1
|
Lee NT, Savvidou I, Selan C, Calvello I, Vuong A, Wright DK, Brkljaca R, Willcox A, Chia JSJ, Wang X, Peter K, Robson SC, Medcalf RL, Nandurkar HH, Sashindranath M. Development of endothelial-targeted CD39 as a therapy for ischemic stroke. J Thromb Haemost 2024; 22:2331-2344. [PMID: 38754782 DOI: 10.1016/j.jtha.2024.04.023] [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: 12/21/2023] [Revised: 04/11/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Ischemic stroke is characterized by a necrotic lesion in the brain surrounded by an area of dying cells termed the penumbra. Salvaging the penumbra either with thrombolysis or mechanical retrieval is the cornerstone of stroke management. At-risk neuronal cells release extracellular adenosine triphosphate, triggering microglial activation and causing a thromboinflammatory response, culminating in endothelial activation and vascular disruption. This is further aggravated by ischemia-reperfusion injury that follows all reperfusion therapies. The ecto-enzyme CD39 regulates extracellular adenosine triphosphate by hydrolyzing it to adenosine, which has antithrombotic and anti-inflammatory properties and reverses ischemia-reperfusion injury. OBJECTIVES The objective off the study was to determine the efficacy of our therapeutic, anti-VCAM-CD39 in ischaemic stroke. METHODS We developed anti-VCAM-CD39 that targets the antithrombotic and anti-inflammatory properties of recombinant CD39 to the activated endothelium of the penumbra by binding to vascular cell adhesion molecule (VCAM)-1. Mice were subjected to 30 minutes of middle cerebral artery occlusion and analyzed at 24 hours. Anti-VCAM-CD39 or control agents (saline, nontargeted CD39, or anti-VCAM-inactive CD39) were given at 3 hours after middle cerebral artery occlusion. RESULTS Anti-VCAM-CD39 treatment reduced neurologic deficit; magnetic resonance imaging confirmed significantly smaller infarcts together with an increase in cerebrovascular perfusion. Anti-VCAM-CD39 also restored blood-brain barrier integrity and reduced microglial activation. Coadministration of anti-VCAM-CD39 with thrombolytics (tissue plasminogen activator [tPA]) further reduced infarct volumes and attenuated blood-brain barrier permeability with no associated increase in intracranial hemorrhage. CONCLUSION Anti-VCAM-CD39, uniquely targeted to endothelial cells, could be a new stroke therapy even when administered 3 hours postischemia and may further synergize with thrombolytic therapy to improve stroke outcomes.
Collapse
Affiliation(s)
- Natasha Ting Lee
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia; Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia
| | - Ioanna Savvidou
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - Carly Selan
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - Ilaria Calvello
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - Amy Vuong
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - David K Wright
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Robert Brkljaca
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia
| | - Abbey Willcox
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - Joanne S J Chia
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - Xiaowei Wang
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia; Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Simon C Robson
- Center for Inflammation Research, Department of Anesthesia, Critical Care & Pain Medicine and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert L Medcalf
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - Harshal H Nandurkar
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - Maithili Sashindranath
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia.
| |
Collapse
|
2
|
Pontius MHH, Ku CJ, Osmond MJ, Disharoon D, Liu Y, Warnock M, Lawrence DA, Marr DWM, Neeves KB, Shavit JA. Magnetically powered microwheel thrombolysis of occlusive thrombi in zebrafish. Proc Natl Acad Sci U S A 2024; 121:e2315083121. [PMID: 38408253 PMCID: PMC10927521 DOI: 10.1073/pnas.2315083121] [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: 09/15/2023] [Accepted: 01/09/2024] [Indexed: 02/28/2024] Open
Abstract
Tissue plasminogen activator (tPA) is the only FDA-approved treatment for ischemic stroke but carries significant risks, including major hemorrhage. Additional options are needed, especially in small vessel thrombi which account for ~25% of ischemic strokes. We have previously shown that tPA-functionalized colloidal microparticles can be assembled into microwheels (µwheels) and manipulated under the control of applied magnetic fields to enable rapid thrombolysis of fibrin gels in microfluidic models of thrombosis. Transparent zebrafish larvae have a highly conserved coagulation cascade that enables studies of hemostasis and thrombosis in the context of intact vasculature, clotting factors, and blood cells. Here, we show that tPA-functionalized µwheels can perform rapid and targeted recanalization in vivo. This effect requires both tPA and µwheels, as minimal to no recanalization is achieved with tPA alone, µwheels alone, or tPA-functionalized microparticles in the absence of a magnetic field. We evaluated tPA-functionalized µwheels in CRISPR-generated plasminogen (plg) heterozygous and homozygous mutants and confirmed that tPA-functionalized µwheels are dose-dependent on plasminogen for lysis. We have found that magnetically powered µwheels as a targeted tPA delivery system are dramatically more efficient at plasmin-mediated thrombolysis than systemic delivery in vivo. Further development of this system in fish and mammalian models could enable a less invasive strategy for alleviating ischemia that is safer than directed thrombectomy or systemic infusion of tPA.
Collapse
Affiliation(s)
| | - Chia-Jui Ku
- Department of Pediatrics, University of Michigan, Ann Arbor, MI48109
| | - Matthew J. Osmond
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO80401
| | - Dante Disharoon
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO80401
| | - Yang Liu
- Department of Pediatrics, University of Michigan, Ann Arbor, MI48109
| | - Mark Warnock
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI48109
| | - Daniel A. Lawrence
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI48109
| | - David W. M. Marr
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO80401
| | - Keith B. Neeves
- Department of Bioengineering, University of Colorado, Denver, Aurora, CO80045
- Department of Pediatrics, University of Colorado, Denver, Aurora, CO80045
| | - Jordan A. Shavit
- Department of Pediatrics, University of Michigan, Ann Arbor, MI48109
- Department of Human Genetics, University of Michigan, Ann Arbor, MI48109
| |
Collapse
|
3
|
Yao SQ, Ye Y, Li Q, Wang XY, Yan L, Huo XM, Pan CS, Fu Y, Liu J, Han JY. YangXueQingNaoWan attenuated blood brain barrier disruption after thrombolysis with tissue plasminogen activator in ischemia stroke. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117024. [PMID: 37572928 DOI: 10.1016/j.jep.2023.117024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANT YangXueQingNaoWan (YXQNW), a compound Chinese medicine, has been widely used for dizziness, irritability, insomnia, and dreaminess caused by blood deficiency and liver hyperactivity in China. However, whether YXQNW can inhibit cerebral microvascular exudation and cerebral hemorrhage (CH) caused by blood brain barrier (BBB) damage after tissue plasminogen activator (tPA) still unknown. AIM OF THE RESEARCH To observe the effect of YXQNW on cerebral microvascular exudation and CH after tPA and investigate its mechanism in protecting BBB. MATERIALS AND METHODS Male C57BL/6 N mice suffered from ischemia stroke by mechanical detachment of carotid artery thrombi with the stimulation of ferric chloride. Then mice were treated with tPA (10 mg/kg) and/or YXQNW (0.72 g/kg) at 4.5 h. Cerebral blood flow (CBF), infarct size, survival rate, neurological scores, gait analysis, Evans blue extravasation, cerebral water content, fluorescein isothiocyanate-labeled albumin leakage, hemorrhage, junction and basement membrane proteins expression, leukocyte adhesion and matrix metalloproteinases (MMPs) expression were evaluated 24 h after tPA. Proteomics was used to identify target proteins. RESULTS YXQNW inhibited cerebral infarction, neurobehavioral deficits, decreased survival, Evans blue leakage, albumin leakage, cerebral water content and CH after tPA thrombolysis; improved CBF, low-expression and degradation of junction proteins, basement membrane proteins, Arhgap21 and its downstream α-catenin and β-catenin proteins expression; and suppressed the increase of adherent leukocytes and the release of MMP-9 derived from macrophage. CONCLUSION YXQNW relieved BBB damage and attenuated cerebral microvascular exudation and CH after tPA thrombolysis. The effect of YXQNW on cerebral microvascular exudation was associated with the inhibition of the low-expression of junction proteins, especially AJs mediated by Rho GTPase-activating protein 21 (Arhgap21), while the effect on CH was associated with the inhibition of leukocyte adhesion, the release of MMP-9 derived from macrophage, and low-expression and degradation of collagen IV and laminin in the vascular basement membrane.
Collapse
Affiliation(s)
- Shu-Qi Yao
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China; The Key Discipline for Basic Integration of Chinese and Western Medicine (microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing 100191, China
| | - Yang Ye
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Quan Li
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing 100191, China; Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing 100191, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Beijing 100191, China; The Key Discipline for Basic Integration of Chinese and Western Medicine (microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing 100191, China
| | - Xiao-Yi Wang
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China; The Key Discipline for Basic Integration of Chinese and Western Medicine (microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing 100191, China
| | - Li Yan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing 100191, China; Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing 100191, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Beijing 100191, China; The Key Discipline for Basic Integration of Chinese and Western Medicine (microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing 100191, China
| | - Xin-Mei Huo
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing 100191, China; Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing 100191, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Beijing 100191, China; The Key Discipline for Basic Integration of Chinese and Western Medicine (microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing 100191, China
| | - Chun-Shui Pan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing 100191, China; Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing 100191, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Beijing 100191, China; The Key Discipline for Basic Integration of Chinese and Western Medicine (microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing 100191, China
| | - Yu Fu
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
| | - Jian Liu
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China; The Key Discipline for Basic Integration of Chinese and Western Medicine (microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing 100191, China
| | - Jing-Yan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing 100191, China; Key Laboratory of Stasis and Phlegm, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing 100191, China; Key Laboratory of Microcirculation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Beijing 100191, China; State Key Laboratory of Core Technology in Innovative Chinese Medicine, Beijing 100191, China; The Key Discipline for Basic Integration of Chinese and Western Medicine (microcirculation) of the National Administration of Traditional Chinese Medicine, Beijing 100191, China.
| |
Collapse
|
4
|
Pontius MHH, Ku CJ, Osmond M, Disharoon D, Liu Y, Marr DW, Neeves KB, Shavit JA. Magnetically Powered Microwheel Thrombolysis of Occlusive Thrombi in Zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.11.557256. [PMID: 37745422 PMCID: PMC10515822 DOI: 10.1101/2023.09.11.557256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Tissue plasminogen activator (tPA) is the only FDA approved treatment for ischemic stroke but carries significant risks, including major hemorrhage. Additional options are needed, especially in small vessel thrombi which account for ~25% of ischemic strokes. We have previously shown that tPA-functionalized colloidal microparticles can be assembled into microwheels (µwheels) and manipulated under the control of applied magnetic fields to enable rapid thrombolysis of fibrin gels in microfluidic models of thrombosis. Providing a living microfluidic analog, transparent zebrafish larvae have a highly conserved coagulation cascade that enables studies of hemostasis and thrombosis in the context of intact vasculature, clotting factors, and blood cells. Here we show that tPA-functionalized µwheels can perform rapid and targeted recanalization in vivo. This effect requires both tPA and µwheels, as minimal to no recanalization is achieved with tPA alone, µwheels alone, or tPA-functionalized microparticles in the absence of a magnetic field. We evaluated tPA-µwheels in CRISPR-generated plasminogen (plg) heterozygous and homozygous mutants and confirmed that tPA-µwheels are dose-dependent on plasminogen for lysis. We have found that magnetically powered µwheels as a targeted tPA delivery system are dramatically more efficient at plasmin-mediated thrombolysis than systemic delivery in vivo. Further development of this system in fish and mammalian models could enable a less invasive strategy for alleviating ischemia that is safer than directed thrombectomy or systemic infusion of tPA.
Collapse
Affiliation(s)
| | - Chia-Jui Ku
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Matthew Osmond
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO
| | - Dante Disharoon
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO
| | - Yang Liu
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - David W.M. Marr
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO
| | - Keith B. Neeves
- Departments of Bioengineering and Pediatrics, University of Colorado, Denver | Anschutz Medical Campus, Aurora, CO
| | - Jordan A. Shavit
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
| |
Collapse
|
5
|
Barreto-Arce LJ, Kim HA, Chan ST, Lim R, Drummond GR, Ma H, Phan TG, Sobey CG, Zhang SR. Protection against brain injury after ischemic stroke by intravenous human amnion epithelial cells in combination with tissue plasminogen activator. Front Neurosci 2023; 17:1157236. [PMID: 37397458 PMCID: PMC10311557 DOI: 10.3389/fnins.2023.1157236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/25/2023] [Indexed: 07/04/2023] Open
Abstract
Background Thrombolytic agents such as tissue plasminogen activator (tPA) are the only drug class approved to treat ischemic stroke and are usually administered within 4.5 h. However, only ~20% of ischemic stroke patients are eligible to receive the therapy. We previously demonstrated that early intravenous administration of human amnion epithelial cells (hAECs) can limit brain inflammation and infarct growth in experimental stroke. Here, we have tested whether hAECs exert cerebroprotective effects in combination with tPA in mice. Methods Male C57Bl/6 mice were subjected to middle cerebral artery occlusion for 60 min followed by reperfusion. Immediately following reperfusion, vehicle (saline, n = 31) or tPA (10 mg/kg; n = 73) was administered intravenously. After 30 min of reperfusion, tPA-treated mice were injected intravenously with either hAECs (1×106; n = 32) or vehicle (2% human serum albumin; n = 41). A further 15 sham-operated mice were treated with vehicle (n = 7) or tPA + vehicle (n = 8). Mice were designated to be euthanised at 3, 6 or 24 h post-stroke (n = 21, 31, and 52, respectively), and brains were collected to assess infarct volume, blood-brain barrier (BBB) disruption, intracerebral bleeding and inflammatory cell content. Results There was no mortality within 6 h of stroke onset, but a high mortality occurred in tPA + saline-treated mice between 6 h and 24 h post-stroke in comparison to mice treated with tPA + hAECs (61% vs. 27%, p = 0.04). No mortality occurred within 24 h of sham surgery in mice treated with tPA + vehicle. We focused on early infarct expansion within 6 h of stroke and found that infarction was ~50% larger in tPA + saline- than in vehicle-treated mice (23 ± 3 mm3 vs. 15 ± 2 mm3, p = 0.02) but not in mice receiving tPA + hAECs (13 ± 2 mm3, p < 0.01 vs. tPA + saline) in which intracerebral hAECs were detected. Similar to the profiles of infarct expansion, BBB disruption and intracerebral bleeding in tPA + saline-treated mice at 6 h was 50-60% greater than in vehicle-treated controls (2.6 ± 0.5 vs. 1.6 ± 0.2, p = 0.05) but not after tPA + hAECs treatment (1.7 ± 0.2, p = 0.10 vs. tPA + saline). No differences in inflammatory cell content were detected between treatment groups. Conclusion When administered following tPA in acute stroke, hAECs improve safety and attenuate infarct growth in association with less BBB disruption and lower 24 h mortality.
Collapse
Affiliation(s)
- Liz J. Barreto-Arce
- Department of Microbiology, Anatomy, Physiology, and Pharmacology and Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Hyun Ah Kim
- Department of Microbiology, Anatomy, Physiology, and Pharmacology and Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Siow Teng Chan
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Rebecca Lim
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Grant R. Drummond
- Department of Microbiology, Anatomy, Physiology, and Pharmacology and Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Henry Ma
- Clinical Trials, Imaging and Informatics (CTI) Division, Stroke and Ageing Research (STARC), Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Thanh G. Phan
- Clinical Trials, Imaging and Informatics (CTI) Division, Stroke and Ageing Research (STARC), Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Christopher G. Sobey
- Department of Microbiology, Anatomy, Physiology, and Pharmacology and Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| | - Shenpeng R. Zhang
- Department of Microbiology, Anatomy, Physiology, and Pharmacology and Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
6
|
Alsayejh B, Kietsiriroje N, Almutairi M, Simmons K, Pechlivani N, Ponnambalam S, Ajjan RA. Plasmin Inhibitor in Health and Diabetes: Role of the Protein as a Therapeutic Target. TH OPEN 2022; 6:e396-e407. [PMID: 36452200 PMCID: PMC9674435 DOI: 10.1055/a-1957-6817] [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: 03/05/2022] [Accepted: 09/23/2022] [Indexed: 11/21/2022] Open
Abstract
The vascular obstructive thrombus is composed of a mesh of fibrin fibers with blood cells trapped in these networks. Enhanced fibrin clot formation and/or suppression of fibrinolysis are associated with an increased risk of vascular occlusive events. Inhibitors of coagulation factors and activators of plasminogen have been clinically used to limit fibrin network formation and enhance lysis. While these agents are effective at reducing vascular occlusion, they carry a significant risk of bleeding complications. Fibrin clot lysis, essential for normal hemostasis, is controlled by several factors including the incorporation of antifibrinolytic proteins into the clot. Plasmin inhibitor (PI), a key antifibrinolytic protein, is cross-linked into fibrin networks with higher concentrations of PI documented in fibrin clots and plasma from high vascular risk individuals. This review is focused on exploring PI as a target for the prevention and treatment of vascular occlusive disease. We first discuss the relationship between the PI structure and antifibrinolytic activity, followed by describing the function of the protein in normal physiology and its role in pathological vascular thrombosis. Subsequently, we describe in detail the potential use of PI as a therapeutic target, including the array of methods employed for the modulation of protein activity. Effective and safe inhibition of PI may prove to be an alternative and specific way to reduce vascular thrombotic events while keeping bleeding risk to a minimum. Key Points Plasmin inhibitor (PI) is a key protein that inhibits fibrinolysis and stabilizes the fibrin network.This review is focused on discussing mechanistic pathways for PI action, role of the molecule in disease states, and potential use as a therapeutic target.
Collapse
Affiliation(s)
- Basmah Alsayejh
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, Leeds, United Kingdom
- Ministry of Education, Riyadh, Kingdom of Saudi Arabia
| | - Noppadol Kietsiriroje
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, Leeds, United Kingdom
- Endocrinology and Metabolism Unit, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Hatyai, Songkhla, Thailand
| | - Mansour Almutairi
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, Leeds, United Kingdom
- General Directorate of Medical Services, Ministry of Interior, Kingdom of Saudi Arabia
| | - Katie Simmons
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, Leeds, United Kingdom
| | - Nikoletta Pechlivani
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, Leeds, United Kingdom
| | - Sreenivasan Ponnambalam
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, UK
| | - Ramzi A. Ajjan
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, Leeds, United Kingdom
| |
Collapse
|
7
|
Pintér P, Alpár A. The Role of Extracellular Matrix in Human Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms231911085. [PMID: 36232390 PMCID: PMC9569603 DOI: 10.3390/ijms231911085] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
The dense neuropil of the central nervous system leaves only limited space for extracellular substances free. The advent of immunohistochemistry, soon followed by advanced diagnostic tools, enabled us to explore the biochemical heterogeneity and compartmentalization of the brain extracellular matrix in exploratory and clinical research alike. The composition of the extracellular matrix is critical to shape neuronal function; changes in its assembly trigger or reflect brain/spinal cord malfunction. In this study, we focus on extracellular matrix changes in neurodegenerative disorders. We summarize its phenotypic appearance and biochemical characteristics, as well as the major enzymes which regulate and remodel matrix establishment in disease. The specifically built basement membrane of the central nervous system, perineuronal nets and perisynaptic axonal coats can protect neurons from toxic agents, and biochemical analysis revealed how the individual glycosaminoglycan and proteoglycan components interact with these molecules. Depending on the site, type and progress of the disease, select matrix components can either proactively trigger the formation of disease-specific harmful products, or reactively accumulate, likely to reduce tissue breakdown and neuronal loss. We review the diagnostic use and the increasing importance of medical screening of extracellular matrix components, especially enzymes, which informs us about disease status and, better yet, allows us to forecast illness.
Collapse
Affiliation(s)
- Panka Pintér
- Department of Anatomy, Semmelweis University, 1113 Budapest, Hungary
| | - Alán Alpár
- Department of Anatomy, Semmelweis University, 1113 Budapest, Hungary
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, 1051 Budapest, Hungary
- Correspondence:
| |
Collapse
|
8
|
Brainer Clares de Andrade J, Mohr JP, Oliveira Lima F, José de Freitas Carvalho J, Andre Castro de Oliveira R, Coelho Maia Barros L, Sampaio Silva G. Predictors of hemorrhagic transformation differences between patients treated or not with reperfusion therapy. J Clin Neurosci 2022; 101:9-15. [DOI: 10.1016/j.jocn.2022.04.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 02/08/2022] [Accepted: 04/23/2022] [Indexed: 11/25/2022]
|
9
|
Tang MY, Gorin FA, Lein PJ. Review of evidence implicating the plasminogen activator system in blood-brain barrier dysfunction associated with Alzheimer's disease. AGEING AND NEURODEGENERATIVE DISEASES 2022; 2. [PMID: 35156107 PMCID: PMC8830591 DOI: 10.20517/and.2022.05] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Elucidating the pathogenic mechanisms of Alzheimer’s disease (AD) to identify therapeutic targets has been the focus of many decades of research. While deposition of extracellular amyloid-beta plaques and intraneuronal neurofibrillary tangles of hyperphosphorylated tau have historically been the two characteristic hallmarks of AD pathology, therapeutic strategies targeting these proteinopathies have not been successful in the clinics. Neuroinflammation has been gaining more attention as a therapeutic target because increasing evidence implicates neuroinflammation as a key factor in the early onset of AD disease progression. The peripheral immune response has emerged as an important contributor to the chronic neuroinflammation associated with AD pathophysiology. In this context, the plasminogen activator system (PAS), also referred to as the vasculature’s fibrinolytic system, is emerging as a potential factor in AD pathogenesis. Evolving evidence suggests that the PAS plays a role in linking chronic peripheral inflammatory conditions to neuroinflammation in the brain. While the PAS is better known for its peripheral functions, components of the PAS are expressed in the brain and have been demonstrated to alter neuroinflammation and blood-brain barrier (BBB) permeation. Here, we review plasmin-dependent and -independent mechanisms by which the PAS modulates the BBB in AD pathogenesis and discuss therapeutic implications of these observations.
Collapse
Affiliation(s)
- Mei-Yun Tang
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Fredric A Gorin
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA.,Department of Neurology, School of Medicine, University of California, Davis, CA 95616, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| |
Collapse
|
10
|
Gubern-Mérida C, Comajoan P, Huguet G, García-Yebenes I, Lizasoain I, Moro MA, Puig-Parnau I, Sánchez JM, Serena J, Kádár E, Castellanos M. Cav-1 Protein Levels in Serum and Infarcted Brain Correlate with Hemorrhagic Volume in a Mouse Model of Thromboembolic Stroke, Independently of rt-PA Administration. Mol Neurobiol 2022; 59:1320-1332. [PMID: 34984586 DOI: 10.1007/s12035-021-02644-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 11/11/2021] [Indexed: 12/27/2022]
Abstract
Thrombolytic therapy with recombinant tissue plasminogen activator (rt-PA) is currently the only FDA-approved drug for acute ischemic stroke. However, its administration is still limited due to the associated increased risk of hemorrhagic transformation (HT). rt-PA may exacerbate blood-brain barrier (BBB) injury by several mechanisms that have not been fully elucidated. Caveolin-1 (Cav-1), a major structural protein of caveolae, has been linked to the endothelial barrier function. The effects of rt-PA on Cav-1 expression remain largely unknown. Here, Cav-1 protein expression after ischemic conditions, with or without rt-PA administration, was analyzed in a murine thromboembolic middle cerebral artery occlusion (MCAO) and in brain microvascular endothelial bEnd.3 cells subjected to oxygen/glucose deprivation (OGD). Our results show that Cav-1 is overexpressed in endothelial cells of infarcted area and in bEnd.3 cell line after ischemia but there is disagreement regarding rt-PA effects on Cav-1 expression between both experimental models. Delayed rt-PA administration significantly reduced Cav-1 total levels from 24 to 72 h after reoxygenation and increased pCav-1/Cav-1 at 72 h in the bEnd.3 cells while it did not modify Cav-1 immunoreactivity in the infarcted area at 24 h post-MCAO. Importantly, tissue Cav-1 positively correlated with Cav-1 serum levels at 24 h post-MCAO and negatively correlated with the volume of hemorrhage after infarction, the latter supporting a protective role of Cav-1 in cerebral ischemia. In addition, the negative association between baseline serum Cav-1 levels and hemorrhagic volume points to a potential usefulness of baseline serum Cav-1 levels to predict hemorrhagic volume, independently of rt-PA administration.
Collapse
Affiliation(s)
- Carme Gubern-Mérida
- Cerebrovascular Pathology Research Group, Department of Neurology, Girona Biomedical Research Institute (IDIBGI), Parc Hospitalari Martí i Julià, C/Dr. Castany s/n, M2 Building, 17190, Salt, Girona, Spain.,Cellular and Molecular Neurobiology Research Group, Department of Biology, University of Girona (UdG), Aulari Comú building, C/Maria Aurèlia Capmany 40, 17003, Girona, Spain
| | - Pau Comajoan
- Cerebrovascular Pathology Research Group, Department of Neurology, Girona Biomedical Research Institute (IDIBGI), Parc Hospitalari Martí i Julià, C/Dr. Castany s/n, M2 Building, 17190, Salt, Girona, Spain.,Cellular and Molecular Neurobiology Research Group, Department of Biology, University of Girona (UdG), Aulari Comú building, C/Maria Aurèlia Capmany 40, 17003, Girona, Spain
| | - Gemma Huguet
- Cerebrovascular Pathology Research Group, Department of Neurology, Girona Biomedical Research Institute (IDIBGI), Parc Hospitalari Martí i Julià, C/Dr. Castany s/n, M2 Building, 17190, Salt, Girona, Spain.,Cellular and Molecular Neurobiology Research Group, Department of Biology, University of Girona (UdG), Aulari Comú building, C/Maria Aurèlia Capmany 40, 17003, Girona, Spain
| | - Isaac García-Yebenes
- Neurovascular Research Unit, Department of Pharmacology and Toxicology and Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Hospital 12 de Octubre (i+12), Complutense University of Madrid (UCM), Pza. Ramón y Cajal s/n, 28040, Madrid, Spain
| | - Ignacio Lizasoain
- Neurovascular Research Unit, Department of Pharmacology and Toxicology and Instituto Universitario de Investigación en Neuroquímica (IUIN), Instituto de Investigación Hospital 12 de Octubre (i+12), Complutense University of Madrid (UCM), Pza. Ramón y Cajal s/n, 28040, Madrid, Spain
| | - María Angeles Moro
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Irene Puig-Parnau
- Cellular and Molecular Neurobiology Research Group, Department of Biology, University of Girona (UdG), Aulari Comú building, C/Maria Aurèlia Capmany 40, 17003, Girona, Spain
| | - Juan Manuel Sánchez
- Cerebrovascular Pathology Research Group, Department of Neurology, Girona Biomedical Research Institute (IDIBGI), Parc Hospitalari Martí i Julià, C/Dr. Castany s/n, M2 Building, 17190, Salt, Girona, Spain.,Analytical and Environmental Chemistry Research Group, Department of Chemistry, University of Girona (UdG), C/Maria Aurèlia Capmany 69, 17003, Girona, Spain
| | - Joaquín Serena
- Cerebrovascular Pathology Research Group, Department of Neurology, Girona Biomedical Research Institute (IDIBGI), Parc Hospitalari Martí i Julià, C/Dr. Castany s/n, M2 Building, 17190, Salt, Girona, Spain.,Cellular and Molecular Neurobiology Research Group, Department of Biology, University of Girona (UdG), Aulari Comú building, C/Maria Aurèlia Capmany 40, 17003, Girona, Spain
| | - Elisabet Kádár
- Cerebrovascular Pathology Research Group, Department of Neurology, Girona Biomedical Research Institute (IDIBGI), Parc Hospitalari Martí i Julià, C/Dr. Castany s/n, M2 Building, 17190, Salt, Girona, Spain. .,Cellular and Molecular Neurobiology Research Group, Department of Biology, University of Girona (UdG), Aulari Comú building, C/Maria Aurèlia Capmany 40, 17003, Girona, Spain.
| | - Mar Castellanos
- Department of Neurology, A Coruña University Hospital/A Coruña Biomedical Research Institute, Xubias de Arriba 84, 15006A, Coruña, Spain.
| |
Collapse
|
11
|
Han S, Zhang D, Dong Q, Wang X, Wang L. Deficiency in Neuroserpin Exacerbates CoCl 2 Induced Hypoxic Injury in the Zebrafish Model by Increased Oxidative Stress. Front Pharmacol 2021; 12:632662. [PMID: 33737878 PMCID: PMC7960655 DOI: 10.3389/fphar.2021.632662] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/27/2021] [Indexed: 01/17/2023] Open
Abstract
Protective strategy against hypoxic-ischemic (H/I) induced injury has been intensively discussed. Neuroserpin, an inhibitor for tissue plasminogen activator (tPA), has been proved a vital neuroprotective agent in cerebral ischemia mouse model and oxygen-glucose deprivation and reoxygenation (OGD/R) cell model. Neuroserpin is a promising therapeutic hint for neonatal hypoxic-ischemia injury. Here, we established a neuroserpin deficient zebrafish to study its role in CoCl2 chemically induced hypoxic injury. CoCl2 exposure was beginning at the embryonic stage. Development defects, neuronal loss, and vascular malformation was assessed by imaging microscopy. Neuroserpin deficient zebrafish showed more development defects, neuronal loss and vascular malformation compared to wide-type. Apoptosis and oxidative stress were evaluated to further identify the possible mechanisms. These findings indicate that neuroserpin could protective against CoCl2 induced hypoxic injury by alleviating oxidative stress.
Collapse
Affiliation(s)
- Sha Han
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Dongyang Zhang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xu Wang
- Cancer Metabolism Laboratory, Cancer Research Institute Fudan University Shanghai Cancer Center, Shanghai, China
| | - Liang Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Ma H, Jiang Z, Xu J, Liu J, Guo ZN. Targeted nano-delivery strategies for facilitating thrombolysis treatment in ischemic stroke. Drug Deliv 2021; 28:357-371. [PMID: 33517820 PMCID: PMC8725844 DOI: 10.1080/10717544.2021.1879315] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Ischemic stroke is one of the major causes of severe disability and death worldwide. It is mainly caused by a sudden reduction in cerebral blood flow due to obstruction of the supplying vessel by thrombi and subsequent initiation of a complex cascade of pathophysiological changes, which ultimately lead to brain ischemia and even irreversible infarction. Thus, timely and effective thrombolysis therapy remains a mainstay for acute ischemic stroke treatment. Tissue plasminogen activator (tPA), the only thrombolytic agent approved globally, provides substantial benefits by exerting a fibrinolysis effect, recovering the blood supply in occluded vessels and, thereby, salvaging the ischemic tissue. However, the clinical application of tPA was limited because of a few unsolved issues, such as a narrow therapeutic window, hemorrhagic complications, and limited thrombolytic efficacy, especially, for large thrombi. With the prosperous development of nanotechnology, a series of targeted delivery strategies and nanocomposites have been extensively investigated for delivering thrombolytic agents to facilitate thrombolysis treatment. Excitingly, numerous novel attempts have been reported to be effective in extending the half-life, targeting the thrombus site, and improving the thrombolytic efficacy in preclinical models. This article begins with a brief introduction to ischemic stroke, then describes the current state of thrombolysis treatment and, finally, introduces the application of various nanotechnology-based strategies for targeted delivery of thrombolytic agents. Representative studies are reviewed according to diverse strategies and nano-formulations, with the aim of providing integrated and up-to-date information and to improve the development of thrombolysis treatment for stroke patients.
Collapse
Affiliation(s)
- Hongyin Ma
- Department of Neurology, The First Hospital of Jilin University, ChangChun, China
| | - Zhenmin Jiang
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, ChangChun, China
| | - Jiayun Xu
- State Key Lab of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, China.,College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou, China
| | - Junqiu Liu
- State Key Lab of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, China.,College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou, China
| | - Zhen-Ni Guo
- Department of Neurology, The First Hospital of Jilin University, ChangChun, China
| |
Collapse
|
13
|
JLX001 attenuates blood-brain barrier dysfunction in MCAO/R rats via activating the Wnt/β-catenin signaling pathway. Life Sci 2020; 260:118221. [PMID: 32768578 DOI: 10.1016/j.lfs.2020.118221] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/28/2020] [Accepted: 08/03/2020] [Indexed: 12/19/2022]
Abstract
JLX001, a new dihydrochloride of Cyclovirobuxine D (CVB-D), has bioactivities against ischemia injury. The blood-brain barrier (BBB) disruption is involved in the pathogeneses of ischemic stroke. This study was designed to explore the effect and potential mechanism of JLX001 on the BBB after ischemic stroke. Rats were subjected to middle cerebral artery occlusion/reperfusion (MCAO/R) to mimic cerebral ischemia in vivo. In vitro, rat primary brain microvascular endothelial cells (PBMECs) were cultured and exposed to oxygen-glucose deprivation/reoxygenation (OGD/R). Posttreatment of JLX001 for 15 days after MCAO/R improved the behavior, learning and memory ability. Pretreatment of JLX001 for 3 days significantly attenuated infarct volume, lessened brain edema, mitigated BBB disruption and decreased the neurological deficit score in MCAO/R rats. Moreover, JLX001 increased cell viability and reduced sodium fluorescein leakage after OGD/R injury. In addition, JLX001 increased the expressions of Claudin-5 and Occludin, decreased the expression of MMP-9 both in vivo and in vitro. Moreover, immunofluorescence staining and western immunoblotting results showed that JLX001 increased the expressions of tight junction proteins via activating Wnt/β-catenin signal pathway in vivo and in vitro, which may be associated with the activation of PI3K/Akt signaling. Besides, XAV939 (an inhibitor of the Wnt/β-catenin pathway) proved the connection of JLX001 and Wnt/β-catenin pathway. These results suggest that JLX001 alleviates BBB disruption after MCAO/R and OGD/R possibly by alleviating MMP-9 and activating the Wnt/β-catenin signaling pathway.
Collapse
|
14
|
Safety and effectiveness of lumbar cerebrospinal fluid drainage to prevent delayed cerebral ischemia after Fisher grade 3 subarachnoid hemorrhage with minimal intraventricular hemorrhage. Neurochirurgie 2020; 66:225-231. [DOI: 10.1016/j.neuchi.2020.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/22/2020] [Accepted: 03/07/2020] [Indexed: 01/29/2023]
|
15
|
Welcome MO, Mastorakis NE. Stress-induced blood brain barrier disruption: Molecular mechanisms and signaling pathways. Pharmacol Res 2020; 157:104769. [PMID: 32275963 DOI: 10.1016/j.phrs.2020.104769] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/09/2020] [Accepted: 03/19/2020] [Indexed: 02/07/2023]
Abstract
Stress is a nonspecific response to a threat or noxious stimuli with resultant damaging consequences. Stress is believed to be an underlying process that can trigger central nervous system disorders such as depression, anxiety, and post-traumatic stress disorder. Though the pathophysiological basis is not completely understood, data have consistently shown a pivotal role of inflammatory mediators and hypothalamo-pituitary-adrenal (HPA) axis activation in stress induced disorders. Indeed emerging experimental evidences indicate a concurrent activation of inflammatory signaling pathways and not only the HPA axis, but also, peripheral and central renin-angiotensin system (RAS). Furthermore, recent experimental data indicate that the HPA and RAS are coupled to the signaling of a range of central neuro-transmitter, -mediator and -peptide molecules that are also regulated, at least in part, by inflammatory signaling cascades and vice versa. More recently, experimental evidences suggest a critical role of stress in disruption of the blood brain barrier (BBB), a neurovascular unit that regulates the movement of substances and blood-borne immune cells into the brain parenchyma, and prevents peripheral injury to the brain substance. However, the mechanisms underlying stress-induced BBB disruption are not exactly known. In this review, we summarize studies conducted on the effects of stress on the BBB and integrate recent data that suggest possible molecular mechanisms and signaling pathways underlying stress-induced BBB disruption. Key molecular targets and pharmacological candidates for treatment of stress and related illnesses are also summarized.
Collapse
Affiliation(s)
- Menizibeya O Welcome
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Nile University of Nigeria, Abuja, Nigeria.
| | | |
Collapse
|
16
|
Mizrachi T, Gur-Wahnon D, Al-Roof Higazi A, Brenner T. Role of tissue plasminogen activator in clinical aggravation of experimental autoimmune encephalomyelitis and its therapeutic potential. Cell Immunol 2020; 348:104040. [PMID: 31955841 DOI: 10.1016/j.cellimm.2020.104040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/05/2020] [Accepted: 01/09/2020] [Indexed: 10/25/2022]
Abstract
Tissue plasminogen activator (tPA), a component of the plasminogen activator (PA) system, is elevated in inflammatory neurological disorders. In the present study, we explored the immunomodulatory activity of tPA in experimental autoimmune encephalomyelitis (EAE). The EAE was treated with two catalytic inactive tPA variant proteins: S(481)A and S(481)A + KHRR(296-299)AAAA. EAE-induced tPA-/- mice presented with markedly more severe disease than wt EAE mice. Further, treatment with tPA variants, demonstrated a significant suppression of disease severity in tPA-/- and wt mice. Immunological evaluation showed that specific T-cell reactivity was markedly reduced in the tPA-/- animals, as indicated by decreased T-cell reactivity and reduction in T-regulatory cells. The current findings indicate that tPA plays a role in the pathogenesis of EAE. Moreover, successful amelioration of EAE was achieved by administration of tPA variant proteins. This might mean that these proteins have potential for the immunomodulation of neuroinflammation.
Collapse
Affiliation(s)
- Tehila Mizrachi
- Laboratory of Neuroimmunology, Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, PO Box 12000, Jerusalem, Israel
| | - Devorah Gur-Wahnon
- Laboratory of Neuroimmunology, Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, PO Box 12000, Jerusalem, Israel
| | - Abd Al-Roof Higazi
- Department of Biochemistry, Hadassah-Hebrew University Medical Center, PO Box 12000, Jerusalem, Israel
| | - Talma Brenner
- Laboratory of Neuroimmunology, Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, PO Box 12000, Jerusalem, Israel.
| |
Collapse
|
17
|
Disharoon D, Marr DW, Neeves KB. Engineered microparticles and nanoparticles for fibrinolysis. J Thromb Haemost 2019; 17:2004-2015. [PMID: 31529593 PMCID: PMC6893081 DOI: 10.1111/jth.14637] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/09/2019] [Accepted: 09/12/2019] [Indexed: 12/28/2022]
Abstract
Fibrinolytic agents including plasmin and plasminogen activators improve outcomes in acute ischemic stroke and thrombosis by recanalizing occluded vessels. In the decades since their introduction into clinical practice, several limitations of have been identified in terms of both efficacy and bleeding risk associated with these agents. Engineered nanoparticles and microparticles address some of these limitations by improving circulation time, reducing inhibition and degradation in circulation, accelerating recanalization, improving targeting to thrombotic occlusions, and reducing off-target effects; however, many particle-based approaches have only been used in preclinical studies to date. This review covers four advances in coupling fibrinolytic agents with engineered particles: (a) modifications of plasminogen activators with macromolecules, (b) encapsulation of plasminogen activators and plasmin in polymer and liposomal particles, (c) triggered release of encapsulated fibrinolytic agents and mechanical disruption of clots with ultrasound, and (d) enhancing targeting with magnetic particles and magnetic fields. Technical challenges for the translation of these approaches to the clinic are discussed.
Collapse
Affiliation(s)
- Dante Disharoon
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO
| | - David W.M. Marr
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO
| | - Keith B. Neeves
- Departments of Bioengineering and Pediatrics, Hemophilia and Thrombosis Center, University of Colorado Denver | Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
18
|
Yang E, Cai Y, Yao X, Liu J, Wang Q, Jin W, Wu Q, Fan W, Qiu L, Kang C, Wu J. Tissue plasminogen activator disrupts the blood-brain barrier through increasing the inflammatory response mediated by pericytes after cerebral ischemia. Aging (Albany NY) 2019; 11:10167-10182. [PMID: 31740626 PMCID: PMC6914411 DOI: 10.18632/aging.102431] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/28/2019] [Indexed: 12/20/2022]
Abstract
Pericytes, important elements of the blood-brain barrier (BBB), play critical roles in maintaining BBB integrity and modulating hemostasis, angiogenesis, inflammation and phagocytic function. We investigated whether pericytes are involved in the recombinant tissue plasminogen activator (rt-PA)-induced inflammatory response, which disrupts the BBB, and investigated the potential mechanisms. Middle cerebral artery occlusion (MCAO) and oxygen-glucose deprivation (OGD) were employed to mimic hypoxic-ischemic conditions. Rt-PA was intravenously injected into mice 1 h after 1 h MCAO, and Rt-PA was added to the culture medium after 4 h OGD. Rt-PA treatment aggravated the disruption of the BBB compared with hypoxia treatment, and etanercept (TNF-α inhibitor) combined with rt-PA alleviated the rt-PA-induced BBB disruption in vivo and in vitro. Rt-PA treatment increased the TNF-α and MCP-1 levels and decreased the TGF-β, p-Smad2/3 and PDGFR-β levels compared with hypoxia treatment in vivo and vitro. TGF-β combined with rt-PA decreased TNF-α and MCP-1 secretion and alleviated BBB disruption compared with rt-PA; these changes were abrogated by TPO427736 HCL (a TGF-β/p-Smad2/3 pathway inhibitor) cotreatment in vitro. Rt-PA did not decrease TGF-β and p-Smad2/3 expression in PDGFR-β-overexpressing pericytes after OGD. These findings identify PDGFR-β/TGF-β/p-Smad2/3 signaling in pericytes as a new therapeutic target for the treatment of rt-PA-induced BBB damage.
Collapse
Affiliation(s)
- Eryan Yang
- Graduate School of Tianjin Medical University, Tianjin 300070, China.,Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China.,Department of Neurology, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Ying Cai
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Xiuhua Yao
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Ji Liu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Qixue Wang
- Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Weili Jin
- Graduate School of Tianjin Medical University, Tianjin 300070, China.,Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Qiaoli Wu
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Weijia Fan
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Lina Qiu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Chunsheng Kang
- Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jialing Wu
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China.,Department of Neurology, Tianjin Huanhu Hospital, Tianjin 300350, China
| |
Collapse
|
19
|
Bhogal P, AlMatter M, Hellstern V, Pérez MA, Ganslandt O, Bäzner H, Henkes H. Mechanical thrombectomy for recurrent large vessel occlusion. J Clin Neurosci 2019; 66:107-112. [PMID: 31113697 DOI: 10.1016/j.jocn.2019.05.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 04/13/2019] [Accepted: 05/07/2019] [Indexed: 11/16/2022]
Abstract
Although mechanical thrombectomy (MT) has been shown in numerous trials to be a successful treatment option for patients with large vessel occlusion (LVO), there is limited information on the safety and effectiveness of the technique in cases of recurrent LVO. To this end, we performed a retrospective review of our prospectively maintained database to identify all patients that had undergone more than one MT procedure January 2008 and January 2018. The data collected on these patients included baseline demographics and any history of diabetes mellitus, hypercholesterolaemia, hypertension, atrial fibrillation or smoking. We also recorded when the symptoms had started, the patient's NIHSS and ASPECT scores, the number of passes taken, the patient's final TICI score, any complications which arose and the patient's mRS at 90 days. Our dataset encompassed 25 patients (of which 17 (68%) were female) who had undergone a total number of 52 MT's. Average age at 1st presentation was 70 ± 12.8 years. The median time between one stroke and the next was 71 days (range 1-1059, IQR 183 days). The majority of the strokes were deemed cardioembolic (86.5%) in origin. There was no significant difference in the procedure times, number of passes or TICI scores. There was a significant difference in the mRS after the 1st and 2nd events (p = 0.014) but no significant difference if the 2nd event occurred contralateral to the 1st event (p = 0.66) (n = 22). It is therefore concluded that recurrent thrombectomy can be safely performed with no significant difference in the technical aspects of the procedure.
Collapse
Affiliation(s)
- P Bhogal
- The Royal London Hospital, Whitechapel Road, London, UK.
| | - M AlMatter
- Neurological Clinic, Neurocenter, Klinikum Stuttgart, Germany
| | - V Hellstern
- Neurological Clinic, Neurocenter, Klinikum Stuttgart, Germany
| | - M Aguilar Pérez
- Neurological Clinic, Neurocenter, Klinikum Stuttgart, Germany
| | - O Ganslandt
- Neurosurgical Clinic, Neurocenter, Klinikum Stuttgart, Germany
| | - H Bäzner
- Neurological Clinic, Neurocenter, Klinikum Stuttgart, Germany
| | - H Henkes
- Neurological Clinic, Neurocenter, Klinikum Stuttgart, Germany; Medical Faculty, University Duisburg-Essen, Germany
| |
Collapse
|
20
|
Canonical Wnt Pathway Maintains Blood-Brain Barrier Integrity upon Ischemic Stroke and Its Activation Ameliorates Tissue Plasminogen Activator Therapy. Mol Neurobiol 2019; 56:6521-6538. [PMID: 30852795 DOI: 10.1007/s12035-019-1539-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/22/2019] [Indexed: 12/22/2022]
Abstract
Stroke induces blood-brain barrier (BBB) breakdown, which promotes complications like oedema and hemorrhagic transformation. Administration of recombinant tissue plasminogen activator (rtPA) within a therapeutic time window of 4.5 h after stroke onset constitutes the only existing treatment. Beyond this time window, rtPA worsens BBB breakdown. Canonical Wnt pathway induces BBB formation and maturation during ontogeny. We hypothesized that the pathway is required to maintain BBB functions after stroke; thus, its activation might improve rtPA therapy. Therefore, we first assessed pathway activity in the brain of mice subjected to transient middle cerebral artery occlusion (MCAo). Next, we evaluated the effect of pathway deactivation early after stroke onset on BBB functions. Finally, we assessed the impact of pathway activation on BBB breakdown associated to delayed administration of rtPA. Our results show that pathway activity is induced predominately in endothelial cells early after ischemic stroke. Early deactivation of the pathway using a potent inhibitor, XAV939, aggravates BBB breakdown and increases hemorrhagic transformation incidence. On the other hand, pathway activation using a potent activator, 6-bromoindirubin-3'-oxime (6-BIO), reduces the incidence of hemorrhagic transformation associated to delayed rtPA administration by attenuating BBB breakdown via promotion of tight junction formation and repressing endothelial basal permeability independently of rtPA proteolytic activity. BBB preservation upon pathway activation limited the deleterious effects of delayed rtPA administration. Our study demonstrates that activation of the canonical Wnt pathway constitutes a clinically relevant strategy to extend the therapeutic time window of rtPA by attenuating BBB breakdown via regulation of BBB-specific mechanisms.
Collapse
|
21
|
González-Miguel J, Valero MA, Reguera-Gomez M, Mas-Bargues C, Bargues MD, Simón F, Mas-Coma S. Numerous Fasciola plasminogen-binding proteins may underlie blood-brain barrier leakage and explain neurological disorder complexity and heterogeneity in the acute and chronic phases of human fascioliasis. Parasitology 2019; 146:284-298. [PMID: 30246668 PMCID: PMC6402360 DOI: 10.1017/s0031182018001464] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/17/2022]
Abstract
Human fascioliasis is a worldwide, pathogenic food-borne trematodiasis. Impressive clinical pictures comprising puzzling polymorphisms, manifestation multifocality, disease evolution changes, sequelae and mortality, have been reported in patients presenting with neurological, meningeal, neuropsychic and ocular disorders caused at distance by flukes infecting the liver. Proteomic and mass spectrometry analyses of the Fasciola hepatica excretome/secretome identified numerous, several new, plasminogen-binding proteins enhancing plasmin generation. This may underlie blood-brain barrier leakage whether by many simultaneously migrating, small-sized juvenile flukes in the acute phase, or by breakage of encapsulating formations triggered by single worm tracks in the chronic phase. Blood-brain barrier leakages may subsequently occur due to a fibrinolytic system-dependent mechanism involving plasmin-dependent generation of the proinflammatory peptide bradykinin and activation of bradykinin B2 receptors, after different plasminogen-binding protein agglomeration waves. Interactions between diverse parasitic situations and non-imbalancing fibrinolysis system alterations are for the first time proposed that explain the complexity, heterogeneity and timely variations of neurological disorders. Additionally, inflammation and dilation of blood vessels may be due to contact system-dependent generation bradykinin. This baseline allows for search of indicators to detect neurological risk in fascioliasis patients and experimental work on antifibrinolytic treatments or B2 receptor antagonists for preventing blood-brain barrier leakage.
Collapse
Affiliation(s)
- J. González-Miguel
- Laboratorio de Parasitología, Instituto de Recursos Naturales y Agrobiología de Salamanca (IRNASA-CSIC), Cordel de Merinas 40-52, 37008 Salamanca, Spain
| | - M. A. Valero
- Departamento de Parasitología, Facultad de Farmacia, Universidad de Valencia, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Valencia, Spain
| | - M. Reguera-Gomez
- Departamento de Parasitología, Facultad de Farmacia, Universidad de Valencia, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Valencia, Spain
| | - C. Mas-Bargues
- Departamento de Fisiología, Facultad de Medicina, Universidad de Valencia, Av. Blasco Ibáñez No. 15, 46010 Valencia, Spain
| | - M. D. Bargues
- Departamento de Parasitología, Facultad de Farmacia, Universidad de Valencia, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Valencia, Spain
| | - F. Simón
- Área de Parasitología, Facultad de Farmacia, Universidad de Salamanca, Av. Licenciado Méndez Nieto s/n, 37007 Salamanca, Spain
| | - S. Mas-Coma
- Departamento de Parasitología, Facultad de Farmacia, Universidad de Valencia, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Valencia, Spain
| |
Collapse
|
22
|
de Oliveira JR, Camargo SEA, de Oliveira LD. Rosmarinus officinalis L. (rosemary) as therapeutic and prophylactic agent. J Biomed Sci 2019; 26:5. [PMID: 30621719 PMCID: PMC6325740 DOI: 10.1186/s12929-019-0499-8] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 01/02/2019] [Indexed: 12/22/2022] Open
Abstract
Rosmarinus officinalis L. (rosemary) is a medicinal plant native to the Mediterranean region and cultivated around the world. Besides the therapeutic purpose, it is commonly used as a condiment and food preservative. R. officinalis L. is constituted by bioactive molecules, the phytocompounds, responsible for implement several pharmacological activities, such as anti-inflammatory, antioxidant, antimicrobial, antiproliferative, antitumor and protective, inhibitory and attenuating activities. Thus, in vivo and in vitro studies were presented in this Review, approaching the therapeutic and prophylactic effects of R. officinalis L. on some physiological disorders caused by biochemical, chemical or biological agents. In this way, methodology, mechanisms, results, and conclusions were described. The main objective of this study was showing that plant products could be equivalent to the available medicines.
Collapse
Affiliation(s)
- Jonatas Rafael de Oliveira
- Departamento de Biociências e Diagnóstico Bucal, Instituto de Ciência e Tecnologia, Universidade Estadual Paulista (UNESP), Av. Engenheiro Francisco José Longo, 777 - Jardim São Dimas, São José dos Campos, SP, CEP 12245-000, Brazil.
| | | | - Luciane Dias de Oliveira
- Departamento de Biociências e Diagnóstico Bucal, Instituto de Ciência e Tecnologia, Universidade Estadual Paulista (UNESP), Av. Engenheiro Francisco José Longo, 777 - Jardim São Dimas, São José dos Campos, SP, CEP 12245-000, Brazil
| |
Collapse
|
23
|
Gong P, Li M, Zou C, Tian Q, Xu Z. Tissue Plasminogen Activator Causes Brain Microvascular Endothelial Cell Injury After Oxygen Glucose Deprivation by Inhibiting Sonic Hedgehog Signaling. Neurochem Res 2018; 44:441-449. [PMID: 30552546 PMCID: PMC6394519 DOI: 10.1007/s11064-018-2697-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 11/22/2018] [Accepted: 12/10/2018] [Indexed: 11/29/2022]
Abstract
The thrombolytic activity of tissue plasminogen activator (tPA) has undisputed benefits. However, the documented neurotoxicity of tPA raises important issues. Currently, common treatments for stroke might not be optimum if exogenous tPA can pass through the blood–brain barrier and enter the brain, thus adding to the deleterious effects of tPA within the cerebral parenchyma. Here, we determined whether tPA could damage brain microvascular endothelial cells (BMECs) during cerebral ischemia. We showed that treatment of BMECs with tPA decreased trans-endothelial electrical resistance and cell proliferation, and blocked the cell cycle at the G0–G1 phase. In addition, the Sonic hedgehog (Shh) signaling pathway was involved in tPA-induced BMECs dysfunction. However, tPA-enhanced oxygen glucose deprivation-induced BMECs dysfunction was eliminated by Shh administration and the effects could be reversed by Shh inhibitors. Taken together, these results demonstrate that tPA administration might result in damage to the endothelial barrier owing to blocked Shh signaling pathway.
Collapse
Affiliation(s)
- Pian Gong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, No. 9 Zhangzhidong Road, Wuchang District, Wuhan, 430072, Hubei Province, China
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, No. 9 Zhangzhidong Road, Wuchang District, Wuhan, 430072, Hubei Province, China.
| | - Changlin Zou
- Department of Neurosurgery, Renmin Hospital of Wuhan University, No. 9 Zhangzhidong Road, Wuchang District, Wuhan, 430072, Hubei Province, China
| | - Qi Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, No. 9 Zhangzhidong Road, Wuchang District, Wuhan, 430072, Hubei Province, China
| | - Zhou Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, No. 9 Zhangzhidong Road, Wuchang District, Wuhan, 430072, Hubei Province, China
| |
Collapse
|
24
|
Li F, Yang B, Li T, Gong X, Zhou F, Hu Z. HSPB8 over-expression prevents disruption of blood-brain barrier by promoting autophagic flux after cerebral ischemia/reperfusion injury. J Neurochem 2018; 148:97-113. [PMID: 30422312 DOI: 10.1111/jnc.14626] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/13/2018] [Accepted: 10/24/2018] [Indexed: 12/24/2022]
Abstract
Heat-shock protein B8 (HSPB8) has been recently reported to confer neuroprotection against ischemia/reperfusion (I/R)-induced cerebral injury in vivo and in vitro. However, the molecular mechanism is still elusive. This study focused on the effect of intracerebroventricular (i.c.v) delivery of lenti-HSPB8 virus against neurological injury in a rat model of cerebral I/R and explored the underlying mechanism. We found that lentivirus i.c.v injection-induced HSPB8 over-expression strongly alleviated infarct volume, improved neurobehavioral outcomes, and reduced brain edema in rat middle cerebral artery occlusion/reperfusion (MCAO/R) model. Concomitantly, HSPB8 over-expression noticeably prevented blood-brain barrier (BBB) disruption after cerebral I/R injury as indicated by the reduction in Evans blue leakage and IgG detection in the ipsilateral hemisphere compared with the vehicle group. Moreover, immunoblotting and immunofluorescence staining of tight junction proteins claudin-5 and occludin showed that HSPB8 over-expression prevented the degradation of these proteins induced by MCAO/R, which indicated the protective effect of HSPB8 on BBB. Western blotting and immunostaining techniques were also utilized to analyze the expression of the markers of autophagy. We found that HSPB8 over-expression promoted autophagic flux, evidenced by increased ratio of LC3 I/II, accumulation of Beclin-1 expression and enhanced p62 degradation. i.c.v injection of 15 μg autophagy inhibitor 3-methyladenine (3-MA) was applied at the onset of reperfusion. The results showed that 3-MA elicited a significant loss of the protective effect of HSPB8 against MCAO/R-induced neurological defect, Evans blue extravasation, and the loss tight junction proteins, suggesting that the BBB protective role of HSPB8 was, at least in part, mediated through autophagy. Collectively, HSPB8 may represent a potential therapeutic agent for preserving BBB integrity following cerebral I/R injury. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/. Cover Image for this issue: doi: 10.1111/jnc.14488.
Collapse
Affiliation(s)
- Fazhao Li
- Department of General Surgery, 2nd Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Binbin Yang
- Department of Neurology, 2nd Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Ting Li
- Department of Neurology, 2nd Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xiyu Gong
- Department of Neurology, 2nd Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Fangfang Zhou
- Department of Neurology, 2nd Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Zhiping Hu
- Department of Neurology, 2nd Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
25
|
Henderson SJ, Weitz JI, Kim PY. Fibrinolysis: strategies to enhance the treatment of acute ischemic stroke. J Thromb Haemost 2018; 16:1932-1940. [PMID: 29953716 DOI: 10.1111/jth.14215] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Indexed: 02/03/2023]
Abstract
Stroke is a major cause of disability worldwide, and is the second leading cause of death after ischemic heart disease. Until recently, tissue-type plasminogen activator (t-PA) was the only treatment for acute ischemic stroke. If administered within 4.5 h of symptom onset, t-PA improves the outcome in stroke patients. Mechanical thrombectomy is now the preferred treatment for patients with acute ischemic stroke resulting from a large-artery occlusion in the anterior circulation. However, the widespread use of mechanical thrombectomy is limited by two factors. First, only ⁓ 10% of patients with acute ischemic stroke have a proximal large-artery occlusion in the anterior circulation and present early enough to undergo mechanical thrombectomy within 6 h; an additional 9-10% of patients presenting within the 6-24-h time window may also qualify for the procedure. Second, not all stroke centers have the resources or expertise to perform mechanical thrombectomy. Nonetheless, patients who present to hospitals where thrombectomy is not an option can receive intravenous t-PA, and those with qualifying anterior circulation strokes can then be transferred to tertiary stroke centers where thrombectomy is available. Therefore, despite the advances afforded by mechanical thrombectomy, there remains a need for treatments that improve the efficacy and safety of thrombolytic therapy. In this review, we discuss: (i) current treatment options for acute ischemic stroke; (ii) the mechanism of action of fibrinolytic agents; and (iii) potential strategies to manipulate the fibrinolytic system to promote endogenous fibrinolysis or to enhance the efficacy of fibrinolytic therapy.
Collapse
Affiliation(s)
- S J Henderson
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
| | - J I Weitz
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
- Department of Medical Sciences, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - P Y Kim
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
- Department of Medical Sciences, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
26
|
Bandla A, Liao LD, Chan SJ, Ling JM, Liu YH, Shih YYI, Pan HC, Wong PTH, Lai HY, King NKK, Chen YY, Ng WH, Thakor NV. Simultaneous functional photoacoustic microscopy and electrocorticography reveal the impact of rtPA on dynamic neurovascular functions after cerebral ischemia. J Cereb Blood Flow Metab 2018; 38:980-995. [PMID: 28685662 PMCID: PMC5999003 DOI: 10.1177/0271678x17712399] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The advance of thrombolytic therapy has been hampered by the lack of optimization of the therapy during the hyperacute phase of focal ischemia. Here, we investigate neurovascular dynamics using a custom-designed hybrid electrocorticography (ECoG)-functional photoacoustic microscopy (fPAM) imaging system during the hyperacute phase (first 6 h) of photothrombotic ischemia (PTI) in male Wistar rats following recombinant tissue plasminogen activator (rtPA)-mediated thrombolysis. We reported, for the first time, the changes in neural activity and cerebral hemodynamic responses following rtPA infusion at different time points post PTI. Interestingly, very early administration of rtPA (< 1 h post PTI) resulted in only partial recovery of neurovascular dynamics (specifically , neural activity recovered to 71 ± 3.5% of baseline and hemodynamics to only 52 ± 2.6% of baseline) and late administration of rtPA (> 4 h post PTI) resulted in the deterioration of neurovascular function. A therapeutic window between 1 and 3 h post PTI was found to improve recovery of neurovascular function (i.e. significant restoration of neural activity to 93 ± 4.2% of baseline and hemodynamics to 81 ± 2.1% of baseline, respectively). The novel combination of fPAM and ECoG enables direct mapping of neurovascular dynamics and serves as a platform to evaluate potential interventions for stroke.
Collapse
Affiliation(s)
- Aishwarya Bandla
- 1 Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore.,2 Department of Biomedical Engineering, National University of Singapore, Singapore
| | - Lun-De Liao
- 1 Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore.,3 Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Taiwan, R.O.C
| | - Su Jing Chan
- 4 Department of Radiology, Massachusetts General Hospital and Harvard Medical School, MA, USA.,5 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ji Min Ling
- 1 Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore.,6 Department of Neurosurgery, National Neuroscience Institute, Singapore.,7 SingHealth Duke-NUS Neuroscience Academic Clinical Program, National Neuroscience Institute, Singapore
| | - Yu-Hang Liu
- 1 Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore.,8 Department of Electrical and Computer Engineering, National University of Singapore, Singapore
| | - Yen-Yu Ian Shih
- 9 Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Han-Chi Pan
- 3 Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Taiwan, R.O.C
| | - Peter Tsun-Hon Wong
- 5 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Hsin-Yi Lai
- 10 Interdisciplinary Institute of Neuroscience and Technology, Qiushi Academy for Advanced Studies, Zhejiang University, China
| | | | - You-Yin Chen
- 11 Department of Biomedical Engineering, National Yang Ming University, Taiwan, R.O.C
| | - Wai Hoe Ng
- 6 Department of Neurosurgery, National Neuroscience Institute, Singapore.,7 SingHealth Duke-NUS Neuroscience Academic Clinical Program, National Neuroscience Institute, Singapore
| | - Nitish V Thakor
- 1 Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore.,2 Department of Biomedical Engineering, National University of Singapore, Singapore.,8 Department of Electrical and Computer Engineering, National University of Singapore, Singapore.,12 Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
27
|
Fandler S, Deutschmann H, Fazekas F, Gattringer T. Repeated Endovascular Treatment of Early Recurrent Proximal Middle Cerebral Artery Occlusion: Case Report and Brief Review of the Literature. Front Neurol 2018; 9:289. [PMID: 29774008 PMCID: PMC5943549 DOI: 10.3389/fneur.2018.00289] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 04/16/2018] [Indexed: 01/19/2023] Open
Abstract
Mechanical thrombectomy (MT) is the gold standard treatment for large vessel occlusion (LVO) stroke of the anterior circulation. Whether MT can also be effectively and safely performed in early recurrent LVO is largely unclear. We present the case of a middle-aged patient who was successfully treated by MT for right proximal middle cerebral artery (MCA) occlusion with excellent outcome. One day after discharge (9 days after the first MT), the patient was readmitted with wake-up stroke. MRI again revealed right proximal MCA occlusion with severe diffusion–perfusion mismatch. Repeat MT was performed and once more led to almost full recovery. The recurrent strokes were attributed to ulcerated non-stenosing plaques in the ipsilateral internal carotid artery, which prompted thromboendarterectomy. In an 18-months follow-up period, no further vascular events occurred. In conclusion, repeated MT for early recurrent LVO appears feasible in carefully selected patients. The collection of similar cases via registries would be desirable.
Collapse
Affiliation(s)
- Simon Fandler
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Hannes Deutschmann
- Division of Neuroradiology, Vascular and Interventional Radiology, Department of Radiology, Medical University of Graz, Graz, Austria
| | - Franz Fazekas
- Department of Neurology, Medical University of Graz, Graz, Austria
| | | |
Collapse
|
28
|
Wu J, Zhu H, Yang G, He J, Wang Y, Zhao S, Zhang X, Gui L, Zhao M, Peng S. Design and synthesis of nanoscaled IQCA-TAVV as a delivery system capable of antiplatelet activation, targeting arterial thrombus and releasing IQCA. Int J Nanomedicine 2018; 13:1139-1158. [PMID: 29520141 PMCID: PMC5833776 DOI: 10.2147/ijn.s150205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Arterial thrombosis has been associated with a series of pathological conditions, and the discovery of arterial thrombosis inhibitor is of clinical importance. METHODS By analyzing the pharmacophores of anti-platelet agents, thrombus targeting peptide and anti-thrombotic nano-systems 3S-1,2,3,4-tetrahydroisoquino-line-3-carbonyl-Thr-Ala-Arg-Gly-Asp(Val)-Val (IQCA-TAVV) was designed and prepared as a nano-scaled arterial thrombosis inhibitor. RESULTS In vitro the nanoparticles of IQCA-TAVV were able to adhere onto the surface of activated platelets, attenuate activated platelets to extend pseudopodia and inhibit activated platelets to form aggregators. In vivo IQCA-TAVV targeted arterial thrombus, dose dependently inhibited arterial thrombosis with a 1 nmol/kg of minimal effective dose, and the activity waŝ1670 folds of that of aspirin. CONCLUSION IQCA-TAVV represented the design, preparation and application of nanomedicine capable of adhering on the surface of activated platelets, attenuating platelet activation, targeting arterial thrombus and inhibiting arterial thrombosis.
Collapse
Affiliation(s)
- Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Haimei Zhu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Guodong Yang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
| | - Jianhong He
- Department of Internal Medicine of TCM, The First Affiliated Hospital of Guanxi University of Chinese Medicine, Nanning, China
| | - Yuji Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Shurui Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Xiaoyi Zhang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Lin Gui
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
29
|
Iglesias-Rey R, Rodríguez-Yáñez M, Rodríguez-Castro E, Pumar JM, Arias S, Santamaría M, López-Dequidt I, Hervella P, Correa-Paz C, Sobrino T, Vivien D, Campos F, Castellanos M, Castillo J. Worse Outcome in Stroke Patients Treated with rt-PA Without Early Reperfusion: Associated Factors. Transl Stroke Res 2017; 9:347-355. [PMID: 29116527 PMCID: PMC6061244 DOI: 10.1007/s12975-017-0584-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/23/2017] [Accepted: 10/30/2017] [Indexed: 02/07/2023]
Abstract
Based on preclinical studies suggesting that recombinant tissue plasminogen activator (rt-PA) may promote ischemic brain injuries, we investigated in patients the possible risk of worse clinical outcome after rt-PA treatment as a result of its inability to resolve cerebral ischemia. Here, we designed a cohort study using a retrospective analysis of patients who received treatment with intravenous (4.5-h window) or intraarterial rt-PA, without or with thrombectomy. Controls were consecutive patients who did not receive recanalization treatment, who met all inclusion criteria. As a marker of reperfusion, we defined the variable of early neurological improvement as the difference between the score of the National Institute of Health Stroke Scale (NIHSS) (at admission and 24 h). The main variable was worsening of the patient’s functional situation in the first 3 months. To compare quantitative variables, we used Student’s t test or the Mann-Whitney test. To estimate the odds ratios of each independent variable in the patient’s worsening in the first 3 months, we used a logistic regression model. We included 1154 patients; 577 received rt-PA, and 577 served as controls. In the group of patients treated with rt-PA, 39.4% who did not present clinical reperfusion data developed worsening within 3 months after stroke compared with 3.5% of patients with reperfusion (P < 0.0001). These differences were not significant in the control group. In summary, administration of rt-PA intravenously or intraarterially without reperfusion within the first 24 h may be associated with a higher risk of functional deterioration in the first 3 months.
Collapse
Affiliation(s)
- Ramón Iglesias-Rey
- Clinical Neurosciences Research Laboratory, Department of Neurology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain.
| | - Manuel Rodríguez-Yáñez
- Clinical Neurosciences Research Laboratory, Department of Neurology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Emilio Rodríguez-Castro
- Clinical Neurosciences Research Laboratory, Department of Neurology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - José Manuel Pumar
- Department of Neuroradiology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Susana Arias
- Clinical Neurosciences Research Laboratory, Department of Neurology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - María Santamaría
- Clinical Neurosciences Research Laboratory, Department of Neurology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Iria López-Dequidt
- Clinical Neurosciences Research Laboratory, Department of Neurology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Pablo Hervella
- Clinical Neurosciences Research Laboratory, Department of Neurology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Clara Correa-Paz
- Clinical Neurosciences Research Laboratory, Department of Neurology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory, Department of Neurology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Denis Vivien
- Inserm, Inserm, UMR-S U1237, Physiopathology and Imaging of Neurological diseases, GIP Cyceron, Caen Normandie University, 14073, Caen, France
- CHU de Caen, Department of Clinical Research, Caen University Hospital, 14000, Caen, France
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory, Department of Neurology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| | - Mar Castellanos
- Department of Neurology, Biomedical Research Institute, University Hospital A Coruña, 15006, Corunna, Spain
| | - José Castillo
- Clinical Neurosciences Research Laboratory, Department of Neurology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), 15706, Santiago de Compostela, Spain
| |
Collapse
|
30
|
Liu YH, Chan SJ, Pan HC, Bandla A, King NKK, Wong PTH, Chen YY, Ng WH, Thakor NV, Liao LD. Integrated treatment modality of cathodal-transcranial direct current stimulation with peripheral sensory stimulation affords neuroprotection in a rat stroke model. NEUROPHOTONICS 2017; 4:045002. [PMID: 29021986 PMCID: PMC5627795 DOI: 10.1117/1.nph.4.4.045002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/12/2017] [Indexed: 05/03/2023]
Abstract
Cathodal-transcranial direct current stimulation induces therapeutic effects in animal ischemia models by preventing the expansion of ischemic injury during the hyperacute phase of ischemia. However, its efficacy is limited by an accompanying decrease in cerebral blood flow. On the other hand, peripheral sensory stimulation can increase blood flow to specific brain areas resulting in rescue of neurovascular functions from ischemic damage. Therefore, the two modalities appear to complement each other to form an integrated treatment modality. Our results showed that hemodynamics was improved in a photothrombotic ischemia model, as cerebral blood volume and hemoglobin oxygen saturation ([Formula: see text]) recovered to 71% and 76% of the baseline values, respectively. Furthermore, neural activities, including somatosensory-evoked potentials (110% increase), the alpha-to-delta ratio (27% increase), and the [Formula: see text] ratio (27% decrease), were also restored. Infarct volume was reduced by 50% with a 2-fold preservation in the number of neurons and a 6-fold reduction in the number of active microglia in the infarct region compared with the untreated group. Grip strength was also better preserved (28% higher) compared with the untreated group. Overall, this nonpharmacological, nonintrusive approach could be prospectively developed into a clinical treatment modality.
Collapse
Affiliation(s)
- Yu-Hang Liu
- National University of Singapore, Singapore Institute for Neurotechnology (SINAPSE), Singapore, Singapore
- National University of Singapore, Department of Electrical and Computer Engineering, Singapore, Singapore
| | - Su Jing Chan
- Massachusetts General Hospital and Harvard Medical School, Department of Radiology, Boston, Massachusetts, United States
| | - Han-Chi Pan
- National Health Research Institutes, Institute of Biomedical Engineering and Nanomedicine, Miaoli, Taiwan
| | - Aishwarya Bandla
- National University of Singapore, Singapore Institute for Neurotechnology (SINAPSE), Singapore, Singapore
| | - Nicolas K. K. King
- National Neuroscience Institute (NNI), Department of Neurosurgery, Singapore, Singapore
- National Neuroscience Institute (NNI), SingHealth Duke-NUS Neuroscience Academic Clinical Program, Singapore, Singapore
| | - Peter Tsun Hon Wong
- National University of Singapore, Department of Pharmacology, Singapore, Singapore
| | - You-Yin Chen
- National Yang Ming University, Department of Biomedical Engineering, Taipei, Taiwan
| | - Wai Hoe Ng
- National Neuroscience Institute (NNI), Department of Neurosurgery, Singapore, Singapore
- National Neuroscience Institute (NNI), SingHealth Duke-NUS Neuroscience Academic Clinical Program, Singapore, Singapore
| | - Nitish V. Thakor
- National University of Singapore, Singapore Institute for Neurotechnology (SINAPSE), Singapore, Singapore
- National University of Singapore, Department of Electrical and Computer Engineering, Singapore, Singapore
- Johns Hopkins University, Department of Biomedical Engineering, Baltimore, Maryland, United States
| | - Lun-De Liao
- National University of Singapore, Singapore Institute for Neurotechnology (SINAPSE), Singapore, Singapore
- National Health Research Institutes, Institute of Biomedical Engineering and Nanomedicine, Miaoli, Taiwan
- Address all correspondence to: Lun-De Liao, E-mail:
| |
Collapse
|
31
|
Michinaga S, Koyama Y. Protection of the Blood–Brain Barrier as a Therapeutic Strategy for Brain Damage. Biol Pharm Bull 2017; 40:569-575. [DOI: 10.1248/bpb.b16-00991] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Shotaro Michinaga
- Laboratory of Pharmacology, Faculty of Pharmacy, Osaka Ohtani University
| | - Yutaka Koyama
- Laboratory of Pharmacology, Faculty of Pharmacy, Osaka Ohtani University
| |
Collapse
|
32
|
Chen Y, Huang L, Wang L, Chen L, Ren W, Zhou W. Differential expression of microRNAs contributed to the health efficacy of EGCG inin vitrosubarachnoid hemorrhage model. Food Funct 2017; 8:4675-4683. [PMID: 29160895 DOI: 10.1039/c7fo01064h] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
(1) EGCG prevented miRNA dysregulation after SAH; (2) multi-target mechanisms of EGCG might rely on its regulatory roles on miRNAs expression, such as those miRNAs targeting p38, Ca2+, and autophagic activation; (3) the differential expression of miRNAs might determine the therapeutic efficacy of different concentration of EGCG.
Collapse
Affiliation(s)
- Ying Chen
- College of Life Science
- Henan Normal University
- Xinxiang 453007
- PR China
| | - Liyong Huang
- Department of Neurosurgery
- the First Affiliated Hospital of Xinxiang Medical University
- Weihui
- China
| | - Lei Wang
- Department of Neurosurgery
- the First Affiliated Hospital of Xinxiang Medical University
- Weihui
- China
| | - Lingyun Chen
- Department of Neurosurgery
- the First Affiliated Hospital of Xinxiang Medical University
- Weihui
- China
| | - Wenhua Ren
- Genomic and Microarray Core
- University of Colorado
- Anschutz Medical Campus
- Aurora
- USA
| | - Wenke Zhou
- Department of Neurosurgery
- the First Affiliated Hospital of Xinxiang Medical University
- Weihui
- China
| |
Collapse
|
33
|
Freese C, Hanada S, Fallier-Becker P, Kirkpatrick CJ, Unger RE. Identification of neuronal and angiogenic growth factors in an in vitro blood-brain barrier model system: Relevance in barrier integrity and tight junction formation and complexity. Microvasc Res 2016; 111:1-11. [PMID: 27988246 DOI: 10.1016/j.mvr.2016.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 11/18/2016] [Accepted: 12/03/2016] [Indexed: 12/01/2022]
Abstract
We previously demonstrated that the co-cultivation of endothelial cells with neural cells resulted in an improved integrity of the in vitro blood-brain barrier (BBB), and that this model could be useful to evaluate the transport properties of potential central nervous system disease drugs through the microvascular brain endothelial. In this study we have used real-time PCR, fluorescent microscopy, protein arrays and enzyme-linked immunosorbent assays to determine which neural- and endothelial cell-derived factors are produced in the co-culture and improve the integrity of the BBB. In addition, a further improvement of the BBB integrity was achieved by adjusting serum concentrations and growth factors or by the addition of brain pericytes. Under specific conditions expression of angiogenic, angiostatic and neurotrophic factors such as endostatin, pigment epithelium derived factor (PEDF/serpins-F1), tissue inhibitor of metalloproteinases (TIMP-1), and vascular endothelial cell growth factor (VEGF) closely mimicked the in vivo situation. Freeze-fracture analysis of these cultures demonstrated the quality and organization of the endothelial tight junction structures and their association to the two different lipidic leaflets of the membrane. Finally, a multi-cell culture model of the BBB with a transendothelial electrical resistance up to 371 (±15) Ω×cm2 was developed, which may be useful for preliminary screening of drug transport across the BBB and to evaluate cellular crosstalk of cells involved in the neurovascular unit.
Collapse
Affiliation(s)
- Christian Freese
- REPAIR-lab, Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Langenbeckstr. 1, 55131 Mainz, Germany.
| | - Sanshiro Hanada
- REPAIR-lab, Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Langenbeckstr. 1, 55131 Mainz, Germany.
| | - Petra Fallier-Becker
- Institute of Pathology and Neuropathology, University of Tuebingen, Geschwister-Scholl-Platz, 72074 Tuebingen, Germany.
| | - C James Kirkpatrick
- REPAIR-lab, Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Langenbeckstr. 1, 55131 Mainz, Germany.
| | - Ronald E Unger
- REPAIR-lab, Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Langenbeckstr. 1, 55131 Mainz, Germany.
| |
Collapse
|
34
|
Dong MX, Hu QC, Shen P, Pan JX, Wei YD, Liu YY, Ren YF, Liang ZH, Wang HY, Zhao LB, Xie P. Recombinant Tissue Plasminogen Activator Induces Neurological Side Effects Independent on Thrombolysis in Mechanical Animal Models of Focal Cerebral Infarction: A Systematic Review and Meta-Analysis. PLoS One 2016; 11:e0158848. [PMID: 27387385 PMCID: PMC4936748 DOI: 10.1371/journal.pone.0158848] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 06/22/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Recombinant tissue plasminogen activator (rtPA) is the only effective drug approved by US FDA to treat ischemic stroke, and it contains pleiotropic effects besides thrombolysis. We performed a meta-analysis to clarify effect of tissue plasminogen activator (tPA) on cerebral infarction besides its thrombolysis property in mechanical animal stroke. METHODS Relevant studies were identified by two reviewers after searching online databases, including Pubmed, Embase, and ScienceDirect, from 1979 to 2016. We identified 6, 65, 17, 12, 16, 12 and 13 comparisons reporting effect of endogenous tPA on infarction volume and effects of rtPA on infarction volume, blood-brain barrier, brain edema, intracerebral hemorrhage, neurological function and mortality rate in all 47 included studies. Standardized mean differences for continuous measures and risk ratio for dichotomous measures were calculated to assess the effects of endogenous tPA and rtPA on cerebral infarction in animals. The quality of included studies was assessed using the Stroke Therapy Academic Industry Roundtable score. Subgroup analysis, meta-regression and sensitivity analysis were performed to explore sources of heterogeneity. Funnel plot, Trim and Fill method and Egger's test were obtained to detect publication bias. RESULTS We found that both endogenous tPA and rtPA had not enlarged infarction volume, or deteriorated neurological function. However, rtPA would disrupt blood-brain barrier, aggravate brain edema, induce intracerebral hemorrhage and increase mortality rate. CONCLUSIONS This meta-analysis reveals rtPA can lead to neurological side effects besides thrombolysis in mechanical animal stroke, which may account for clinical exacerbation for stroke patients that do not achieve vascular recanalization with rtPA.
Collapse
Affiliation(s)
- Mei-Xue Dong
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Qing-Chuan Hu
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Peng Shen
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun-Xi Pan
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - You-Dong Wei
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi-Yun Liu
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi-Fei Ren
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zi-Hong Liang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hai-Yang Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Li-Bo Zhao
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Xie
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| |
Collapse
|
35
|
Yang S, Krug SM, Heitmann J, Hu L, Reinhold AK, Sauer S, Bosten J, Sommer C, Fromm M, Brack A, Rittner HL. Analgesic drug delivery via recombinant tissue plasminogen activator and microRNA-183-triggered opening of the blood-nerve barrier. Biomaterials 2016; 82:20-33. [DOI: 10.1016/j.biomaterials.2015.11.053] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/15/2015] [Accepted: 11/29/2015] [Indexed: 01/07/2023]
|
36
|
Release of Matrix Metalloproteinases-2 and 9 by S-Nitrosylated Caveolin-1 Contributes to Degradation of Extracellular Matrix in tPA-Treated Hypoxic Endothelial Cells. PLoS One 2016; 11:e0149269. [PMID: 26881424 PMCID: PMC4755609 DOI: 10.1371/journal.pone.0149269] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 01/30/2016] [Indexed: 11/19/2022] Open
Abstract
Intracranial hemorrhage remains the most feared complication in tissue plasminogen activator (tPA) thrombolysis for ischemic stroke. However, the underlying molecular mechanisms are still poorly elucidated. In this study, we reported an important role of caveolin-1 (Cav-1) s-nitrosylation in matrix metalloproteinase (MMP)-2 and 9 secretion from tPA-treated ischemic endothelial cells. Brain vascular endothelial cells (bEND3) were exposed to oxygen-glucose deprivation (OGD) for 2 h before adding recombinant human tPA for 6 h. This treatment induced a significant increase of MMP2 and 9 in the media of bEND3 cells and a simultaneous degradation of fibronectin and laminin β-1, the two main components of extracellular matrix (ECM). Inhibition of MMP2 and 9 with SB-3CT completely blocked the degradation of fibronectin and laminin β-1. ODG+tPA treatment led to Cav-1 shedding from bEND3 cells into the media. Notably, OGD triggered nitric oxide (NO) production and S-nitrosylationof Cav-1 (SNCav-1). Meanwhile tPA induced activation of ERK signal pathway and stimulates the secretion of SNCav-1. Pretreatment of bEND3 cells with C-PTIO (a NO scavenger) or U0126 (a specific ERK inhibitor) significantly reduced OGD-induced S-nitrosylation of Cav-1 in cells and blocked the secretion of Cav-1 and MMP2 and 9 into the media as well as the degradation of fibronectin and laminin β-1 in OGD and tPA-treated cells. These data indicate that OGD-triggered Cav-1 S-nitrosylation interacts with tPA-induced ERK activation to augment MMP2 and 9 secretion and subsequent ECM degradation, which may account for the exacerbation of ischemic blood brain barrier damage following tPA thrombolysis for ischemic stroke.
Collapse
|
37
|
The Neuroprotective Effect of Rosemary ( Rosmarinus officinalis L.) Hydro-alcoholic Extract on Cerebral Ischemic Tolerance in Experimental Stroke. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2016; 15:875-883. [PMID: 28243285 PMCID: PMC5316267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The prevention of BBB breakdown and the subsequent vasogenic edema are important parts of the medical management of ischemic stroke. The purpose of this study was to investigate the ischemic tolerance effect of Rosmarinus officinalis leaf hydro-alcoholic extract (RHE). Five groups of animals were designed: sham (underwent surgery without MCAO) and MCAO groups, the MCAO groups were pretreated orally by gavages with RHE (50, 75, and 100 mg/Kg/day), daily for 30 days. Two hours after the last dose, serum lipid levels were determined and then the rats were subjected to 60 min of middle cerebral artery occlusion followed by 24 h of reperfusion. Subsequently, brain infarct size, brain edema and Evans Blue dye extravasations were measured and neurological deficits were scored. Dietary RHE could significantly reduce cortical and sub-cortical infarct volumes (211.55 ± 24.88 mm3vs. 40.59 ± 10.04 mm3vs. 29.96 ± 12.19 mm3vs. 6.58 ± 3.2 mm3), neurologic deficit scores, cerebral edema (82.34 ± 0.42% vs. 79.92 ± 0.49% vs. 79.45 ± 0.26% vs. 79.30 ± 0.19%), blood-brain barrier (BBB) permeability (7.73 ± 0.4 μg/g tissue vs. 4.1 ± 0.23 μg/g tissue vs. 3.58 ± 0.3 μg/g tissue vs. 3.38 ± 0.25 μg/g tissue) in doses of 50, 75 and 100 mg/Kg/day as compared with the control group in the transient model of focal cerebral ischemia. Although pretreatment with RHE plays an important role in the generation of tolerance against cerebral I/R injury, further studies are needed to clarify the mechanism of the ischemic tolerance.
Collapse
|
38
|
Bardehle S, Rafalski VA, Akassoglou K. Breaking boundaries-coagulation and fibrinolysis at the neurovascular interface. Front Cell Neurosci 2015; 9:354. [PMID: 26441525 PMCID: PMC4584986 DOI: 10.3389/fncel.2015.00354] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 08/24/2015] [Indexed: 12/20/2022] Open
Abstract
Blood proteins at the neurovascular unit (NVU) are emerging as important molecular determinants of communication between the brain and the immune system. Over the past two decades, roles for the plasminogen activation (PA)/plasmin system in fibrinolysis have been extended from peripheral dissolution of blood clots to the regulation of central nervous system (CNS) functions in physiology and disease. In this review, we discuss how fibrin and its proteolytic degradation affect neuroinflammatory, degenerative and repair processes. In particular, we focus on novel functions of fibrin—the final product of the coagulation cascade and the main substrate of plasmin—in the activation of immune responses and trafficking of immune cells into the brain. We also comment on the suitability of the coagulation and fibrinolytic systems as potential biomarkers and drug targets in diseases, such as multiple sclerosis (MS), Alzheimer’s disease (AD) and stroke. Studying coagulation and fibrinolysis as major molecular pathways that regulate cellular functions at the NVU has the potential to lead to the development of novel strategies for the detection and treatment of neurologic diseases.
Collapse
Affiliation(s)
- Sophia Bardehle
- Gladstone Institute of Neurological Disease, University of California, San Francisco San Francisco, CA, USA
| | - Victoria A Rafalski
- Gladstone Institute of Neurological Disease, University of California, San Francisco San Francisco, CA, USA
| | - Katerina Akassoglou
- Gladstone Institute of Neurological Disease, University of California, San Francisco San Francisco, CA, USA ; Department of Neurology, University of California, San Francisco San Francisco, CA, USA
| |
Collapse
|
39
|
Liao LD, Liu YH, Lai HY, Bandla A, Shih YYI, Chen YY, Thakor NV. Rescue of cortical neurovascular functions during the hyperacute phase of ischemia by peripheral sensory stimulation. Neurobiol Dis 2015; 75:53-63. [DOI: 10.1016/j.nbd.2014.12.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 12/07/2014] [Accepted: 12/23/2014] [Indexed: 10/24/2022] Open
|
40
|
Merino-Zamorano C, Hernández-Guillamon M, Jullienne A, Le Béhot A, Bardou I, Parés M, Fernández-Cadenas I, Giralt D, Carrera C, Ribó M, Vivien D, Ali C, Rosell A, Montaner J. NURR1 involvement in recombinant tissue-type plasminogen activator treatment complications after ischemic stroke. Stroke 2014; 46:477-84. [PMID: 25503547 DOI: 10.1161/strokeaha.114.006826] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND PURPOSE Despite the effectiveness of recombinant tissue-type plasminogen activator (r-tPA) during the acute phase of ischemic stroke, the therapy remains limited by a narrow time window and the occurrence of occasional vascular side effects, particularly symptomatic hemorrhages. Our aim was to investigate the mechanisms underlying the endothelial damage resulting from r-tPA treatment in ischemic-like conditions. METHODS Microarray analyses were performed on cerebral endothelial cells submitted to r-tPA treatment during oxygen and glucose deprivation to identify novel biomarker candidates. Validation was then performed in vivo in a mouse model of thromboembolic stroke and culminated in an analysis in a clinical cohort of patients with ischemic stroke treated with thrombolysis. RESULTS The transcription factor NURR1 (NR4A2) was identified as a downstream target induced by r-tPA during oxygen and glucose deprivation. Silencing NURR1 expression reversed the endothelial-toxicity induced by the combined stimuli, a protective effect attributable to reduced levels of proinflammatory mediators, such as nuclear factor-kappa-beta 2 (NF-κ-B2), interleukin 1 alpha (IL1α), intercellular adhesion molecule 1 (ICAM1), SMAD family member 3 (SMAD3), colony stimulating factor 2 (granulocyte-macrophage; CSF2). The detrimental effect of delayed thrombolysis, in conditions in which NURR1 gene expression was enhanced, was confirmed in the preclinical stroke model. Finally, we determined that patients with stroke who had a symptomatic hemorrhagic transformation after r-tPA treatment exhibited higher baseline serum NURR1 levels than did patients with an asymptomatic or absence of cerebral bleedings. CONCLUSIONS Our results suggest that NURR1 upregulation by r-tPA during ischemic stroke is associated with endothelial dysfunction and inflammation and the enhancement of hemorrhagic complications associated to thrombolysis.
Collapse
Affiliation(s)
- Cristina Merino-Zamorano
- From the Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain (C.M.-Z., M.H.-G., M.P., I.F.-C., D.G., C.C., A.R., J.M.); INSERM UMR-S U919, GIP Cyceron, University of Caen, Caen, France (A.J., A.L.B., I.B., D.V., C.A.); and Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain (M.R., J.M.)
| | - Mar Hernández-Guillamon
- From the Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain (C.M.-Z., M.H.-G., M.P., I.F.-C., D.G., C.C., A.R., J.M.); INSERM UMR-S U919, GIP Cyceron, University of Caen, Caen, France (A.J., A.L.B., I.B., D.V., C.A.); and Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain (M.R., J.M.).
| | - Amandine Jullienne
- From the Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain (C.M.-Z., M.H.-G., M.P., I.F.-C., D.G., C.C., A.R., J.M.); INSERM UMR-S U919, GIP Cyceron, University of Caen, Caen, France (A.J., A.L.B., I.B., D.V., C.A.); and Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain (M.R., J.M.)
| | - Audrey Le Béhot
- From the Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain (C.M.-Z., M.H.-G., M.P., I.F.-C., D.G., C.C., A.R., J.M.); INSERM UMR-S U919, GIP Cyceron, University of Caen, Caen, France (A.J., A.L.B., I.B., D.V., C.A.); and Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain (M.R., J.M.)
| | - Isabelle Bardou
- From the Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain (C.M.-Z., M.H.-G., M.P., I.F.-C., D.G., C.C., A.R., J.M.); INSERM UMR-S U919, GIP Cyceron, University of Caen, Caen, France (A.J., A.L.B., I.B., D.V., C.A.); and Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain (M.R., J.M.)
| | - Mireia Parés
- From the Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain (C.M.-Z., M.H.-G., M.P., I.F.-C., D.G., C.C., A.R., J.M.); INSERM UMR-S U919, GIP Cyceron, University of Caen, Caen, France (A.J., A.L.B., I.B., D.V., C.A.); and Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain (M.R., J.M.)
| | - Israel Fernández-Cadenas
- From the Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain (C.M.-Z., M.H.-G., M.P., I.F.-C., D.G., C.C., A.R., J.M.); INSERM UMR-S U919, GIP Cyceron, University of Caen, Caen, France (A.J., A.L.B., I.B., D.V., C.A.); and Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain (M.R., J.M.)
| | - Dolors Giralt
- From the Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain (C.M.-Z., M.H.-G., M.P., I.F.-C., D.G., C.C., A.R., J.M.); INSERM UMR-S U919, GIP Cyceron, University of Caen, Caen, France (A.J., A.L.B., I.B., D.V., C.A.); and Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain (M.R., J.M.)
| | - Caty Carrera
- From the Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain (C.M.-Z., M.H.-G., M.P., I.F.-C., D.G., C.C., A.R., J.M.); INSERM UMR-S U919, GIP Cyceron, University of Caen, Caen, France (A.J., A.L.B., I.B., D.V., C.A.); and Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain (M.R., J.M.)
| | - Marc Ribó
- From the Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain (C.M.-Z., M.H.-G., M.P., I.F.-C., D.G., C.C., A.R., J.M.); INSERM UMR-S U919, GIP Cyceron, University of Caen, Caen, France (A.J., A.L.B., I.B., D.V., C.A.); and Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain (M.R., J.M.)
| | - Denis Vivien
- From the Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain (C.M.-Z., M.H.-G., M.P., I.F.-C., D.G., C.C., A.R., J.M.); INSERM UMR-S U919, GIP Cyceron, University of Caen, Caen, France (A.J., A.L.B., I.B., D.V., C.A.); and Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain (M.R., J.M.)
| | - Carine Ali
- From the Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain (C.M.-Z., M.H.-G., M.P., I.F.-C., D.G., C.C., A.R., J.M.); INSERM UMR-S U919, GIP Cyceron, University of Caen, Caen, France (A.J., A.L.B., I.B., D.V., C.A.); and Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain (M.R., J.M.)
| | - Anna Rosell
- From the Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain (C.M.-Z., M.H.-G., M.P., I.F.-C., D.G., C.C., A.R., J.M.); INSERM UMR-S U919, GIP Cyceron, University of Caen, Caen, France (A.J., A.L.B., I.B., D.V., C.A.); and Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain (M.R., J.M.)
| | - Joan Montaner
- From the Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain (C.M.-Z., M.H.-G., M.P., I.F.-C., D.G., C.C., A.R., J.M.); INSERM UMR-S U919, GIP Cyceron, University of Caen, Caen, France (A.J., A.L.B., I.B., D.V., C.A.); and Neurovascular Unit, Department of Neurology, Vall d'Hebron Hospital, Barcelona, Spain (M.R., J.M.)
| |
Collapse
|
41
|
Balakathiresan NS, Chandran R, Bhomia M, Jia M, Li H, Maheshwari RK. Serum and amygdala microRNA signatures of posttraumatic stress: fear correlation and biomarker potential. J Psychiatr Res 2014; 57:65-73. [PMID: 24998397 DOI: 10.1016/j.jpsychires.2014.05.020] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 05/21/2014] [Accepted: 05/29/2014] [Indexed: 12/11/2022]
Abstract
Exposure to acute traumatic stress can cause permanent changes in neurological circuitry and may lead to the development of an anxiety disorder known as posttraumatic stress disorder (PTSD). Current diagnosis of PTSD is based on clinical or behavioral symptom assessment, however, these are not definitive due to overlapping symptoms with other psychiatric disorders or mild traumatic brain injury (mTBI). No FDA approved diagnostic tests or biomarkers are currently available for diagnosis of PTSD. Recently, circulating miRNAs have emerged as novel biomarkers of many diseases. In this study, we have examined the altered expression of serum and amygdala miRNAs in an animal model of PTSD. Differentially expressed and statistically significant miRNAs in serum were validated for their presence in amygdala of corresponding animals. A panel of nine stress-responsive miRNAs viz., miR-142-5p, miR-19b, miR-1928, miR-223-3p, miR-322∗, miR-324, miR-421-3p and miR-463∗ and miR-674∗ were identified, and may have potential as biomarker(s) for PTSD. Further validations by bioinformatics and system biology approaches indicate that five miRNAs such as miR-142-5p, miR-19b, miR-1928, miR-223 and miR-421-3p may play a potential role in the regulation of genes associated with delayed and exaggerated fear. To the best of our knowledge, this is the first report demonstrating the plausibility of using circulating miRNAs as biomarkers of PTSD.
Collapse
Affiliation(s)
- Nagaraja S Balakathiresan
- Department of Pathology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Raghavendar Chandran
- Department of Pathology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; Biological Sciences Group, Birla Institute of Technology and Science, Pilani 333031, Rajasthan, India
| | - Manish Bhomia
- Department of Pathology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Min Jia
- Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - He Li
- Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Radha K Maheshwari
- Department of Pathology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA.
| |
Collapse
|
42
|
Mosnier LO, Zlokovic BV, Griffin JH. Cytoprotective-selective activated protein C therapy for ischaemic stroke. Thromb Haemost 2014; 112:883-92. [PMID: 25230930 DOI: 10.1160/th14-05-0448] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 08/05/2014] [Indexed: 12/14/2022]
Abstract
Despite years of research and efforts to translate stroke research to clinical therapy, ischaemic stroke remains a major cause of death, disability, and diminished quality of life. Primary and secondary preventive measures combined with improved quality of care have made significant progress. However, no novel drug for ischaemic stroke therapy has been approved in the past decade. Numerous studies have shown beneficial effects of activated protein C (APC) in rodent stroke models. In addition to its natural anticoagulant functions, APC conveys multiple direct cytoprotective effects on many different cell types that involve multiple receptors including protease activated receptor (PAR) 1, PAR3, and the endothelial protein C receptor (EPCR). Application of molecular engineered APC variants with altered selectivity profiles to rodent stroke models demonstrated that the beneficial effects of APC primarily require its cytoprotective activities but not its anticoagulant activities. Extensive basic, preclinical, and clinical research provided a compelling rationale based on strong evidence for translation of APC therapy that has led to the clinical development of the cytoprotective-selective APC variant, 3K3A-APC, for ischaemic stroke. Recent identification of non-canonical PAR1 and PAR3 activation by APC that give rise to novel tethered-ligands capable of inducing biased cytoprotective signalling as opposed to the canonical signalling provides a mechanistic explanation for how APC-mediated PAR activation can selectively induce cytoprotective signalling pathways. Collectively, these paradigm-shifting discoveries provide detailed insights into the receptor targets and the molecular mechanisms for neuroprotection by cytoprotective-selective 3K3A-APC, which is currently a biologic drug in clinical trials for ischaemic stroke.
Collapse
Affiliation(s)
- Laurent O Mosnier
- Laurent O. Mosnier, PhD, Department of Molecular and Experimental Medicine (MEM-180), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California, USA, Tel.: +1 858 784 2227, Fax: +1 858 784 2243, E-mail:
| | | | | |
Collapse
|
43
|
Plasmin-dependent modulation of the blood-brain barrier: a major consideration during tPA-induced thrombolysis? J Cereb Blood Flow Metab 2014; 34:1283-96. [PMID: 24896566 PMCID: PMC4126105 DOI: 10.1038/jcbfm.2014.99] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 05/09/2014] [Accepted: 05/09/2014] [Indexed: 01/16/2023]
Abstract
Plasmin, the principal downstream product of tissue-type plasminogen activator (tPA), is known for its potent fibrin-degrading capacity but is also recognized for many non-fibrinolytic activities. Curiously, plasmin has not been conclusively linked to blood-brain barrier (BBB) disruption during recombinant tPA (rtPA)-induced thrombolysis in ischemic stroke. This is surprising given the substantial involvement of tPA in the modulation of BBB permeability and the co-existence of tPA and plasminogen in both blood and brain throughout the ischemic event. Here, we review the work that argues a role for plasmin together with endogenous tPA or rtPA in BBB alteration, presenting the overall controversy around the topic yet creating a rational case for an involvement of plasmin in this process.
Collapse
|
44
|
Gur-Wahnon D, Mizrachi T, Wald-Altman S, Higazi AAR, Brenner T. Tissue plasminogen activator involvement in experimental autoimmune myasthenia gravis: Aggravation and therapeutic potential. J Autoimmun 2014; 52:36-43. [DOI: 10.1016/j.jaut.2013.12.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 12/12/2013] [Indexed: 02/01/2023]
|
45
|
Niego B, Medcalf RL. Improved method for the preparation of a human cell-based, contact model of the blood-brain barrier. J Vis Exp 2013:e50934. [PMID: 24300849 DOI: 10.3791/50934] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The blood-brain barrier (BBB) comprises impermeable but adaptable brain capillaries which tightly control the brain environment. Failure of the BBB has been implied in the etiology of many brain pathologies, creating a need for development of human in vitro BBB models to assist in clinically-relevant research. Among the numerous BBB models thus far described, a static (without flow), contact BBB model, where astrocytes and brain endothelial cells (BECs) are cocultured on the opposite sides of a porous membrane, emerged as a simplified yet authentic system to simulate the BBB with high throughput screening capacity. Nevertheless the generation of such model presents few technical challenges. Here, we describe a protocol for preparation of a contact human BBB model utilizing a novel combination of primary human BECs and immortalized human astrocytes. Specifically, we detail an innovative method for cell-seeding on inverted inserts as well as specify insert staining techniques and exemplify how we use our model for BBB-related research.
Collapse
Affiliation(s)
- Be'eri Niego
- Australian Centre for Blood Diseases, Monash University
| | | |
Collapse
|
46
|
Armstead WM, Bohman LE, Riley J, Yarovoi S, Higazi AAR, Cines DB. tPA-S(481)A prevents impairment of cerebrovascular autoregulation by endogenous tPA after traumatic brain injury by upregulating p38 MAPK and inhibiting ET-1. J Neurotrauma 2013; 30:1898-907. [PMID: 23731391 DOI: 10.1089/neu.2013.2962] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Traumatic brain injury (TBI) is associated with loss of cerebrovascular autoregulation, which leads to cerebral hypoperfusion. Mitogen activated protein kinase (MAPK) isoforms ERK, p38, and JNK and endothelin-1 (ET-1) are mediators of impaired cerebral hemodynamics after TBI. Excessive tissue plasminogen activator (tPA) released after TBI may cause loss of cerebrovascular autoregulation either by over-activating N-methyl-D-aspartate receptors (NMDA-Rs) or by predisposing to intracranial hemorrhage. Our recent work shows that a catalytically inactive tPA variant (tPA-S(481)A) that competes with endogenous wild type (wt) tPA for binding to NMDA-R through its receptor docking site but that cannot activate it, prevents activation of ERK by wt tPA and impairment of autoregulation when administered 30 min after fluid percussion injury (FPI). We investigated the ability of variants that lack proteolytic activity but bind/block activation of NMDA-Rs by wt tPA (tPA-S(481)A), do not bind/block activation of NMDA-Rs but are proteolytic (tPA-A(296-299)), or neither bind/block NMDA-Rs nor are proteolytic (tPA-A(296-299)S(481)A) to prevent impairment of autoregulation after TBI and the role of MAPK and ET-1 in such effects. Results show that tPA-S(481)A given 3 h post-TBI, but not tPA-A(296-299) or tPA-A(296-299)S(481)A prevents impaired autoregulation by upregulating p38 and inhibiting ET-1, suggesting that tPA-S(481)A has a realistic therapeutic window and focuses intervention on NMDA-Rs to improve outcome.
Collapse
Affiliation(s)
- William M Armstead
- 1 Department of Anesthesiology and Critical Care, University of Pennsylvania , Philadelphia, Pennsylvania
| | | | | | | | | | | |
Collapse
|
47
|
Krakovsky M, Polianski V, Nimrod A, Higazi A, Leker RR, Lamensdorf I. THR-18, a 18-mer peptide derived from PAI-1, is neuroprotective and improves thrombolysis by tPA in rat stroke models. Neurol Res 2013; 33:983-90. [DOI: 10.1179/1743132811y.0000000018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
48
|
Schmidt A, Minnerup J, Kleinschnitz C. Emerging neuroprotective drugs for the treatment of acute ischaemic stroke. Expert Opin Emerg Drugs 2013; 18:109-20. [DOI: 10.1517/14728214.2013.790363] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
49
|
Ström JO, Ingberg E, Theodorsson A, Theodorsson E. Method parameters' impact on mortality and variability in rat stroke experiments: a meta-analysis. BMC Neurosci 2013; 14:41. [PMID: 23548160 PMCID: PMC3637133 DOI: 10.1186/1471-2202-14-41] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 03/22/2013] [Indexed: 12/14/2022] Open
Abstract
Background Even though more than 600 stroke treatments have been shown effective in preclinical studies, clinically proven treatment alternatives for cerebral infarction remain scarce. Amongst the reasons for the discrepancy may be methodological shortcomings, such as high mortality and outcome variability, in the preclinical studies. A common approach in animal stroke experiments is that A) focal cerebral ischemia is inflicted, B) some type of treatment is administered and C) the infarct sizes are assessed. However, within this paradigm, the researcher has to make numerous methodological decisions, including choosing rat strain and type of surgical procedure. Even though a few studies have attempted to address the questions experimentally, a lack of consensus regarding the optimal methodology remains. Methods We therefore meta-analyzed data from 502 control groups described in 346 articles to find out how rat strain, procedure for causing focal cerebral ischemia and the type of filament coating affected mortality and infarct size variability. Results The Wistar strain and intraluminal filament procedure using a silicone coated filament was found optimal in lowering infarct size variability. The direct and endothelin methods rendered lower mortality rate, whereas the embolus method increased it compared to the filament method. Conclusions The current article provides means for researchers to adjust their middle cerebral artery occlusion (MCAo) protocols to minimize infarct size variability and mortality.
Collapse
Affiliation(s)
- Jakob O Ström
- Department of Clinical and Experimental Medicine, Clinical Chemistry, Faculty of Health Sciences, Linköping University, County Council of Östergötland, Linköping, Sweden.
| | | | | | | |
Collapse
|
50
|
Ishrat T, Soliman S, Guan W, Saler M, Fagan SC. Vascular protection to increase the safety of tissue plasminogen activator for stroke. Curr Pharm Des 2012; 18:3677-84. [PMID: 22574982 DOI: 10.2174/138161212802002779] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 01/24/2012] [Indexed: 12/22/2022]
Abstract
Thrombolytic therapy with tissue plasminogen activator (tPA) remains the most effective treatment for acute ischemic stroke, but can cause vascular damage leading to edema formation and hemorrhagic transformation (HT). In this review, we discuss how tPA contributes to the pathogenesis of vascular damage and highlight evidence to support combination therapy of tPA with pharmacological agents that are vascular protective. There is an unmet need to develop therapeutic interventions which target the underlying mechanisms of vascular damage after acute ischemic stroke in order to prevent HT and improve the safety and impact of tPA.
Collapse
Affiliation(s)
- Tauheed Ishrat
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, 1120 15th St., Augusta, GA 30912, USA
| | | | | | | | | |
Collapse
|