1
|
Chen M, Joshi KC, Kolb B, Sitton CW, Pujara DK, Abraham MG, Ortega-Gutierrez S, Kasner SE, Hussain SM, Churilov L, Blackburn S, Sundararajan S, Hu YC, Herial N, Arenillas JF, Tsai JP, Budzik RF, Hicks W, Kozak O, Yan B, Cordato D, Manning NW, Parsons M, Hanel RA, Aghaebrahim A, Wu T, Cardona Portela P, Gandhi CD, Al-Mufti F, Perez de la Ossa N, Schaafsma J, Blasco J, Sangha N, Warach S, Kleinig TJ, Johns H, Shaker F, Abdulrazzak MA, Ray A, Sunshine J, Opaskar A, Duncan KR, Xiong W, Al-Shaibi FK, Samaniego EA, Nguyen TN, Fifi JT, Tjoumakaris SI, Jabbour P, Mendes Pereira V, Lansberg MG, Sila C, Bambakidis NC, Davis S, Wechsler L, Albers GW, Grotta JC, Ribo M, Hassan AE, Campbell B, Hill MD, Sarraj A. Clinical relevance of intracranial hemorrhage after thrombectomy versus medical management for large core infarct: a secondary analysis of the SELECT2 randomized trial. J Neurointerv Surg 2025; 17:120-127. [PMID: 38471760 DOI: 10.1136/jnis-2023-021219] [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: 11/02/2023] [Accepted: 02/18/2024] [Indexed: 03/14/2024]
Abstract
BACKGROUND The incidence of intracerebral hemorrhage (ICH) and its effect on the outcomes after endovascular thrombectomy (EVT) for patients with large core infarcts have not been well-characterized. METHODS SELECT2 trial follow-up imaging was evaluated using the Heidelberg Bleeding Classification (HBC) to define hemorrhage grade. The association of ICH with clinical outcomes and treatment effect was examined. RESULTS Of 351 included patients, 194 (55%) and 189 (54%) demonstrated intracranial and intracerebral hemorrhage, respectively, with a higher incidence in EVT (134 (75%) and 130 (73%)) versus medical management (MM) (60 (35%) and 59 (34%), both P<0.001). Hemorrhagic infarction type 1 (HBC=1a) and type 2 (HBC=1b) accounted for 93% of all hemorrhages. Parenchymal hematoma (PH) type 1 (HBC=1c) and type 2 (HBC=2) were observed in 1 (0.6%) EVT-treated and 4 (2.2%) MM patients. Symptomatic ICH (sICH) (SITS-MOST definition) was seen in 0.6% EVT patients and 1.2% MM patients. No trend for ICH with core volumes (P=0.10) or Alberta Stroke Program Early CT Score (ASPECTS) (P=0.74) was observed. Among EVT patients, the presence of any ICH did not worsen clinical outcome (modified Rankin Scale (mRS) at 90 days: 4 (3-6) vs 4 (3-6); adjusted generalized OR 1.00, 95% CI 0.68 to 1.47, P>0.99) or modify EVT treatment effect (Pinteraction=0.77). CONCLUSIONS ICH was present in 75% of the EVT population, but PH or sICH were infrequent. The presence of any ICH did not worsen functional outcomes or modify EVT treatment effect at 90-day follow-up. The high rate of hemorrhages overall still represents an opportunity for adjunctive therapies in EVT patients with a large ischemic core.
Collapse
Affiliation(s)
- Michael Chen
- Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Krishna C Joshi
- Neurological Surgery, Rush University Medical Center, Chicago, Illinois, USA
| | - Bradley Kolb
- Rush University Medical Center, Chicago, Illinois, USA
| | - Clark W Sitton
- Diagnostic & Interventional Imaging, UT Houston, Houston, Texas, USA
| | | | - Michael G Abraham
- Neurology and Radiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Santiago Ortega-Gutierrez
- Neuroloy, Neurosurgery and Radiology, The University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Scott E Kasner
- Neurology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shazam M Hussain
- Cleveland Clinic Stroke Program, Cleveland Clinic, Cleveland Heights, Ohio, USA
| | | | - Spiros Blackburn
- Neurosurgery, University of Texas Medical School at Houston Vivian L Smith Department of Neurosurgery, Houston, Texas, USA
| | | | - Yin C Hu
- Neurosurgery, University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | - Nabeel Herial
- Neurological Surgery, Thomas Jefferson University Hospitals, Philadelphia, Pennsylvania, USA
| | - Juan F Arenillas
- Stroke Unit, Neurology Department, Stroke Unit. Neurology Department. Universitary Hospital, Valladolid (Spain), Valladolid, Spain
| | - Jenny P Tsai
- Cerebrovascular Center, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - William Hicks
- OhioHealth Neurological Physicians, Riverside Methodist Hospital, Columbus, Ohio, USA
| | - Osman Kozak
- Neurosurgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- Abington Memorial Hospital, Abington, Pennsylvania, USA
| | - Bernard Yan
- The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Dennis Cordato
- Department of Neurology and Neurophysiology, Liverpool Hospital, Liverpool, New South Wales, Australia
- Ingham Institute of Applied Medical Research, Sydney, New South Wales, Australia
| | - Nathan W Manning
- Institute of Neurosciences, UNSW Prince of Wales Clinical School, Sydney, New South Wales, Australia
- Advanced Endovascular Therapy, Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia
| | - Mark Parsons
- Liverpool Hospital, Liverpool, New South Wales, Australia
| | | | - Amin Aghaebrahim
- Neurological Institute, Lyerley Neurosurgery, Baptist Medical Center Jacksonville, Jacksonville, Florida, USA
| | - Teddy Wu
- Neurology, Christchurch Hospital, Christchurch, Canterbury, New Zealand
| | | | - Chirag D Gandhi
- Neurosurgery, Westchester Medical Center, Valhalla, New York, USA
| | - Fawaz Al-Mufti
- Neurology and Neurosurgery, Westchester Medical Center, Valhalla, New York, USA
| | | | - Joanna Schaafsma
- Medicine - Div. Neurology, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Jordi Blasco
- Neurointerventional Department C.D.I, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Navdeep Sangha
- Kaiser Permanente Southern California, Los Angeles, California, USA
| | - Steven Warach
- University of Texas at Austin Dell Seton Medical Center, Austin, Texas, USA
| | - Timothy J Kleinig
- Neurology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Hannah Johns
- University of Melbourne, Parkville, Victoria, Australia
| | - Faris Shaker
- Neurology, University of Texas McGovern Medical School, Houston, Texas, USA
| | | | - Abhishek Ray
- Neurological Surgery, University Hospitals, Cleveland, Ohio, USA
| | - Jeffery Sunshine
- Radiology, University Hospitals-Case Medical Center, Cleveland, Ohio, USA
| | - Amanda Opaskar
- University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Kelsey R Duncan
- University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Wei Xiong
- University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | | | - Edgar A Samaniego
- Neurology, The University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Thanh N Nguyen
- Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Johanna T Fifi
- Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Pascal Jabbour
- Neurological Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Vitor Mendes Pereira
- Division of Neuroradiology, Department of Medical Imaging and Division of Neurosurgery, Department of Surgery, University Health Network - Toronto Western Hospital, Toronto, Ontario, Canada
| | - Maarten G Lansberg
- Department of Neurology, Stanford Stroke Center, Stanford University, Stanford, California, USA
| | - Cathy Sila
- Neurological Institute, University Hospitals- Case Medical Center, Cleveland, Ohio, USA
- Neurology, Case Western Reserve University School of Medicine
| | - Nicholas C Bambakidis
- Neurological Surgery, University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | - Stephen Davis
- The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Lawrence Wechsler
- University of Pittsburgh Medical Center Health System, Pittsburgh, Pennsylvania, USA
| | - Gregory W Albers
- Neurology & Neurological Sciences, Stanford University, Stanford, California, USA
| | - James C Grotta
- Neurology, Memorial Hermann Hospital/UT Houston, Houston, Texas, USA
| | - Marc Ribo
- Stroke Unit, Neurology, Hospital Vall d'Hebron, Barcelona, Spain
- Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ameer E Hassan
- Department of Neurology, University of Texas Rio Grande Valley, Harlingen, Texas, USA
| | - Bruce Campbell
- The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Michael D Hill
- Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Clinical Neurosciences, Foothills Medical Centre, Calgary, Alberta, Canada
| | - Amrou Sarraj
- Neurology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
2
|
Zohdy YM, Garzon-Muvdi T, Grossberg JA, Barrow DL, Howard BM, Pradilla G, Kobeissy FH, Tomlinson S, Alawieh AM. Complement inhibition targets a rich-club within the neuroinflammatory network after stroke to improve radiographic and functional outcomes. J Neuroinflammation 2025; 22:1. [PMID: 39754245 PMCID: PMC11699776 DOI: 10.1186/s12974-024-03316-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 11/29/2024] [Indexed: 01/06/2025] Open
Abstract
Following recent advances in post-thrombectomy stroke care, the role of neuroinflammation and neuroprotective strategies in mitigating secondary injury has gained prominence. Yet, while neuroprotection and anti-inflammatory agents have re-emerged in clinical trials, their success has been limited. The neuroinflammatory response in cerebral ischemia is robust and multifactorial, complicating therapeutic approaches targeting single pathways. In this study, we aimed to characterize early inflammatory gene dysregulation following ischemic stroke using translational in-silico and in-vivo approaches. Using an in vivo ischemic stroke model, transcriptomic analysis revealed significant dysregulation of inflammatory genes. Graph theory analysis then showed a rich-club organization among stroke-related genes, with highly connected core nodes. The expression levels of the core genes identified within this network significantly explained radiological outcomes, including T2-signal hyperintensity (R2 = 0.57, P < 0.001), mean diffusivity (R2 = 0.52, P < 0.001), and mean kurtosis (R2 = 0.65, P < 0.001), correlating more strongly than non-core genes. Similar findings were observed with functional and cognitive outcomes, showing R2 values of 0.58, 0.7, 0.54, and 0.7 for neurological severity scores, corner tasks, passive avoidance, and novel object recognition tasks, respectively (P < 0.001). Using in-silico analysis, we identified a set of upstream regulators directly interacting with core network nodes, leading to simulations that highlighted C3-targeted therapy as a potential treatment. This hypothesis was then confirmed in vivo using a targeted C3 inhibitor (CR2-fH), which reversed gene dysregulation in the neuroinflammatory network and improved radiological and functional outcomes. Our findings underscore the significance of neuroinflammation in stroke pathology, supporting network-based therapeutic targeting and demonstrating the benefits of targeted complement inhibition in enhancing outcomes through modulation of the neuroinflammatory network core. This study's approach, combining graph theory analysis along with in-silico modeling, offers a promising translational pipeline applicable to stroke and other complex diseases.
Collapse
Affiliation(s)
- Youssef M Zohdy
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Tomas Garzon-Muvdi
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jonathan A Grossberg
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Daniel L Barrow
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Brian M Howard
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Gustavo Pradilla
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Firas H Kobeissy
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Ralph Johnson VA Medical Center, Charleston, SC, USA
| | - Ali M Alawieh
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
3
|
Balaji PG, Bhimrao LS, Yadav AK. Revolutionizing Stroke Care: Nanotechnology-Based Brain Delivery as a Novel Paradigm for Treatment and Diagnosis. Mol Neurobiol 2025; 62:184-220. [PMID: 38829514 DOI: 10.1007/s12035-024-04215-3] [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: 03/19/2024] [Accepted: 05/03/2024] [Indexed: 06/05/2024]
Abstract
Stroke, a severe medical condition arising from abnormalities in the coagulation-fibrinolysis cycle and metabolic processes, results in brain cell impairment and injury due to blood flow obstruction within the brain. Prompt and efficient therapeutic approaches are imperative to control and preserve brain functions. Conventional stroke medications, including fibrinolytic agents, play a crucial role in facilitating reperfusion to the ischemic brain. However, their clinical efficacy is hampered by short plasma half-lives, limited brain tissue distribution attributed to the blood-brain barrier (BBB), and lack of targeted drug delivery to the ischemic region. To address these challenges, diverse nanomedicine strategies, such as vesicular systems, polymeric nanoparticles, dendrimers, exosomes, inorganic nanoparticles, and biomimetic nanoparticles, have emerged. These platforms enhance drug pharmacokinetics by facilitating targeted drug accumulation at the ischemic site. By leveraging nanocarriers, engineered drug delivery systems hold the potential to overcome challenges associated with conventional stroke medications. This comprehensive review explores the pathophysiological mechanism underlying stroke and BBB disruption in stroke. Additionally, this review investigates the utilization of nanocarriers for current therapeutic and diagnostic interventions in stroke management. By addressing these aspects, the review aims to provide insight into potential strategies for improving stroke treatment and diagnosis through a nanomedicine approach.
Collapse
Affiliation(s)
- Paul Gajanan Balaji
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli (An Institute of National Importance under Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, GOI), A Transit Campus at Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow, 226002, Uttar Pradesh, India
| | - Londhe Sachin Bhimrao
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli (An Institute of National Importance under Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, GOI), A Transit Campus at Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow, 226002, Uttar Pradesh, India
| | - Awesh K Yadav
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli (An Institute of National Importance under Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, GOI), A Transit Campus at Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow, 226002, Uttar Pradesh, India.
| |
Collapse
|
4
|
Wang Y, Maeda T, You S, Chen C, Liu L, Zhou Z, Robinson TG, Lindley RI, Delcourt C, Mair G, Wardlaw JM, Chalmers JP, Arima H, Huang Y, Kim JS, Lavados PM, Lee TH, Levi C, Parsons MW, Martins SC, Pandian JD, Pontes-Neto OM, Sharma VK, Nguyen TH, Wang J, Wu S, Liu M, Anderson CS, Chen X. Patterns and Clinical Implications of Hemorrhagic Transformation After Thrombolysis in Acute Ischemic Stroke: Results From the ENCHANTED Study. Neurology 2024; 103:e210020. [PMID: 39541551 DOI: 10.1212/wnl.0000000000210020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Hemorrhagic transformation may be a potentially devastating complication of IV thrombolysis (IVT) in acute ischemic stroke, but what degree of hemorrhage indicates the greatest negative effect is not known. We aimed to define the associations between hemorrhagic transformation patterns, classified according to clinical and imaging categories, and clinical outcomes after IVT. METHODS We conducted a post hoc analysis from the international Enhanced Control of Hypertension and Thrombolysis Stroke Study. Symptomatic intracerebral hemorrhage (sICH) was defined based on established criteria, such as the Safe Implementation of Thrombolysis in Stroke-Monitoring Study (SITS-MOST) criteria. Asymptomatic intracerebral hemorrhage (aICH) was defined as any intracerebral hemorrhage that did not meet the criteria for sICH. Imaging subtypes of hemorrhagic transformation were assessed using the Heidelberg Bleeding Classification system. The primary outcome was death or major disability, defined by modified Rankin scale (mRS) scores 3-6 at 90 days. Secondary outcomes included death, death or disability (mRS 2-6), and poor health-related quality of life (HRQoL), defined as an overall heath utility score ≤0.7 (mean). RESULTS Of the 4,370 participants, 779 (17.8%) developed any intracranial hemorrhage (ICH), with a median time from randomization to hemorrhage of 23.5 hours (interquartile range 18.92-26.07). According to the SITS-MOST criteria, 62 patients (1.4% of 4,370) were classified as sICH, and 717 patients (16.4% of 4,370) were classified as aICH. sICH per SITS-MOST criteria was associated with death or major disability (odds ratio [OR] 23.05, 95% CI 8.97-59.23), death (OR 20.14, 95% CI 11.32-35.84), death or disability (OR 61.36, 95% CI 8.40-448.01), and poor HRQoL (OR 17.87, 95% CI 7.47-42.71). Similarly, aICH per SITS-MOST criteria was also associated with death or major disability (OR 2.23, 95% CI 1.84-2.70), death (OR 1.82, 95% CI 1.39-2.38), death or disability (OR 2.29, 95% CI 1.87-2.80), and poor HRQoL (OR 1.81, 95% CI 1.50-2.18). Comparable associations were observed for sICH and aICH defined by other criteria, as well as for imaging subtypes based on Heidelberg Bleeding Classification system. DISCUSSION All forms of post-IVT hemorrhagic transformation in acute ischemic stroke are associated with increased odds of poor clinical outcomes. Of note, aICH after IVT should not be considered clinically innocuous. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov (NCT01422616).
Collapse
Affiliation(s)
- Yanan Wang
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Toshiki Maeda
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Shoujiang You
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Chen Chen
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Leibo Liu
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Zien Zhou
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Thompson G Robinson
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Richard Iain Lindley
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Candice Delcourt
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Grant Mair
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Joanna M Wardlaw
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - John Philip Chalmers
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Hisatomi Arima
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Yining Huang
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Jong S Kim
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Pablo M Lavados
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Tsong-Hai Lee
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Christopher Levi
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Mark W Parsons
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Sheila Co Martins
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Jeyaraj Durai Pandian
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Octavio M Pontes-Neto
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Vijay K Sharma
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Thang Huy Nguyen
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Jiguang Wang
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Simiao Wu
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Ming Liu
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Craig S Anderson
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| | - Xiaoying Chen
- From the Department of Neurology (Y.W., S.W., M.L.), West China Hospital, Sichuan University, Chengdu, China; The George Institute for Global Health (Y.W., T.M., S.Y., C.C., L.L., Z.Z., C.D., J.P.C., C.S.A., X.C.), Faculty of Medicine, University of New South Wales, Sydney, Australia; Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre (T.G.R.), Leicester, United Kingdom; University of Sydney (R.I.L.); Department of Clinical Medicine (C.D.), Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Division of Neuroimaging Sciences (G.M., J.M.W.), Centre for Clinical Brain Sciences and Centre in the UK Dementia Research Institute, University of Edinburgh, United Kingdom; Department of Neurology (J.P.C., C.S.A.), Royal Prince Alfred Hospital, Sydney Health Partners, Australia; Department of Preventive Medicine and Public Health (H.A.), Faculty of Medicine, Fukuoka University, Fukuoka, Japan; Neurology Department (Y.H.), Peking University First Hospital, Beijing, China; Neurology Department (J.S.K.), Asan Medical Centre, Seoul, South Korea; Neurology and Psychiatry Department (P.M.L.), Clinica Alemana de Santiago, Facultad de Medicina Universidad del Desarrollo, Chile; Neurology Department (T.-H.L.), Chang Gung Memorial Hospital, Taipei, Taiwan; Neurology Department (C.L., M.W.P.), John Hunter Hospital and Hunter Medical Research Institute, Newcastle, Australia; Brazilian Stroke Network (S.C.M.), Hospital de Clínicas de Porto Alegre, Hospital Moinhos de Vento, Brazil; Neurology Department (J.D.P.), Christian Medical College, Ludhiana, India; Department of Neuroscience and Behavioral Sciences (O.M.P.-N.), Ribeirao Preto School of Medicine, Brazil; School of Medicine (V.K.S.), National University of Singapore and Division of Neurology (V.K.S.), National University Hospital, Singapore; Department of Cerebrovascular Disease (T.H.N.), 115 Hospital, Ho Chi Minh City, Vietnam; Shanghai Institute for Hypertension (J.W.), Rui Jin Hospital and Shanghai Jiaotong University; and Institute of Science and Technology for Brain-inspired Intelligence (C.S.A.), Fudan University, Shanghai, China
| |
Collapse
|
5
|
Li Q, Ye J, Li Z, Xiao Q, Tan S, Hu B, Jin H. The role of neutrophils in tPA thrombolysis after stroke: a malicious troublemaker. Front Immunol 2024; 15:1477669. [PMID: 39606238 PMCID: PMC11598929 DOI: 10.3389/fimmu.2024.1477669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Acute ischemic stroke represents a critical, life-threatening condition affecting the central nervous system. Intravenous thrombolysis with tissue plasminogen activator (tPA) remains a cornerstone for achieving vascular recanalization in such patients; however, its therapeutic utility is limited, with only approximately 10% of patients benefiting due to the narrow therapeutic window and significant risk of hemorrhagic transformation. Enhancing the efficacy of tPA thrombolysis is therefore imperative. Neutrophils have been identified as key modulators of thrombolytic outcomes, interacting with tPA post-stroke to influence treatment effectiveness. The binding of tPA to low-density lipoprotein receptor-related protein 1 (LRP-1) on neutrophil surfaces induces degranulation and formation of neutrophil extracellular traps (NETs). Conversely, neutrophils impede the thrombolytic action of tPA by obstructing its interaction with fibrin and activating platelets. These findings suggest that targeting neutrophils may hold promise for improving thrombolysis outcomes. This review explores the role of neutrophils in tPA-mediated thrombolysis following acute ischemic stroke, examines neutrophil-associated biomarkers, and outlines potential strategies for enhancing tPA efficacy.
Collapse
Affiliation(s)
| | | | | | | | | | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of
Science and Technology, Wuhan, China
| | - Huijuan Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of
Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Crawford AM, Lorenzi EC, Saville BR, Lewis RJ, Anderson CS. Adaptive Clinical Trials in Stroke. Stroke 2024; 55:2731-2741. [PMID: 39435555 DOI: 10.1161/strokeaha.124.046125] [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] [Indexed: 10/23/2024]
Abstract
Designing a clinical trial to evaluate the efficacy of an intervention is often complicated by uncertainty over aspects of the study population, potential treatment effects, most relevant outcomes, dropouts, and other factors. However, once participants begin to be enrolled and partial trial data become available, this level of uncertainty is reduced. Adaptive clinical trials are designed to take advantage of the accumulating data during the conduct of a trial to make changes according to prespecified decision rules to increase the likelihood of success or statistical efficiency. Common adaptive rules address early stopping for benefit or futility, sample size reestimation, adding or dropping treatment arms or altering randomization ratios, and changing the eligibility criteria to focus on responder patient subgroups. Adaptive clinical trials are gaining popularity for clinical stroke research. We provide an overview of the methods, practical considerations, challenges and limitations, and potential future role of adaptive clinical trials in advancing knowledge and practice in stroke.
Collapse
Affiliation(s)
- Amy M Crawford
- Berry Consultants LLC, Austin, TX (A.M.C., E.C.L., R.J.L.)
| | | | - Benjamin R Saville
- Adaptix Trials LLC, Austin, TX (B.R.S.)
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN (B.R.S.)
| | - Roger J Lewis
- Berry Consultants LLC, Austin, TX (A.M.C., E.C.L., R.J.L.)
- Department of Emergency Medicine, Harbor-UCLA Medical Center, Torrance, CA (R.J.L.)
| | - Craig S Anderson
- George Institute for Global Health, University of New South Wales, Sydney, Australia (C.S.A.)
- Institute for Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China (C.S.A.)
| |
Collapse
|
7
|
Monjazeb S, Chang HV, Lyden PD. Before, during, and after: An Argument for Safety and Improved Outcome of Thrombolysis in Acute Ischemic Stroke with Direct Oral Anticoagulant Treatment. Ann Neurol 2024; 96:871-886. [PMID: 39258443 PMCID: PMC11496014 DOI: 10.1002/ana.27058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/01/2024] [Accepted: 08/03/2024] [Indexed: 09/12/2024]
Abstract
Direct oral anticoagulants are the primary stroke prevention option in patients with atrial fibrillation. Anticoagulant use before stroke, however, might inhibit clinician comfort with thrombolysis if a stroke does occur. Resuming anticoagulants after ischemic stroke is also problematic for fear of hemorrhage. We describe extensive literature showing that thrombolysis is safe after stroke with direct anticoagulant use. Early reinstitution of direct anticoagulant treatment is associated with lower risk of embolic recurrence and lower hemorrhage risk. The use of direct anticoagulants before, during, and after thrombolysis appears to be safe and is likely to promote improved outcomes after ischemic stroke. ANN NEUROL 2024;96:871-886.
Collapse
Affiliation(s)
- Sanaz Monjazeb
- Department of Neurology, Zilkha Neurogenetic Institute of the Keck School of Medicine, 1501 San Pablo Ave, ZNI 245, Los Angeles, CA 90089
| | - Heather V. Chang
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute of the Keck School of Medicine, 1501 San Pablo Ave, ZNI 245, Los Angeles, CA 90089
| | - Patrick D. Lyden
- Department of Neurology, Zilkha Neurogenetic Institute of the Keck School of Medicine, 1501 San Pablo Ave, ZNI 245, Los Angeles, CA 90089
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute of the Keck School of Medicine, 1501 San Pablo Ave, ZNI 245, Los Angeles, CA 90089
| |
Collapse
|
8
|
Gillani SA, Ma X, Huang Y, Bains NK, Fakih R, Siddiq F, French BR, Gomez CR, Lyden PD, Qureshi AI. Effect of post thrombolytic intracerebral hemorrhage volume on 90-day outcomes in acute ischemic stroke patients. J Stroke Cerebrovasc Dis 2024; 33:107962. [PMID: 39191317 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107962] [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: 03/26/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND AND PURPOSE Post thrombolytic intracerebral hemorrhage (ICH) is associated with higher rate of death or disability in acute ischemic stroke patients. We investigated the relationship between post thrombolytic ICH volume and change in volume and death or disability at 90 days in acute ischemic stroke patients. METHODS We analyzed 110 patents recruited in the Safety Evaluation of 3K3A-APC in Ischemic Stroke (RHAPSODY) trial who received intravenous tissue plasminogen activator (tPA) followed by mechanical thrombectomy (if indicated) and 3K3A-APC or placebo. ICH volume was measured at Day 2 and Day 7 using susceptibility weighted sequence (SWI) on magnetic resonance imaging (MRI). We also calculated the post thrombolytic ICH volume change between Day 2 and Day 7. Outcomes were determined by using utility weighted modified Rankin scale (UW-mRs) at 90-days, Outcomes were determined by using utility weighted modified Rankin scale (UW-mRS) at 90 days. To minimize interpretation bias, outcome assessors were blinded to the treatment allocation and clinical data.We adjusted for age, gender, National Institutes of Health Stroke Scale (NIHSS) score (<10,10-19 and ≥20), location of hemorrhage (single basal ganglia hemorrhage, single lobar, single cerebellum, and multiple sites) in multivariate regression analysis. RESULTS A total of 88 (80%) of 110 patients had post thrombolytic ICH (mean volume 28.3 ml ± SD 62 ml). The strata of ICH volume were not associated with UW-mRs at 90 days: <20 cc (regression coefficient (RC)-0.05, p= 0.58), 20-39 cc (RC-0.22, p=0.17), or ≥40 cc (RC-0.34, p= 0.083) compared with no ICH after adjusting for potential confounders. Change in ICH mean volume 26.78 ml ±59.68, 52 had increase in volume) between Day 2 and day 7 was not associated with UW-mRS at 90 days (RC -67.71, p= 0.06). CONCLUSIONS We did not observe any independent effect of post thrombolytic ICH volume on death or disability in acute ischemic stroke patients. Although further studies must be done, our data suggest that strategies to prevent ICH expansion such as antifibrinolytic medications and reduction in ICH volume such as surgical evacuation may not reduce death or disability in acute ischemic stroke patients with post thrombolytic ICH.
Collapse
Affiliation(s)
- Syed A Gillani
- Zeenat Qureshi Stroke Institute and Department of Neurology, University of Missouri, Columbia, MO, USA.
| | - Xiaoyu Ma
- Department of Statistics, University of Missouri, Columbia, MO, USA.
| | - Yilun Huang
- Department of Statistics, University of Missouri, Columbia, MO, USA.
| | - Navpreet K Bains
- Zeenat Qureshi Stroke Institute and Department of Neurology, University of Missouri, Columbia, MO, USA.
| | - Rami Fakih
- Zeenat Qureshi Stroke Institute and Department of Neurology, University of Missouri, Columbia, MO, USA.
| | - Farhan Siddiq
- Department of Neurosurgery, University of Missouri, Columbia, MO, USA.
| | - Brandi R French
- Zeenat Qureshi Stroke Institute and Department of Neurology, University of Missouri, Columbia, MO, USA.
| | - Camilo R Gomez
- Zeenat Qureshi Stroke Institute and Department of Neurology, University of Missouri, Columbia, MO, USA.
| | - Patrick D Lyden
- Department of Physiology and Neuroscience and the Department of Neurology, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA.
| | - Adnan I Qureshi
- Zeenat Qureshi Stroke Institute and Department of Neurology, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
9
|
Gu Z, Li S, Liu J, Zhang X, Pang C, Ding L, Cao C. Protection of blood-brain barrier by endothelial DAPK1 deletion after stroke. Biochem Biophys Res Commun 2024; 724:150216. [PMID: 38851140 DOI: 10.1016/j.bbrc.2024.150216] [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: 05/28/2024] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
Death-associated protein kinase (DAPK) 1 is a critical mediator for neuronal cell death in cerebral ischemia, but its role in blood-brain barrier (BBB) disruption is incompletely understood. Here, we found that endothelial-specific deletion of Dapk1 using Tie2 Cre protected the brain of Dapk1fl/fl mice against middle cerebral artery occlusion (MCAO), characterized by mitigated Evans blue dye (EBD) extravasation, reduced infarct size and improved behavior. In vitro experiments also indicated that DAPK1 deletion inhibited oxygen-glucose deprivation (OGD)-induced tight junction alteration between cerebral endothelial cells (CECs). Mechanistically, we revealed that DAPK1-DAPK3 interaction activated cytosolic phospholipase A2 (cPLA2) in OGD-stimulated CECs. Our results thus suggest that inhibition of endothelial DAPK1 specifically prevents BBB damage after stroke.
Collapse
Affiliation(s)
- Zhijiang Gu
- Department of Neurosurgery, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian, 223300, China
| | - Shaoxun Li
- Department of Neurosurgery, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian, 223300, China
| | - Jiyu Liu
- Huaian Clinical College of Xuzhou Medical University, Huaian, 223300, China
| | - Xiaotian Zhang
- Department of Neurosurgery, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian, 223300, China
| | - Cong Pang
- Department of Neurosurgery, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian, 223300, China
| | - Lianshu Ding
- Department of Neurosurgery, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian, 223300, China.
| | - Changchun Cao
- Department of Pharmacy, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huaian, 223300, China; Huaian Clinical College of Xuzhou Medical University, Huaian, 223300, China.
| |
Collapse
|
10
|
Xu X, Chen M, Zhu D. Reperfusion and cytoprotective agents are a mutually beneficial pair in ischaemic stroke therapy: an overview of pathophysiology, pharmacological targets and candidate drugs focusing on excitotoxicity and free radical. Stroke Vasc Neurol 2024; 9:351-359. [PMID: 37832977 PMCID: PMC11420919 DOI: 10.1136/svn-2023-002671] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Stroke is the second-leading cause of death and the leading cause of disability in much of the world. In particular, China faces the greatest challenge from stroke, since the population is aged quickly. In decades of clinical trials, no neuroprotectant has had reproducible efficacy on primary clinical end points, because reperfusion is probably a necessity for neuroprotection to be clinically beneficial. Fortunately, the success of thrombolysis and endovascular thrombectomy has taken us into a reperfusion era of acute ischaemic stroke (AIS) therapy. Brain cytoprotective agents can prevent detrimental effects of ischaemia, and therefore 'freeze' ischaemic penumbra before reperfusion, extend the time window for reperfusion therapy. Because reperfusion often leads to reperfusion injury, including haemorrhagic transformation, brain oedema, infarct progression and neurological worsening, cytoprotective agents will enhance the efficacy and safety of reperfusion therapy by preventing or reducing reperfusion injuries. Therefore, reperfusion and cytoprotective agents are a mutually beneficial pair in AIS therapy. In this review, we outline critical pathophysiological events causing cell death within the penumbra after ischaemia or ischaemia/reperfusion in the acute phase of AIS, focusing on excitotoxicity and free radicals. We discuss key pharmacological targets for cytoprotective therapy and evaluate the recent advances of cytoprotective agents going through clinical trials, highlighting multitarget cytoprotective agents that intervene at multiple levels of the ischaemic and reperfusion cascade.
Collapse
Affiliation(s)
- Xiumei Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mingyu Chen
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dongya Zhu
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
11
|
Verbout NG, Lorentz CU, Markway BD, Wallisch M, Marbury TC, Di Cera E, Shatzel JJ, Gruber A, Tucker EI. Safety and tolerability of the protein C activator AB002 in end-stage renal disease patients on hemodialysis: a randomized phase 2 trial. COMMUNICATIONS MEDICINE 2024; 4:153. [PMID: 39060370 PMCID: PMC11282208 DOI: 10.1038/s43856-024-00575-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND The protein C system regulates blood coagulation, inflammation, and vascular integrity. AB002 is an injectable protein C activating enzyme under investigation to safely prevent and treat thrombosis. In preclinical models, AB002 is antithrombotic, cytoprotective, and anti-inflammatory. Since prophylactic use of heparin is contraindicated during hemodialysis in some end-stage renal disease (ESRD) patients, we propose using AB002 as a short-acting alternative to safely limit blood loss due to clotting in the dialysis circuit. METHODS This phase 2, randomized, double-blind, placebo-controlled, single-dose study evaluates the safety and tolerability of AB002 administered into the hemodialysis line of ESRD patients during hemodialysis at one study center in the United States (ClinicalTrials.gov: NCT03963895). In this study, 36 patients were sequentially enrolled into two cohorts and randomized to AB002 or placebo in a 2:1 ratio. In cohort 1, patients received 1.5 µg/kg AB002 (n = 12) or placebo (n = 6); in cohort 2, patients received 3 µg/kg AB002 (n = 12) or placebo (n = 6). Patients underwent five heparin-free hemodialysis sessions over 10 days and were dosed with AB002 or placebo during session four. RESULTS Here we show that AB002 is safe and well-tolerated in ESRD patients, with no treatment-related adverse events. Clinically relevant bleeding did not occur in any patient, and the time to hemostasis at the vascular access sites is not affected by AB002. CONCLUSIONS As far as we are aware, this proof-of-concept study is the first clinical trial assessing the therapeutic potential of protein C activation. The results herein support additional investigation of AB002 to safely prevent and treat thrombosis in at-risk populations.
Collapse
Affiliation(s)
- Norah G Verbout
- Aronora, Inc., Portland, OR, USA.
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.
| | - Christina U Lorentz
- Aronora, Inc., Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | | | - Michael Wallisch
- Aronora, Inc., Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | | | - Enrico Di Cera
- Edward A. Doisy Department of Biochemistry and Molecular Biology, School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Joseph J Shatzel
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | | | - Erik I Tucker
- Aronora, Inc., Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
12
|
Arnalich-Montiel A, Burgos-Santamaría A, Pazó-Sayós L, Quintana-Villamandos B. Comprehensive Management of Stroke: From Mechanisms to Therapeutic Approaches. Int J Mol Sci 2024; 25:5252. [PMID: 38791292 PMCID: PMC11120719 DOI: 10.3390/ijms25105252] [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: 03/10/2024] [Revised: 04/29/2024] [Accepted: 05/05/2024] [Indexed: 05/26/2024] Open
Abstract
Acute ischemic stroke (AIS) is a challenging disease, which needs urgent comprehensive management. Endovascular thrombectomy (EVT), alone or combined with iv thrombolysis, is currently the most effective therapy for patients with acute ischemic stroke (AIS). However, only a limited number of patients are eligible for this time-sensitive treatment. Even though there is still significant room for improvement in the management of this group of patients, up until now there have been no alternative therapies approved for use in clinical practice. However, there is still hope, as clinical research with novel emerging therapies is now generating promising results. These drugs happen to stop or palliate some of the underlying molecular mechanisms involved in cerebral ischemia and secondary brain damage. The aim of this review is to provide a deep understanding of these mechanisms and the pathogenesis of AIS. Later, we will discuss the potential therapies that have already demonstrated, in preclinical or clinical studies, to improve the outcomes of patients with AIS.
Collapse
Affiliation(s)
- Ana Arnalich-Montiel
- Department of Anaesthesia and Intensive Care, Gregorio Marañón’s University Hospital, 28007 Madrid, Spain; (A.B.-S.); (B.Q.-V.)
- Department of Pharmacology, College of Medicine, Complutense University, 28040 Madrid, Spain
| | - Alba Burgos-Santamaría
- Department of Anaesthesia and Intensive Care, Gregorio Marañón’s University Hospital, 28007 Madrid, Spain; (A.B.-S.); (B.Q.-V.)
| | - Laia Pazó-Sayós
- Department of Anaesthesia and Intensive Care, Gregorio Marañón’s University Hospital, 28007 Madrid, Spain; (A.B.-S.); (B.Q.-V.)
| | - Begoña Quintana-Villamandos
- Department of Anaesthesia and Intensive Care, Gregorio Marañón’s University Hospital, 28007 Madrid, Spain; (A.B.-S.); (B.Q.-V.)
- Department of Pharmacology, College of Medicine, Complutense University, 28040 Madrid, Spain
| |
Collapse
|
13
|
Fang H, Bo Y, Hao Z, Mang G, Jin J, Wang H. A promising frontier: targeting NETs for stroke treatment breakthroughs. Cell Commun Signal 2024; 22:238. [PMID: 38654328 PMCID: PMC11036592 DOI: 10.1186/s12964-024-01563-4] [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: 11/22/2023] [Accepted: 03/07/2024] [Indexed: 04/25/2024] Open
Abstract
Stroke is a prevalent global acute cerebrovascular condition, with ischaemic stroke being the most frequently occurring type. After a stroke, neutrophils accumulate in the brain and subsequently generate and release neutrophil extracellular traps (NETs). The accumulation of NETs exacerbates the impairment of the blood‒brain barrier (BBB), hampers neovascularization, induces notable neurological deficits, worsens the prognosis of stroke patients, and can facilitate the occurrence of t-PA-induced cerebral haemorrhage subsequent to ischaemic stroke. Alternative approaches to pharmacological thrombolysis or endovascular thrombectomy are being explored, and targeting NETs is a promising treatment that warrants further investigation.
Collapse
Affiliation(s)
- Huijie Fang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Yunfei Bo
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Zhongfei Hao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Ge Mang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiaqi Jin
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Hongjun Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
14
|
Fraser JF, Pahwa S, Maniskas M, Michas C, Martinez M, Pennypacker KR, Dornbos D. Now that the door is open: an update on ischemic stroke pharmacotherapeutics for the neurointerventionalist. J Neurointerv Surg 2024; 16:425-428. [PMID: 37258227 DOI: 10.1136/jnis-2022-019293] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/22/2023] [Indexed: 06/02/2023]
Abstract
The last 10 years have seen a major shift in management of large vessel ischemic stroke with changes towards ever-expanding use of reperfusion therapies (intravenous thrombolysis and mechanical thrombectomy). These strategies 'open the door' to acute therapeutics for ischemic tissue, and we should investigate novel therapeutic approaches to enhance survival of recently reperfused brain. Key insights into new approaches have been provided through translational research models and preclinical paradigms, and through detailed research on ischemic mechanisms. Additional recent clinical trials offer exciting salvos into this new strategy of pairing reperfusion with neuroprotective therapy. This pairing strategy can be employed using drugs that have shown neuroprotective efficacy; neurointerventionalists can administer these during or immediately after reperfusion therapy. This represents a crucial moment when we emphasize reperfusion, and have the technological capability along with the clinical trial experience to lead the way in multiprong approaches to stroke treatment.
Collapse
Affiliation(s)
- Justin F Fraser
- Department of Neurological Surgery, University of Kentucky, Lexington, Kentucky, USA
- Department of Neurology, University of Kentucky, Lexington, Kentucky, USA
- Department of Radiology, University of Kentucky, Lexington, Kentucky, USA
| | - Shivani Pahwa
- Department of Neurological Surgery, University of Kentucky, Lexington, Kentucky, USA
- Department of Radiology, University of Kentucky, Lexington, Kentucky, USA
| | - Michael Maniskas
- Department of Neurology, The University of Texas Health Science Center at Houston John P and Katherine G McGovern Medical School, Houston, Texas, USA
| | - Christopher Michas
- Department of Neurological Surgery, University of Kentucky, Lexington, Kentucky, USA
| | - Mesha Martinez
- Department of Neurointerventional Radiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Keith R Pennypacker
- Department of Neurology, University of Kentucky, Lexington, Kentucky, USA
- University of Kentucky, Lexington, Kentucky, USA
| | - David Dornbos
- Department of Neurological Surgery, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
15
|
Jung JM, Gruber A, Heseltine P, Rajamani K, Ameriso SF, Fisher MJ. New Directions in Infection-Associated Ischemic Stroke. J Clin Neurol 2024; 20:140-152. [PMID: 38330416 PMCID: PMC10921058 DOI: 10.3988/jcn.2023.0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 09/06/2023] [Accepted: 11/12/2023] [Indexed: 02/10/2024] Open
Abstract
The relationship between infections and stroke has not been fully characterized, probably delaying the development of specific treatments. This narrative review addresses mechanisms of stroke linked to infections, including hypercoagulability, endothelial dysfunction, vasculitis, and impaired thrombolysis. SARS-CoV-2, the virus that causes COVID-19, may promote the development of stroke, which may represent its most severe neurological complication. The development of specific therapies for infection-associated stroke remains a profound challenge. Perhaps the most important remaining issue is the distinction between infections that trigger a stroke versus infections that are truly incidental. This distinction likely requires the establishment of appropriate biomarkers, candidates of which are elevated levels of fibrin D-dimer and anticardiolipin/antiphospholipid antibodies. These candidate biomarkers might have potential use in identifying pathogenic infections preceding stroke, which is a precursor to establishing specific therapies for this syndrome.
Collapse
Affiliation(s)
- Jin-Man Jung
- Department of Neurology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea; Korea University Zebrafish, Translational Medical Research Center, Ansan, Korea
| | | | - Peter Heseltine
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Kumar Rajamani
- Department of Neurology, Wayne State University-Detroit Medical Center, Detroit, MI, USA
| | - Sebastián F Ameriso
- Division of Vascular Neurology, Department of Neurology, Fleni, Autonomous City of Buenos Aires, Argentina
| | - Mark J Fisher
- Department of Neurology, University of California Irvine Medical Center, Orange, CA, USA.
| |
Collapse
|
16
|
Fatmi MK, Wang H, Slotabec L, Wen C, Seale B, Zhao B, Li J. Single-Cell RNA-seq reveals transcriptomic modulation of Alzheimer's disease by activated protein C. Aging (Albany NY) 2024; 16:3137-3159. [PMID: 38385967 PMCID: PMC10929801 DOI: 10.18632/aging.205624] [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: 06/27/2023] [Accepted: 01/09/2024] [Indexed: 02/23/2024]
Abstract
Single-Cell RNA sequencing reveals changes in cell population in Alzheimer's disease (AD) model 5xFAD (5x Familial AD mutation) versus wild type (WT) mice. The returned sequencing data was processed through the 10x Genomics CellRanger platform to perform alignment and form corresponding matrix to perform bioinformatic analysis. Alterations in glial cells occurred in 5xFAD versus WT, especially increases in microglia proliferation were profound in 5xFAD. Differential expression testing of glial cells in 5xFAD versus WT revealed gene regulation. Globally, the critical genes implicated in AD progression are upregulated such as Apoe, Ctsb, Trem2, and Tyrobp. Using this differential expression data, GO term enrichment was completed to observe possible biological processes impacted by AD progression. Utilizing anti-inflammatory and cyto-protective recombinant Activated Protein C (APC), we uncover inflammatory processes to be downregulated by APC treatment in addition to recuperation of nervous system processes. Moreover, animal studies demonstrated that administration of recombinant APC significantly attenuated Aβ burden and improved cognitive function of 5xFAD mice. The downregulation of highly expressed AD biomarkers in 5xFAD could provide insight into the mechanisms by which APC administration benefits AD.
Collapse
Affiliation(s)
- Mohammad Kasim Fatmi
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Hao Wang
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Lily Slotabec
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Changhong Wen
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Blaise Seale
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Bi Zhao
- Genomics Program, College of Public Health, University of South Florida, Tampa, FL 33612, USA
| | - Ji Li
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
17
|
Harker P, Aziz YN, Vranic J, Chulluncuy‐Rivas R, Previtera M, Yaghi S, DeHavenon AH, Tsivgoulis GK, Khatri V, Mistry AM, Khatri P, Mistry EA. Asymptomatic Intracerebral Hemorrhage Following Endovascular Stroke Therapy Is Not Benign: A Systematic Review and Meta-Analysis. J Am Heart Assoc 2024; 13:e031749. [PMID: 38348800 PMCID: PMC11010099 DOI: 10.1161/jaha.123.031749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/11/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND Asymptomatic intracerebral hemorrhage (aICH) occurs in approximately 35% of patients with acute ischemic stroke after endovascular thrombectomy. Unlike symptomatic ICH, studies evaluating the effect of aICH on outcomes have been inconclusive. We performed a systematic review and meta-analysis to evaluate the long-term effects of postendovascular thrombectomy aICH. METHODS AND RESULTS The meta-analysis protocol was submitted to the International Prospective Register of Systematic Reviews a priori. PubMed, Scopus, and Web of Science were searched from inception through September 2023, yielding 312 studies. Two authors independently reviewed all abstracts. Included studies contained adult patients with ischemic stroke undergoing endovascular thrombectomy with follow-up imaging assessment of ICH reporting comparative outcomes according to aICH versus no ICH. After screening, 60 papers were fully reviewed, and 10 studies fulfilled inclusion criteria (n=5723 patients total, 1932 with aICH). Meta-analysis was performed using Cochrane RevMan v5.4. Effects were estimated by a random-effects model to estimate summary odds ratio (OR) of the effect of aICH versus no ICH on primary outcomes of 90-day modified Rankin Scale 3 to 6 and mortality. The presence of aICH was associated with a higher odds of 90-day mRS 3 to 6 (OR, 2.17 [95% CI, 1.81-2.60], P<0.0001, I2 46% Q 19.15) and mortality (OR, 1.72 [95% CI, 1.17-2.53], P:0.005, I2 79% Q 27.59) compared with no ICH. This difference was maintained following subgroup analysis according to hemorrhage classification and recanalization status. CONCLUSIONS The presence of aICH is associated with worse 90-day functional outcomes and higher mortality. Further studies to evaluate the factors predicting aICH and treatments aimed at reducing its occurrence are warranted.
Collapse
Affiliation(s)
- Pablo Harker
- Department of Neurology and Rehabilitation MedicineUniversity of CincinnatiCincinnatiOH
| | - Yasmin N. Aziz
- Department of Neurology and Rehabilitation MedicineUniversity of CincinnatiCincinnatiOH
| | - Justin Vranic
- Department of NeurosurgeryMassachusetts General HospitalBostonMA
| | | | - Melissa Previtera
- Department of Neurology and Rehabilitation MedicineUniversity of CincinnatiCincinnatiOH
| | - Shadi Yaghi
- Department of NeurologyBrown UniversityProvidenceRI
| | | | | | | | | | - Pooja Khatri
- Department of Neurology and Rehabilitation MedicineUniversity of CincinnatiCincinnatiOH
| | - Eva A. Mistry
- Department of Neurology and Rehabilitation MedicineUniversity of CincinnatiCincinnatiOH
| |
Collapse
|
18
|
Mavridis T, Choratta T, Papadopoulou A, Sawafta A, Archontakis-Barakakis P, Laou E, Sakellakis M, Chalkias A. Protease-Activated Receptors (PARs): Biology and Therapeutic Potential in Perioperative Stroke. Transl Stroke Res 2024:10.1007/s12975-024-01233-0. [PMID: 38326662 DOI: 10.1007/s12975-024-01233-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/12/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024]
Abstract
Perioperative stroke is a devastating complication that occurs during surgery or within 30 days following the surgical procedure. Its prevalence ranges from 0.08 to 10% although it is most likely an underestimation, as sedatives and narcotics can substantially mask symptomatology and clinical presentation. Understanding the underlying pathophysiology and identifying potential therapeutic targets are of paramount importance. Protease-activated receptors (PARs), a unique family of G-protein-coupled receptors, are widely expressed throughout the human body and play essential roles in various physiological and pathological processes. This review elucidates the biology and significance of PARs, outlining their diverse functions in health and disease, and their intricate involvement in cerebrovascular (patho)physiology and neuroprotection. PARs exhibit a dual role in cerebral ischemia, which underscores their potential as therapeutic targets to mitigate the devastating effects of stroke in surgical patients.
Collapse
Affiliation(s)
- Theodoros Mavridis
- Department of Neurology, Tallaght University Hospital (TUH)/The Adelaide and Meath Hospital, Dublin, incorporating the National Children's Hospital (AMNCH), Dublin, D24 NR0A, Ireland
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528, Athens, Greece
| | - Theodora Choratta
- Department of General Surgery, Metaxa Hospital, 18537, Piraeus, Greece
| | - Androniki Papadopoulou
- Department of Anesthesiology, G. Gennimatas General Hospital, 54635, Thessaloniki, Greece
| | - Assaf Sawafta
- Department of Cardiology, University Hospital of Larisa, 41110, Larisa, Greece
| | | | - Eleni Laou
- Department of Anesthesiology, Agia Sophia Children's Hospital, 15773, Athens, Greece
| | - Minas Sakellakis
- Department of Medicine, Jacobi Medical Center-North Central Bronx Hospital, Bronx, NY, 10467, USA
| | - Athanasios Chalkias
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104-5158, USA.
- Outcomes Research Consortium, Cleveland, OH, 44195, USA.
| |
Collapse
|
19
|
Xue M, Jackson CJ, Lin H, Zhao R, Liang HPH, Weiler H, Griffin JH, March L. Endothelial Protein C Receptor and 3K3A-Activated Protein C Protect Mice from Allergic Contact Dermatitis in a Contact Hypersensitivity Model. Int J Mol Sci 2024; 25:1255. [PMID: 38279255 PMCID: PMC10816322 DOI: 10.3390/ijms25021255] [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: 12/10/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/28/2024] Open
Abstract
Endothelial protein C receptor (EPCR) is a receptor for the natural anti-coagulant activated protein C (aPC). It mediates the anti-inflammatory and barrier-protective functions of aPC through the cleavage of protease-activated receptor (PAR)1/2. Allergic contact dermatitis is a common skin disease characterized by inflammation and defective skin barrier. This study investigated the effect of EPCR and 3K3A-aPC on allergic contact dermatitis using a contact hypersensitivity (CHS) model. CHS was induced using 1-Fluoro-2,4-dinitrobenzene in EPCR-deficient (KO) and matched wild-type mice and mice treated with 3K3A-aPC, a mutant form of aPC with diminished anti-coagulant activity. Changes in clinical and histological features, cytokines, and immune cells were examined. EPCRKO mice displayed more severe CHS, with increased immune cell infiltration in the skin and higher levels of inflammatory cytokines and IgE than wild-type mice. EPCR, aPC, and PAR1/2 were expressed by the skin epidermis, with EPCR presenting almost exclusively in the basal layer. EPCRKO increased the epidermal expression of aPC and PAR1, whereas in CHS, their expression was reduced compared to wild-type mice. 3K3A-aPC reduced CHS severity in wild-type and EPCRKO mice by suppressing immune cell infiltration/activation and inflammatory cytokines. In summary, EPCRKO exacerbated CHS, whereas 3K3A-aPC could reduce the severity of CHS in both EPCRKO and wild-type mice.
Collapse
Affiliation(s)
- Meilang Xue
- Sutton Arthritis Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2065, Australia; (H.L.); (R.Z.); (H.P.H.L.)
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2065, Australia;
| | - Christopher J. Jackson
- Sutton Arthritis Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2065, Australia; (H.L.); (R.Z.); (H.P.H.L.)
| | - Haiyan Lin
- Sutton Arthritis Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2065, Australia; (H.L.); (R.Z.); (H.P.H.L.)
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2065, Australia;
| | - Ruilong Zhao
- Sutton Arthritis Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2065, Australia; (H.L.); (R.Z.); (H.P.H.L.)
| | - Hai Po H. Liang
- Sutton Arthritis Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2065, Australia; (H.L.); (R.Z.); (H.P.H.L.)
| | - Hartmut Weiler
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA;
- Department of Physiology, The Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA;
| | - Lyn March
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2065, Australia;
| |
Collapse
|
20
|
Dammavalam V, Lin S, Nessa S, Daksla N, Stefanowski K, Costa A, Bergese S. Neuroprotection during Thrombectomy for Acute Ischemic Stroke: A Review of Future Therapies. Int J Mol Sci 2024; 25:891. [PMID: 38255965 PMCID: PMC10815099 DOI: 10.3390/ijms25020891] [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: 11/30/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Stroke is a major cause of death and disability worldwide. Endovascular thrombectomy has been impactful in decreasing mortality. However, many clinical results continue to show suboptimal functional outcomes despite high recanalization rates. This gap in recanalization and symptomatic improvement suggests a need for adjunctive therapies in post-thrombectomy care. With greater insight into ischemia-reperfusion injury, recent preclinical testing of neuroprotective agents has shifted towards preventing oxidative stress through upregulation of antioxidants and downstream effectors, with positive results. Advances in multiple neuroprotective therapies, including uric acid, activated protein C, nerinetide, otaplimastat, imatinib, verapamil, butylphthalide, edaravone, nelonemdaz, ApTOLL, regional hypothermia, remote ischemic conditioning, normobaric oxygen, and especially nuclear factor erythroid 2-related factor 2, have promising evidence for improving stroke care. Sedation and blood pressure management in endovascular thrombectomy also play crucial roles in improved stroke outcomes. A hand-in-hand approach with both endovascular therapy and neuroprotection may be the key to targeting disability due to stroke.
Collapse
Affiliation(s)
- Vikalpa Dammavalam
- Department of Neurology, Stony Brook University Hospital, Stony Brook, NY 11794, USA; (V.D.); (K.S.)
| | - Sandra Lin
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA; (S.L.); (N.D.); (A.C.)
| | - Sayedatun Nessa
- Department of Neurology, Stony Brook University Hospital, Stony Brook, NY 11794, USA; (V.D.); (K.S.)
| | - Neil Daksla
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA; (S.L.); (N.D.); (A.C.)
| | - Kamil Stefanowski
- Department of Neurology, Stony Brook University Hospital, Stony Brook, NY 11794, USA; (V.D.); (K.S.)
| | - Ana Costa
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA; (S.L.); (N.D.); (A.C.)
| | - Sergio Bergese
- Department of Anesthesiology, Stony Brook University Hospital, Stony Brook, NY 11794, USA; (S.L.); (N.D.); (A.C.)
| |
Collapse
|
21
|
Thielen O, Mitra S, Debot M, Schaid T, Hallas W, Gallagher LT, Erickson C, Cralley A, Stafford P, Silliman C, D'Alessandro A, Hansen K, Sauaia A, Moore E, Mosnier L, Griffin J, Cohen M. Mitigation of trauma-induced endotheliopathy by activated protein C: A potential therapeutic for postinjury thromboinflammation. J Trauma Acute Care Surg 2024; 96:116-122. [PMID: 37733304 PMCID: PMC10841096 DOI: 10.1097/ta.0000000000004142] [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] [Indexed: 09/22/2023]
Abstract
BACKGROUND Activated Protein C (aPC) plays dual roles after injury, driving both trauma-induced coagulopathy (TIC) by cleaving, and thus inactivating, factors Va and VIIIa and depressing fibrinolysis while also mediating an inflammomodulatory milieu via protease activated receptor-1 (PAR-1) cytoprotective signaling. Because of this dual role, it represents and ideal target for study and therapeutics after trauma. A known aPC variant, 3K3A-aPC, has been engineered to preserve cytoprotective activity while retaining minimal anticoagulant activity rendering it potentially ideal as a cytoprotective therapeutic after trauma. We hypothesized that 3K3A-aPC would mitigate the endotheliopathy of trauma by protecting against endothelial permeability. METHODS We used electric cell-substrate impedance sensing to measure permeability changes in real time in primary endothelial cells. These were cultured, grown to confluence, and treated with a 2 μg/mL solution of 3K3A-aPC at 180 minutes, 120 minutes, 60 minutes, 30 minutes prior to stimulation with ex vivo plasma taken from severely injured trauma patients (Injury Severity Score > 15 and BD < -6) (trauma plasma [TP]). Cells treated with thrombin and untreated cells were included in this study as control groups. Permeability changes were recorded in real time via electric cell-substrate impedance sensing for 30 minutes after treatment with TP. We quantified permeability changes in the control and treatment groups as area under the curve (AUC). Rac1/RhoA activity was also compared between these groups. Statistical significance was determined by one-way ANOVA followed by a post hoc analysis using Tukey's multiple comparison's test. RESULTS Treatment with aPC mitigated endothelial permeability induced by ex vivo trauma plasma at all pre-treatment time points. The AUC of the 30-minute 3K3A-aPC pretreatment group was higher than TP alone (mean diff. 22.12 95% CI [13.75, 30.49], p < 0.0001) (Figure). Moreover, the AUC of the 60-minute, 120-minute, and 180-minute pretreatment groups was also higher than TP alone (mean diff., 16.30; 95% confidence interval [CI], 7.93-24.67; 19.43; 95% CI, 11.06-27.80, and 18.65; 95% CI, 10.28-27.02;, all p < 0.0001, respectively). Rac1/RhoA activity was higher in the aPC pretreatment group when compared with all other groups ( p < 0.01). CONCLUSION Pretreatment with 3K3A-aPC, which retains its cytoprotective function but has only ~5% of its anticoagulant function, abrogates the effects of trauma-induced endotheliopathy. This represents a potential therapeutic treatment for dysregulated thromboinflammation for injured patients by minimizing aPC's role in trauma-induced coagulopathy while concurrently amplifying its essential cytoprotective function. LEVEL OF EVIDENCE Prognostic and Epidemiological; Level III.
Collapse
Affiliation(s)
- Otto Thielen
- From the Department of Gastrointestinal, Trauma, and Endocrine Surgery (O.T., S.M., M.D., T.S., W.H., L.T.G., C.E., A.C., P.S., C.S., A.D'A., K.H., A.S., E.M., M.C.), University of Colorado, Denver, Colorado; Department of Surgery (A.S., E.M.), Denver Health Medical Center, Denver, Colorado; and Department of Molecular Medicine (L.M., J.G.), Scripps Research, La Jolla, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Ronaldson PT, Williams EI, Betterton RD, Stanton JA, Nilles KL, Davis TP. CNS Drug Delivery in Stroke: Improving Therapeutic Translation From the Bench to the Bedside. Stroke 2024; 55:190-202. [PMID: 38134249 PMCID: PMC10752297 DOI: 10.1161/strokeaha.123.043764] [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] [Indexed: 12/24/2023]
Abstract
Drug development for ischemic stroke is challenging as evidenced by the paucity of therapeutics that have advanced beyond a phase III trial. There are many reasons for this lack of clinical translation including factors related to the experimental design of preclinical studies. Often overlooked in therapeutic development for ischemic stroke is the requirement of effective drug delivery to the brain, which is critical for neuroprotective efficacy of several small and large molecule drugs. Advancing central nervous system drug delivery technologies implies a need for detailed comprehension of the blood-brain barrier (BBB) and neurovascular unit. Such knowledge will permit the innate biology of the BBB/neurovascular unit to be leveraged for improved bench-to-bedside translation of novel stroke therapeutics. In this review, we will highlight key aspects of BBB/neurovascular unit pathophysiology and describe state-of-the-art approaches for optimization of central nervous system drug delivery (ie, passive diffusion, mechanical opening of the BBB, liposomes/nanoparticles, transcytosis, intranasal drug administration). Additionally, we will discuss how endogenous BBB transporters represent the next frontier of drug delivery strategies for stroke. Overall, this review will provide cutting edge perspective on how central nervous system drug delivery must be considered for the advancement of new stroke drugs toward human trials.
Collapse
Affiliation(s)
- Patrick T Ronaldson
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| | - Erica I Williams
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| | - Robert D Betterton
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| | - Joshua A Stanton
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| | - Kelsy L Nilles
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| | - Thomas P Davis
- Department of Pharmacology, College of Medicine (P.T.R., E.I.C., R.D.B., J.A.S., T.P.D.) and Graduate Interdisciplinary Program in Neuroscience (P.T.R., K.L.N., T.P.D.), University of Arizona, Tucson
| |
Collapse
|
23
|
Zhao R, Xue M, Lin H, Smith M, Liang H, Weiler H, Griffin JH, Jackson CJ. A recombinant signalling-selective activated protein C that lacks anticoagulant activity is efficacious and safe in cutaneous wound preclinical models. Wound Repair Regen 2024; 32:90-103. [PMID: 38155595 DOI: 10.1111/wrr.13148] [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: 06/24/2023] [Revised: 12/09/2023] [Accepted: 12/18/2023] [Indexed: 12/30/2023]
Abstract
Various preclinical and clinical studies have demonstrated the robust wound healing capacity of the natural anticoagulant activated protein C (APC). A bioengineered APC variant designated 3K3A-APC retains APC's cytoprotective cell signalling actions with <10% anticoagulant activity. This study was aimed to provide preclinical evidence that 3K3A-APC is efficacious and safe as a wound healing agent. 3K3A-APC, like wild-type APC, demonstrated positive effects on proliferation of human skin cells (keratinocytes, endothelial cells and fibroblasts). Similarly it also increased matrix metollaproteinase-2 activation in keratinocytes and fibroblasts. Topical 3K3A-APC treatment at 10 or 30 μg both accelerated mouse wound healing when culled on Day 11. And at 10 μg, it was superior to APC and had half the dermal wound gape compared to control. Further testing was conducted in excisional porcine wounds due to their congruence to human skin. Here, 3K3A-APC advanced macroscopic healing in a dose-dependent manner (100, 250 and 500 μg) when culled on Day 21. This was histologically corroborated by greater collagen maturity, suggesting more advanced remodelling. A non-interference arm of this study found no evidence that topical 3K3A-APC caused either any significant systemic side-effects or any significant leakage into the circulation. However the female pigs exhibited transient and mild local reactions after treatments in week three, which did not impact healing. Overall these preclinical studies support the hypothesis that 3K3A-APC merits future human wound studies.
Collapse
Affiliation(s)
- Ruilong Zhao
- Sutton Laboratory, Kolling Institute of Medical Research, Sydney, New South Wales, Australia
| | - Meilang Xue
- Sutton Laboratory, Kolling Institute of Medical Research, Sydney, New South Wales, Australia
| | - Haiyan Lin
- Sutton Laboratory, Kolling Institute of Medical Research, Sydney, New South Wales, Australia
| | - Margaret Smith
- Raymond Purves Laboratory, Kolling Institute of Medical Research, Sydney, New South Wales, Australia
| | - Helena Liang
- Sutton Laboratory, Kolling Institute of Medical Research, Sydney, New South Wales, Australia
| | - Hartmut Weiler
- Department of Physiology, Blood Research Institute, Milwaukee, Wisconsin, USA
| | - John H Griffin
- Department of Molecular Medicine, The Scripps Research Institute, San Diego, California, USA
| | - Christopher J Jackson
- Sutton Laboratory, Kolling Institute of Medical Research, Sydney, New South Wales, Australia
| |
Collapse
|
24
|
Schleicher RL, Vorasayan P, McCabe ME, Bevers MB, Davis TP, Griffin JH, Hinduja A, Jadhav AP, Lee JM, Sawyer RN, Zlokovic BV, Sheth KN, Fedler JK, Lyden P, Kimberly WT. Analysis of brain edema in RHAPSODY. Int J Stroke 2024; 19:68-75. [PMID: 37382409 PMCID: PMC10789908 DOI: 10.1177/17474930231187268] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
BACKGROUND Cerebral edema is a secondary complication of acute ischemic stroke, but its time course and imaging markers are not fully understood. Recently, net water uptake (NWU) has been proposed as a novel marker of edema. AIMS Studying the RHAPSODY trial cohort, we sought to characterize the time course of edema and test the hypothesis that NWU provides distinct information when added to traditional markers of cerebral edema after stroke by examining its association with other markers. METHODS A total of 65 patients had measurable supratentorial ischemic lesions. Patients underwent head computed tomography (CT), brain magnetic resonance imaging (MRI) scans, or both at the baseline visit and after 2, 7, 30, and 90 days following enrollment. CT and MRI scans were used to measure four imaging markers of edema: midline shift (MLS), hemisphere volume ratio (HVR), cerebrospinal fluid (CSF) volume, and NWU using semi-quantitative threshold analysis. Trajectories of the markers were summarized, as available. Correlations of the markers of edema were computed and the markers compared by clinical outcome. Regression models were used to examine the effect of 3K3A-activated protein C (APC) treatment. RESULTS Two measures of mass effect, MLS and HVR, could be measured on all imaging modalities, and had values available across all time points. Accordingly, mass effect reached a maximum level by day 7, normalized by day 30, and then reversed by day 90 for both measures. In the first 2 days after stroke, the change in CSF volume was associated with MLS (ρ = -0.57, p = 0.0001) and HVR (ρ = -0.66, p < 0.0001). In contrast, the change in NWU was not associated with the other imaging markers (all p ⩾ 0.49). While being directionally consistent, we did not observe a difference in the edema markers by clinical outcome. In addition, baseline stroke volume was associated with all markers (MLS (p < 0.001), HVR (p < 0.001), change in CSF volume (p = 0.003)) with the exception of NWU (p = 0.5). Exploratory analysis did not reveal a difference in cerebral edema markers by treatment arm. CONCLUSIONS Existing cerebral edema imaging markers potentially describe two distinct processes, including lesional water concentration (i.e. NWU) and mass effect (MLS, HVR, and CSF volume). These two types of imaging markers may represent distinct aspects of cerebral edema, which could be useful for future trials targeting this process.
Collapse
Affiliation(s)
- Riana L. Schleicher
- Division of Neurocritical Care and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Pongpat Vorasayan
- Division of Neurocritical Care and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Neurology, Department of Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Megan E. McCabe
- Department of Biostatistics, University of Iowa, Iowa City, IA, USA
| | - Matthew B. Bevers
- Divisions of Stroke, Cerebrovascular and Critical Care Neurology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Thomas P. Davis
- Department of Pharmacology, University of Arizona Health Sciences, Tucson, AZ, USA
| | - John H. Griffin
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, USA
| | - Archana Hinduja
- Department of Neurology, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Jin-Moo Lee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Robert N. Sawyer
- Department of Neurology, University of Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | - Berislav V. Zlokovic
- Department of Physiology & Neuroscience, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Kevin N. Sheth
- Division of Neurocritical Care, Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Janel K. Fedler
- Department of Biostatistics, University of Iowa, Iowa City, IA, USA
| | - Patrick Lyden
- Department of Physiology & Neuroscience, Keck School of Medicine of USC, Los Angeles, CA, USA
- Department of Neurology, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - W. Taylor Kimberly
- Division of Neurocritical Care and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
25
|
O'Donnell JS, Fleming H, Noone D, Preston RJS. Unraveling coagulation factor-mediated cellular signaling. J Thromb Haemost 2023; 21:3342-3353. [PMID: 37391097 DOI: 10.1016/j.jtha.2023.06.019] [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: 03/02/2023] [Revised: 05/15/2023] [Accepted: 06/12/2023] [Indexed: 07/02/2023]
Abstract
Blood coagulation is initiated in response to blood vessel injury or proinflammatory stimuli, which activate coagulation factors to coordinate complex biochemical and cellular responses necessary for clot formation. In addition to these critical physiologic functions, plasma protein factors activated during coagulation mediate a spectrum of signaling responses via receptor-binding interactions on different cell types. In this review, we describe examples and mechanisms of coagulation factor signaling. We detail the molecular basis for cell signaling mediated by coagulation factor proteases via the protease-activated receptor family, considering new insights into the role of protease-specific cleavage sites, cofactor and coreceptor interactions, and distinct signaling intermediate interactions in shaping protease-activated receptor signaling diversity. Moreover, we discuss examples of how injury-dependent conformational activation of other coagulation proteins, such as fibrin(ogen) and von Willebrand factor, decrypts their signaling potential, unlocking their capacity to contribute to aberrant proinflammatory signaling. Finally, we consider the role of coagulation factor signaling in disease development and the status of pharmacologic approaches to either attenuate or enhance coagulation factor signaling for therapeutic benefit, emphasizing new approaches to inhibit deleterious coagulation factor signaling without impacting hemostatic activity.
Collapse
Affiliation(s)
- James S O'Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland; National Children's Research Centre, Children's Health Ireland, Crumlin, Dublin, Ireland. https://twitter.com/profJSOdonnell
| | - Harry Fleming
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland. https://www.twitter.com/PrestonLab_RCSI
| | - David Noone
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland. https://www.twitter.com/PrestonLab_RCSI
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland; National Children's Research Centre, Children's Health Ireland, Crumlin, Dublin, Ireland.
| |
Collapse
|
26
|
Wechsler LR, Adeoye O, Alemseged F, Bahr-Hosseini M, Deljkich E, Favilla C, Fisher M, Grotta J, Hill MD, Kamel H, Khatri P, Lyden P, Mirza M, Nguyen TN, Samaniego E, Schwamm L, Selim M, Silva G, Yavagal DR, Yenari MA, Zachrison KS, Boltze J, Yaghi S. Most Promising Approaches to Improve Stroke Outcomes: The Stroke Treatment Academic Industry Roundtable XII Workshop. Stroke 2023; 54:3202-3213. [PMID: 37886850 DOI: 10.1161/strokeaha.123.044279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023]
Abstract
The Stroke Treatment Academic Industry Roundtable XII included a workshop to discuss the most promising approaches to improve outcome from acute stroke. The workshop brought together representatives from academia, industry, and government representatives. The discussion examined approaches in 4 epochs: pre-reperfusion, reperfusion, post-reperfusion, and access to acute stroke interventions. The participants identified areas of priority for developing new and existing treatments and approaches to improve stroke outcomes. Although many advances in acute stroke therapy have been achieved, more work is necessary for reperfusion therapies to benefit the most possible patients. Prioritization of promising approaches should help guide the use of resources and investigator efforts.
Collapse
Affiliation(s)
- Lawrence R Wechsler
- University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, PA (L.R.W.)
| | - Opeolu Adeoye
- Washington University School of Medicine, St. Louis, MO (O.A.)
| | | | | | | | | | - Marc Fisher
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (M.F.)
| | | | | | - Hooman Kamel
- Weill Cornel School of Medicine, New York, NY (H.K.)
| | - Pooja Khatri
- University of Cincinnati Medical Center, OH (P.K.)
| | - Patrick Lyden
- University of Southern California, Los Angeles, CA (P.L.)
| | | | | | | | - Lee Schwamm
- Massachusetts General Hospital, Boston (L.S.)
| | - Magdy Selim
- Beth Israel Deaconess Medical Center, Boston, MA (M.S.)
| | | | | | | | | | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, United Kingdom (J.B.)
| | | |
Collapse
|
27
|
Misra S, Khan EI, Lam TT, Mazumder R, Gururangan K, Hickman LB, Goswami V, Funaro MC, Eldem E, Sansing LH, Sico JJ, Quinn TJ, Liebeskind DS, Montaner J, Kwan P, Mishra NK. Common Pathways of Epileptogenesis in Patients With Epilepsy Post-Brain Injury: Findings From a Systematic Review and Meta-analysis. Neurology 2023; 101:e2243-e2256. [PMID: 37550071 PMCID: PMC10727219 DOI: 10.1212/wnl.0000000000207749] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 09/13/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Epilepsy may result from various brain injuries, including stroke (ischemic and hemorrhagic), traumatic brain injury, and infections. Identifying shared common biological pathways and biomarkers of the epileptogenic process initiated by the different injuries may lead to novel targets for preventing the development of epilepsy. We systematically reviewed biofluid biomarkers to test their association with the risk of post-brain injury epilepsy. METHODS We searched articles until January 25, 2022, in MEDLINE, Embase, PsycInfo, Web of Science, and Cochrane. The primary outcome was the difference in mean biomarker levels in patients with and without post-brain injury epilepsy. We used the modified quality score on prognostic studies for risk of bias assessment. We calculated each biomarker's pooled standardized mean difference (SMD) and 95% CI. Molecular interaction network and enrichment analyses were conducted in Cytoscape (PROSPERO CRD42021297110). RESULTS We included 22 studies with 1,499 cases with post-brain injury epilepsy and 7,929 controls without post-brain injury epilepsy. Forty-five biomarkers in the blood or CSF were investigated with samples collected at disparate time points. Of 22 studies, 21 had a moderate-to-high risk of bias. Most of the biomarkers (28/45) were investigated in single studies; only 9 provided validation data, and studies used variable definitions for early-onset and late-onset seizures. A meta-analysis was possible for 19 biomarkers. Blood glucose levels in 4 studies were significantly higher in patients with poststroke epilepsy (PSE) than those without PSE (SMD 0.44; CI 0.19-0.69). From individual studies, 15 biomarkers in the blood and 7 in the CSF were significantly associated with post-brain injury epilepsy. Enrichment analysis identified that the significant biomarkers (interleukin [IL]-6, IL-1β]) were predominantly inflammation related. DISCUSSION We cannot yet recommend using the reported biomarkers for designing antiepileptogenesis trials or use in the clinical setting because of methodological heterogeneity, bias in the included studies, and insufficient validation studies. Although our analyses indicate the plausible role of inflammation in epileptogenesis, this is likely not the only mechanism. For example, an individual's genetic susceptibilities might contribute to his/her risk of epileptogenesis after brain injury. Rigorously designed biomarker studies with methods acceptable to the regulatory bodies should be conducted.
Collapse
Affiliation(s)
- Shubham Misra
- From the Department of Neurology (S.M., E.E., L.H.S., J.J.S., N.K.M.), Yale University School of Medicine, New Haven, CT; Medical School (E.I.K.), B.J. Medical College and Civil Hospital, Ahmedabad, India; Keck MS & Proteomics Resource (T.T.L.), Yale University School of Medicine, New Haven, CT; Department of Neurology (R.M., K.G., L.B.H., D.S.L.), University of California, Los Angeles; Department of Neurology (K.G.), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (V.G.), Tower Health, Philadelphia, PA; Harvey Cushing/John Hay Whitney Medical Library (M.C.F.), Yale University, New Haven, CT; Department of Immunobiology (L.H.S.), Yale University School of Medicine, New Haven, CT; Institute of Cardiovascular and Medical Sciences (T.J.Q.), University of Glasgow, Scotland, UK; Institute de Biomedicine of Seville (J.M.), IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville; Department of Neurology (J.M.), Hospital Universitario Virgen Macarena, Seville; Neurovascular Research Laboratory Vall d'Hebron Institute of Research (VHIR) (J.M.), Barcelona, Spain; and Department of Neuroscience (P.K.), Central Clinical School, Monash University, Melbourne, Australia
| | - Erum I Khan
- From the Department of Neurology (S.M., E.E., L.H.S., J.J.S., N.K.M.), Yale University School of Medicine, New Haven, CT; Medical School (E.I.K.), B.J. Medical College and Civil Hospital, Ahmedabad, India; Keck MS & Proteomics Resource (T.T.L.), Yale University School of Medicine, New Haven, CT; Department of Neurology (R.M., K.G., L.B.H., D.S.L.), University of California, Los Angeles; Department of Neurology (K.G.), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (V.G.), Tower Health, Philadelphia, PA; Harvey Cushing/John Hay Whitney Medical Library (M.C.F.), Yale University, New Haven, CT; Department of Immunobiology (L.H.S.), Yale University School of Medicine, New Haven, CT; Institute of Cardiovascular and Medical Sciences (T.J.Q.), University of Glasgow, Scotland, UK; Institute de Biomedicine of Seville (J.M.), IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville; Department of Neurology (J.M.), Hospital Universitario Virgen Macarena, Seville; Neurovascular Research Laboratory Vall d'Hebron Institute of Research (VHIR) (J.M.), Barcelona, Spain; and Department of Neuroscience (P.K.), Central Clinical School, Monash University, Melbourne, Australia
| | - TuKiet T Lam
- From the Department of Neurology (S.M., E.E., L.H.S., J.J.S., N.K.M.), Yale University School of Medicine, New Haven, CT; Medical School (E.I.K.), B.J. Medical College and Civil Hospital, Ahmedabad, India; Keck MS & Proteomics Resource (T.T.L.), Yale University School of Medicine, New Haven, CT; Department of Neurology (R.M., K.G., L.B.H., D.S.L.), University of California, Los Angeles; Department of Neurology (K.G.), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (V.G.), Tower Health, Philadelphia, PA; Harvey Cushing/John Hay Whitney Medical Library (M.C.F.), Yale University, New Haven, CT; Department of Immunobiology (L.H.S.), Yale University School of Medicine, New Haven, CT; Institute of Cardiovascular and Medical Sciences (T.J.Q.), University of Glasgow, Scotland, UK; Institute de Biomedicine of Seville (J.M.), IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville; Department of Neurology (J.M.), Hospital Universitario Virgen Macarena, Seville; Neurovascular Research Laboratory Vall d'Hebron Institute of Research (VHIR) (J.M.), Barcelona, Spain; and Department of Neuroscience (P.K.), Central Clinical School, Monash University, Melbourne, Australia
| | - Rajarshi Mazumder
- From the Department of Neurology (S.M., E.E., L.H.S., J.J.S., N.K.M.), Yale University School of Medicine, New Haven, CT; Medical School (E.I.K.), B.J. Medical College and Civil Hospital, Ahmedabad, India; Keck MS & Proteomics Resource (T.T.L.), Yale University School of Medicine, New Haven, CT; Department of Neurology (R.M., K.G., L.B.H., D.S.L.), University of California, Los Angeles; Department of Neurology (K.G.), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (V.G.), Tower Health, Philadelphia, PA; Harvey Cushing/John Hay Whitney Medical Library (M.C.F.), Yale University, New Haven, CT; Department of Immunobiology (L.H.S.), Yale University School of Medicine, New Haven, CT; Institute of Cardiovascular and Medical Sciences (T.J.Q.), University of Glasgow, Scotland, UK; Institute de Biomedicine of Seville (J.M.), IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville; Department of Neurology (J.M.), Hospital Universitario Virgen Macarena, Seville; Neurovascular Research Laboratory Vall d'Hebron Institute of Research (VHIR) (J.M.), Barcelona, Spain; and Department of Neuroscience (P.K.), Central Clinical School, Monash University, Melbourne, Australia
| | - Kapil Gururangan
- From the Department of Neurology (S.M., E.E., L.H.S., J.J.S., N.K.M.), Yale University School of Medicine, New Haven, CT; Medical School (E.I.K.), B.J. Medical College and Civil Hospital, Ahmedabad, India; Keck MS & Proteomics Resource (T.T.L.), Yale University School of Medicine, New Haven, CT; Department of Neurology (R.M., K.G., L.B.H., D.S.L.), University of California, Los Angeles; Department of Neurology (K.G.), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (V.G.), Tower Health, Philadelphia, PA; Harvey Cushing/John Hay Whitney Medical Library (M.C.F.), Yale University, New Haven, CT; Department of Immunobiology (L.H.S.), Yale University School of Medicine, New Haven, CT; Institute of Cardiovascular and Medical Sciences (T.J.Q.), University of Glasgow, Scotland, UK; Institute de Biomedicine of Seville (J.M.), IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville; Department of Neurology (J.M.), Hospital Universitario Virgen Macarena, Seville; Neurovascular Research Laboratory Vall d'Hebron Institute of Research (VHIR) (J.M.), Barcelona, Spain; and Department of Neuroscience (P.K.), Central Clinical School, Monash University, Melbourne, Australia
| | - L Brian Hickman
- From the Department of Neurology (S.M., E.E., L.H.S., J.J.S., N.K.M.), Yale University School of Medicine, New Haven, CT; Medical School (E.I.K.), B.J. Medical College and Civil Hospital, Ahmedabad, India; Keck MS & Proteomics Resource (T.T.L.), Yale University School of Medicine, New Haven, CT; Department of Neurology (R.M., K.G., L.B.H., D.S.L.), University of California, Los Angeles; Department of Neurology (K.G.), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (V.G.), Tower Health, Philadelphia, PA; Harvey Cushing/John Hay Whitney Medical Library (M.C.F.), Yale University, New Haven, CT; Department of Immunobiology (L.H.S.), Yale University School of Medicine, New Haven, CT; Institute of Cardiovascular and Medical Sciences (T.J.Q.), University of Glasgow, Scotland, UK; Institute de Biomedicine of Seville (J.M.), IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville; Department of Neurology (J.M.), Hospital Universitario Virgen Macarena, Seville; Neurovascular Research Laboratory Vall d'Hebron Institute of Research (VHIR) (J.M.), Barcelona, Spain; and Department of Neuroscience (P.K.), Central Clinical School, Monash University, Melbourne, Australia
| | - Vaibhav Goswami
- From the Department of Neurology (S.M., E.E., L.H.S., J.J.S., N.K.M.), Yale University School of Medicine, New Haven, CT; Medical School (E.I.K.), B.J. Medical College and Civil Hospital, Ahmedabad, India; Keck MS & Proteomics Resource (T.T.L.), Yale University School of Medicine, New Haven, CT; Department of Neurology (R.M., K.G., L.B.H., D.S.L.), University of California, Los Angeles; Department of Neurology (K.G.), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (V.G.), Tower Health, Philadelphia, PA; Harvey Cushing/John Hay Whitney Medical Library (M.C.F.), Yale University, New Haven, CT; Department of Immunobiology (L.H.S.), Yale University School of Medicine, New Haven, CT; Institute of Cardiovascular and Medical Sciences (T.J.Q.), University of Glasgow, Scotland, UK; Institute de Biomedicine of Seville (J.M.), IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville; Department of Neurology (J.M.), Hospital Universitario Virgen Macarena, Seville; Neurovascular Research Laboratory Vall d'Hebron Institute of Research (VHIR) (J.M.), Barcelona, Spain; and Department of Neuroscience (P.K.), Central Clinical School, Monash University, Melbourne, Australia
| | - Melissa C Funaro
- From the Department of Neurology (S.M., E.E., L.H.S., J.J.S., N.K.M.), Yale University School of Medicine, New Haven, CT; Medical School (E.I.K.), B.J. Medical College and Civil Hospital, Ahmedabad, India; Keck MS & Proteomics Resource (T.T.L.), Yale University School of Medicine, New Haven, CT; Department of Neurology (R.M., K.G., L.B.H., D.S.L.), University of California, Los Angeles; Department of Neurology (K.G.), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (V.G.), Tower Health, Philadelphia, PA; Harvey Cushing/John Hay Whitney Medical Library (M.C.F.), Yale University, New Haven, CT; Department of Immunobiology (L.H.S.), Yale University School of Medicine, New Haven, CT; Institute of Cardiovascular and Medical Sciences (T.J.Q.), University of Glasgow, Scotland, UK; Institute de Biomedicine of Seville (J.M.), IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville; Department of Neurology (J.M.), Hospital Universitario Virgen Macarena, Seville; Neurovascular Research Laboratory Vall d'Hebron Institute of Research (VHIR) (J.M.), Barcelona, Spain; and Department of Neuroscience (P.K.), Central Clinical School, Monash University, Melbourne, Australia
| | - Ece Eldem
- From the Department of Neurology (S.M., E.E., L.H.S., J.J.S., N.K.M.), Yale University School of Medicine, New Haven, CT; Medical School (E.I.K.), B.J. Medical College and Civil Hospital, Ahmedabad, India; Keck MS & Proteomics Resource (T.T.L.), Yale University School of Medicine, New Haven, CT; Department of Neurology (R.M., K.G., L.B.H., D.S.L.), University of California, Los Angeles; Department of Neurology (K.G.), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (V.G.), Tower Health, Philadelphia, PA; Harvey Cushing/John Hay Whitney Medical Library (M.C.F.), Yale University, New Haven, CT; Department of Immunobiology (L.H.S.), Yale University School of Medicine, New Haven, CT; Institute of Cardiovascular and Medical Sciences (T.J.Q.), University of Glasgow, Scotland, UK; Institute de Biomedicine of Seville (J.M.), IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville; Department of Neurology (J.M.), Hospital Universitario Virgen Macarena, Seville; Neurovascular Research Laboratory Vall d'Hebron Institute of Research (VHIR) (J.M.), Barcelona, Spain; and Department of Neuroscience (P.K.), Central Clinical School, Monash University, Melbourne, Australia
| | - Lauren H Sansing
- From the Department of Neurology (S.M., E.E., L.H.S., J.J.S., N.K.M.), Yale University School of Medicine, New Haven, CT; Medical School (E.I.K.), B.J. Medical College and Civil Hospital, Ahmedabad, India; Keck MS & Proteomics Resource (T.T.L.), Yale University School of Medicine, New Haven, CT; Department of Neurology (R.M., K.G., L.B.H., D.S.L.), University of California, Los Angeles; Department of Neurology (K.G.), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (V.G.), Tower Health, Philadelphia, PA; Harvey Cushing/John Hay Whitney Medical Library (M.C.F.), Yale University, New Haven, CT; Department of Immunobiology (L.H.S.), Yale University School of Medicine, New Haven, CT; Institute of Cardiovascular and Medical Sciences (T.J.Q.), University of Glasgow, Scotland, UK; Institute de Biomedicine of Seville (J.M.), IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville; Department of Neurology (J.M.), Hospital Universitario Virgen Macarena, Seville; Neurovascular Research Laboratory Vall d'Hebron Institute of Research (VHIR) (J.M.), Barcelona, Spain; and Department of Neuroscience (P.K.), Central Clinical School, Monash University, Melbourne, Australia
| | - Jason J Sico
- From the Department of Neurology (S.M., E.E., L.H.S., J.J.S., N.K.M.), Yale University School of Medicine, New Haven, CT; Medical School (E.I.K.), B.J. Medical College and Civil Hospital, Ahmedabad, India; Keck MS & Proteomics Resource (T.T.L.), Yale University School of Medicine, New Haven, CT; Department of Neurology (R.M., K.G., L.B.H., D.S.L.), University of California, Los Angeles; Department of Neurology (K.G.), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (V.G.), Tower Health, Philadelphia, PA; Harvey Cushing/John Hay Whitney Medical Library (M.C.F.), Yale University, New Haven, CT; Department of Immunobiology (L.H.S.), Yale University School of Medicine, New Haven, CT; Institute of Cardiovascular and Medical Sciences (T.J.Q.), University of Glasgow, Scotland, UK; Institute de Biomedicine of Seville (J.M.), IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville; Department of Neurology (J.M.), Hospital Universitario Virgen Macarena, Seville; Neurovascular Research Laboratory Vall d'Hebron Institute of Research (VHIR) (J.M.), Barcelona, Spain; and Department of Neuroscience (P.K.), Central Clinical School, Monash University, Melbourne, Australia
| | - Terence J Quinn
- From the Department of Neurology (S.M., E.E., L.H.S., J.J.S., N.K.M.), Yale University School of Medicine, New Haven, CT; Medical School (E.I.K.), B.J. Medical College and Civil Hospital, Ahmedabad, India; Keck MS & Proteomics Resource (T.T.L.), Yale University School of Medicine, New Haven, CT; Department of Neurology (R.M., K.G., L.B.H., D.S.L.), University of California, Los Angeles; Department of Neurology (K.G.), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (V.G.), Tower Health, Philadelphia, PA; Harvey Cushing/John Hay Whitney Medical Library (M.C.F.), Yale University, New Haven, CT; Department of Immunobiology (L.H.S.), Yale University School of Medicine, New Haven, CT; Institute of Cardiovascular and Medical Sciences (T.J.Q.), University of Glasgow, Scotland, UK; Institute de Biomedicine of Seville (J.M.), IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville; Department of Neurology (J.M.), Hospital Universitario Virgen Macarena, Seville; Neurovascular Research Laboratory Vall d'Hebron Institute of Research (VHIR) (J.M.), Barcelona, Spain; and Department of Neuroscience (P.K.), Central Clinical School, Monash University, Melbourne, Australia
| | - David S Liebeskind
- From the Department of Neurology (S.M., E.E., L.H.S., J.J.S., N.K.M.), Yale University School of Medicine, New Haven, CT; Medical School (E.I.K.), B.J. Medical College and Civil Hospital, Ahmedabad, India; Keck MS & Proteomics Resource (T.T.L.), Yale University School of Medicine, New Haven, CT; Department of Neurology (R.M., K.G., L.B.H., D.S.L.), University of California, Los Angeles; Department of Neurology (K.G.), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (V.G.), Tower Health, Philadelphia, PA; Harvey Cushing/John Hay Whitney Medical Library (M.C.F.), Yale University, New Haven, CT; Department of Immunobiology (L.H.S.), Yale University School of Medicine, New Haven, CT; Institute of Cardiovascular and Medical Sciences (T.J.Q.), University of Glasgow, Scotland, UK; Institute de Biomedicine of Seville (J.M.), IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville; Department of Neurology (J.M.), Hospital Universitario Virgen Macarena, Seville; Neurovascular Research Laboratory Vall d'Hebron Institute of Research (VHIR) (J.M.), Barcelona, Spain; and Department of Neuroscience (P.K.), Central Clinical School, Monash University, Melbourne, Australia
| | - Joan Montaner
- From the Department of Neurology (S.M., E.E., L.H.S., J.J.S., N.K.M.), Yale University School of Medicine, New Haven, CT; Medical School (E.I.K.), B.J. Medical College and Civil Hospital, Ahmedabad, India; Keck MS & Proteomics Resource (T.T.L.), Yale University School of Medicine, New Haven, CT; Department of Neurology (R.M., K.G., L.B.H., D.S.L.), University of California, Los Angeles; Department of Neurology (K.G.), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (V.G.), Tower Health, Philadelphia, PA; Harvey Cushing/John Hay Whitney Medical Library (M.C.F.), Yale University, New Haven, CT; Department of Immunobiology (L.H.S.), Yale University School of Medicine, New Haven, CT; Institute of Cardiovascular and Medical Sciences (T.J.Q.), University of Glasgow, Scotland, UK; Institute de Biomedicine of Seville (J.M.), IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville; Department of Neurology (J.M.), Hospital Universitario Virgen Macarena, Seville; Neurovascular Research Laboratory Vall d'Hebron Institute of Research (VHIR) (J.M.), Barcelona, Spain; and Department of Neuroscience (P.K.), Central Clinical School, Monash University, Melbourne, Australia
| | - Patrick Kwan
- From the Department of Neurology (S.M., E.E., L.H.S., J.J.S., N.K.M.), Yale University School of Medicine, New Haven, CT; Medical School (E.I.K.), B.J. Medical College and Civil Hospital, Ahmedabad, India; Keck MS & Proteomics Resource (T.T.L.), Yale University School of Medicine, New Haven, CT; Department of Neurology (R.M., K.G., L.B.H., D.S.L.), University of California, Los Angeles; Department of Neurology (K.G.), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (V.G.), Tower Health, Philadelphia, PA; Harvey Cushing/John Hay Whitney Medical Library (M.C.F.), Yale University, New Haven, CT; Department of Immunobiology (L.H.S.), Yale University School of Medicine, New Haven, CT; Institute of Cardiovascular and Medical Sciences (T.J.Q.), University of Glasgow, Scotland, UK; Institute de Biomedicine of Seville (J.M.), IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville; Department of Neurology (J.M.), Hospital Universitario Virgen Macarena, Seville; Neurovascular Research Laboratory Vall d'Hebron Institute of Research (VHIR) (J.M.), Barcelona, Spain; and Department of Neuroscience (P.K.), Central Clinical School, Monash University, Melbourne, Australia
| | - Nishant K Mishra
- From the Department of Neurology (S.M., E.E., L.H.S., J.J.S., N.K.M.), Yale University School of Medicine, New Haven, CT; Medical School (E.I.K.), B.J. Medical College and Civil Hospital, Ahmedabad, India; Keck MS & Proteomics Resource (T.T.L.), Yale University School of Medicine, New Haven, CT; Department of Neurology (R.M., K.G., L.B.H., D.S.L.), University of California, Los Angeles; Department of Neurology (K.G.), Icahn School of Medicine at Mount Sinai, New York, NY; Department of Neurology (V.G.), Tower Health, Philadelphia, PA; Harvey Cushing/John Hay Whitney Medical Library (M.C.F.), Yale University, New Haven, CT; Department of Immunobiology (L.H.S.), Yale University School of Medicine, New Haven, CT; Institute of Cardiovascular and Medical Sciences (T.J.Q.), University of Glasgow, Scotland, UK; Institute de Biomedicine of Seville (J.M.), IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville; Department of Neurology (J.M.), Hospital Universitario Virgen Macarena, Seville; Neurovascular Research Laboratory Vall d'Hebron Institute of Research (VHIR) (J.M.), Barcelona, Spain; and Department of Neuroscience (P.K.), Central Clinical School, Monash University, Melbourne, Australia.
| |
Collapse
|
28
|
de Araújo CN, Santiago PB, Causin Vieira G, Silva GDS, Moura RP, Bastos IMD, de Santana JM. The biotechnological potential of proteases from hematophagous arthropod vectors. Front Cell Infect Microbiol 2023; 13:1287492. [PMID: 37965257 PMCID: PMC10641018 DOI: 10.3389/fcimb.2023.1287492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/09/2023] [Indexed: 11/16/2023] Open
Affiliation(s)
- Carla Nunes de Araújo
- Host-Pathogen Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, DF, Brazil
- Faculty of Ceilândia, University of Brasília, Brasília, DF, Brazil
| | - Paula Beatriz Santiago
- Host-Pathogen Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, DF, Brazil
| | - Giulia Causin Vieira
- Host-Pathogen Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, DF, Brazil
| | - Gabriel dos Santos Silva
- Host-Pathogen Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, DF, Brazil
| | - Renan Pereira Moura
- Host-Pathogen Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, DF, Brazil
| | - Izabela Marques Dourado Bastos
- Host-Pathogen Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, DF, Brazil
| | - Jaime Martins de Santana
- Host-Pathogen Interface Laboratory, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, DF, Brazil
| |
Collapse
|
29
|
Liu Q, Shi K, Wang Y, Shi FD. Neurovascular Inflammation and Complications of Thrombolysis Therapy in Stroke. Stroke 2023; 54:2688-2697. [PMID: 37675612 DOI: 10.1161/strokeaha.123.044123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Intravenous thrombolysis via tPA (tissue-type plasminogen activator) is the only approved pharmacological treatment for acute ischemic stroke, but its benefits are limited by hemorrhagic transformation. Emerging evidence reveals that tPA swiftly mobilizes immune cells which extravasate into the brain parenchyma via the cerebral vasculature, augmenting neurovascular inflammation, and tissue injury. In this review, we summarize the pronounced alterations of immune cells induced by tPA in patients with stroke and experimental stroke models. We argue that neuroinflammation, triggered by ischemia-induced cell death and exacerbated by tPA, compromises neurovascular integrity and the microcirculation, leading to hemorrhagic transformation. Finally, we discuss current and future approaches to attenuate thrombolysis-associated hemorrhagic transformation via uncoupling immune cells from the neurovascular unit.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Neurology, Tianjin Medical University General Hospital, China (Q.L., F.-D.S.)
| | - Kaibin Shi
- Department of Neurology, National Clinical Research Center for Neurological Diseases of China, Beijing Tiantan Hospital, Capital Medical University (K.S., Y.W., F.-D.S.)
| | - Yongjun Wang
- Department of Neurology, National Clinical Research Center for Neurological Diseases of China, Beijing Tiantan Hospital, Capital Medical University (K.S., Y.W., F.-D.S.)
| | - Fu-Dong Shi
- Department of Neurology, Tianjin Medical University General Hospital, China (Q.L., F.-D.S.)
- Department of Neurology, National Clinical Research Center for Neurological Diseases of China, Beijing Tiantan Hospital, Capital Medical University (K.S., Y.W., F.-D.S.)
| |
Collapse
|
30
|
Sim DS, Shukla M, Mallari CR, Fernández JA, Xu X, Schneider D, Bauzon M, Hermiston TW, Mosnier LO. Selective modulation of activated protein C activities by a nonactive site-targeting nanobody library. Blood Adv 2023; 7:3036-3048. [PMID: 36735416 PMCID: PMC10331410 DOI: 10.1182/bloodadvances.2022008740] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 01/13/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023] Open
Abstract
Activated protein C (APC) is a pleiotropic coagulation protease with anticoagulant, anti-inflammatory, and cytoprotective activities. Selective modulation of these APC activities contributes to our understanding of the regulation of these physiological mechanisms and permits the development of therapeutics for the pathologies associated with these pathways. An antibody library targeting the nonactive site of APC was generated using llama antibodies (nanobodies). Twenty-one nanobodies were identified that selectively recognize APC compared with the protein C zymogen. Overall, 3 clusters of nanobodies were identified based on the competition for APC in biolayer interferometry studies. APC functional assays for anticoagulant activity, histone H3 cleavage, and protease-activated receptor 1 (PAR1) cleavage were used to understand their diversity. These functional assays revealed 13 novel nanobody-induced APC activity profiles via the selective modulation of APC pleiotropic activities, with the potential to regulate specific mechanisms for therapeutic purposes. Within these, 3 nanobodies (LP2, LP8, and LP17) inhibited all 3 APC functions. Four nanobodies (LP1, LP5, LP16, and LP20) inhibited only 2 of the 3 functions. Monofunction inhibition specific to APC anticoagulation activity was observed only by 2 nanobodies (LP9 and LP11). LP11 was also found to shift the ratio of APC cleavage of PAR1 at R46 relative to R41, which results in APC-mediated biased PAR1 signaling and APC cytoprotective effects. Thus, LP11 has an activity profile that could potentially promote hemostasis and cytoprotection in bleedings associated with hemophilia or coagulopathy by selectively modulating APC anticoagulation and PAR1 cleavage profile.
Collapse
Affiliation(s)
- Derek S. Sim
- Coagulant Therapeutics Corporation, Berkeley, CA
| | - Meenal Shukla
- Department of Molecular Medicine, Scripps Research, La Jolla, CA
| | | | | | - Xiao Xu
- Department of Molecular Medicine, Scripps Research, La Jolla, CA
| | | | - Maxine Bauzon
- Consultants for Coagulant Therapeutics, Berkeley, CA
| | | | | |
Collapse
|
31
|
Weinberger Y, Budnik I, Nisgav Y, Palevski D, Ben-David G, Fernández JA, Margalit SN, Levy-Mendelovich S, Kenet G, Weinberger D, Griffin JH, Livnat T. 3K3A-Activated Protein C Inhibits Choroidal Neovascularization Growth and Leakage and Reduces NLRP3 Inflammasome, IL-1β, and Inflammatory Cell Accumulation in the Retina. Int J Mol Sci 2023; 24:10642. [PMID: 37445820 PMCID: PMC10341424 DOI: 10.3390/ijms241310642] [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: 05/25/2023] [Revised: 06/20/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
3K3A-Activated Protein C (APC) is a recombinant variant of the physiological anticoagulant APC with cytoprotective properties and reduced bleeding risks. We studied the potential use of 3K3A-APC as a multi-target therapeutic option for choroidal neovascularization (CNV), a common cause of vision loss in age-related macular degeneration. CNV was induced by laser photocoagulation in a murine model, and 3K3A-APC was intravitreally injected. The impact of 3K3A-APC treatment on myeloid and microglia cell activation and recruitment and on NLRP3 inflammasome, IL-1β, and VEGF levels was assessed using cryosection, retinal flat-mount immunohistochemistry and vascular imaging. Additionally, we evaluated the use of fluorescein angiography as a surrogate marker for in vivo evaluation of the efficacy of 3K3A-APC treatment against leaking CNV lesions. Our results demonstrated that 3K3A-APC treatment significantly reduced the accumulation and activation of myeloid cells and microglia in the CNV area and decreased the NLRP3 and IL-1β levels at the CNV site and the surrounding retina. Furthermore, 3K3A-APC treatment resulted in leakage regression and CNV growth suppression. These findings indicate that the anti-inflammatory activities of 3K3A-APC contribute to CNV inhibition. Our study suggests the potential use of 3K3A-APC as a novel multi-target treatment for CNV.
Collapse
Affiliation(s)
- Yehonatan Weinberger
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research Felsenstein Medical Research Center, Petah-Tikva 5251108, Israel
- Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Ivan Budnik
- Department of Internal Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - Yael Nisgav
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research Felsenstein Medical Research Center, Petah-Tikva 5251108, Israel
| | - Dahlia Palevski
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research Felsenstein Medical Research Center, Petah-Tikva 5251108, Israel
- Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Gil Ben-David
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research Felsenstein Medical Research Center, Petah-Tikva 5251108, Israel
- Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - José A. Fernández
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Shany Nivinsky Margalit
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research Felsenstein Medical Research Center, Petah-Tikva 5251108, Israel
| | - Sarina Levy-Mendelovich
- Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
- Sheba Medical Center, The Amalia Biron Thrombosis and Hemostasis Research Institute, Tel-Hashomer 52621, Israel
| | - Gili Kenet
- Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
- Sheba Medical Center, The Amalia Biron Thrombosis and Hemostasis Research Institute, Tel-Hashomer 52621, Israel
| | - Dov Weinberger
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research Felsenstein Medical Research Center, Petah-Tikva 5251108, Israel
- Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tami Livnat
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research Felsenstein Medical Research Center, Petah-Tikva 5251108, Israel
- Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
- Sheba Medical Center, The Amalia Biron Thrombosis and Hemostasis Research Institute, Tel-Hashomer 52621, Israel
| |
Collapse
|
32
|
Schneider AM, Regenhardt RW, Dmytriw AA, Patel AB, Hirsch JA, Buchan AM. Cerebroprotection in the endovascular era: an update. J Neurol Neurosurg Psychiatry 2023; 94:267-271. [PMID: 36600581 DOI: 10.1136/jnnp-2022-330379] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 11/19/2022] [Indexed: 12/12/2022]
Abstract
Despite advances in clinical diagnosis and increasing numbers of patients eligible for revascularisation, ischaemic stroke remains a significant public health concern accounting for 3.3 million deaths annually. In addition to recanalisation therapy, patient outcomes could be improved through cerebroprotection, but all translational attempts have remained unsuccessful. In this narrative review, we discuss potential reasons for those failures. We then outline the diverse, multicellular effects of ischaemic stroke and the complex temporal sequences of the pathophysiological cascade during and following ischaemia, reperfusion, and recovery. This evidence is linked with findings from prior cerebroprotective trials and interpreted for the modern endovascular era. Future cerebroprotective agents that are multimodal and multicellular, promoting cellular and metabolic health to different targets at time points that are most responsive to treatment, might prove more successful.
Collapse
Affiliation(s)
- Anna M Schneider
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.,Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Robert W Regenhardt
- Neuroendovascular Program, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Adam A Dmytriw
- Neuroendovascular Program, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aman B Patel
- Neuroendovascular Program, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joshua Adam Hirsch
- Neuroendovascular Program, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alastair M Buchan
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
33
|
Pajor MJ, Adeoye OM. Evolving Stroke Systems of Care: Stroke Diagnosis and Treatment in the Post-Thrombectomy Era. Neurotherapeutics 2023; 20:655-663. [PMID: 36977818 PMCID: PMC10047478 DOI: 10.1007/s13311-023-01371-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Thrombectomy became the gold-standard treatment of acute ischemic stroke caused by large-vessel occlusions (LVO) in 2015 after five clinical trials published that year demonstrated significantly improved patient outcomes. In subsequent years, advances in stroke systems of care have centered around improving access to and expanding patient eligibility for thrombectomy. The prehospital and acute stroke treatment settings have had the greatest emphasis. Numerous prehospital stroke scales now provide emergency medical services with focused physical exams to identify LVOs, and many devices to non-invasively detect LVO are undergoing clinical testing. Mobile stroke units deployed throughout Western Europe and the USA also show promising results by bringing elements of acute stroke care directly to the patient. Numerous clinical trials since 2015 have aimed to increase candidates for thrombectomy by expanding indications and the eligibility time window. Further optimizations of thrombectomy treatment have focused on the role of thrombolytics and other adjunctive therapies that may promote neuroprotection and neurorecovery. While many of these approaches require further clinical investigation, the next decade shows significant potential for further advances in stroke care.
Collapse
Affiliation(s)
- Michael J. Pajor
- Department of Emergency Medicine, Washington University School of Medicine, 660 S. Euclid Ave, Campus Box 8072, St. Louis, MO 63110 USA
| | - Opeolu M. Adeoye
- Department of Emergency Medicine, Washington University School of Medicine, 660 S. Euclid Ave, Campus Box 8072, St. Louis, MO 63110 USA
| |
Collapse
|
34
|
Thrombolysis for acute ischaemic stroke: current status and future perspectives. Lancet Neurol 2023; 22:418-429. [PMID: 36907201 DOI: 10.1016/s1474-4422(22)00519-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 03/14/2023]
Abstract
Alteplase is currently the only approved thrombolytic agent for treatment of acute ischaemic stroke, but interest is burgeoning in the development of new thrombolytic agents for systemic reperfusion with an improved safety profile, increased efficacy, and convenient delivery. Tenecteplase has emerged as a potential alternative thrombolytic agent that might be preferred over alteplase because of its ease of administration and reported efficacy in patients with large vessel occlusion. Ongoing research efforts are also looking at potential improvements in recanalisation with the use of adjunct therapies to intravenous thrombolysis. New treatment strategies are also emerging that aim to reduce the risk of vessel reocclusion after intravenous thrombolysis administration. Other research endeavors are looking at the use of intra-arterial thrombolysis after mechanical thrombectomy to induce tissue reperfusion. The growing implementation of mobile stroke units and advanced neuroimaging could boost the number of patients who can receive intravenous thrombolysis by shortening onset-to-treatment times and identifying patients with salvageable penumbra. Continued improvements in this area will be essential to facilitate the ongoing research endeavors and to improve delivery of new interventions.
Collapse
|
35
|
Neuroprotective Strategies for Ischemic Stroke-Future Perspectives. Int J Mol Sci 2023; 24:ijms24054334. [PMID: 36901765 PMCID: PMC10002358 DOI: 10.3390/ijms24054334] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
Ischemic stroke is the main cause of death and the most common cause of acquired physical disability worldwide. Recent demographic changes increase the relevance of stroke and its sequelae. The acute treatment for stroke is restricted to causative recanalization and restoration of cerebral blood flow, including both intravenous thrombolysis and mechanical thrombectomy. Still, only a limited number of patients are eligible for these time-sensitive treatments. Hence, new neuroprotective approaches are urgently needed. Neuroprotection is thus defined as an intervention resulting in the preservation, recovery, and/or regeneration of the nervous system by interfering with the ischemic-triggered stroke cascade. Despite numerous preclinical studies generating promising data for several neuroprotective agents, successful bench-to-bedside translations are still lacking. The present study provides an overview of current approaches in the research field of neuroprotective stroke treatment. Aside from "traditional" neuroprotective drugs focusing on inflammation, cell death, and excitotoxicity, stem-cell-based treatment methods are also considered. Furthermore, an overview of a prospective neuroprotective method using extracellular vesicles that are secreted from various stem cell sources, including neural stem cells and bone marrow stem cells, is also given. The review concludes with a short discussion on the microbiota-gut-brain axis that may serve as a potential target for future neuroprotective therapies.
Collapse
|
36
|
Morais A, Locascio JJ, Sansing LH, Lamb J, Nagarkatti K, Imai T, van Leyen K, Aronowski J, Koenig JI, Bosetti F, Lyden P, Ayata C. Embracing Heterogeneity in The Multicenter Stroke Preclinical Assessment Network (SPAN) Trial. Stroke 2023; 54:620-631. [PMID: 36601951 PMCID: PMC9870939 DOI: 10.1161/strokeaha.122.040638] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The Stroke Preclinical Assessment Network (SPAN) is a multicenter preclinical trial platform using rodent models of transient focal cerebral ischemia to address translational failure in experimental stroke. In addition to centralized randomization and blinding and large samples, SPAN aimed to introduce heterogeneity to simulate the heterogeneity embodied in clinical trials for robust conclusions. Here, we report the heterogeneity introduced by allowing the 6 SPAN laboratories to vary most of the biological and experimental model variables and the impact of this heterogeneity on middle cerebral artery occlusion (MCAo) performance. We included the modified intention-to-treat population of the control mouse cohort of the first SPAN trial (n=421) and examined the biological and procedural independent variables and their covariance. We then determined their impact on the dependent variables cerebral blood flow drop during MCAo, time to achieve MCAo, and total anesthesia duration using multivariable analyses. We found heterogeneity in biological and procedural independent variables introduced mainly by the site. Consequently, all dependent variables also showed heterogeneity among the sites. Multivariable analyses with the site as a random effect variable revealed filament choice as an independent predictor of cerebral blood flow drop after MCAo. Comorbidity, sex, use of laser Doppler flow to monitor cerebral blood flow, days after trial onset, and maintaining anesthesia throughout the MCAo emerged as independent predictors of time to MCAo. Total anesthesia duration was predicted by most independent variables. We present with high granularity the heterogeneity introduced by the biological and model selections by the testing sites in the first trial of cerebroprotection in rodent transient filament MCAo by SPAN. Rather than trying to homogenize all variables across all sites, we embraced the heterogeneity to better approximate clinical trials. Awareness of the heterogeneity, its sources, and how it impacts the study performance may further improve the study design and statistical modeling for future multicenter preclinical trials.
Collapse
Affiliation(s)
- Andreia Morais
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Joseph J. Locascio
- Department of Biostatistics, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Lauren H. Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, CT USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT USA
| | - Jessica Lamb
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Los Angeles, CA USA
| | - Karisma Nagarkatti
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Los Angeles, CA USA
| | - Takahiko Imai
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Klaus van Leyen
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Jaroslaw Aronowski
- Department of Neurology, McGovern Medical School, University of Texas HSC, Houston, TX, USA
| | - James I. Koenig
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD USA
| | - Francesca Bosetti
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD USA
| | - Patrick Lyden
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Los Angeles, CA USA
- Department of Neurology, Keck School of Medicine at USC, Los Angeles, CA USA
| | - Cenk Ayata
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | | |
Collapse
|
37
|
Surugiu R, Burdusel D, Ruscu MA, Cercel A, Hermann DM, Cadenas IF, Popa-Wagner A. Clinical Ageing. Subcell Biochem 2023; 103:437-458. [PMID: 37120476 DOI: 10.1007/978-3-031-26576-1_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Ageing is generally characterised by the declining ability to respond to stress, increasing homeostatic imbalance, and increased risk of ageing-associated diseases . Mechanistically, the lifelong accumulation of a wide range of molecular and cellular impairments leads to organismal senescence. The aging population poses a severe medical concern due to the burden it places on healthcare systems and the general public as well as the prevalence of diseases and impairments associated with old age. In this chapter, we discuss organ failure during ageing as well as ageing of the hypothalamic-pituitary-adrenal axis and drugs that can regulate it. A much-debated subject is about ageing and regeneration. With age, there is a gradual decline in the regenerative properties of most tissues. The goal of regenerative medicine is to restore cells, tissues, and structures that are lost or damaged after disease, injury, or ageing. The question arises as to whether this is due to the intrinsic ageing of stem cells or, rather, to the impairment of stem-cell function in the aged tissue environment. The risk of having a stroke event doubles each decade after the age of 55. Therefore, it is of great interest to develop neurorestorative therapies for stroke which occurs mostly in elderly people. Initial enthusiasm for stimulating restorative processes in the ischaemic brain with cell-based therapies has meanwhile converted into a more balanced view, recognising impediments related to survival, migration, differentiation, and integration of therapeutic cells in the hostile aged brain environment. Therefore, a current lack of understanding of the fate of transplanted cells means that the safety of cell therapy in stroke patients is still unproven. Another issue associated with ischaemic stroke is that patients at risk for these sequels of stroke are not duly diagnosed and treated due to the lack of reliable biomarkers. However, recently neurovascular unit-derived exosomes in response to Stroke and released into serum are new plasma genetic and proteomic biomarkers associated with ischaemic stroke. The second valid option, which is also more economical, is to invest in prevention.
Collapse
Affiliation(s)
- Roxana Surugiu
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Craiova, Romania
- University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Daiana Burdusel
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Craiova, Romania
- University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Mihai-Andrei Ruscu
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Andreea Cercel
- Stroke Pharmacogenomics and Genetics Group, Sant Pau Hospital Institute of Research, Barcelona, Spain
| | - Dirk M Hermann
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Craiova, Romania
- University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Israel Fernandez Cadenas
- Stroke Pharmacogenomics and Genetics Group, Sant Pau Hospital Institute of Research, Barcelona, Spain
| | - Aurel Popa-Wagner
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, Craiova, Romania.
- University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
38
|
Peach CJ, Edgington-Mitchell LE, Bunnett NW, Schmidt BL. Protease-activated receptors in health and disease. Physiol Rev 2023; 103:717-785. [PMID: 35901239 PMCID: PMC9662810 DOI: 10.1152/physrev.00044.2021] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 07/08/2022] [Accepted: 07/10/2022] [Indexed: 11/22/2022] Open
Abstract
Proteases are signaling molecules that specifically control cellular functions by cleaving protease-activated receptors (PARs). The four known PARs are members of the large family of G protein-coupled receptors. These transmembrane receptors control most physiological and pathological processes and are the target of a large proportion of therapeutic drugs. Signaling proteases include enzymes from the circulation; from immune, inflammatory epithelial, and cancer cells; as well as from commensal and pathogenic bacteria. Advances in our understanding of the structure and function of PARs provide insights into how diverse proteases activate these receptors to regulate physiological and pathological processes in most tissues and organ systems. The realization that proteases and PARs are key mediators of disease, coupled with advances in understanding the atomic level structure of PARs and their mechanisms of signaling in subcellular microdomains, has spurred the development of antagonists, some of which have advanced to the clinic. Herein we review the discovery, structure, and function of this receptor system, highlight the contribution of PARs to homeostatic control, and discuss the potential of PAR antagonists for the treatment of major diseases.
Collapse
Affiliation(s)
- Chloe J Peach
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Department of Neuroscience and Physiology and Neuroscience Institute, Grossman School of Medicine, New York University, New York, New York
| | - Laura E Edgington-Mitchell
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Bluestone Center for Clinical Research, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, New York
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Department of Neuroscience and Physiology and Neuroscience Institute, Grossman School of Medicine, New York University, New York, New York
| | - Brian L Schmidt
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Bluestone Center for Clinical Research, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, New York
| |
Collapse
|
39
|
Palevski D, Ben-David G, Weinberger Y, Haj Daood R, Fernández JA, Budnik I, Levy-Mendelovich S, Kenet G, Nisgav Y, Weinberger D, Griffin JH, Livnat T. 3K3A-Activated Protein C Prevents Microglia Activation, Inhibits NLRP3 Inflammasome and Limits Ocular Inflammation. Int J Mol Sci 2022; 23:ijms232214196. [PMID: 36430674 PMCID: PMC9694680 DOI: 10.3390/ijms232214196] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/07/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
3K3A-Activated Protein C (APC) is a recombinant variant of the physiological anticoagulant APC with pleiotropic cytoprotective properties albeit without the bleeding risks. The anti-inflammatory activities of 3K3A-APC were demonstrated in multiple preclinical injury models, including various neurological disorders. We determined the ability of 3K3A-APC to inhibit ocular inflammation in a murine model of lipopolysaccharide (LPS)-induced uveitis. Leukocyte recruitment, microglia activation, NLRP3 inflammasome and IL-1β levels were assessed using flow cytometry, retinal cryosection histology, retinal flatmount immunohistochemistry and vascular imaging, with and without 3K3A-APC treatment. LPS triggered robust inflammatory cell recruitment in the posterior chamber. The 3K3A-APC treatment significantly decreased leukocyte numbers and inhibited leukocyte extravasation from blood vessels into the retinal parenchyma to a level similar to controls. Resident microglia, which underwent an inflammatory transition following LPS injection, remained quiescent in eyes treated with 3K3A-APC. An inflammation-associated increase in retinal thickness, observed in LPS-injected eyes, was diminished by 3K3A-APC treatment, suggesting its clinical relevancy. Finally, 3K3A-APC treatment inhibited inflammasome activation, determined by lower levels of NLRP3 and its downstream effector IL-1β. Our results highlight the anti-inflammatory properties of 3K3A-APC in ocular inflammation and suggest its potential use as a novel treatment for retinal diseases associated with inflammation.
Collapse
Affiliation(s)
- Dahlia Palevski
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Gil Ben-David
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Yehonatan Weinberger
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Rabeei Haj Daood
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - José A. Fernández
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ivan Budnik
- Sheba Medical Center, The Amalia Biron Thrombosis Research Institute, Tel-Hashomer 52621, Israel
| | - Sarina Levy-Mendelovich
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
- Sheba Medical Center, The Amalia Biron Thrombosis Research Institute, Tel-Hashomer 52621, Israel
| | - Gili Kenet
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
- Sheba Medical Center, The Amalia Biron Thrombosis Research Institute, Tel-Hashomer 52621, Israel
| | - Yael Nisgav
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
| | - Dov Weinberger
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - John H. Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tami Livnat
- Rabin Medical Center, Ophthalmology Department and Laboratory of Eye Research, Felsenstein Medical Research Center, Petah-Tikva 49100, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
- Sheba Medical Center, The Amalia Biron Thrombosis Research Institute, Tel-Hashomer 52621, Israel
- Correspondence:
| |
Collapse
|
40
|
Mishra A, Malik R, Hachiya T, Jürgenson T, Namba S, Posner DC, Kamanu FK, Koido M, Le Grand Q, Shi M, He Y, Georgakis MK, Caro I, Krebs K, Liaw YC, Vaura FC, Lin K, Winsvold BS, Srinivasasainagendra V, Parodi L, Bae HJ, Chauhan G, Chong MR, Tomppo L, Akinyemi R, Roshchupkin GV, Habib N, Jee YH, Thomassen JQ, Abedi V, Cárcel-Márquez J, Nygaard M, Leonard HL, Yang C, Yonova-Doing E, Knol MJ, Lewis AJ, Judy RL, Ago T, Amouyel P, Armstrong ND, Bakker MK, Bartz TM, Bennett DA, Bis JC, Bordes C, Børte S, Cain A, Ridker PM, Cho K, Chen Z, Cruchaga C, Cole JW, de Jager PL, de Cid R, Endres M, Ferreira LE, Geerlings MI, Gasca NC, Gudnason V, Hata J, He J, Heath AK, Ho YL, Havulinna AS, Hopewell JC, Hyacinth HI, Inouye M, Jacob MA, Jeon CE, Jern C, Kamouchi M, Keene KL, Kitazono T, Kittner SJ, Konuma T, Kumar A, Lacaze P, Launer LJ, Lee KJ, Lepik K, Li J, Li L, Manichaikul A, Markus HS, Marston NA, Meitinger T, Mitchell BD, Montellano FA, Morisaki T, Mosley TH, Nalls MA, Nordestgaard BG, O'Donnell MJ, Okada Y, Onland-Moret NC, Ovbiagele B, Peters A, Psaty BM, Rich SS, Rosand J, Sabatine MS, Sacco RL, Saleheen D, Sandset EC, Salomaa V, Sargurupremraj M, Sasaki M, Satizabal CL, Schmidt CO, Shimizu A, Smith NL, Sloane KL, Sutoh Y, Sun YV, Tanno K, Tiedt S, Tatlisumak T, Torres-Aguila NP, Tiwari HK, Trégouët DA, Trompet S, Tuladhar AM, Tybjærg-Hansen A, van Vugt M, Vibo R, Verma SS, Wiggins KL, Wennberg P, Woo D, Wilson PWF, Xu H, Yang Q, Yoon K, Millwood IY, Gieger C, Ninomiya T, Grabe HJ, Jukema JW, Rissanen IL, Strbian D, Kim YJ, Chen PH, Mayerhofer E, Howson JMM, Irvin MR, Adams H, Wassertheil-Smoller S, Christensen K, Ikram MA, Rundek T, Worrall BB, Lathrop GM, Riaz M, Simonsick EM, Kõrv J, França PHC, Zand R, Prasad K, Frikke-Schmidt R, de Leeuw FE, Liman T, Haeusler KG, Ruigrok YM, Heuschmann PU, Longstreth WT, Jung KJ, Bastarache L, Paré G, Damrauer SM, Chasman DI, Rotter JI, Anderson CD, Zwart JA, Niiranen TJ, Fornage M, Liaw YP, Seshadri S, Fernández-Cadenas I, Walters RG, Ruff CT, Owolabi MO, Huffman JE, Milani L, Kamatani Y, Dichgans M, Debette S. Stroke genetics informs drug discovery and risk prediction across ancestries. Nature 2022; 611:115-123. [PMID: 36180795 PMCID: PMC9524349 DOI: 10.1038/s41586-022-05165-3] [Citation(s) in RCA: 199] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 07/29/2022] [Indexed: 01/29/2023]
Abstract
Previous genome-wide association studies (GWASs) of stroke - the second leading cause of death worldwide - were conducted predominantly in populations of European ancestry1,2. Here, in cross-ancestry GWAS meta-analyses of 110,182 patients who have had a stroke (five ancestries, 33% non-European) and 1,503,898 control individuals, we identify association signals for stroke and its subtypes at 89 (61 new) independent loci: 60 in primary inverse-variance-weighted analyses and 29 in secondary meta-regression and multitrait analyses. On the basis of internal cross-ancestry validation and an independent follow-up in 89,084 additional cases of stroke (30% non-European) and 1,013,843 control individuals, 87% of the primary stroke risk loci and 60% of the secondary stroke risk loci were replicated (P < 0.05). Effect sizes were highly correlated across ancestries. Cross-ancestry fine-mapping, in silico mutagenesis analysis3, and transcriptome-wide and proteome-wide association analyses revealed putative causal genes (such as SH3PXD2A and FURIN) and variants (such as at GRK5 and NOS3). Using a three-pronged approach4, we provide genetic evidence for putative drug effects, highlighting F11, KLKB1, PROC, GP1BA, LAMC2 and VCAM1 as possible targets, with drugs already under investigation for stroke for F11 and PROC. A polygenic score integrating cross-ancestry and ancestry-specific stroke GWASs with vascular-risk factor GWASs (integrative polygenic scores) strongly predicted ischaemic stroke in populations of European, East Asian and African ancestry5. Stroke genetic risk scores were predictive of ischaemic stroke independent of clinical risk factors in 52,600 clinical-trial participants with cardiometabolic disease. Our results provide insights to inform biology, reveal potential drug targets and derive genetic risk prediction tools across ancestries.
Collapse
Affiliation(s)
- Aniket Mishra
- Bordeaux Population Health Research Center, University of Bordeaux, Inserm, UMR 1219, Bordeaux, France
| | - Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Tsuyoshi Hachiya
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, Japan
| | - Tuuli Jürgenson
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia
| | - Shinichi Namba
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Daniel C Posner
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - Frederick K Kamanu
- TIMI Study Group, Boston, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Masaru Koido
- Division of Molecular Pathology, Institute of Medical Sciences, The University of Tokyo, Tokyo, Japan
- Laboratory of Complex Trait Genomics, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Quentin Le Grand
- Bordeaux Population Health Research Center, University of Bordeaux, Inserm, UMR 1219, Bordeaux, France
| | - Mingyang Shi
- Laboratory of Complex Trait Genomics, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Yunye He
- Laboratory of Complex Trait Genomics, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Marios K Georgakis
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ilana Caro
- Bordeaux Population Health Research Center, University of Bordeaux, Inserm, UMR 1219, Bordeaux, France
| | - Kristi Krebs
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Yi-Ching Liaw
- Laboratory of Clinical Genome Sequencing, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung, Taiwan
| | - Felix C Vaura
- Department of Internal Medicine, University of Turku, Turku, Finland
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Turku, Finland
| | - Kuang Lin
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Bendik Slagsvold Winsvold
- Department of Research and Innovation, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Vinodh Srinivasasainagendra
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Livia Parodi
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hee-Joon Bae
- Department of Neurology and Cerebrovascular Disease Center, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | | | - Michael R Chong
- Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Liisa Tomppo
- Department of Neurology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Rufus Akinyemi
- Center for Genomic and Precision Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Neuroscience and Ageing Research Unit Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Gennady V Roshchupkin
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Naomi Habib
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yon Ho Jee
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Jesper Qvist Thomassen
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Vida Abedi
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Health System, Danville, VA, USA
- Department of Public Health Sciences, College of Medicine, The Pennsylvania State University, State College, PA, USA
| | - Jara Cárcel-Márquez
- Stroke Pharmacogenomics and Genetics Laboratory, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marianne Nygaard
- The Danish Twin Registry, Department of Public Health, University of Southern Denmark, Odense, Denmark
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Hampton L Leonard
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Glen Echo, MD, USA
| | - Chaojie Yang
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Ekaterina Yonova-Doing
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Maria J Knol
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Adam J Lewis
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Renae L Judy
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Tetsuro Ago
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Philippe Amouyel
- University of Lille, INSERM U1167, RID-AGE, LabEx DISTALZ, Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France
- CHU Lille, Public Health Department, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Nicole D Armstrong
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mark K Bakker
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Constance Bordes
- Bordeaux Population Health Research Center, University of Bordeaux, Inserm, UMR 1219, Bordeaux, France
| | - Sigrid Børte
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Research and Communication Unit for Musculoskeletal Health (FORMI), Department of Research and Innovation, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
| | - Anael Cain
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - Zhengming Chen
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - John W Cole
- VA Maryland Health Care System, Baltimore, MD, USA
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Phil L de Jager
- Program in Medical and Population Genetics, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Rafael de Cid
- GenomesForLife-GCAT Lab Group, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Matthias Endres
- Klinik und Hochschulambulanz für Neurologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), partner site Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Leslie E Ferreira
- Post-Graduation Program on Health and Environment, Department of Medicine and Joinville Stroke Biobank, University of the Region of Joinville, Santa Catarina, Brazil
| | - Mirjam I Geerlings
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Natalie C Gasca
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Jun Hata
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jing He
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alicia K Heath
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Yuk-Lam Ho
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - Aki S Havulinna
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Institute for Molecular Medicine Finland, FIMM-HiLIFE, Helsinki, Finland
| | - Jemma C Hopewell
- Clinical Trial Service and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Hyacinth I Hyacinth
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael Inouye
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
| | - Mina A Jacob
- Department of Neurology, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Christina E Jeon
- Los Angeles County Department of Public Health, Los Angeles, CA, USA
| | - Christina Jern
- Institute of Biomedicine, Department of Laboratory Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Masahiro Kamouchi
- Department of Health Care Administration and Management, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keith L Keene
- Department of Biology, Brody School of Medicine Center for Health Disparities, East Carolina University, Greenville, NC, USA
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Steven J Kittner
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Neurology and Geriatric Research and Education Clinical Center, VA Maryland Health Care System, Baltimore, MD, USA
| | - Takahiro Konuma
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Amit Kumar
- Rajendra Institute of Medical Sciences, Ranchi, India
| | - Paul Lacaze
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Lenore J Launer
- Intramural Research Program, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Keon-Joo Lee
- Department of Neurology, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Kaido Lepik
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- University Center for Primary Care and Public Health, Lausanne, Switzerland
| | - Jiang Li
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Health System, Danville, VA, USA
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Hugh S Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Nicholas A Marston
- TIMI Study Group, Boston, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas Meitinger
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Braxton D Mitchell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Felipe A Montellano
- Institute of Clinical Epidemiology and Biometry, University of Würzburg, Würzburg, Germany
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Takayuki Morisaki
- Division of Molecular Pathology, Institute of Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - Thomas H Mosley
- The MIND Center, University of Mississippi Medical Center, Jackson, MS, USA
| | - Mike A Nalls
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Glen Echo, MD, USA
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Martin J O'Donnell
- College of Medicine Nursing and Health Science, NUI Galway, Galway, Ireland
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
| | - N Charlotte Onland-Moret
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Bruce Ovbiagele
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München,, German Research Center for Environmental Health, Neuherberg, Germany
- Institute for Medical Information Processing, Biometry and Epidemiology, Ludwig Maximilian University Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich, Munich, Germany
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jonathan Rosand
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Marc S Sabatine
- TIMI Study Group, Boston, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ralph L Sacco
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
- Evelyn F. McKnight Brain Institute, Gainesville, FL, USA
| | - Danish Saleheen
- Division of Cardiology, Department of Medicine, Columbia University, New York, NY, USA
| | - Else Charlotte Sandset
- Stroke Unit, Department of Neurology, Oslo University Hospital, Oslo, Norway
- Research and Development, The Norwegian Air Ambulance Foundation, Oslo, Norway
| | - Veikko Salomaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Muralidharan Sargurupremraj
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Makoto Sasaki
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, Japan
| | - Claudia L Satizabal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Carsten O Schmidt
- University Medicine Greifswald, Institute for Community Medicine, SHIP/KEF, Greifswald, Germany
| | - Atsushi Shimizu
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, Japan
| | - Nicholas L Smith
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, USA
- Department of Veterans Affairs Office of Research and Development, Seattle Epidemiologic Research and Information Center, Seattle, WA, USA
| | - Kelly L Sloane
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Yoichi Sutoh
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, Japan
| | - Yan V Sun
- Atlanta VA Health Care System, Decatur, GA, USA
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Kozo Tanno
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, Japan
| | - Steffen Tiedt
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Turgut Tatlisumak
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Unviersity Hospital, Gothenburg, Sweden
| | - Nuria P Torres-Aguila
- Stroke Pharmacogenomics and Genetics Laboratory, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Hemant K Tiwari
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David-Alexandre Trégouët
- Bordeaux Population Health Research Center, University of Bordeaux, Inserm, UMR 1219, Bordeaux, France
| | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anil Man Tuladhar
- Department of Neurology, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Marion van Vugt
- Division Heart & Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Riina Vibo
- Department of Neurology and Neurosurgery, University of Tartu, Tartu, Estonia
| | - Shefali S Verma
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Patrik Wennberg
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Daniel Woo
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Peter W F Wilson
- Atlanta VA Health Care System, Decatur, GA, USA
- Department of Medicine, Division of Cardiovascular Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Huichun Xu
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Qiong Yang
- Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Kyungheon Yoon
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju, Republic of Korea
| | - Iona Y Millwood
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - Christian Gieger
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), site Rostock/Greifswald, Rostock, Germany
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands
| | - Ina L Rissanen
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Daniel Strbian
- Department of Neurology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Young Jin Kim
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju, Republic of Korea
| | - Pei-Hsin Chen
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung, Taiwan
| | - Ernst Mayerhofer
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Joanna M M Howson
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Marguerite R Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hieab Adams
- Department of Clinical Genetics, Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Latin American Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Sylvia Wassertheil-Smoller
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, New York, NY, USA
| | - Kaare Christensen
- The Danish Twin Registry, Department of Public Health, University of Southern Denmark, Odense, Denmark
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Mohammad A Ikram
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tatjana Rundek
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
- Evelyn F. McKnight Brain Institute, Gainesville, FL, USA
| | - Bradford B Worrall
- Department of Neurology, University of Virginia, Charlottesville, VA, USA
- Department of Public Health Science, University of Virginia, Charlottesville, VA, USA
| | | | - Moeen Riaz
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Eleanor M Simonsick
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Janika Kõrv
- Department of Neurology and Neurosurgery, University of Tartu, Tartu, Estonia
| | - Paulo H C França
- Post-Graduation Program on Health and Environment, Department of Medicine and Joinville Stroke Biobank, University of the Region of Joinville, Santa Catarina, Brazil
| | - Ramin Zand
- Geisinger Neuroscience Institute, Geisinger Health System, Danville, PA, USA
- Department of Neurology, College of Medicine, The Pennsylvania State University, State College, PA, USA
| | | | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Frank-Erik de Leeuw
- Department of Neurology, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thomas Liman
- Center for Stroke Research Berlin, Berlin, Germany
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Klinik für Neurologie, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
| | | | - Ynte M Ruigrok
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Peter Ulrich Heuschmann
- Institute of Clinical Epidemiology and Biometry, University of Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
- Clinical Trial Center, University Hospital Würzburg, Würzburg, Germany
| | - W T Longstreth
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Keum Ji Jung
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Republic of Korea
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Guillaume Paré
- Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada
| | - Scott M Damrauer
- Department of Surgery and Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Christopher D Anderson
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - John-Anker Zwart
- Department of Research and Innovation, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Teemu J Niiranen
- Department of Internal Medicine, University of Turku, Turku, Finland
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yung-Po Liaw
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
- Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Israel Fernández-Cadenas
- Stroke Pharmacogenomics and Genetics Laboratory, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Robin G Walters
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - Christian T Ruff
- TIMI Study Group, Boston, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mayowa O Owolabi
- Center for Genomic and Precision Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Department of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Jennifer E Huffman
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| | - Lili Milani
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Yoichiro Kamatani
- Laboratory of Complex Trait Genomics, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
- Munich Cluster for Systems Neurology, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
| | - Stephanie Debette
- Bordeaux Population Health Research Center, University of Bordeaux, Inserm, UMR 1219, Bordeaux, France.
- Department of Neurology, Institute for Neurodegenerative Diseases, CHU de Bordeaux, Bordeaux, France.
| |
Collapse
|
41
|
Su EJ, Lawrence DA. Diabetes and the treatment of ischemic stroke. J Diabetes Complications 2022; 36:108318. [PMID: 36228562 DOI: 10.1016/j.jdiacomp.2022.108318] [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: 08/10/2022] [Accepted: 09/23/2022] [Indexed: 11/23/2022]
Abstract
This white paper examines the current challenges for treating ischemic stroke in diabetic patients. The need for a greater understanding of the mechanisms that underlie the relationship between diabetes and the cerebral vascular responses to ischemia is discussed. The critical need to improve the efficacy and safety of thrombolysis is addressed, as is the need for a better characterization the off-target actions of tPA, the only currently approved thrombolytic for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Enming J Su
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Daniel A Lawrence
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
42
|
Matossian V, Starkman S, Sanossian N, Stratton S, Eckstein M, Conwit R, Liebeskind DS, Sharma L, Tenser MK, Saver JL. Quantifying the amount of greater brain ischemia protection time with pre-hospital vs. in-hospital neuroprotective agent start. Front Neurol 2022; 13:990339. [PMID: 36176566 PMCID: PMC9514007 DOI: 10.3389/fneur.2022.990339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
The objective of this study is to quantify the increase in brain-under-protection time that may be achieved with pre-hospital compared with the post-arrival start of neuroprotective therapy among patients undergoing endovascular thrombectomy. In order to do this, a comparative analysis was performed of two randomized trials of neuroprotective agents: (1) pre-hospital strategy: Field administration of stroke therapy-magnesium (FAST-MAG) Trial; (2) in-hospital strategy: Efficacy and safety of nerinetide for the treatment of acute ischemic stroke (ESCAPE-NA1) Trial. In the FAST-MAG trial, among 1,041 acute ischemic stroke patients, 44 were treated with endovascular reperfusion therapy (ERT), including 32 treated with both intravenous thrombolysis and ERT and 12 treated with ERT alone. In the ESCAPE-NA1 trial, among 1,105 acute ischemic stroke patients, 659 were treated with both intravenous thrombolysis and ERT, and 446 were treated with ERT alone. The start of the neuroprotective agent was sooner after onset with pre-hospital vs. in-hospital start: 45 m (IQR 38-56) vs. 122 m. The neuroprotective agent in FAST-MAG was started 8 min prior to ED arrival compared with 64 min after arrival in ESCAPE-NA1. Projecting modern endovascular workflows to FAST-MAG, the total time of "brain under protection" (neuroprotective agent start to reperfusion) was greater with pre-hospital than in-hospital start: 94 m (IQR 90-98) vs. 22 m. Initiating a neuroprotective agent in the pre-hospital setting enables a faster treatment start, yielding 72 min additional brain protection time for patients with acute ischemic stroke. These findings provide support for the increased performance of ambulance-based, pre-hospital treatment trials in the development of neuroprotective stroke therapies.
Collapse
Affiliation(s)
- Vartan Matossian
- MSTAR Program, Department of Geriatrics, University of California, Los Angeles, Los Angeles, CA, United States,*Correspondence: Vartan Matossian
| | - Sidney Starkman
- Stroke Center and Department of Emergency Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nerses Sanossian
- Department of Neurology, University of Southern California, Los Angeles, CA, United States
| | - Samuel Stratton
- Department of Emergency Medicine, University Harbor-UCLA Medical Center, Los Angeles, CA, United States
| | - Marc Eckstein
- Department of Emergency Medicine, University of Southern California, Los Angeles, CA, United States
| | - Robin Conwit
- Division of Extramural Research, National Institutes of Health/National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
| | - David S. Liebeskind
- Stroke Center and Department of Neurology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Latisha Sharma
- Stroke Center and Department of Neurology, University of California, Los Angeles, Los Angeles, CA, United States
| | - May-Kim Tenser
- Department of Neurology, University of Southern California, Los Angeles, CA, United States
| | - Jeffrey L. Saver
- Stroke Center and Department of Neurology, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
43
|
Tiedt S, Buchan AM, Dichgans M, Lizasoain I, Moro MA, Lo EH. The neurovascular unit and systemic biology in stroke - implications for translation and treatment. Nat Rev Neurol 2022; 18:597-612. [PMID: 36085420 DOI: 10.1038/s41582-022-00703-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2022] [Indexed: 12/24/2022]
Abstract
Ischaemic stroke is a leading cause of disability and death for which no acute treatments exist beyond recanalization. The development of novel therapies has been repeatedly hindered by translational failures that have changed the way we think about tissue damage after stroke. What was initially a neuron-centric view has been replaced with the concept of the neurovascular unit (NVU), which encompasses neuronal, glial and vascular compartments, and the biphasic nature of neural-glial-vascular signalling. However, it is now clear that the brain is not the private niche it was traditionally thought to be and that the NVU interacts bidirectionally with systemic biology, such as systemic metabolism, the peripheral immune system and the gut microbiota. Furthermore, these interactions are profoundly modified by internal and external factors, such as ageing, temperature and day-night cycles. In this Review, we propose an extension of the concept of the NVU to include its dynamic interactions with systemic biology. We anticipate that this integrated view will lead to the identification of novel mechanisms of stroke pathophysiology, potentially explain previous translational failures, and improve stroke care by identifying new biomarkers of and treatment targets in stroke.
Collapse
Affiliation(s)
- Steffen Tiedt
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA), . .,Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.
| | - Alastair M Buchan
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA).,Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Martin Dichgans
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA).,Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Ignacio Lizasoain
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA).,Department of Pharmacology and Toxicology, Complutense Medical School, Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Maria A Moro
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA).,Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain
| | - Eng H Lo
- Consortium International pour la Recherche Circadienne sur l'AVC (CIRCA), . .,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW To provide an overview of the state-of-the-art in protein C (PC) pathway research. RECENT FINDINGS The PC pathway is crucial for maintaining hemostasis to prevent venous thromboembolism. This is evident from genetic mutations that result in impaired PC pathway activity and contribute to increased venous thromboembolism risk in affected individuals. In addition to its anticoagulant role, activated PC (APC) also mediates a complex, pleiotropic role in the maintenance of vascular cell health, which it achieves via anti-inflammatory and antiapoptotic cell signaling on endothelial cells. Emerging data have demonstrated that cell signaling by APC, mediated by multiple receptor interactions on different cell types, also confers cytoprotective and anti-inflammatory benefits. Defects in both arms of the PC pathway are associated with increased susceptibility to thrombo-inflammatory disease in various preclinical thrombotic, proinflammatory and neurological disease models. Moreover, recent studies have identified attenuation of anticoagulant PC pathway activity as an exciting therapeutic opportunity to promote hemostasis in patients with inherited or acquired bleeding disorders. SUMMARY In this review, we provide an overview of some recent developments in our understanding of the PC pathways.
Collapse
Affiliation(s)
- Gemma Leon
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Crumlin, Dublin 12, Ireland
| | - Aisling M Rehill
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Crumlin, Dublin 12, Ireland
| |
Collapse
|
45
|
Rana R, Manoharan J, Gupta A, Gupta D, Elwakiel A, Khawaja H, Fatima S, Zimmermann S, Singh K, Ambreen S, Gadi I, Biemann R, Jiang S, Shahzad K, Kohli S, Isermann B. Activated Protein C Ameliorates Tubular Mitochondrial Reactive Oxygen Species and Inflammation in Diabetic Kidney Disease. Nutrients 2022; 14:nu14153138. [PMID: 35956315 PMCID: PMC9370435 DOI: 10.3390/nu14153138] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023] Open
Abstract
Diabetic kidney disease (DKD) is an emerging pandemic, paralleling the worldwide increase in obesity and diabetes mellitus. DKD is now the most frequent cause of end-stage renal disease and is associated with an excessive risk of cardiovascular morbidity and mortality. DKD is a consequence of systemic endothelial dysfunction. The endothelial-dependent cytoprotective coagulation protease activated protein C (aPC) ameliorates glomerular damage in DKD, in part by reducing mitochondrial ROS generation in glomerular cells. Whether aPC reduces mitochondrial ROS generation in the tubular compartment remains unknown. Here, we conducted expression profiling of kidneys in diabetic mice (wild-type and mice with increased plasma levels of aPC, APChigh mice). The top induced pathways were related to metabolism and in particular to oxidoreductase activity. In tubular cells, aPC maintained the expression of genes related to the electron transport chain, PGC1-α expression, and mitochondrial mass. These effects were associated with reduced mitochondrial ROS generation. Likewise, NLRP3 inflammasome activation and sterile inflammation, which are known to be linked to excess ROS generation in DKD, were reduced in diabetic APChigh mice. Thus, aPC reduces mitochondrial ROS generation in tubular cells and dampens the associated renal sterile inflammation. These studies support approaches harnessing the cytoprotective effects of aPC in DKD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Berend Isermann
- Correspondence: ; Tel.: +49-(0)341-972-2200; Fax: 49-(0)341-972-2379
| |
Collapse
|
46
|
Ronaldson PT, Davis TP. Transport Mechanisms at the Blood-Brain Barrier and in Cellular Compartments of the Neurovascular Unit: Focus on CNS Delivery of Small Molecule Drugs. Pharmaceutics 2022; 14:1501. [PMID: 35890396 PMCID: PMC9324459 DOI: 10.3390/pharmaceutics14071501] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 02/06/2023] Open
Abstract
Ischemic stroke is a primary origin of morbidity and mortality in the United States and around the world. Indeed, several research projects have attempted to discover new drugs or repurpose existing therapeutics to advance stroke pharmacotherapy. Many of these preclinical stroke studies have reported positive results for neuroprotective agents; however, only one compound (3K3A-activated protein C (3K3A-APC)) has advanced to Phase III clinical trial evaluation. One reason for these many failures is the lack of consideration of transport mechanisms at the blood-brain barrier (BBB) and neurovascular unit (NVU). These endogenous transport processes function as a "gateway" that is a primary determinant of efficacious brain concentrations for centrally acting drugs. Despite the knowledge that some neuroprotective agents (i.e., statins and memantine) are substrates for these endogenous BBB transporters, preclinical stroke studies have largely ignored the role of transporters in CNS drug disposition. Here, we review the current knowledge on specific BBB transporters that either limit drug uptake into the brain (i.e., ATP-binding cassette (ABC) transporters) or can be targeted for optimized drug delivery (i.e., solute carrier (SLC) transporters). Additionally, we highlight the current knowledge on transporter expression in astrocytes, microglia, pericytes, and neurons with an emphasis on transport mechanisms in these cell types that can influence drug distribution within the brain.
Collapse
Affiliation(s)
- Patrick T. Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724-5050, USA;
| | | |
Collapse
|
47
|
Gallego-Fabrega C, Muiño E, Cárcel-Márquez J, Llucià-Carol L, Lledós M, Martín-Campos JM, Cullell N, Fernández-Cadenas I. Genome-Wide Studies in Ischaemic Stroke: Are Genetics Only Useful for Finding Genes? Int J Mol Sci 2022; 23:6840. [PMID: 35743317 PMCID: PMC9224543 DOI: 10.3390/ijms23126840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 02/07/2023] Open
Abstract
Ischaemic stroke is a complex disease with some degree of heritability. This means that heritability factors, such as genetics, could be risk factors for ischaemic stroke. The era of genome-wide studies has revealed some of these heritable risk factors, although the data generated by these studies may also be useful in other disciplines. Analysis of these data can be used to understand the biological mechanisms associated with stroke risk and stroke outcome, to determine the causality between stroke and other diseases without the need for expensive clinical trials, or to find potential drug targets with higher success rates than other strategies. In this review we will discuss several of the most relevant studies regarding the genetics of ischaemic stroke and the potential use of the data generated.
Collapse
Affiliation(s)
- Cristina Gallego-Fabrega
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.G.-F.); (E.M.); (J.C.-M.); (L.L.-C.); (M.L.); (J.M.M.-C.); (N.C.)
| | - Elena Muiño
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.G.-F.); (E.M.); (J.C.-M.); (L.L.-C.); (M.L.); (J.M.M.-C.); (N.C.)
| | - Jara Cárcel-Márquez
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.G.-F.); (E.M.); (J.C.-M.); (L.L.-C.); (M.L.); (J.M.M.-C.); (N.C.)
| | - Laia Llucià-Carol
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.G.-F.); (E.M.); (J.C.-M.); (L.L.-C.); (M.L.); (J.M.M.-C.); (N.C.)
- Institute for Biomedical Research of Barcelona (IIBB), National Spanish Research Council (CSIC), 08036 Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Miquel Lledós
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.G.-F.); (E.M.); (J.C.-M.); (L.L.-C.); (M.L.); (J.M.M.-C.); (N.C.)
| | - Jesús M. Martín-Campos
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.G.-F.); (E.M.); (J.C.-M.); (L.L.-C.); (M.L.); (J.M.M.-C.); (N.C.)
| | - Natalia Cullell
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.G.-F.); (E.M.); (J.C.-M.); (L.L.-C.); (M.L.); (J.M.M.-C.); (N.C.)
| | - Israel Fernández-Cadenas
- Stroke Pharmacogenomics and Genetics Group, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (C.G.-F.); (E.M.); (J.C.-M.); (L.L.-C.); (M.L.); (J.M.M.-C.); (N.C.)
- Stroke Pharmacogenomics and Genetics Group, Fundació MútuaTerrassa per la Docència i la Recerca, 08221 Terrassa, Spain
| |
Collapse
|
48
|
Ghozy S, Reda A, Varney J, Elhawary AS, Shah J, Murry K, Sobeeh MG, Nayak SS, Azzam AY, Brinjikji W, Kadirvel R, Kallmes DF. Neuroprotection in Acute Ischemic Stroke: A Battle Against the Biology of Nature. Front Neurol 2022; 13:870141. [PMID: 35711268 PMCID: PMC9195142 DOI: 10.3389/fneur.2022.870141] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/21/2022] [Indexed: 12/22/2022] Open
Abstract
Stroke is the second most common cause of global death following coronary artery disease. Time is crucial in managing stroke to reduce the rapidly progressing insult of the ischemic penumbra and the serious neurologic deficits that might follow it. Strokes are mainly either hemorrhagic or ischemic, with ischemic being the most common of all types of strokes. Thrombolytic therapy with recombinant tissue plasminogen activator and endovascular thrombectomy are the main types of management of acute ischemic stroke (AIS). In addition, there is a vital need for neuroprotection in the setting of AIS. Neuroprotective agents are important to investigate as they may reduce mortality, lessen disability, and improve quality of life after AIS. In our review, we will discuss the main types of management and the different modalities of neuroprotection, their mechanisms of action, and evidence of their effectiveness after ischemic stroke.
Collapse
Affiliation(s)
- Sherief Ghozy
- Department of Neuroradiology, Mayo Clinic, Rochester, MN, United States.,Nuffield Department of Primary Care Health Sciences and Department for Continuing Education (EBHC Program), Oxford University, Oxford, United Kingdom
| | - Abdullah Reda
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Joseph Varney
- School of Medicine, American University of the Caribbean, Philipsburg, Sint Maarten
| | | | - Jaffer Shah
- Medical Research Center, Kateb University, Kabul, Afghanistan
| | | | - Mohamed Gomaa Sobeeh
- Faculty of Physical Therapy, Sinai University, Cairo, Egypt.,Faculty of Physical Therapy, Cairo University, Giza, Egypt
| | - Sandeep S Nayak
- Department of Internal Medicine, NYC Health + Hospitals/Metropolitan, New York, NY, United States
| | - Ahmed Y Azzam
- Faculty of Medicine, October 6 University, Giza, Egypt
| | - Waleed Brinjikji
- Department of Neurosurgery, Mayo Clinic Rochester, Rochester, MN, United States
| | | | - David F Kallmes
- Department of Neuroradiology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
49
|
Abstract
Intravenous thrombolytics and endovascular thrombectomy for ischemic stroke have evolved in parallel. However, the best approach to combine these reperfusion therapies in patients eligible for both strategies remains uncertain. Initial randomized trials of endovascular thrombectomy included administration of intravenous thrombolytics to all eligible patients. However, whether that is of net benefit has been questioned and parallels drawn with treatment of ST-segment-elevation myocardial infarction, where intravenous thrombolytics are only given if first medical contact to percutaneous intervention is expected to be >90 minutes. Six randomized trials of a direct thrombectomy approach versus intravenous thrombolytics followed by endovascular thrombectomy have now reported their results. With exception of a minority of patients in one trial, the trials all used alteplase rather than potentially more effective tenecteplase. This review examines the current state of evidence and implications for clinical practice. Importantly, these trials only apply to patients who present to a hospital with immediate access to endovascular thrombectomy and are not relevant to patients who receive thrombolytic and are then transferred to an endovascular-capable hospital. Although 2 of the 6 randomized trials met their prespecified noninferiority margin, these margins were large compared with the absolute benefit of alteplase. Overall, functional outcome was similar, with slight trends favoring bridging thrombolytics and a significant increase in final reperfusion. Symptomatic hemorrhage was increased by ≈1.8% in the bridging group but death was nonsignificantly lower. The workflow in direct thrombectomy trials involved delaying thrombolytic administration until eligibility for thrombectomy and the trials was established and randomization completed. This reduced the time available for thrombolytics to occur prethrombectomy compared with standard practice. We conclude that, pending individual-patient data meta-analyses, intravenous thrombolytics retain an important role alongside endovascular thrombectomy. Further efforts to accelerate and enhance reperfusion with thrombolytics and perform individual patient-level pooled subgroup analyses are warranted.
Collapse
Affiliation(s)
- Bruce C V Campbell
- Department of Medicine and Neurology, Melbourne Brain Centre at the Royal Melbourne Hospital (B.C.V.C.), University of Melbourne, Parkville, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health (B.C.V.C.), University of Melbourne, Parkville, Victoria, Australia
| | - Manon Kappelhof
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands (M.K.)
| | - Urs Fischer
- Department of Neurology, Universitätsspital Basel, Switzerland (U.F.)
- Department of Neurology, University Hospital Bern, University of Bern, Switzerland (U.F.)
| |
Collapse
|
50
|
Mechtouff L, Eker OF, Nighoghossian N, Cho TH. Fisiopatologia dell’ischemia cerebrale. Neurologia 2022. [DOI: 10.1016/s1634-7072(22)46428-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|