs associated with CeAD at a genome-wide significance level of P < 5 × 10- 8. Among them, 10 SNPs were annotated in or near (in the upstream and downstream regions of ± 500Kb) 10 functional genes. rs34508376 (OR2L13) played a suggestive role in CeAD pathophysiology which was in line with previous observations in aortic aneurysms. The other nine genes were first-time associations in CeAD cases. GO enrichment analyses showed that these 10 genes have known roles in 20 important GO terms clustered into two groups: (1) cellular biological processes (BP); (2) molecular function (MF). We used genome-wide association data to compute PRS including 32 independent SNPs and constructed predictive model for CeAD by using age, sex and PRS as predictors both in training and validation test. The area under curve (AUC) of PRS predictive model for CeAD reached 99% and 95% in the training test and validation test respectively, which were significantly larger than the age and sex models of 83% and 86%. CONCLUSIONS Our study showed that ten risk loci were associated with CeAD susceptibility, and annotated functional genes had roles in 20 important GO terms clustered into biological process and molecular function. The PRS derived from risk variants was associated with CeAD incidence after adjusting for age and sex both in training test and validation.
Collapse
Grants
- No. 8227052180 National Natural Science Foundation of China
- No. 8227052180 National Natural Science Foundation of China
- No. 8227052180 National Natural Science Foundation of China
- No. 8227052180 National Natural Science Foundation of China
- No. 8227052180 National Natural Science Foundation of China
- No. 8227052180 National Natural Science Foundation of China
- No. 8227052180 National Natural Science Foundation of China
- No. 8227052180 National Natural Science Foundation of China
- No. 8227052180 National Natural Science Foundation of China
- No. 8227052180 National Natural Science Foundation of China
- No. 8227052180 National Natural Science Foundation of China
- none State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan university.
- none State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan university.
- none State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan university.
- none State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan university.
- none State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan university.
- none State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan university.
- none State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan university.
- none State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan university.
- none State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan university.
- none State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan university.
- none State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan university.
- none The Cerebrovascular Disease Management Project of Sailing Foundation of China Stroke Association.
- none The Cerebrovascular Disease Management Project of Sailing Foundation of China Stroke Association.
- none The Cerebrovascular Disease Management Project of Sailing Foundation of China Stroke Association.
- none The Cerebrovascular Disease Management Project of Sailing Foundation of China Stroke Association.
- none The Cerebrovascular Disease Management Project of Sailing Foundation of China Stroke Association.
- none The Cerebrovascular Disease Management Project of Sailing Foundation of China Stroke Association.
- none The Cerebrovascular Disease Management Project of Sailing Foundation of China Stroke Association.
- none The Cerebrovascular Disease Management Project of Sailing Foundation of China Stroke Association.
- none The Cerebrovascular Disease Management Project of Sailing Foundation of China Stroke Association.
- none The Cerebrovascular Disease Management Project of Sailing Foundation of China Stroke Association.
- none The Cerebrovascular Disease Management Project of Sailing Foundation of China Stroke Association.
- NO. HIGHER2022107 Heart and Brain Health Public Welfare Project of Buchang Zhiyuan Foundation
- NO. HIGHER2022107 Heart and Brain Health Public Welfare Project of Buchang Zhiyuan Foundation
- NO. HIGHER2022107 Heart and Brain Health Public Welfare Project of Buchang Zhiyuan Foundation
- NO. HIGHER2022107 Heart and Brain Health Public Welfare Project of Buchang Zhiyuan Foundation
- NO. HIGHER2022107 Heart and Brain Health Public Welfare Project of Buchang Zhiyuan Foundation
- NO. HIGHER2022107 Heart and Brain Health Public Welfare Project of Buchang Zhiyuan Foundation
- NO. HIGHER2022107 Heart and Brain Health Public Welfare Project of Buchang Zhiyuan Foundation
- NO. HIGHER2022107 Heart and Brain Health Public Welfare Project of Buchang Zhiyuan Foundation
- NO. HIGHER2022107 Heart and Brain Health Public Welfare Project of Buchang Zhiyuan Foundation
- NO. HIGHER2022107 Heart and Brain Health Public Welfare Project of Buchang Zhiyuan Foundation
- NO. HIGHER2022107 Heart and Brain Health Public Welfare Project of Buchang Zhiyuan Foundation
Collapse
Affiliation(s)
- Shufan Zhang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Dongliang Zhu
- State Key Laboratory of Genetic Engineering, School of life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Zhengyu Wu
- Department of Geriatrics, Huashan Hospital, Fudan University, Shanghai, China
| | - Shilin Yang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuanzeng Liu
- Gu Mei Community Health Service Center of Minhang District, Shanghai, China
| | - Xiaocui Kang
- Department of Neurology, Shanghai Shidong Hospital, Shanghai, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering, School of life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Zhu Zhu
- Department of Neurology, Indianan University Health, Bloomington, IN, USA
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Chen Suo
- State Key Laboratory of Genetic Engineering, School of life Sciences, Human Phenome Institute, Fudan University, Shanghai, China.
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China.
| | - Xiang Han
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Yang J, Lin X, Wang A, Meng X, Zhao X, Jing J, Zhang Y, Li H, Wang Y. Derivation and Validation of a Scoring System for Predicting Poor Outcome After Posterior Circulation Ischemic Stroke in China. Neurology 2024; 102:e209312. [PMID: 38759139 DOI: 10.1212/wnl.0000000000209312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Guidelines for posterior circulation ischemic stroke (PCIS) treatment are lacking and outcome prediction is crucial for patients and clinicians. We aimed to develop and validate a prognostic score to predict the poor outcome for patients with PCIS. METHODS The score was developed from a prospective derivation cohort named the Third China National Stroke Registry (August 2015-March 2018) and validated in a spatiotemporal independent validation cohort (December 2017-March 2023) in China. Patients with PCIS with acute infarctions defined as hyperintense lesions on diffusion-weighted imaging were included in this study. The poor outcome was measured as modified Rankin scale (mRS) score 3-6 at 3 months after PCIS. Multivariable logistic regression analysis was used to identify predictors for poor outcome. The prognostic score, namely PCIS Outcome Score (PCISOS), was developed by assigning points to variables based on their relative β-coefficients in the logistic model. RESULTS The PCISOS was derived from 3,294 patients (median age 62 [interquartile range (IQR) 55-70] years; 2,250 [68.3%] men) and validated in 501 patients (median age 61 [IQR 53-68] years; 404 [80.6%] men). Among them, 384 (11.7%) and 64 (12.8%) had poor outcome 3 months after stroke in respective cohorts. Age, mRS before admission, NIH Stroke Scale on admission, ischemic stroke history, infarction distribution, basilar artery, and posterior cerebral artery stenosis or occlusion were identified as independent predictors for poor outcome and included in PCISOS. This easy-to-use integer scoring system identified a marked risk gradient between 4 risk groups. PCISOS performed better than previous scores, with an excellent discrimination (C statistic) of 0.80 (95% CI 0.77-0.83) in the derivation cohort and 0.81 (95% CI 0.77-0.84) in the validation cohort. Calibration test showed high agreement between the predicted and observed outcomes in both cohorts. DISCUSSION PCISOS can be applied for patients with PCIS with acute infarctions to predict functional outcome at 3 months post-PCIS. This simple tool helps clinicians to identify patients with PCIS with higher risk of poor outcome and provides reliable outcome expectations for patients. This information might be used for personalized rehabilitation plan and patient selection for future clinical trials to reduce disability and mortality.
Collapse
Affiliation(s)
- Jialei Yang
- From the Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University
| | - Xiaoyu Lin
- From the Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University
| | - Anxin Wang
- From the Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University
| | - Xia Meng
- From the Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University
| | - Xingquan Zhao
- From the Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University
| | - Jing Jing
- From the Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University
| | - Yijun Zhang
- From the Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University
| | - Hao Li
- From the Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University
| | - Yongjun Wang
- From the Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University
| |
Collapse
|
7
|
Carnwath TP, Demel SL, Prestigiacomo CJ. Genetics of ischemic stroke functional outcome. J Neurol 2024; 271:2345-2369. [PMID: 38502340 PMCID: PMC11055934 DOI: 10.1007/s00415-024-12263-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 03/21/2024]
Abstract
Ischemic stroke, which accounts for 87% of cerebrovascular accidents, is responsible for massive global burden both in terms of economic cost and personal hardship. Many stroke survivors face long-term disability-a phenotype associated with an increasing number of genetic variants. While clinical variables such as stroke severity greatly impact recovery, genetic polymorphisms linked to functional outcome may offer physicians a unique opportunity to deliver personalized care based on their patient's genetic makeup, leading to improved outcomes. A comprehensive catalogue of the variants at play is required for such an approach. In this review, we compile and describe the polymorphisms associated with outcome scores such as modified Rankin Scale and Barthel Index. Our search identified 74 known genetic polymorphisms spread across 48 features associated with various poststroke disability metrics. The known variants span diverse biological systems and are related to inflammation, vascular homeostasis, growth factors, metabolism, the p53 regulatory pathway, and mitochondrial variation. Understanding how these variants influence functional outcome may be helpful in maximizing poststroke recovery.
Collapse
Affiliation(s)
- Troy P Carnwath
- University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| | - Stacie L Demel
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Charles J Prestigiacomo
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| |
Collapse
|
8
|
Phillips CM, Stamatovic SM, Keep RF, Andjelkovic AV. Epigenetics and stroke: role of DNA methylation and effect of aging on blood-brain barrier recovery. Fluids Barriers CNS 2023; 20:14. [PMID: 36855111 PMCID: PMC9972738 DOI: 10.1186/s12987-023-00414-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/10/2023] [Indexed: 03/02/2023] Open
Abstract
Incomplete recovery of blood-brain barrier (BBB) function contributes to stroke outcomes. How the BBB recovers after stroke remains largely unknown. Emerging evidence suggests that epigenetic factors play a significant role in regulating post-stroke BBB recovery. This study aimed to evaluate the epigenetic and transcriptional profile of cerebral microvessels after thromboembolic (TE) stroke to define potential causes of limited BBB recovery. RNA-sequencing and reduced representation bisulfite sequencing (RRBS) analyses were performed using microvessels isolated from young (6 months) and old (18 months) mice seven days poststroke compared to age-matched sham controls. DNA methylation profiling of poststroke brain microvessels revealed 11,287 differentially methylated regions (DMR) in old and 9818 DMR in young mice, corresponding to annotated genes. These DMR were enriched in genes encoding cell structural proteins (e.g., cell junction, and cell polarity, actin cytoskeleton, extracellular matrix), transporters and channels (e.g., potassium transmembrane transporter, organic anion and inorganic cation transporters, calcium ion transport), and proteins involved in endothelial cell processes (e.g., angiogenesis/vasculogenesis, cell signaling and transcription regulation). Integrated analysis of methylation and RNA sequencing identified changes in cell junctions (occludin), actin remodeling (ezrin) as well as signaling pathways like Rho GTPase (RhoA and Cdc42ep4). Aging as a hub of aberrant methylation affected BBB recovery processes by profound alterations (hypermethylation and repression) in structural protein expression (e.g., claudin-5) as well as activation of a set of genes involved in endothelial to mesenchymal transformation (e.g., Sox9, Snai1), repression of angiogenesis and epigenetic regulation. These findings revealed that DNA methylation plays an important role in regulating BBB repair after stroke, through regulating processes associated with BBB restoration and prevalently with processes enhancing BBB injury.
Collapse
Affiliation(s)
- Chelsea M Phillips
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Svetlana M Stamatovic
- Department of Pathology, Medical School, University of Michigan, 7520A MSRB I, 1150 W Medical Center Dr, Ann Arbor, MI, 48109-5602, USA
| | - Richard F Keep
- Department of Neurosurgery, Medical School, University of Michigan, 7520A MSRB I, 1150 W Medical Center Dr, Ann Arbor, MI, 48109-5602, USA.,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Anuska V Andjelkovic
- Department of Pathology, Medical School, University of Michigan, 7520A MSRB I, 1150 W Medical Center Dr, Ann Arbor, MI, 48109-5602, USA. .,Department of Neurosurgery, Medical School, University of Michigan, 7520A MSRB I, 1150 W Medical Center Dr, Ann Arbor, MI, 48109-5602, USA.
| |
Collapse
|
9
|
Phillips C, Stamatovic S, Keep R, Andjelkovic A. Epigenetics and stroke: role of DNA methylation and effect of aging on blood-brain barrier recovery. RESEARCH SQUARE 2023:rs.3.rs-2444060. [PMID: 36711725 PMCID: PMC9882686 DOI: 10.21203/rs.3.rs-2444060/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Incomplete recovery of blood-brain barrier (BBB) function contributes to stroke outcomes. How the BBB recovers after stroke remains largely unknown. Emerging evidence suggests that epigenetic factors play a significant role in regulating post-stroke BBB recovery. This study aimed to evaluate the epigenetic and transcriptional profile of cerebral microvessels after thromboembolic (TE) stroke to define potential causes of limited BBB recovery. RNA-sequencing and reduced representation bisulfite sequencing (RRBS) analyses were performed using microvessels isolated from young (6 months) and old (18 months) mice seven days poststroke compared to age-matched sham controls. DNA methylation profiling of poststroke brain microvessels revealed 11287 differentially methylated regions (DMR) in old and 9818 DMR in young mice, corresponding to annotated genes. These DMR were enriched in genes encoding cell structural proteins (e.g., cell junction, and cell polarity, actin cytoskeleton, extracellular matrix), transporters and channels (e.g., potassium transmembrane transporter, organic anion and inorganic cation transporters, calcium ion transport), and proteins involved in endothelial cell processes (e.g., angiogenesis/vasculogenesis, cell signaling and transcription regulation). Integrated analysis of methylation and RNA sequencing identified changes in cell junctions (occludin), actin remodeling (ezrin) as well as signaling pathways like Rho GTPase (RhoA and Cdc42ep4). Aging as a hub of aberrant methylation affected BBB recovery processes by profound alterations (hypermethylation and repression) in structural protein expression (e.g., claudin-5) as well as activation of a set of genes involved in endothelial to mesenchymal transformation (e.g., Sox17 , Snail1 ), repression of angiogenesis and epigenetic regulation. These findings revealed that DNA methylation plays an important role in regulating BBB repair after stroke, through regulating processes associated with BBB restoration and prevalently with processes enhancing BBB injury.
Collapse
|
10
|
Guo Y, Yang Y, Cao F, Li W, Wang M, Luo Y, Guo J, Zaman A, Zeng X, Miu X, Li L, Qiu W, Kang Y. Novel Survival Features Generated by Clinical Text Information and Radiomics Features May Improve the Prediction of Ischemic Stroke Outcome. Diagnostics (Basel) 2022; 12:1664. [PMID: 35885568 PMCID: PMC9324145 DOI: 10.3390/diagnostics12071664] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/17/2022] [Accepted: 07/05/2022] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Accurate outcome prediction is of great clinical significance in customizing personalized treatment plans, reducing the situation of poor recovery, and objectively and accurately evaluating the treatment effect. This study intended to evaluate the performance of clinical text information (CTI), radiomics features, and survival features (SurvF) for predicting functional outcomes of patients with ischemic stroke. METHODS SurvF was constructed based on CTI and mRS radiomics features (mRSRF) to improve the prediction of the functional outcome in 3 months (90-day mRS). Ten machine learning models predicted functional outcomes in three situations (2-category, 4-category, and 7-category) using seven feature groups constructed by CTI, mRSRF, and SurvF. RESULTS For 2-category, ALL (CTI + mRSRF+ SurvF) performed best, with an mAUC of 0.884, mAcc of 0.864, mPre of 0.877, mF1 of 0.86, and mRecall of 0.864. For 4-category, ALL also achieved the best mAuc of 0.787, while CTI + SurvF achieved the best score with mAcc = 0.611, mPre = 0.622, mF1 = 0.595, and mRe-call = 0.611. For 7-category, CTI + SurvF performed best, with an mAuc of 0.788, mPre of 0.519, mAcc of 0.529, mF1 of 0.495, and mRecall of 0.47. CONCLUSIONS The above results indicate that mRSRF + CTI can accurately predict functional outcomes in ischemic stroke patients with proper machine learning models. Moreover, combining SurvF will improve the prediction effect compared with the original features. However, limited by the small sample size, further validation on larger and more varied datasets is necessary.
Collapse
Affiliation(s)
- Yingwei Guo
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China; (Y.G.); (Y.Y.); (F.C.); (X.M.)
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, China; (W.L.); (A.Z.); (X.Z.); (L.L.); (W.Q.)
| | - Yingjian Yang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China; (Y.G.); (Y.Y.); (F.C.); (X.M.)
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, China; (W.L.); (A.Z.); (X.Z.); (L.L.); (W.Q.)
| | - Fengqiu Cao
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China; (Y.G.); (Y.Y.); (F.C.); (X.M.)
| | - Wei Li
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, China; (W.L.); (A.Z.); (X.Z.); (L.L.); (W.Q.)
| | - Mingming Wang
- Department of Radiology, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai 200434, China;
| | - Yu Luo
- Department of Radiology, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai 200434, China;
| | - Jia Guo
- Department of Psychiatry, Columbia University, New York, NY 10027, USA;
| | - Asim Zaman
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, China; (W.L.); (A.Z.); (X.Z.); (L.L.); (W.Q.)
- Engineering Research Centre of Medical Imaging and Intelligent Analysis, Ministry of Education, Shenyang 110169, China
| | - Xueqiang Zeng
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, China; (W.L.); (A.Z.); (X.Z.); (L.L.); (W.Q.)
- Engineering Research Centre of Medical Imaging and Intelligent Analysis, Ministry of Education, Shenyang 110169, China
| | - Xiaoqiang Miu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China; (Y.G.); (Y.Y.); (F.C.); (X.M.)
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, China; (W.L.); (A.Z.); (X.Z.); (L.L.); (W.Q.)
| | - Longyu Li
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, China; (W.L.); (A.Z.); (X.Z.); (L.L.); (W.Q.)
| | - Weiyan Qiu
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, China; (W.L.); (A.Z.); (X.Z.); (L.L.); (W.Q.)
| | - Yan Kang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China; (Y.G.); (Y.Y.); (F.C.); (X.M.)
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, China; (W.L.); (A.Z.); (X.Z.); (L.L.); (W.Q.)
- Engineering Research Centre of Medical Imaging and Intelligent Analysis, Ministry of Education, Shenyang 110169, China
- School of Applied Technology, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
11
|
Stroke Genomics: Current Knowledge, Clinical Applications and Future Possibilities. Brain Sci 2022; 12:brainsci12030302. [PMID: 35326259 PMCID: PMC8946102 DOI: 10.3390/brainsci12030302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/19/2022] [Accepted: 02/22/2022] [Indexed: 12/01/2022] Open
Abstract
The pathophysiology of stoke involves many complex pathways and risk factors. Though there are several ongoing studies on stroke, treatment options are limited, and the prevalence of stroke is continuing to increase. Understanding the genomic variants and biological pathways associated with stroke could offer novel therapeutic alternatives in terms of drug targets and receptor modulations for newer treatment methods. It is challenging to identify individual causative mutations in a single gene because many alleles are responsible for minor effects. Therefore, multiple factorial analyses using single nucleotide polymorphisms (SNPs) could be used to gain new insight by identifying potential genetic risk factors. There are many studies, such as Genome-Wide Association Studies (GWAS) and Phenome-Wide Association Studies (PheWAS) which have identified numerous independent loci associated with stroke, which could be instrumental in developing newer drug targets and novel therapies. Additionally, using analytical techniques, such as meta-analysis and Mendelian randomization could help in evaluating stroke risk factors and determining treatment priorities. Combining SNPs into polygenic risk scores and lifestyle risk factors could detect stroke risk at a very young age and help in administering preventive interventions.
Collapse
|
12
|
Johansson M, Pedersen A, Cole JW, Lagging C, Lindgren A, Maguire JM, Rost NS, Söderholm M, Worrall BB, Stanne TM, Jern C. Genetic Predisposition to Mosaic Chromosomal Loss Is Associated With Functional Outcome After Ischemic Stroke. NEUROLOGY-GENETICS 2021; 7:e634. [PMID: 34786478 PMCID: PMC8589264 DOI: 10.1212/nxg.0000000000000634] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 09/15/2021] [Indexed: 11/27/2022]
Abstract
Background and Objectives To test the hypothesis that a predisposition to acquired genetic alterations is associated with ischemic stroke outcome by investigating the association between a polygenic risk score (PRS) for mosaic loss of chromosome Y (mLOY) and outcome in a large international data set. Methods We used data from the genome-wide association study performed within the Genetics of Ischemic Stroke Functional Outcome network, which included 6,165 patients (3,497 men and 2,668 women) with acute ischemic stroke of mainly European ancestry. We assessed a weighted PRS for mLOY and examined possible associations with the modified Rankin Scale (mRS) score 3 months poststroke in logistic regression models. We investigated the whole study sample as well as men and women separately. Results Increasing PRS for mLOY was associated with poor functional outcome (mRS score >2) with an odds ratio (OR) of 1.11 (95% confidence interval [CI] 1.03–1.19) per 1 SD increase in the PRS after adjustment for age, sex, ancestry, stroke severity (NIH Stroke Scale), smoking, and diabetes mellitus. In sex-stratified analyses, we found a statistically significant association in women (adjusted OR 1.20, 95% CI 1.08–1.33). In men, the association was in the same direction (adjusted OR 1.04, 95% CI 0.95–1.14), and we observed no significant genotype-sex interaction. Discussion In this exploratory study, we found associations between genetic variants predisposing to mLOY and stroke outcome. The significant association in women suggests underlying mechanisms related to genomic instability that operate in both sexes. These findings need replication and mechanistic exploration.
Collapse
Affiliation(s)
- Malin Johansson
- Institute of Biomedicine (M.J., A.P., C.L., T.M.S., C.J.), Sahlgrenska Academy at the University of Gothenburg; Department of Clinical Genetics and Genomics (A.P., C.L., C.J.), Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Neurology (J.W.C.), Baltimore VA Medical Center and University of Maryland School of Medicine, Baltimore, MD; Department of Clinical Sciences Lund (A.L., M.S.), Neurology, Lund University; Department of Neurology (A.L., M.S.), Skåne University Hospital, Lund and Malmö, Sweden; Faculty of Health (J.M.M.), University of Technology Sydney, Australia; Hunter Medical Research Centre (J.M.M.), Newcastle, Australia; J. Philip Kistler Stroke Research Center (N.S.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; and Departments of Neurology and Health Evaluation Sciences (B.B.W.), University of Virginia, Charlottesville, VA
| | - Annie Pedersen
- Institute of Biomedicine (M.J., A.P., C.L., T.M.S., C.J.), Sahlgrenska Academy at the University of Gothenburg; Department of Clinical Genetics and Genomics (A.P., C.L., C.J.), Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Neurology (J.W.C.), Baltimore VA Medical Center and University of Maryland School of Medicine, Baltimore, MD; Department of Clinical Sciences Lund (A.L., M.S.), Neurology, Lund University; Department of Neurology (A.L., M.S.), Skåne University Hospital, Lund and Malmö, Sweden; Faculty of Health (J.M.M.), University of Technology Sydney, Australia; Hunter Medical Research Centre (J.M.M.), Newcastle, Australia; J. Philip Kistler Stroke Research Center (N.S.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; and Departments of Neurology and Health Evaluation Sciences (B.B.W.), University of Virginia, Charlottesville, VA
| | - John W Cole
- Institute of Biomedicine (M.J., A.P., C.L., T.M.S., C.J.), Sahlgrenska Academy at the University of Gothenburg; Department of Clinical Genetics and Genomics (A.P., C.L., C.J.), Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Neurology (J.W.C.), Baltimore VA Medical Center and University of Maryland School of Medicine, Baltimore, MD; Department of Clinical Sciences Lund (A.L., M.S.), Neurology, Lund University; Department of Neurology (A.L., M.S.), Skåne University Hospital, Lund and Malmö, Sweden; Faculty of Health (J.M.M.), University of Technology Sydney, Australia; Hunter Medical Research Centre (J.M.M.), Newcastle, Australia; J. Philip Kistler Stroke Research Center (N.S.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; and Departments of Neurology and Health Evaluation Sciences (B.B.W.), University of Virginia, Charlottesville, VA
| | - Cecilia Lagging
- Institute of Biomedicine (M.J., A.P., C.L., T.M.S., C.J.), Sahlgrenska Academy at the University of Gothenburg; Department of Clinical Genetics and Genomics (A.P., C.L., C.J.), Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Neurology (J.W.C.), Baltimore VA Medical Center and University of Maryland School of Medicine, Baltimore, MD; Department of Clinical Sciences Lund (A.L., M.S.), Neurology, Lund University; Department of Neurology (A.L., M.S.), Skåne University Hospital, Lund and Malmö, Sweden; Faculty of Health (J.M.M.), University of Technology Sydney, Australia; Hunter Medical Research Centre (J.M.M.), Newcastle, Australia; J. Philip Kistler Stroke Research Center (N.S.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; and Departments of Neurology and Health Evaluation Sciences (B.B.W.), University of Virginia, Charlottesville, VA
| | - Arne Lindgren
- Institute of Biomedicine (M.J., A.P., C.L., T.M.S., C.J.), Sahlgrenska Academy at the University of Gothenburg; Department of Clinical Genetics and Genomics (A.P., C.L., C.J.), Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Neurology (J.W.C.), Baltimore VA Medical Center and University of Maryland School of Medicine, Baltimore, MD; Department of Clinical Sciences Lund (A.L., M.S.), Neurology, Lund University; Department of Neurology (A.L., M.S.), Skåne University Hospital, Lund and Malmö, Sweden; Faculty of Health (J.M.M.), University of Technology Sydney, Australia; Hunter Medical Research Centre (J.M.M.), Newcastle, Australia; J. Philip Kistler Stroke Research Center (N.S.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; and Departments of Neurology and Health Evaluation Sciences (B.B.W.), University of Virginia, Charlottesville, VA
| | - Jane M Maguire
- Institute of Biomedicine (M.J., A.P., C.L., T.M.S., C.J.), Sahlgrenska Academy at the University of Gothenburg; Department of Clinical Genetics and Genomics (A.P., C.L., C.J.), Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Neurology (J.W.C.), Baltimore VA Medical Center and University of Maryland School of Medicine, Baltimore, MD; Department of Clinical Sciences Lund (A.L., M.S.), Neurology, Lund University; Department of Neurology (A.L., M.S.), Skåne University Hospital, Lund and Malmö, Sweden; Faculty of Health (J.M.M.), University of Technology Sydney, Australia; Hunter Medical Research Centre (J.M.M.), Newcastle, Australia; J. Philip Kistler Stroke Research Center (N.S.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; and Departments of Neurology and Health Evaluation Sciences (B.B.W.), University of Virginia, Charlottesville, VA
| | - Natalia S Rost
- Institute of Biomedicine (M.J., A.P., C.L., T.M.S., C.J.), Sahlgrenska Academy at the University of Gothenburg; Department of Clinical Genetics and Genomics (A.P., C.L., C.J.), Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Neurology (J.W.C.), Baltimore VA Medical Center and University of Maryland School of Medicine, Baltimore, MD; Department of Clinical Sciences Lund (A.L., M.S.), Neurology, Lund University; Department of Neurology (A.L., M.S.), Skåne University Hospital, Lund and Malmö, Sweden; Faculty of Health (J.M.M.), University of Technology Sydney, Australia; Hunter Medical Research Centre (J.M.M.), Newcastle, Australia; J. Philip Kistler Stroke Research Center (N.S.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; and Departments of Neurology and Health Evaluation Sciences (B.B.W.), University of Virginia, Charlottesville, VA
| | - Martin Söderholm
- Institute of Biomedicine (M.J., A.P., C.L., T.M.S., C.J.), Sahlgrenska Academy at the University of Gothenburg; Department of Clinical Genetics and Genomics (A.P., C.L., C.J.), Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Neurology (J.W.C.), Baltimore VA Medical Center and University of Maryland School of Medicine, Baltimore, MD; Department of Clinical Sciences Lund (A.L., M.S.), Neurology, Lund University; Department of Neurology (A.L., M.S.), Skåne University Hospital, Lund and Malmö, Sweden; Faculty of Health (J.M.M.), University of Technology Sydney, Australia; Hunter Medical Research Centre (J.M.M.), Newcastle, Australia; J. Philip Kistler Stroke Research Center (N.S.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; and Departments of Neurology and Health Evaluation Sciences (B.B.W.), University of Virginia, Charlottesville, VA
| | - Bradford B Worrall
- Institute of Biomedicine (M.J., A.P., C.L., T.M.S., C.J.), Sahlgrenska Academy at the University of Gothenburg; Department of Clinical Genetics and Genomics (A.P., C.L., C.J.), Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Neurology (J.W.C.), Baltimore VA Medical Center and University of Maryland School of Medicine, Baltimore, MD; Department of Clinical Sciences Lund (A.L., M.S.), Neurology, Lund University; Department of Neurology (A.L., M.S.), Skåne University Hospital, Lund and Malmö, Sweden; Faculty of Health (J.M.M.), University of Technology Sydney, Australia; Hunter Medical Research Centre (J.M.M.), Newcastle, Australia; J. Philip Kistler Stroke Research Center (N.S.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; and Departments of Neurology and Health Evaluation Sciences (B.B.W.), University of Virginia, Charlottesville, VA
| | - Tara M Stanne
- Institute of Biomedicine (M.J., A.P., C.L., T.M.S., C.J.), Sahlgrenska Academy at the University of Gothenburg; Department of Clinical Genetics and Genomics (A.P., C.L., C.J.), Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Neurology (J.W.C.), Baltimore VA Medical Center and University of Maryland School of Medicine, Baltimore, MD; Department of Clinical Sciences Lund (A.L., M.S.), Neurology, Lund University; Department of Neurology (A.L., M.S.), Skåne University Hospital, Lund and Malmö, Sweden; Faculty of Health (J.M.M.), University of Technology Sydney, Australia; Hunter Medical Research Centre (J.M.M.), Newcastle, Australia; J. Philip Kistler Stroke Research Center (N.S.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; and Departments of Neurology and Health Evaluation Sciences (B.B.W.), University of Virginia, Charlottesville, VA
| | - Christina Jern
- Institute of Biomedicine (M.J., A.P., C.L., T.M.S., C.J.), Sahlgrenska Academy at the University of Gothenburg; Department of Clinical Genetics and Genomics (A.P., C.L., C.J.), Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Neurology (J.W.C.), Baltimore VA Medical Center and University of Maryland School of Medicine, Baltimore, MD; Department of Clinical Sciences Lund (A.L., M.S.), Neurology, Lund University; Department of Neurology (A.L., M.S.), Skåne University Hospital, Lund and Malmö, Sweden; Faculty of Health (J.M.M.), University of Technology Sydney, Australia; Hunter Medical Research Centre (J.M.M.), Newcastle, Australia; J. Philip Kistler Stroke Research Center (N.S.R.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; and Departments of Neurology and Health Evaluation Sciences (B.B.W.), University of Virginia, Charlottesville, VA
| |
Collapse
|
13
|
Franks PW, Melén E, Friedman M, Sundström J, Kockum I, Klareskog L, Almqvist C, Bergen SE, Czene K, Hägg S, Hall P, Johnell K, Malarstig A, Catrina A, Hagström H, Benson M, Gustav Smith J, Gomez MF, Orho-Melander M, Jacobsson B, Halfvarson J, Repsilber D, Oresic M, Jern C, Melin B, Ohlsson C, Fall T, Rönnblom L, Wadelius M, Nordmark G, Johansson Å, Rosenquist R, Sullivan PF. Technological readiness and implementation of genomic-driven precision medicine for complex diseases. J Intern Med 2021; 290:602-620. [PMID: 34213793 DOI: 10.1111/joim.13330] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 03/21/2021] [Accepted: 04/12/2021] [Indexed: 12/20/2022]
Abstract
The fields of human genetics and genomics have generated considerable knowledge about the mechanistic basis of many diseases. Genomic approaches to diagnosis, prognostication, prevention and treatment - genomic-driven precision medicine (GDPM) - may help optimize medical practice. Here, we provide a comprehensive review of GDPM of complex diseases across major medical specialties. We focus on technological readiness: how rapidly a test can be implemented into health care. Although these areas of medicine are diverse, key similarities exist across almost all areas. Many medical areas have, within their standards of care, at least one GDPM test for a genetic variant of strong effect that aids the identification/diagnosis of a more homogeneous subset within a larger disease group or identifies a subset with different therapeutic requirements. However, for almost all complex diseases, the majority of patients do not carry established single-gene mutations with large effects. Thus, research is underway that seeks to determine the polygenic basis of many complex diseases. Nevertheless, most complex diseases are caused by the interplay of genetic, behavioural and environmental risk factors, which will likely necessitate models for prediction and diagnosis that incorporate genetic and non-genetic data.
Collapse
Affiliation(s)
- P W Franks
- From the, Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden.,Department of Nutrition, Harvard School of Public Health, Boston, MA, USA
| | - E Melén
- Department of Clinical Science and Education Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - M Friedman
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - J Sundström
- Department of Cardiology, Akademiska Sjukhuset, Uppsala, Sweden.,George Institute for Global Health, Camperdown, NSW, Australia.,Medical Sciences, Uppsala University, Uppsala, Sweden
| | - I Kockum
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - L Klareskog
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Rheumatology, Karolinska Institutet, Stockholm, Sweden
| | - C Almqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - S E Bergen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - K Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - S Hägg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - P Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Oncology, Södersjukhuset, Stockholm, Sweden
| | - K Johnell
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - A Malarstig
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Pfizer, Worldwide Research and Development, Stockholm, Sweden
| | - A Catrina
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - H Hagström
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,Division of Hepatology, Department of Upper GI, Karolinska University Hospital, Stockholm, Sweden
| | - M Benson
- Department of Pediatrics, Linkopings Universitet, Linkoping, Sweden.,Division of Ear, Nose and Throat Diseases, Department of Clinical Sciences, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - J Gustav Smith
- Department of Cardiology and Wallenberg Center for Molecular Medicine, Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden.,Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - M F Gomez
- From the, Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - M Orho-Melander
- From the, Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - B Jacobsson
- Division of Health Data and Digitalisation, Norwegian Institute of Public Health, Genetics and Bioinformatics, Oslo, Norway.,Department of Obstetrics and Gynecology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Obstetrics and Gynecology, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - J Halfvarson
- School of Medical Sciences, Örebro University, Örebro, Sweden
| | - D Repsilber
- Functional Bioinformatics, Örebro University, Örebro, Sweden
| | - M Oresic
- School of Medical Sciences, Örebro University, Örebro, Sweden.,Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, FI, Finland
| | - C Jern
- Department of Clinical Genetics and Genomics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - B Melin
- Department of Radiation Sciences, Oncology, Umeå Universitet, Umeå, Sweden
| | - C Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre, CBAR, University of Gothenburg, Gothenburg, Sweden.,Department of Drug Treatment, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - T Fall
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, Sweden
| | - L Rönnblom
- Department of Medical Sciences, Rheumatology & Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - M Wadelius
- Department of Medical Sciences, Clinical Pharmacogenomics & Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - G Nordmark
- Department of Medical Sciences, Rheumatology & Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Å Johansson
- Institute for Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - R Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - P F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
14
|
Rost NS, Meschia JF, Gottesman R, Wruck L, Helmer K, Greenberg SM. Cognitive Impairment and Dementia After Stroke: Design and Rationale for the DISCOVERY Study. Stroke 2021; 52:e499-e516. [PMID: 34039035 PMCID: PMC8316324 DOI: 10.1161/strokeaha.120.031611] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Stroke is a leading cause of the adult disability epidemic in the United States, with a major contribution from poststroke cognitive impairment and dementia (PSCID), the rates of which are disproportionally high among the health disparity populations. Despite the PSCID's overwhelming impact on public health, a knowledge gap exists with regard to the complex interaction between the acute stroke event and highly prevalent preexisting brain pathology related to cerebrovascular and Alzheimer disease or related dementia. Understanding the factors that modulate PSCID risk in relation to index stroke event is critically important for developing personalized prognostication of PSCID, targeted interventions to prevent it, and for informing future clinical trial design. The DISCOVERY study (Determinants of Incident Stroke Cognitive Outcomes and Vascular Effects on Recovery), a collaborative network of thirty clinical performance clinical sites with access to acute stroke populations and the expertise and capacity for systematic assessment of PSCID will address this critical challenge. DISCOVERY is a prospective, multicenter, observational, nested-cohort study of 8000 nondemented ischemic and hemorrhagic stroke patients enrolled at the time of index stroke and followed for a minimum of 2 years, with serial cognitive evaluations and assessments of functional outcome, with subsets undergoing research magnetic resonance imaging and positron emission tomography and comprehensive genetic/genomic and fluid biomarker testing. The overall scientific objective of this study is to elucidate mechanisms of brain resilience and susceptibility to PSCID in diverse US populations based on complex interplay between life-course exposure to multiple vascular risk factors, preexisting burden of microvascular and neurodegenerative pathology, the effect of strategic acute stroke lesions, and the mediating effect of genomic and epigenomic variation.
Collapse
Affiliation(s)
- Natalia S. Rost
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | | | | | | | - Karl Helmer
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA
| | - Steven M. Greenberg
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
15
|
Lee JM, Fernandez Cadenas I, Lindgren A. Using Human Genetics to Understand Mechanisms in Ischemic Stroke Outcome: From Early Brain Injury to Long-Term Recovery. Stroke 2021; 52:3013-3024. [PMID: 34399587 PMCID: PMC8938679 DOI: 10.1161/strokeaha.121.032622] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There is a critical need to elucidate molecular mechanisms underlying brain injury, repair, and recovery following ischemic stroke-a global health problem with major social and economic impact. Despite 5 decades of intensive research, there are no widely accepted neuroprotective drugs that mitigate ischemic brain injury, or neuroreparative drugs, or personalized approaches that guide therapies to enhance recovery. We here explore novel reverse translational approaches that will complement traditional forward translational methods in identifying mechanisms relevant to human stroke outcome. Although genome-wide association studies have yielded over 30 genetic loci that influence ischemic stroke risk, only a few genome-wide association studies have been performed for stroke outcome. We discuss important considerations for genetic studies of ischemic stroke outcome-including carefully designed phenotypes that capture injury/recovery mechanisms, anchored in time to stroke onset. We also address recent genome-wide association studies that provide insight into mechanisms underlying brain injury and repair. There are several ongoing initiatives exploring genomic associations with novel phenotypes related to stroke outcome. To improve the understanding of the genetic architecture of ischemic stroke outcome, larger studies using standardized phenotypes, preferably embedded in standard-of-care measures, are needed. Novel techniques beyond genome-wide association studies-including exploiting informatics, multi-omics, and novel analytics-promise to uncover genetic and molecular pathways from which drug targets and other new interventions may be identified.
Collapse
Affiliation(s)
- Jin-Moo Lee
- The Hope Center for Neurological Disorders and the Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Israel Fernandez Cadenas
- Stroke pharmacogenomics and genetics group. Sant Pau Biomedical Research Institute, Barcelona, Spain
| | - Arne Lindgren
- Department of Clinical Sciences Lund, Neurology, Lund University; Department of Neurology, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
16
|
A Clinical-Radiomics Nomogram for Functional Outcome Predictions in Ischemic Stroke. Neurol Ther 2021; 10:819-832. [PMID: 34170502 PMCID: PMC8571444 DOI: 10.1007/s40120-021-00263-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/10/2021] [Indexed: 11/02/2022] Open
Abstract
INTRODUCTION Stroke remains a leading cause of death and disability worldwide. Effective and prompt prognostic evaluation is vital for determining the appropriate management strategy. Radiomics is an emerging noninvasive method used to identify the quantitative imaging indicators for predicting important clinical outcomes. This study was conducted to investigate and validate a radiomics nomogram for predicting ischemic stroke prognosis using the modified Rankin scale (mRS). METHODS A total of 598 consecutive patients with subacute infarction confirmed by diffusion-weighted imaging (DWI), from January 2018 to December 2019, were retrospectively assessed. They were assigned to the good (mRS ≤ 2) and poor (mRS > 2) functional outcome groups, respectively. Then, 399 patients examined by MR scanner 1 and 199 patients scanned by MR scanner 2 were assigned to the training and validation cohorts, respectively. Infarction lesions underwent manual segmentation on DWI, extracting 402 radiomic features. A radiomics nomogram encompassing patient characteristics and the radiomics signature was built using a multivariate logistic regression model. The performance of the nomogram was evaluated in the training and validation cohorts. Ultimately, decision curve analysis was implemented to assess the clinical value of the nomogram. The performance of infarction lesion volume was also evaluated using univariate analysis. RESULTS Stroke lesion volume showed moderate performance, with an area under the curve (AUC) of 0.678. The radiomics signature, including 11 radiomics features, exhibited good prediction performance. The radiomics nomogram, encompassing clinical characteristics (age, hemorrhage, and 24 h National Institutes of Health Stroke Scale score) and the radiomics signature, presented good discriminatory potential in the training cohort [AUC = 0.80; 95% confidence interval (CI) 0.75-0.86], which was validated in the validation cohort (AUC = 0.73; 95% CI 0.63-0.82). In addition, it demonstrated good calibration in the training (p = 0.55) and validation (p = 0.21) cohorts. Decision curve analysis confirmed the clinical value of this nomogram. CONCLUSION This novel noninvasive clinical-radiomics nomogram shows good performance in predicting ischemic stroke prognosis.
Collapse
|
17
|
Lindgren AG, Braun RG, Juhl Majersik J, Clatworthy P, Mainali S, Derdeyn CP, Maguire J, Jern C, Rosand J, Cole JW, Lee JM, Khatri P, Nyquist P, Debette S, Keat Wei L, Rundek T, Leifer D, Thijs V, Lemmens R, Heitsch L, Prasad K, Jimenez Conde J, Dichgans M, Rost NS, Cramer SC, Bernhardt J, Worrall BB, Fernandez-Cadenas I. International stroke genetics consortium recommendations for studies of genetics of stroke outcome and recovery. Int J Stroke 2021; 17:260-268. [PMID: 33739214 PMCID: PMC8864333 DOI: 10.1177/17474930211007288] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Numerous biological mechanisms contribute to outcome after stroke, including
brain injury, inflammation, and repair mechanisms. Clinical genetic studies have
the potential to discover biological mechanisms affecting stroke recovery in
humans and identify intervention targets. Large sample sizes are needed to
detect commonly occurring genetic variations related to stroke brain injury and
recovery. However, this usually requires combining data from multiple studies
where consistent terminology, methodology, and data collection timelines are
essential. Our group of expert stroke and rehabilitation clinicians and
researchers with knowledge in genetics of stroke recovery here present
recommendations for harmonizing phenotype data with focus on measures suitable
for multicenter genetic studies of ischemic stroke brain injury and recovery.
Our recommendations have been endorsed by the International Stroke Genetics
Consortium.
Collapse
Affiliation(s)
- Arne G Lindgren
- Department of Clinical Sciences Lund, Neurology, 5193-->Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Robynne G Braun
- Department of Neurology, University of Maryland, Baltimore, MD, USA
| | | | | | - Shraddha Mainali
- Department of Neurology, 2647-->The Ohio State University, Columbus, OH, USA
| | - Colin P Derdeyn
- Department of Radiology, University of Iowa, Iowa City, IA, USA
| | - Jane Maguire
- Faculty of Health, University of Technology Sydney, Ultimo, NSW, Australia
| | - Christina Jern
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jonathan Rosand
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - John W Cole
- Neurology Service, Baltimore Veterans Affairs Medical Center, Baltimore, MD, USA.,Department of Neurology, 12264-->University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jin-Moo Lee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Pooja Khatri
- Department of Neurology and Rehabilitation Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Paul Nyquist
- Neurology, Anesthesiology/Critical Care Medicine, Neurosurgery, and General Internal Medicine, 1500-->Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Stéphanie Debette
- Bordeaux Population Health, Inserm U1219, University of Bordeaux, Bordeaux, France.,Neurology Department, Bordeaux University Hospital, Bordeaux, France
| | - Loo Keat Wei
- Department of Biological Science, Faculty of Science, Universiti Tunku Abdul Rahman, Perak, Malaysia
| | - Tatjana Rundek
- Department of Neurology, 12235-->University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dana Leifer
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Vincent Thijs
- Stroke Theme, Florey Institute of Neuroscience and Mental Health, Melbourne, Vic, Australia
| | - Robin Lemmens
- Department of Neuroscience, University of Leuven, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Laura Heitsch
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kameshwar Prasad
- Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Jordi Jimenez Conde
- Neurology Department, Neurovascular Research Group, Institut Hospital del Mar d'Investigació Mèdica, Barcelona, Spain.,Neurology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital, LMU, Munich, Germany
| | - Natalia S Rost
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Steven C Cramer
- Department of Neurology, UCLA, Los Angeles, CA, USA.,California Rehabilitation Institute, Los Angeles, CA, USA
| | - Julie Bernhardt
- Stroke Theme, Florey Institute of Neuroscience and Mental Health, Melbourne, Vic, Australia
| | - Bradford B Worrall
- Department of Neurology, University of Virginia, Charlottesville, VA, USA
| | - Israel Fernandez-Cadenas
- Stroke Pharmacogenomics and Genetics Group, Sant Pau Biomedical Research Institute, Barcelona, Spain
| | | |
Collapse
|
18
|
Cole JW, Adigun T, Akinyemi R, Akpa OM, Bell S, Chen B, Jimenez Conde J, Lazcano Dobao U, Fernandez I, Fornage M, Gallego-Fabrega C, Jern C, Krawczak M, Lindgren A, Markus HS, Melander O, Owolabi M, Schlicht K, Söderholm M, Srinivasasainagendra V, Soriano Tárraga C, Stenman M, Tiwari H, Corasaniti M, Fecteau N, Guizzardi B, Lopez H, Nguyen K, Gaynor B, O’Connor T, Stine OC, Kittner SJ, McArdle P, Mitchell BD, Xu H, Grond-Ginsbach C. The copy number variation and stroke (CaNVAS) risk and outcome study. PLoS One 2021; 16:e0248791. [PMID: 33872305 PMCID: PMC8055008 DOI: 10.1371/journal.pone.0248791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 03/04/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND AND PURPOSE The role of copy number variation (CNV) variation in stroke susceptibility and outcome has yet to be explored. The Copy Number Variation and Stroke (CaNVAS) Risk and Outcome study addresses this knowledge gap. METHODS Over 24,500 well-phenotyped IS cases, including IS subtypes, and over 43,500 controls have been identified, all with readily available genotyping on GWAS and exome arrays, with case measures of stroke outcome. To evaluate CNV-associated stroke risk and stroke outcome it is planned to: 1) perform Risk Discovery using several analytic approaches to identify CNVs that are associated with the risk of IS and its subtypes, across the age-, sex- and ethnicity-spectrums; 2) perform Risk Replication and Extension to determine whether the identified stroke-associated CNVs replicate in other ethnically diverse datasets and use biomarker data (e.g. methylation, proteomic, RNA, miRNA, etc.) to evaluate how the identified CNVs exert their effects on stroke risk, and lastly; 3) perform outcome-based Replication and Extension analyses of recent findings demonstrating an inverse relationship between CNV burden and stroke outcome at 3 months (mRS), and then determine the key CNV drivers responsible for these associations using existing biomarker data. RESULTS The results of an initial CNV evaluation of 50 samples from each participating dataset are presented demonstrating that the existing GWAS and exome chip data are excellent for the planned CNV analyses. Further, some samples will require additional considerations for analysis, however such samples can readily be identified, as demonstrated by a sample demonstrating clonal mosaicism. CONCLUSION The CaNVAS study will cost-effectively leverage the numerous advantages of using existing case-control data sets, exploring the relationships between CNV and IS and its subtypes, and outcome at 3 months, in both men and women, in those of African and European-Caucasian descent, this, across the entire adult-age spectrum.
Collapse
Affiliation(s)
- John W. Cole
- Veterans Affairs Maryland Health Care System, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | | | | | | | - Steven Bell
- Unversity of Cambridge, Cambridge, England, United Kingdom
| | - Bowang Chen
- National Center for Cardiovascular Diseases, Beijing, China
| | | | - Uxue Lazcano Dobao
- IMIM-Hospital del Mar; Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Israel Fernandez
- Institute of Research Hospital de la Santa Creu I Sant Pau, Barcelona, Spain
| | - Myriam Fornage
- University of Texas Health Science at Houston, Institute of Molecular Medicine & School of Public Health, Houston, TX, United States of America
| | | | | | - Michael Krawczak
- Institute of Medical Statistics and Informatics, University of Kiel, Kiel, Germany
| | | | - Hugh S. Markus
- Unversity of Cambridge, Cambridge, England, United Kingdom
| | | | | | - Kristina Schlicht
- Institute of Medical Statistics and Informatics, University of Kiel, Kiel, Germany
| | - Martin Söderholm
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital Malmö and Lund, Lund, Sweden
| | | | | | | | - Hemant Tiwari
- School of Public Health, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Margaret Corasaniti
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Natalie Fecteau
- Veterans Affairs Maryland Health Care System, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Beth Guizzardi
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Haley Lopez
- Veterans Affairs Maryland Health Care System, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Kevin Nguyen
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Brady Gaynor
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Timothy O’Connor
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - O. Colin Stine
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Steven J. Kittner
- Veterans Affairs Maryland Health Care System, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Patrick McArdle
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Braxton D. Mitchell
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Huichun Xu
- University of Maryland School of Medicine, Baltimore, MD, United States of America
| | | |
Collapse
|
19
|
Wang H, Lin J, Zheng L, Zhao J, Song B, Dai Y. Texture analysis based on ADC maps and T2-FLAIR images for the assessment of the severity and prognosis of ischaemic stroke. Clin Imaging 2020; 67:152-159. [PMID: 32739735 DOI: 10.1016/j.clinimag.2020.06.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/12/2020] [Accepted: 06/07/2020] [Indexed: 10/24/2022]
Abstract
OBJECTIVES To explore the feasibility of texture analysis based on T2-weighted fluid-attenuated inversion recovery (T2-FLAIR) images and apparent diffusion coefficient (ADC) maps in the assessment of the severity and prognosis of ischaemic stroke using the National Institutes of Health Stroke Scale (NIHSS) and modified Rankin scale (mRS) scores, respectively. METHODS Overall, 116 patients diagnosed with subacute ischaemic stroke were included in this retrospective study. Based on T2-FLAIR images and ADC maps, 15 texture features were extracted from the ROIs of each patient using grey-level co-occurrence matrix (GLCM) and local binary pattern histogram Fourier (LBP-HF) methods. The correlations of NIHSS score on admission (NIHSSbaseline), NIHSS score 24 h after stroke onset (NIHSS24h) and mRS score with the texture features were evaluated using Spearman's partial correlations. The receiver operating characteristic (ROC) curve was used to compare the performance of the selected texture features in the evaluation of stroke severity and prognosis. RESULTS Texture features derived from the T2-FLAIR images and ADC maps were correlated with NIHSS score and mRS score. EntropyADC and 0.75QuantileT2-FLAIR showed the best diagnostic performance for assessing stroke severity. The combination of EntropyADC and 0.75QuantileT2-FLAIR achieved a better performance in the evaluation of stroke severity (AUC = 0.7, p = 0.01) than either feature alone. Only 0.05QuantileT2-FLAIR was found to be correlated with mRS score, and none of the texture features were predictive of mRS score. CONCLUSION Texture features derived from T2-FLAIR images and ADC maps might serve as biomarkers to evaluate stroke severity, but were insufficient to predict stroke prognosis.
Collapse
Affiliation(s)
- Hao Wang
- Department of Radiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Jixian Lin
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai, China; Department of Electronic Engineering, Fudan University, Shanghai, China
| | - Liyun Zheng
- Central Research Institute, United Imaging Healthcare, Shanghai, China
| | - Jing Zhao
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai, China
| | - Bin Song
- Department of Radiology, Minhang Hospital, Fudan University, Shanghai, China.
| | - Yongming Dai
- Central Research Institute, United Imaging Healthcare, Shanghai, China
| |
Collapse
|
20
|
Multilevel omics for the discovery of biomarkers and therapeutic targets for stroke. Nat Rev Neurol 2020; 16:247-264. [PMID: 32322099 DOI: 10.1038/s41582-020-0350-6] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2020] [Indexed: 02/07/2023]
Abstract
Despite many years of research, no biomarkers for stroke are available to use in clinical practice. Progress in high-throughput technologies has provided new opportunities to understand the pathophysiology of this complex disease, and these studies have generated large amounts of data and information at different molecular levels. The integration of these multi-omics data means that thousands of proteins (proteomics), genes (genomics), RNAs (transcriptomics) and metabolites (metabolomics) can be studied simultaneously, revealing interaction networks between the molecular levels. Integrated analysis of multi-omics data will provide useful insight into stroke pathogenesis, identification of therapeutic targets and biomarker discovery. In this Review, we detail current knowledge on the pathology of stroke and the current status of biomarker research in stroke. We summarize how proteomics, metabolomics, transcriptomics and genomics are all contributing to the identification of new candidate biomarkers that could be developed and used in clinical stroke management.
Collapse
|
21
|
Affiliation(s)
- James F Meschia
- From the Department of Neurology, Mayo Clinic in Florida, Jacksonville
| |
Collapse
|