1
|
Sun F, Zhou J, Chen X, Yang T, Wang G, Ge J, Zhang Z, Mei Z. No-reflow after recanalization in ischemic stroke: From pathomechanisms to therapeutic strategies. J Cereb Blood Flow Metab 2024; 44:857-880. [PMID: 38420850 PMCID: PMC11318407 DOI: 10.1177/0271678x241237159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 01/07/2024] [Accepted: 02/18/2024] [Indexed: 03/02/2024]
Abstract
Endovascular reperfusion therapy is the primary strategy for acute ischemic stroke. No-reflow is a common phenomenon, which is defined as the failure of microcirculatory reperfusion despite clot removal by thrombolysis or mechanical embolization. It has been reported that up to 25% of ischemic strokes suffer from no-reflow, which strongly contributes to an increased risk of poor clinical outcomes. No-reflow is associated with functional and structural alterations of cerebrovascular microcirculation, and the injury to the microcirculation seriously hinders the neural functional recovery following macrovascular reperfusion. Accumulated evidence indicates that pathology of no-reflow is linked to adhesion, aggregation, and rolling of blood components along the endothelium, capillary stagnation with neutrophils, astrocytes end-feet, and endothelial cell edema, pericyte contraction, and vasoconstriction. Prevention or treatment strategies aim to alleviate or reverse these pathological changes, including targeted therapies such as cilostazol, adhesion molecule blocking antibodies, peroxisome proliferator-activated receptors (PPARs) activator, adenosine, pericyte regulators, as well as adjunctive therapies, such as extracorporeal counterpulsation, ischemic preconditioning, and alternative or complementary therapies. Herein, we provide an overview of pathomechanisms, predictive factors, diagnosis, and intervention strategies for no-reflow, and attempt to convey a new perspective on the clinical management of no-reflow post-ischemic stroke.
Collapse
Affiliation(s)
- Feiyue Sun
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jing Zhou
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xiangyu Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Tong Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Guozuo Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Academy of Chinese Medicine, Changsha, Hunan, China
| | - Zhanwei Zhang
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
2
|
Tudor T, Spinazzi EF, Alexander JE, Mandigo GK, Lavine SD, Grinband J, Connolly ES. Progressive microvascular failure in acute ischemic stroke: A systematic review, meta-analysis, and time-course analysis. J Cereb Blood Flow Metab 2024; 44:192-208. [PMID: 38016953 PMCID: PMC10993872 DOI: 10.1177/0271678x231216766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 09/01/2023] [Accepted: 10/02/2023] [Indexed: 11/30/2023]
Abstract
This systematic review, meta-analysis, and novel time course analysis examines microvascular failure in the treatment of acute ischemic stroke (AIS) patients undergoing endovascular therapy (EVT) and/or thrombolytic administration for stroke management. A systematic review and meta-analysis following PRIMSA-2020 guidelines was conducted along with a novel curve-of-best fit analysis to elucidate the time-course of microvascular failure. Scopus and PubMed were searched using relevant keywords to identify studies that examine recanalization and reperfusion assessment of AIS patients following large vessel occlusion. Meta-analysis was conducted using a random-effects model. Curve-of-best-fit analysis of microvascular failure rate was performed with a negative exponential model. Twenty-seven studies with 1151 patients were included. Fourteen studies evaluated patients within a standard stroke onset-to-treatment time window (≤6 hours after last known normal) and thirteen studies had an extended time window (>6 hours). Our analysis yields a 22% event rate of microvascular failure following successful recanalization (95% CI: 16-30%). A negative exponential curve modeled a microvascular failure rate asymptote of 28.5% for standard time window studies, with no convergence of the model for extended time window studies. Progressive microvascular failure is a phenomenon that is increasingly identified in clinical studies of AIS patients undergoing revascularization treatment.
Collapse
Affiliation(s)
- Thilan Tudor
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Eleonora F Spinazzi
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Julia E Alexander
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Grace K Mandigo
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Sean D Lavine
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Jack Grinband
- Departments of Psychiatry and Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - E Sander Connolly
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
3
|
Jia M, Jin F, Li S, Ren C, Ruchi M, Ding Y, Zhao W, Ji X. No-reflow after stroke reperfusion therapy: An emerging phenomenon to be explored. CNS Neurosci Ther 2024; 30:e14631. [PMID: 38358074 PMCID: PMC10867879 DOI: 10.1111/cns.14631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 01/02/2024] [Accepted: 01/21/2024] [Indexed: 02/16/2024] Open
Abstract
In the field of stroke thrombectomy, ineffective clinical and angiographic reperfusion after successful recanalization has drawn attention. Partial or complete microcirculatory reperfusion failure after the achievement of full patency of a former obstructed large vessel, known as the "no-reflow phenomenon" or "microvascular obstruction," was first reported in the 1960s and was later detected in both experimental models and patients with stroke. The no-reflow phenomenon (NRP) was reported to result from intraluminal occlusions formed by blood components and extraluminal constriction exerted by the surrounding structures of the vessel wall. More recently, an emerging number of clinical studies have estimated the prevalence of the NRP in stroke patients following reperfusion therapy, ranging from 3.3% to 63% depending on its evaluation methods or study population. Studies also demonstrated its detrimental effects on infarction progress and neurological outcomes. In this review, we discuss the research advances, underlying pathogenesis, diagnostic techniques, and management approaches concerning the no-reflow phenomenon in the stroke population to provide a comprehensive understanding of this phenomenon and offer references for future investigations.
Collapse
Affiliation(s)
- Milan Jia
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Feiyang Jin
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Sijie Li
- Department of Emergency, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Mangal Ruchi
- Department of NeurosurgeryWayne State University School of MedicineDetroitMichiganUSA
| | - Yuchuan Ding
- Department of NeurosurgeryWayne State University School of MedicineDetroitMichiganUSA
| | - Wenbo Zhao
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
4
|
Zhang L, Luo H, Li C, Teng H, Powell B, Lu M, Chopp M, Zhang ZG. Treatment of stroke in aged male and female rats with Vepoloxamer and tPA reduces neurovascular damage. Front Neurol 2023; 14:1282736. [PMID: 37869138 PMCID: PMC10587547 DOI: 10.3389/fneur.2023.1282736] [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: 08/24/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
Stroke is a leading cause of death and disability worldwide, mainly affecting the elderly. Unfortunately, current treatments for acute ischemic stroke warrant improvement. To date, tissue plasminogen activator (tPA) is of limited use in stroke patients mainly due to its narrow therapeutic window and potential for hemorrhagic complication. The adjuvant treatment with Vepoloxamer, a purified amphipathic polymer has been shown to enhance the thrombolytic efficacy of tPA treatment in young adult male rats after embolic stroke. However, most stroke patients are aged; therefore, the current study investigated the therapeutic effect of the combined tPA and Vepoloxamer treatment in aged male and female rats subjected to embolic stroke. Methods Male and female Wistar rats at 18 months of age were subjected to embolic middle cerebral artery occlusion and treated either with monotherapy of tPA or Vepoloxamer, a combination of these two agents, or saline at 4 h after stroke onset. Neurological outcomes were evaluated with a battery of behavioral tests including adhesive removal, foot-fault, and modified neurological severity score tests at 1 and 7 days after stroke onset, followed by histopathological analysis of infarct volume. Residual clot size and vascular patency and integrity were analyzed. Results The combination treatment with Vepoloxamer and tPA significantly reduced infarct volume and neurological deficits in male and female rats compared to rats treated with saline and the monotherapies of tPA and Vepoloxamer. While Vepoloxamer monotherapy moderately reduced neurological deficits, monotherapies with tPA and Vepoloxamer failed to reduce infarct volume compared to saline treatment. Furthermore, the combination treatment with tPA and Vepoloxamer accelerated thrombolysis, reduced ischemia and tPA-potentiated microvascular disruption, and concomitantly improved cerebrovascular integrity and perfusion in the male ischemic rats. Conclusion Combination treatment with tPA and Vepoloxamer at 4 h after stroke onset effectively reduces ischemic neurovascular damage by accelerating thrombolysis and reducing ischemia and tPA potentiated side effects in the aged rats. This funding suggests that the combination treatment with tPA and Vepoloxamer represents a promising strategy to potentially apply to the general population of stroke patients.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Hao Luo
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Chao Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Hua Teng
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Brianna Powell
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Mei Lu
- Department of Biostatistics and Research Epidemiology, Henry Ford Hospital, Detroit, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| |
Collapse
|
5
|
Torrente D, Su EJ, Fredriksson L, Warnock M, Bushart D, Mann KM, Emal CD, Lawrence DA. Compartmentalized Actions of the Plasminogen Activator Inhibitors, PAI-1 and Nsp, in Ischemic Stroke. Transl Stroke Res 2022; 13:801-815. [PMID: 35122213 PMCID: PMC9349468 DOI: 10.1007/s12975-022-00992-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/22/2021] [Accepted: 01/26/2022] [Indexed: 02/08/2023]
Abstract
Tissue plasminogen activator (tPA) is a multifunctional protease. In blood tPA is best understood for its role in fibrinolysis, whereas in the brain tPA is reported to regulate blood-brain barrier (BBB) function and to promote neurodegeneration. Thrombolytic tPA is used for the treatment of ischemic stroke. However, its use is associated with an increased risk of hemorrhagic transformation. In blood the primary regulator of tPA activity is plasminogen activator inhibitor 1 (PAI-1), whereas in the brain, its primary inhibitor is thought to be neuroserpin (Nsp). In this study, we compare the effects of PAI-1 and Nsp deficiency in a mouse model of ischemic stroke and show that tPA has both beneficial and harmful effects that are differentially regulated by PAI-1 and Nsp. Following ischemic stroke Nsp deficiency in mice leads to larger strokes, increased BBB permeability, and increased spontaneous intracerebral hemorrhage. In contrast, PAI-1 deficiency results in smaller infarcts and increased cerebral blood flow recovery. Mechanistically, our data suggests that these differences are largely due to the compartmentalized action of PAI-1 and Nsp, with Nsp deficiency enhancing tPA activity in the CNS which increases BBB permeability and worsens stroke outcomes, while PAI-1 deficiency enhances fibrinolysis and improves recovery. Finally, we show that treatment with a combination therapy that enhances endogenous fibrinolysis by inhibiting PAI-1 with MDI-2268 and reduces BBB permeability by inhibiting tPA-mediated PDGFRα signaling with imatinib significantly reduces infarct size compared to vehicle-treated mice and to mice with either treatment alone.
Collapse
Affiliation(s)
- Daniel Torrente
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Enming Joseph Su
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-0644, USA
| | - Linda Fredriksson
- Biomedicum, Karolinska Institute, Solnavägen 9, Quarter 6D, 17165, Solna, Sweden
| | - Mark Warnock
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-0644, USA
| | - David Bushart
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-0644, USA
- Current affiliation: Ohio State University College of Medicine, Columbus, OH, USA
| | - Kris M Mann
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-0644, USA
| | - Cory D Emal
- Department of Chemistry, Eastern Michigan University, Ypsilanti, MI, 48197, USA
| | - Daniel A Lawrence
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-0644, USA.
| |
Collapse
|
6
|
Jin R, Wang M, Zhong W, Kissinger CR, Villafranca JE, Li G. J147 Reduces tPA-Induced Brain Hemorrhage in Acute Experimental Stroke in Rats. Front Neurol 2022; 13:821082. [PMID: 35309561 PMCID: PMC8925862 DOI: 10.3389/fneur.2022.821082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
Background and purpose J147, a novel neurotrophic compound, was originally developed to treat aging-associated neurological diseases. Based on the broad spectrum of cytoprotective effects exhibited by this compound, we investigated whether J147 has cerebroprotection for acute ischemic stroke and whether it can enhance the effectiveness of thrombolytic therapy with tissue plasminogen activator (tPA). Methods Rats were subjected to transient occlusion of the middle cerebral artery (tMCAO) by insertion of an intraluminal suture or embolic middle cerebral artery occlusion (eMCAO), and treated intravenously with J147 alone or in combination with tPA. Results We found that J147 treatment significantly reduced infarct volume when administered at 2 h after stroke onset in the tMCAO model, but had no effect in eMCAO without tPA. However, combination treatment with J147 plus tPA at 4 h after stroke onset significantly reduced infarct volume and neurological deficits at 72 h after stroke compared with saline or tPA alone groups in the eMCAO model. Importantly, the combination treatment significantly reduced delayed tPA-associated brain hemorrhage and secondary microvascular thrombosis. These protective effects were associated with J147-mediated inhibition of matrix metalloproteinase-9 (MMP9), 15-lipoxygenase-1, and plasminogen activator inhibitor (PAI) expression in the ischemic hemispheres (predominantly in ischemic cerebral endothelium). Moreover, the combination treatment significantly reduced circulating platelet activation and platelet-leukocyte aggregation compared with saline or tPA alone groups at 24 h after stroke, which might also contribute to reduced microvascular thrombosis and neuroinflammation (as demonstrated by reduced neutrophil brain infiltration and microglial activation). Conclusion Our results demonstrate that J147 treatment alone exerts cerebral cytoprotective effects in a suture model of acute ischemic stroke, while in an embolic stroke model co-administration of J147 with tPA reduces delayed tPA-induced intracerebral hemorrhage and confers cerebroprotection. These findings suggest that J147-tPA combination therapy could be a promising approach to improving the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Rong Jin
- Department of Neurosurgery and Neuroscience Institute, Penn State Hershey Medical Center, Hershey, PA, United States
| | - Min Wang
- Department of Neurosurgery and Neuroscience Institute, Penn State Hershey Medical Center, Hershey, PA, United States
| | - Wei Zhong
- Department of Neurosurgery and Neuroscience Institute, Penn State Hershey Medical Center, Hershey, PA, United States
| | | | | | - Guohong Li
- Department of Neurosurgery and Neuroscience Institute, Penn State Hershey Medical Center, Hershey, PA, United States
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, United States
| |
Collapse
|
7
|
El Amki M, Glück C, Binder N, Middleham W, Wyss MT, Weiss T, Meister H, Luft A, Weller M, Weber B, Wegener S. Neutrophils Obstructing Brain Capillaries Are a Major Cause of No-Reflow in Ischemic Stroke. Cell Rep 2021; 33:108260. [PMID: 33053341 DOI: 10.1016/j.celrep.2020.108260] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 08/18/2020] [Accepted: 09/21/2020] [Indexed: 12/29/2022] Open
Abstract
Despite successful clot retrieval in large vessel occlusion stroke, ∼50% of patients have an unfavorable clinical outcome. The mechanisms underlying this functional reperfusion failure remain unknown, and therapeutic options are lacking. In the thrombin-model of middle cerebral artery (MCA) stroke in mice, we show that, despite successful thrombolytic recanalization of the proximal MCA, cortical blood flow does not fully recover. Using in vivo two-photon imaging, we demonstrate that this is due to microvascular obstruction of ∼20%-30% of capillaries in the infarct core and penumbra by neutrophils adhering to distal capillary segments. Depletion of circulating neutrophils using an anti-Ly6G antibody restores microvascular perfusion without increasing the rate of hemorrhagic complications. Strikingly, infarct size and functional deficits are smaller in mice treated with anti-Ly6G. Thus, we propose neutrophil stalling of brain capillaries to contribute to reperfusion failure, which offers promising therapeutic avenues for ischemic stroke.
Collapse
Affiliation(s)
- Mohamad El Amki
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Chaim Glück
- Experimental Imaging and Neuroenergetics, Institute of Pharmacology and Toxicology, University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Nadine Binder
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - William Middleham
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Matthias T Wyss
- Experimental Imaging and Neuroenergetics, Institute of Pharmacology and Toxicology, University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Tobias Weiss
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Hanna Meister
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Andreas Luft
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Bruno Weber
- Experimental Imaging and Neuroenergetics, Institute of Pharmacology and Toxicology, University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland.
| | - Susanne Wegener
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland.
| |
Collapse
|
8
|
Zhang Y, Li C, Qin Y, Cepparulo P, Millman M, Chopp M, Kemper A, Szalad A, Lu X, Wang L, Zhang ZG. Small extracellular vesicles ameliorate peripheral neuropathy and enhance chemotherapy of oxaliplatin on ovarian cancer. J Extracell Vesicles 2021; 10:e12073. [PMID: 33728031 PMCID: PMC7931803 DOI: 10.1002/jev2.12073] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 01/07/2021] [Accepted: 02/13/2021] [Indexed: 12/17/2022] Open
Abstract
There are no effective treatments for chemotherapy induced peripheral neuropathy (CIPN). Small extracellular vesicles (sEVs) facilitate intercellular communication and mediate nerve function and tumour progression. We found that the treatment of mice bearing ovarian tumour with sEVs derived from cerebral endothelial cells (CEC-sEVs) in combination with a chemo-drug, oxaliplatin, robustly reduced oxaliplatin-induced CIPN by decreasing oxaliplatin-damaged myelination and nerve fibres of the sciatic nerve and significantly amplified chemotherapy of oxaliplatin by reducing tumour size. The combination therapy substantially increased a set of sEV cargo-enriched miRNAs, but significantly reduced oxaliplatin-increased proteins in the sciatic nerve and tumour tissues. Bioinformatics analysis revealed the altered miRNAs and proteins formed two distinct networks that regulate neuropathy and tumour growth, respectively. Intravenously administered CEC-sEVs were internalized by axons of the sciatic nerve and cancer cells. Reduction of CEC-sEV cargo miRNAs abolished the effects of CEC-sEVs on oxaliplatin-inhibited axonal growth and on amplification of the anti-cancer effect in ovarian cancer cells, suggesting that alterations in the networks of miRNAs and proteins in recipient cells contribute to the therapeutic effect of CEC-sEVs on CIPN. Together, the present study demonstrates that CEC-sEVs suppressed CIPN and enhanced chemotherapy of oxaliplatin in the mouse bearing ovarian tumour.
Collapse
Affiliation(s)
- Yi Zhang
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| | - Chao Li
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| | - Yi Qin
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| | | | | | - Michael Chopp
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
- Department of PhysicsOakland UniversityRochesterMichiganUSA
| | - Amy Kemper
- Department of PathologyHenry Ford Health SystemDetroitMichiganUSA
| | - Alexandra Szalad
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| | - Xuerong Lu
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| | - Lei Wang
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| | - Zheng Gang Zhang
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| |
Collapse
|
9
|
Combination of Polyethylene Glycol-Conjugated Urokinase Nanogels and Urokinase for Acute Ischemic Stroke Therapeutic Implications. Transl Stroke Res 2020; 12:844-857. [DOI: 10.1007/s12975-020-00865-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 09/17/2020] [Accepted: 10/11/2020] [Indexed: 12/25/2022]
|
10
|
Bai S, Liao J, Zhang B, Zhao M, You B, Li P, Ran H, Wang Z, Shi R, Zhang G. Multimodal and multifunctional nanoparticles with platelet targeting ability and phase transition efficiency for the molecular imaging and thrombolysis of coronary microthrombi. Biomater Sci 2020; 8:5047-5060. [PMID: 32830839 DOI: 10.1039/d0bm00818d] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In this article, we constructed PLGA-cRGD-PFH-ICG NPs through emulsification process and then the bi-modal imaging of coronary microthrombi in ischemia/reperfusion rat model and thrombolysis of clots in vitro were both successfully completed by these NPs.
Collapse
Affiliation(s)
- Sheng Bai
- Department of Ultrasound
- Xiangya Hospital Central South University
- Changsha
- China
| | - Jintang Liao
- Department of Ultrasound
- Xiangya Hospital Central South University
- Changsha
- China
| | - Bo Zhang
- Department of Ultrasound
- Xiangya Hospital Central South University
- Changsha
- China
| | - Min Zhao
- Department of Nuclear Medicine
- Xiangya Hospital Central South University
- Changsha
- China
| | - Baiyang You
- Cardiac Rehabilitation Center
- Department of Rehabilitation
- Xiangya Hospital Central South University
- Changsha
- China
| | - Pan Li
- Institute of Ultrasound imaging of Chongqing Medical University
- Chongqing 400010
- P. R. China
| | - Haitao Ran
- Institute of Ultrasound imaging of Chongqing Medical University
- Chongqing 400010
- P. R. China
| | - Zhigang Wang
- Institute of Ultrasound imaging of Chongqing Medical University
- Chongqing 400010
- P. R. China
| | - Ruizheng Shi
- Department of Cardiovascular Medicine
- Xiangya Hospital Central South University
- Changsha
- China
| | - Guogang Zhang
- Department of Cardiovascular Medicine
- Xiangya Hospital Central South University
- Changsha
- China
| |
Collapse
|
11
|
Sezer M, van Royen N, Umman B, Bugra Z, Bulluck H, Hausenloy DJ, Umman S. Coronary Microvascular Injury in Reperfused Acute Myocardial Infarction: A View From an Integrative Perspective. J Am Heart Assoc 2019; 7:e009949. [PMID: 30608201 PMCID: PMC6404180 DOI: 10.1161/jaha.118.009949] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Murat Sezer
- 1 Istanbul Faculty of Medicine Istanbul University Istanbul Turkey
| | | | - Berrin Umman
- 1 Istanbul Faculty of Medicine Istanbul University Istanbul Turkey
| | - Zehra Bugra
- 1 Istanbul Faculty of Medicine Istanbul University Istanbul Turkey
| | - Heerajnarain Bulluck
- 3 The Hatter Cardiovascular Institute Institute of Cardiovascular Science University College London London United Kingdom.,4 Papworth Hospital NHS Trust Cambridge United Kingdom
| | - Derek J Hausenloy
- 3 The Hatter Cardiovascular Institute Institute of Cardiovascular Science University College London London United Kingdom.,4 Papworth Hospital NHS Trust Cambridge United Kingdom.,5 National Heart Research Institute Singapore National Heart Centre Singapore Singapore.,6 Cardiovascular and Metabolic Disorders Program Duke-National University of Singapore Singapore.,7 Yong Loo Lin School of Medicine National University Singapore Singapore.,8 The National Institute of Health Research University College London Hospitals Biomedical Research Centre London United Kingdom.,9 Barts Heart Centre St Bartholomew's Hospital London United Kingdom
| | - Sabahattin Umman
- 1 Istanbul Faculty of Medicine Istanbul University Istanbul Turkey
| |
Collapse
|
12
|
Jin R, Xiao AY, Li J, Wang M, Li G. PI3Kγ (Phosphoinositide 3-Kinase-γ) Inhibition Attenuates Tissue-Type Plasminogen Activator-Induced Brain Hemorrhage and Improves Microvascular Patency After Embolic Stroke. Hypertension 2019; 73:206-216. [PMID: 30571560 DOI: 10.1161/hypertensionaha.118.12001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Genetic and pharmacological inhibition of the PI3Kγ (phosphoinositide 3-kinase-γ) exerts anti-inflammatory and protective effects in a number of inflammatory and autoimmune diseases. SHRs (spontaneously hypertensive rats) subjected to embolic middle cerebral occlusion were treated with AS605240 (30 mg/kg) at 2 or 4 hours, tPA (tissue-type plasminogen activator; 10 mg/kg) at 2 or 6 hours, or AS605240 at 4 hours plus tPA at 6 hours. Infarct volume, brain hemorrhage, neurological function, microvascular thrombosis, and cerebral microvessel patency were examined. We found that treatment with AS605240 alone at 2 hours or the combination treatment with AS605240 at 4 hours and tPA at 6 hours significantly reduced infarct volume and neurological deficits at 3 days after stroke compared with ischemic rats treated with saline, AS605240 alone at 4 hours, and tPA alone at 6 hours. Moreover, the combination treatment effectively prevented the delayed tPA-induced cerebral hemorrhage. These protective effects are associated with reduced disruption of the blood-brain barrier, reduced downstream microvascular thrombosis, and improved microvascular patency by AS605240. Inhibition of the NF-κB (nuclear transcription factor-κB)-dependent MMP (matrix metalloproteinase)-9 and PAI-1 (plasminogen activator inhibitor-1) in the ischemic brain endothelium may underlie the neurovascular protective effect of AS605240. In addition, the combination treatment significantly reduced circulating platelet P-selectin expression and platelet-leukocyte aggregation compared with ischemic rats treated with saline or tPA alone at 6 hours. In conclusion, inhibition of PI3Kγ with AS605240 reduces delayed tPA-induced intracerebral hemorrhage and improves microvascular patency, which likely contributes to neuroprotective effect of the combination treatment.
Collapse
Affiliation(s)
- Rong Jin
- From the Department of Neurosurgery, Neuroscience Institute, Penn State Hershey Medical Center, Hershey, PA (R.J., M.W., G.L.)
| | - Adam Y Xiao
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport (A.Y.X.)
| | | | - Min Wang
- From the Department of Neurosurgery, Neuroscience Institute, Penn State Hershey Medical Center, Hershey, PA (R.J., M.W., G.L.)
| | - Guohong Li
- From the Department of Neurosurgery, Neuroscience Institute, Penn State Hershey Medical Center, Hershey, PA (R.J., M.W., G.L.)
| |
Collapse
|
13
|
Affiliation(s)
- Turgay Dalkara
- From the Department of Neurology, Faculty of Medicine and Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey; and Department of Radiology, Massachusetts General Hospital, Harvard University, Boston
| |
Collapse
|
14
|
Chan SL, Bishop N, Li Z, Cipolla MJ. Inhibition of PAI (Plasminogen Activator Inhibitor)-1 Improves Brain Collateral Perfusion and Injury After Acute Ischemic Stroke in Aged Hypertensive Rats. Stroke 2019; 49:1969-1976. [PMID: 29991657 DOI: 10.1161/strokeaha.118.022056] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background and Purpose- Aging and hypertension, comorbidities prevalent in the stroke population, are associated with poor collateral status and worsened stroke outcome. However, underlying mechanisms by which these conditions affect stroke outcome are not clear. We studied the role of PAI (plasminogen activator inhibitor)-1 that is increased in aging and hypertension on brain and vascular expression of inflammatory factors and perfusion that may contribute to worse stroke outcomes. Methods- Aged (≈50 weeks) and young (≈18 weeks) spontaneously hypertensive rats (SHR) were subjected to ischemia by middle cerebral artery occlusion (2 hours) and reperfusion (2 hours) with or without treatment with the PAI-1 inhibitor TM5441. Changes in middle cerebral artery and collateral perfusion territories were measured by multisite laser Doppler. Reactivity to TM5441 was studied using isolated and pressurized leptomeningeal anastomotic arterioles. Brain injury was determined by 2,3,5-triphenyltetrazolium staining and quantitative immunohistochemistry of amyloid-β-42, PAI-1, and hemoglobin. Circulating inflammatory factors were measured by ELISA. Results- Changes in cerebral blood flow during middle cerebral artery occlusion were similar between groups, with both having poor collateral perfusion and incomplete reperfusion. However, aged SHR had greater brain injury versus young (41±2 versus 23±2%, P<0.05) as well as increased brain deposition of amyloid-β-42 and circulating oxLDL (oxidized low-density lipoprotein). Erythrocyte aggregation and hemorrhage within the injured brain was observed in 50% of aged but no young SHR, with increased circulating PAI-1 in this subgroup of aged SHR (16±3 versus 6±2 ng/mL, P<0.05). PAI-1 inhibition with TM5441 improved brain injury but did not affect hemorrhage. TM5441 increased collateral perfusion by 38±7% and dilated leptomeningeal anastomotic arterioles by 44±10%, which was abolished by nitric oxide synthase inhibition. Conclusions- Increased injury in aged SHR seemed to be related to poor collateral perfusion, hemorrhagic transformation, increased amyloid-β-42, and oxidative stress. PAI-1 inhibition reduced infarction in both groups of SHR that possibly due, in part, to increased collateral perfusion.
Collapse
Affiliation(s)
- Siu-Lung Chan
- From the Departments of Neurological Sciences (S.-L.C., N.B., Z.L., M.J.C.)
| | - Nicole Bishop
- From the Departments of Neurological Sciences (S.-L.C., N.B., Z.L., M.J.C.)
| | - Zhaojin Li
- From the Departments of Neurological Sciences (S.-L.C., N.B., Z.L., M.J.C.)
| | - Marilyn J Cipolla
- From the Departments of Neurological Sciences (S.-L.C., N.B., Z.L., M.J.C.).,Obstetrics, Gynecology and Reproductive Sciences (M.J.C.).,Pharmacology (M.J.C.), University of Vermont Larner College of Medicine, Burlington
| |
Collapse
|
15
|
Griemert EV, Recarte Pelz K, Engelhard K, Schäfer MK, Thal SC. PAI-1 but Not PAI-2 Gene Deficiency Attenuates Ischemic Brain Injury After Experimental Stroke. Transl Stroke Res 2019; 10:372-380. [PMID: 29978354 PMCID: PMC6647425 DOI: 10.1007/s12975-018-0644-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 06/21/2018] [Accepted: 06/25/2018] [Indexed: 11/17/2022]
Abstract
After stroke, secondary brain damage is influenced by the extent of fibrin clot formation. This is counteracted by the endogenous fibrinolysis. Of major interest are the key players of the fibrinolytic plasminogen activator system including the urokinase plasminogen activator (uPA), the tissue-type plasminogen activator (tPA), and their endogenous inhibitors plasminogen activator inhibitor 1 (PAI-1) and PAI-2. The role of PAI-1 in brain injury is well established, whereas the importance of PAI-2 is unknown at present. The objectives of the present were twofold: first, to characterize the time-dependent cerebral mRNA expression of the plasminogen activator system (PAS) after brain ischemia and second, to investigate the impact of PAI-1 and PAI-2 on brain infarct volume using gene-deficient mice. Adult C57Bl/6J mice were subjected to unilateral transient middle cerebral artery occlusion (MCAO) followed by reperfusion for 3, 24, 72, or 120 h. Quantitative PCR revealed that brain mRNA expression levels of the PAS components, and particularly of PAI-1 (237-fold) and PAI-2 (19-fold), peaked at 24 h after stroke. Accordingly, PAI-1 plasma activity was strongly increased. Brain infarct volume in TTC (2,3,5-triphenyltetrazolium chloride)-stained brain sections was significantly smaller 24 h after MCAO in PAI-1-deficient mice (- 31%), but not in PAI-2-deficient mice (- 6%). Thus, endogenous upregulation of PAI-1, but not of PAI-2, might contribute to increased brain damage after acute ischemic stroke. The present study therefore shows that PAI-2 is induced by brain ischemia, but does not play an important or relevant role for secondary brain damage after brain injury.
Collapse
Affiliation(s)
- Eva-Verena Griemert
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Kirsten Recarte Pelz
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Kristin Engelhard
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Michael K Schäfer
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Serge C Thal
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany.
| |
Collapse
|
16
|
Increased volumes of mildly elevated capillary transit time heterogeneity positively predict favorable outcome and negatively predict intracranial hemorrhage in acute ischemic stroke with large vessel occlusion. Eur Radiol 2019; 29:3523-3532. [DOI: 10.1007/s00330-019-06064-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/05/2019] [Accepted: 02/04/2019] [Indexed: 01/11/2023]
|
17
|
Abstract
Recent stroke research has shifted the focus to the microvasculature from neuron-centric views. It is increasingly recognized that a successful neuroprotection is not feasible without microvascular protection. On the other hand, recent studies on pericytes, long-neglected cells on microvessels have provided insight into the regulation of microcirculation. Pericytes play an essential role in matching the metabolic demand of nervous tissue with the blood flow in addition to regulating the development and maintenance of the blood-brain barrier (BBB), leukocyte trafficking across the BBB and angiogenesis. Pericytes appears to be highly vulnerable to injury. Ischemic injury to pericytes on cerebral microvasculature unfavorably impacts the stroke-induced tissue damage and brain edema by disrupting microvascular blood flow and BBB integrity. Strongly supporting this, clinical imaging studies show that tissue reperfusion is not always obtained after recanalization. Therefore, prevention of pericyte dysfunction may improve the outcome of recanalization therapies by promoting microcirculatory reperfusion and preventing hemorrhage and edema. In the peri-infarct tissue, pericytes are detached from microvessels and promote angiogenesis and neurogenesis, and hence positively effect stroke outcome. Expectedly, we will learn more about the place of pericytes in CNS pathologies including stroke and devise approaches to treat them in the next decades.
Collapse
|
18
|
Cheng Q, Tong F, Shen Y, He C, Wang C, Ding F. Achyranthes bidentata polypeptide k improves long-term neurological outcomes through reducing downstream microvascular thrombosis in experimental ischemic stroke. Brain Res 2018; 1706:166-176. [PMID: 30414726 DOI: 10.1016/j.brainres.2018.11.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 11/04/2018] [Accepted: 11/08/2018] [Indexed: 01/07/2023]
Abstract
Achyranthes bidentata Bl. (A. bidentata) occupies an important position in traditional Chinese medicine owing to the property of promoting the circulation of blood and removing stasis. Achyranthes bidentata polypeptide k (ABPPk) is one of the active components isolated from A. bidentata. We previously demonstrated that ABPPk has potent neuroprotective effects against neuronal apoptosis both in vitro and in vivo, but the roles and mechanisms of ABPPk on long-term functional recovery after ischemic stroke remain unknown. In the current study, we investigated the neuroprotective effects of ABPPk on filament transient middle cerebral artery occlusion (tMCAO) rats and found that ABPPk reduced the infarct volume and maintained the neuronal integrity in the ischemic penumbra. Moreover, we found that ABPPk might reduce the formation of downstream microthrombus through preventing ischemic-induced oxidative damage of brain endothelial cells and activation of tissue factor (TF), plasminogen activator inhibitor-1 (PAI-1), and NF-κB. ABPPk also inhibited polymorphonuclear leukocytes (PMNs) infiltration and matrix metalloproteinase-2/-9 (MMP-2/-9) activation in the ischemic penumbra. Morris water maze, foot fault test, and modified neurological severity score were assessed for a period of 6 weeks following tMCAO. ABPPk improved long-term recognition abilities and neurological outcomes after stroke compared with saline-treated rats. Taken together, these results suggested that ABPPk is beneficial to the improvement of long-term outcomes after transient cerebral ischemia injury and can be used as a potential neuroprotective agent.
Collapse
Affiliation(s)
- Qiong Cheng
- School of Biology and Basic Medical Science, Soochow University, Suzhou, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Fang Tong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Chunjiao He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Caiping Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Fei Ding
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.
| |
Collapse
|
19
|
Engedal TS, Hjort N, Hougaard KD, Simonsen CZ, Andersen G, Mikkelsen IK, Boldsen JK, Eskildsen SF, Hansen MB, Angleys H, Jespersen SN, Pedraza S, Cho TH, Serena J, Siemonsen S, Thomalla G, Nighoghossian N, Fiehler J, Mouridsen K, Østergaard L. Transit time homogenization in ischemic stroke - A novel biomarker of penumbral microvascular failure? J Cereb Blood Flow Metab 2018; 38:2006-2020. [PMID: 28758524 PMCID: PMC6259320 DOI: 10.1177/0271678x17721666] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Cerebral ischemia causes widespread capillary no-flow in animal studies. The extent of microvascular impairment in human stroke, however, is unclear. We examined how acute intra-voxel transit time characteristics and subsequent recanalization affect tissue outcome on follow-up MRI in a historic cohort of 126 acute ischemic stroke patients. Based on perfusion-weighted MRI data, we characterized voxel-wise transit times in terms of their mean transit time (MTT), standard deviation (capillary transit time heterogeneity - CTH), and the CTH:MTT ratio (relative transit time heterogeneity), which is expected to remain constant during changes in perfusion pressure in a microvasculature consisting of passive, compliant vessels. To aid data interpretation, we also developed a computational model that relates graded microvascular failure to changes in these parameters. In perfusion-diffusion mismatch tissue, prolonged mean transit time (>5 seconds) and very low cerebral blood flow (≤6 mL/100 mL/min) was associated with high risk of infarction, largely independent of recanalization status. In the remaining mismatch region, low relative transit time heterogeneity predicted subsequent infarction if recanalization was not achieved. Our model suggested that transit time homogenization represents capillary no-flow. Consistent with this notion, low relative transit time heterogeneity values were associated with lower cerebral blood volume. We speculate that low RTH may represent a novel biomarker of penumbral microvascular failure.
Collapse
Affiliation(s)
- Thorbjørn S Engedal
- 1 Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University, Aarhus University Hospital, Aarhus C, Denmark.,2 Department of Neuroradiology, Aarhus University Hospital, Nørrebrogade 44, 8000 Aarhus C, Denmark
| | - Niels Hjort
- 3 Department of Neurology Aarhus University Hospital, Aarhus C, Denmark
| | | | - Claus Z Simonsen
- 3 Department of Neurology Aarhus University Hospital, Aarhus C, Denmark
| | - Grethe Andersen
- 3 Department of Neurology Aarhus University Hospital, Aarhus C, Denmark
| | - Irene Klærke Mikkelsen
- 1 Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University, Aarhus University Hospital, Aarhus C, Denmark
| | - Jens K Boldsen
- 1 Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University, Aarhus University Hospital, Aarhus C, Denmark
| | - Simon F Eskildsen
- 1 Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University, Aarhus University Hospital, Aarhus C, Denmark
| | - Mikkel B Hansen
- 1 Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University, Aarhus University Hospital, Aarhus C, Denmark
| | - Hugo Angleys
- 1 Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University, Aarhus University Hospital, Aarhus C, Denmark
| | - Sune N Jespersen
- 1 Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University, Aarhus University Hospital, Aarhus C, Denmark.,4 Department of Physics and Astronomy, Aarhus University, Aarhus, Denmark
| | | | - Tae H Cho
- 6 Hospices Civils de Lyon, Lyon, France
| | | | | | - Götz Thomalla
- 7 University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | | | - Jens Fiehler
- 7 University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Kim Mouridsen
- 1 Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University, Aarhus University Hospital, Aarhus C, Denmark
| | - Leif Østergaard
- 1 Center of Functionally Integrative Neuroscience and MINDLab, Aarhus University, Aarhus University Hospital, Aarhus C, Denmark.,2 Department of Neuroradiology, Aarhus University Hospital, Nørrebrogade 44, 8000 Aarhus C, Denmark
| |
Collapse
|
20
|
Jin R, Xiao AY, Liu S, Wang M, Li G. Taurine Reduces tPA (Tissue-Type Plasminogen Activator)-Induced Hemorrhage and Microvascular Thrombosis After Embolic Stroke in Rat. Stroke 2018; 49:1708-1718. [PMID: 29844028 DOI: 10.1161/strokeaha.118.020747] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/18/2018] [Accepted: 04/25/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE Taurine (2-aminoethansulfolic amino acid) exerts neuroprotective actions in experimental stroke. Here, we investigated the effect of taurine in combination with delayed tPA (tissue-type plasminogen activator) on embolic stroke. METHODS Rats subjected to embolic middle cerebral artery occlusion were treated with taurine (50 mg/kg) at 4 hours in combination with tPA (10 mg/kg) at 6 hours. Control groups consisted of ischemic rats treated with either taurine (50 mg/kg) or saline at 4 hours or tPA (10 mg/kg) alone at 2 or 6 hours after middle cerebral artery occlusion. RESULTS We found that combination treatment with taurine and tPA robustly reduced infarct volume and neurological deficits 3 days after stroke, whereas treatment with taurine alone had a less-significant protective effect. tPA alone at 6 hours had no effects on infarct volume but instead induced intracerebral hemorrhage. The combination treatment with taurine prevented tPA-associated hemorrhage and reduced intravascular deposition of fibrin/fibrinogen and platelets in downstream microvessels and hence improved microvascular patency. These protective effects are associated with profound inhibition of CD147 (cluster of differentiation 147)-dependent MMP-9 (matrix metalloproteinase-9) pathway in ischemic brain endothelium by taurine. Notably, targeted inhibition of CD147 by intracerebroventricular injection of the rat CD147 siRNA profoundly inhibited ischemia-induced and tPA-enhanced MMP-9 activity in ischemic brain endothelium and blocked tPA-induced cerebral hemorrhage. Finally, the combination treatment with taurine and tPA improved long-term outcome at least 45 days after stroke compared with saline-treated group. CONCLUSIONS Our results suggest that taurine in combination with tPA may be a clinically feasible approach toward future attempts at combination stroke therapy.
Collapse
Affiliation(s)
- Rong Jin
- From the Department of Neurosurgery, Neuroscience Institute, Penn State Hershey Medical Center (R.J., S.L., M.W., G.L.)
| | - Adam Y Xiao
- Department of Molecular and Cellular Physiology (A.Y.X., G.L.), Louisiana State University Health Sciences Center, Shreveport
| | - Shan Liu
- From the Department of Neurosurgery, Neuroscience Institute, Penn State Hershey Medical Center (R.J., S.L., M.W., G.L.)
| | - Min Wang
- From the Department of Neurosurgery, Neuroscience Institute, Penn State Hershey Medical Center (R.J., S.L., M.W., G.L.)
| | - Guohong Li
- From the Department of Neurosurgery, Neuroscience Institute, Penn State Hershey Medical Center (R.J., S.L., M.W., G.L.) .,Department of Neurosurgery (G.L.).,Department of Molecular and Cellular Physiology (A.Y.X., G.L.), Louisiana State University Health Sciences Center, Shreveport
| |
Collapse
|
21
|
El Amki M, Wegener S. Improving Cerebral Blood Flow after Arterial Recanalization: A Novel Therapeutic Strategy in Stroke. Int J Mol Sci 2017; 18:ijms18122669. [PMID: 29232823 PMCID: PMC5751271 DOI: 10.3390/ijms18122669] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 11/30/2017] [Accepted: 12/06/2017] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke is caused by a disruption in blood supply to a region of the brain. It induces dysfunction of brain cells and networks, resulting in sudden neurological deficits. The cause of stroke is vascular, but the consequences are neurological. Decades of research have focused on finding new strategies to reduce the neural damage after cerebral ischemia. However, despite the incredibly huge investment, all strategies targeting neuroprotection have failed to demonstrate clinical efficacy. Today, treatment for stroke consists of dealing with the cause, attempting to remove the occluding blood clot and recanalize the vessel. However, clinical evidence suggests that the beneficial effect of post-stroke recanalization may be hampered by the occurrence of microvascular reperfusion failure. In short: recanalization is not synonymous with reperfusion. Today, clinicians are confronted with several challenges in acute stroke therapy, even after successful recanalization: (1) induce reperfusion, (2) avoid hemorrhagic transformation (HT), and (3) avoid early or late vascular reocclusion. All these parameters impact the restoration of cerebral blood flow after stroke. Recent advances in understanding the molecular consequences of recanalization and reperfusion may lead to innovative therapeutic strategies for improving reperfusion after stroke. In this review, we will highlight the importance of restoring normal cerebral blood flow after stroke and outline molecular mechanisms involved in blood flow regulation.
Collapse
Affiliation(s)
- Mohamad El Amki
- Department of Neurology, University Hospital Zurich and University of Zurich, 8091 Zürich, Switzerland.
| | - Susanne Wegener
- Department of Neurology, University Hospital Zurich and University of Zurich, 8091 Zürich, Switzerland.
| |
Collapse
|
22
|
Jin R, Xiao AY, Chen R, Granger DN, Li G. Inhibition of CD147 (Cluster of Differentiation 147) Ameliorates Acute Ischemic Stroke in Mice by Reducing Thromboinflammation. Stroke 2017; 48:3356-3365. [PMID: 29114092 DOI: 10.1161/strokeaha.117.018839] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/03/2017] [Accepted: 10/11/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND PURPOSE Inflammation and thrombosis currently are recognized as critical contributors to the pathogenesis of ischemic stroke. CD147 (cluster of differentiation 147), also known as extracellular matrix metalloproteinase inducer, can function as a key mediator of inflammatory and immune responses. CD147 expression is increased in the brain after cerebral ischemia, but its role in the pathogenesis of ischemic stroke remains unknown. In this study, we show that CD147 acts as a key player in ischemic stroke by driving thrombotic and inflammatory responses. METHODS Focal cerebral ischemia was induced in C57BL/6 mice by a 60-minute transient middle cerebral artery occlusion. Animals were treated with anti-CD147 function-blocking antibody (αCD147) or isotype control antibody. Blood-brain barrier permeability, thrombus formation, and microvascular patency were assessed 24 hours after ischemia. Infarct size, neurological deficits, and inflammatory cells invaded in the brain were assessed 72 hours after ischemia. RESULTS CD147 expression was rapidly increased in ischemic brain endothelium after transient middle cerebral artery occlusion. Inhibition of CD147 reduced infarct size and improved functional outcome on day 3 after transient middle cerebral artery occlusion. The neuroprotective effects were associated with (1) prevented blood-brain barrier damage, (2) decreased intravascular fibrin and platelet deposition, which in turn reduced thrombosis and increased cerebral perfusion, and (3) reduced brain inflammatory cell infiltration. The underlying mechanism may include reduced NF-κB (nuclear factor κB) activation, MMP-9 (matrix metalloproteinase-9) activity, and PAI-1 (plasminogen activator inhibitor-1) expression in brain microvascular endothelial cells. CONCLUSIONS Inhibition of CD147 ameliorates acute ischemic stroke by reducing thromboinflammation. CD147 might represent a novel and promising therapeutic target for ischemic stroke and possibly other thromboinflammatory disorders.
Collapse
Affiliation(s)
- Rong Jin
- From the Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey (R.J., G.L.); and Department of Neurosurgery (R.C., R.J., G.L.) and the Department of Molecular and Cellular Physiology (A.Y.X., G.L., D.N.G.), Louisiana State University Health Sciences Center, Shreveport
| | - Adam Y Xiao
- From the Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey (R.J., G.L.); and Department of Neurosurgery (R.C., R.J., G.L.) and the Department of Molecular and Cellular Physiology (A.Y.X., G.L., D.N.G.), Louisiana State University Health Sciences Center, Shreveport
| | - Rui Chen
- From the Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey (R.J., G.L.); and Department of Neurosurgery (R.C., R.J., G.L.) and the Department of Molecular and Cellular Physiology (A.Y.X., G.L., D.N.G.), Louisiana State University Health Sciences Center, Shreveport
| | - D Neil Granger
- From the Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey (R.J., G.L.); and Department of Neurosurgery (R.C., R.J., G.L.) and the Department of Molecular and Cellular Physiology (A.Y.X., G.L., D.N.G.), Louisiana State University Health Sciences Center, Shreveport
| | - Guohong Li
- From the Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey (R.J., G.L.); and Department of Neurosurgery (R.C., R.J., G.L.) and the Department of Molecular and Cellular Physiology (A.Y.X., G.L., D.N.G.), Louisiana State University Health Sciences Center, Shreveport.
| |
Collapse
|
23
|
Lu D, Sanberg PR, Mahmood A, Li Y, Wang L, Sanchez-Ramos J, Chopp M. Intravenous Administration of Human Umbilical Cord Blood Reduces Neurological Deficit in the Rat after Traumatic Brain Injury. Cell Transplant 2017. [DOI: 10.3727/096020198389924] [Citation(s) in RCA: 205] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We measured the effect of treatment of traumatic brain injury (TBI) in the rat with human umbilical cord blood (HUCB) administered IV. HUCB cells were injected into the tail vein 24 h after TBI and the rats were sacrificed at day 28 after the treatment. The Rotarod test and the neurological severity score (NSS) were used to evaluate neurological function. The distribution of the donor cells in the brain, heart, lung, kidney, liver, spleen, bone marrow, and muscle were analyzed in recipient rats using immunohistochemical staining and laser confocal microscopy. HUCB cells injected IV significantly reduced motor and neurological deficits compared with control groups by day 28 after the treatment. The cells preferentially entered the brain and migrated into the parenchyma of the injured brain and expressed the neuronal markers, NeuN and MAP-2, and the astrocytic marker, GFAP. Some HUCB cells integrated into the vascular walls within the boundary zone of the injured area. Our data suggest that IV administration of HUCB may be useful in the treatment of TBI.
Collapse
Affiliation(s)
- Dunyue Lu
- Departments of Neurosurgery, Henry Ford Health Sciences Center, Detroit, MI
| | - Paul R. Sanberg
- Center for Aging and Repair, Departments of Neurosurgery, University of South Florida, Tampa, FL
| | - Asim Mahmood
- Departments of Neurosurgery, Henry Ford Health Sciences Center, Detroit, MI
| | - Yi Li
- Departments of Neurology, Henry Ford Health Sciences Center, Detroit, MI
| | - Lei Wang
- Departments of Neurology, Henry Ford Health Sciences Center, Detroit, MI
| | - Juan Sanchez-Ramos
- Center for Aging and Repair, Departments of Neurology, University of South Florida, Tampa, FL
| | - Michael Chopp
- Departments of Neurology, Henry Ford Health Sciences Center, Detroit, MI
- Department of Physics, Oakland University, Rochester, MI
| |
Collapse
|
24
|
Annexin A2 Plus Low-Dose Tissue Plasminogen Activator Combination Attenuates Cerebrovascular Dysfunction After Focal Embolic Stroke of Rats. Transl Stroke Res 2017; 8:549-559. [PMID: 28580536 DOI: 10.1007/s12975-017-0542-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 05/18/2017] [Accepted: 05/23/2017] [Indexed: 02/06/2023]
Abstract
Previous studies showed recombinant annexin A2 (rA2) in combination with low-dose tissue-type plasminogen activator (tPA) improved thrombolytic efficacy and long-term neurological outcomes after embolic focal ischemia in rats. The objective of this study was to investigate the effects and mechanisms of the combination in early BBB integrity and cerebrovascular patency in the rat focal embolic stroke model. Ischemic brain infarct volume and hemorrhagic transformation were quantified at 24 h after stroke. At an earlier time point, 16 h after stroke, BBB integrity was evaluated by IgG extravasation, and the involved mechanisms were assessed for tight junction ZO-1 and adhesion junction ve-cadherin protein expression, matrix metalloproteinase activation, extracellular matrix collagen IV and endothelial barrier antigen expression, and activation of microglia/macrophages and astrocytes. While at the same time point, cerebrovascular patency was assessed by intravascular fibrin and platelet depositions. At 24 h after stroke, the combination showed significant reduction in brain infarction and intracerebral hemorrhage. At 16 h after stroke onset, the combination therapy significantly reduced BBB disruption, and improved preservation of the junction proteins ZO-1 and ve-cadherin, decreased activation of matrix metalloproteinase, inhibited degradation of extracellular matrix collagen IV and endothelial barrier antigen, and reduced microglia/macrophage and astrocytes activations. Meanwhile, the combination also significantly improved cerebrovascular patency by reducing intravascular fibrin and platelet depositions in the peri-infarct brain tissues. These results suggest the beneficial effects of the rA2 plus low-dose tPA combination may be mediated in part by the amelioration of BBB disruption and improvement of cerebrovascular patency.
Collapse
|
25
|
Abstract
Stroke is one of the leading causes of death and disability worldwide. Stroke recovery is orchestrated by a set of highly interactive processes that involve the neurovascular unit and neural stem cells. Emerging data suggest that exosomes play an important role in intercellular communication by transferring exosomal protein and RNA cargo between source and target cells in the brain. Here, we review these advances and their impact on promoting coupled brain remodeling processes after stroke. The use of exosomes for therapeutic applications in stroke is also highlighted.
Collapse
Affiliation(s)
- Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
- Department of Physics, Oakland University, Rochester, Michigan, USA
| |
Collapse
|
26
|
Desilles JP, Loyau S, Syvannarath V, Gonzalez-Valcarcel J, Cantier M, Louedec L, Lapergue B, Amarenco P, Ajzenberg N, Jandrot-Perrus M, Michel JB, Ho-Tin-Noe B, Mazighi M. Alteplase Reduces Downstream Microvascular Thrombosis and Improves the Benefit of Large Artery Recanalization in Stroke. Stroke 2015; 46:3241-8. [PMID: 26443832 DOI: 10.1161/strokeaha.115.010721] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 09/02/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Downstream microvascular thrombosis (DMT) is known to be a contributing factor to incomplete reperfusion in acute ischemic stroke. The aim of this study was to determine the timing of DMT with intravital imaging and to test the hypothesis that intravenous alteplase infusion could reduce DMT in a transient middle cerebral artery occlusion (MCAO) rat stroke model. METHODS Rats were subjected to 60-minute transient MCAO. Alteplase (10 mg/kg) was administered 30 minutes after the beginning of MCAO. Real-time intravital fluorescence microscopy through a dura-sparing craniotomy was used to visualize circulating blood cells and fibrinogen. Cerebral microvessel patency was quantitatively evaluated by fluorescein isothiocyanate-dextran perfusion. RESULTS Immediately after MCAO, platelet and leukocyte accumulation were observed mostly in the venous compartment. Within 30 minutes after MCAO, microthrombi and parietal fibrin deposits were detected in postcapillary microvessels. Alteplase treatment significantly (P=0.006) reduced infarct volume and increased the percentage of perfused vessels during MCAO (P=0.02) compared with saline. Plasma levels of fibrinogen from alteplase-treated rats showed a rapid and profound hypofibrinogenemia. In vitro platelet aggregation demonstrated that alteplase reduced platelet aggregation (P=0.0001) and facilitated platelet disaggregation (P=0.001). These effects were reversible in the presence of exogenous fibrinogen. CONCLUSIONS Our data demonstrate that DMT is an early phenomenon initiated before recanalization. We further show that alteplase-dependent maintenance of downstream perfusion during MCAO improves acute ischemic stroke outcome through a fibrinogen-dependent platelet aggregation reduction. Our results indicate that early targeting of DMT represents a therapeutic strategy to improve the benefit of large artery recanalization in acute ischemic stroke.
Collapse
Affiliation(s)
- Jean-Philippe Desilles
- From the Univ Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France (J.-P.D., S.L., V.S., J.G.-V., M.C., L.L., P.A., N.A., M.J.-P., J.-B.M., B.H.-T.-N., M.M.); Division of Neurology, Stroke Center, Foch Hospital, University Versailles Saint-Quentin en Yvelines, Paris, France (B.L.); Departments of Neurology and Stroke Center (P.A.) and Hematology (N.A.), AP-HP, Bichat Hospital, Paris, France; and Department of Neurology and Stroke Center, AP-HP, Lariboisière Hospital, DHU Neurovasc, Paris, France (M.M.).
| | - Stephane Loyau
- From the Univ Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France (J.-P.D., S.L., V.S., J.G.-V., M.C., L.L., P.A., N.A., M.J.-P., J.-B.M., B.H.-T.-N., M.M.); Division of Neurology, Stroke Center, Foch Hospital, University Versailles Saint-Quentin en Yvelines, Paris, France (B.L.); Departments of Neurology and Stroke Center (P.A.) and Hematology (N.A.), AP-HP, Bichat Hospital, Paris, France; and Department of Neurology and Stroke Center, AP-HP, Lariboisière Hospital, DHU Neurovasc, Paris, France (M.M.)
| | - Varouna Syvannarath
- From the Univ Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France (J.-P.D., S.L., V.S., J.G.-V., M.C., L.L., P.A., N.A., M.J.-P., J.-B.M., B.H.-T.-N., M.M.); Division of Neurology, Stroke Center, Foch Hospital, University Versailles Saint-Quentin en Yvelines, Paris, France (B.L.); Departments of Neurology and Stroke Center (P.A.) and Hematology (N.A.), AP-HP, Bichat Hospital, Paris, France; and Department of Neurology and Stroke Center, AP-HP, Lariboisière Hospital, DHU Neurovasc, Paris, France (M.M.)
| | - Jaime Gonzalez-Valcarcel
- From the Univ Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France (J.-P.D., S.L., V.S., J.G.-V., M.C., L.L., P.A., N.A., M.J.-P., J.-B.M., B.H.-T.-N., M.M.); Division of Neurology, Stroke Center, Foch Hospital, University Versailles Saint-Quentin en Yvelines, Paris, France (B.L.); Departments of Neurology and Stroke Center (P.A.) and Hematology (N.A.), AP-HP, Bichat Hospital, Paris, France; and Department of Neurology and Stroke Center, AP-HP, Lariboisière Hospital, DHU Neurovasc, Paris, France (M.M.)
| | - Marie Cantier
- From the Univ Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France (J.-P.D., S.L., V.S., J.G.-V., M.C., L.L., P.A., N.A., M.J.-P., J.-B.M., B.H.-T.-N., M.M.); Division of Neurology, Stroke Center, Foch Hospital, University Versailles Saint-Quentin en Yvelines, Paris, France (B.L.); Departments of Neurology and Stroke Center (P.A.) and Hematology (N.A.), AP-HP, Bichat Hospital, Paris, France; and Department of Neurology and Stroke Center, AP-HP, Lariboisière Hospital, DHU Neurovasc, Paris, France (M.M.)
| | - Liliane Louedec
- From the Univ Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France (J.-P.D., S.L., V.S., J.G.-V., M.C., L.L., P.A., N.A., M.J.-P., J.-B.M., B.H.-T.-N., M.M.); Division of Neurology, Stroke Center, Foch Hospital, University Versailles Saint-Quentin en Yvelines, Paris, France (B.L.); Departments of Neurology and Stroke Center (P.A.) and Hematology (N.A.), AP-HP, Bichat Hospital, Paris, France; and Department of Neurology and Stroke Center, AP-HP, Lariboisière Hospital, DHU Neurovasc, Paris, France (M.M.)
| | - Bertrand Lapergue
- From the Univ Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France (J.-P.D., S.L., V.S., J.G.-V., M.C., L.L., P.A., N.A., M.J.-P., J.-B.M., B.H.-T.-N., M.M.); Division of Neurology, Stroke Center, Foch Hospital, University Versailles Saint-Quentin en Yvelines, Paris, France (B.L.); Departments of Neurology and Stroke Center (P.A.) and Hematology (N.A.), AP-HP, Bichat Hospital, Paris, France; and Department of Neurology and Stroke Center, AP-HP, Lariboisière Hospital, DHU Neurovasc, Paris, France (M.M.)
| | - Pierre Amarenco
- From the Univ Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France (J.-P.D., S.L., V.S., J.G.-V., M.C., L.L., P.A., N.A., M.J.-P., J.-B.M., B.H.-T.-N., M.M.); Division of Neurology, Stroke Center, Foch Hospital, University Versailles Saint-Quentin en Yvelines, Paris, France (B.L.); Departments of Neurology and Stroke Center (P.A.) and Hematology (N.A.), AP-HP, Bichat Hospital, Paris, France; and Department of Neurology and Stroke Center, AP-HP, Lariboisière Hospital, DHU Neurovasc, Paris, France (M.M.)
| | - Nadine Ajzenberg
- From the Univ Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France (J.-P.D., S.L., V.S., J.G.-V., M.C., L.L., P.A., N.A., M.J.-P., J.-B.M., B.H.-T.-N., M.M.); Division of Neurology, Stroke Center, Foch Hospital, University Versailles Saint-Quentin en Yvelines, Paris, France (B.L.); Departments of Neurology and Stroke Center (P.A.) and Hematology (N.A.), AP-HP, Bichat Hospital, Paris, France; and Department of Neurology and Stroke Center, AP-HP, Lariboisière Hospital, DHU Neurovasc, Paris, France (M.M.)
| | - Martine Jandrot-Perrus
- From the Univ Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France (J.-P.D., S.L., V.S., J.G.-V., M.C., L.L., P.A., N.A., M.J.-P., J.-B.M., B.H.-T.-N., M.M.); Division of Neurology, Stroke Center, Foch Hospital, University Versailles Saint-Quentin en Yvelines, Paris, France (B.L.); Departments of Neurology and Stroke Center (P.A.) and Hematology (N.A.), AP-HP, Bichat Hospital, Paris, France; and Department of Neurology and Stroke Center, AP-HP, Lariboisière Hospital, DHU Neurovasc, Paris, France (M.M.)
| | - Jean-Baptiste Michel
- From the Univ Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France (J.-P.D., S.L., V.S., J.G.-V., M.C., L.L., P.A., N.A., M.J.-P., J.-B.M., B.H.-T.-N., M.M.); Division of Neurology, Stroke Center, Foch Hospital, University Versailles Saint-Quentin en Yvelines, Paris, France (B.L.); Departments of Neurology and Stroke Center (P.A.) and Hematology (N.A.), AP-HP, Bichat Hospital, Paris, France; and Department of Neurology and Stroke Center, AP-HP, Lariboisière Hospital, DHU Neurovasc, Paris, France (M.M.)
| | - Benoit Ho-Tin-Noe
- From the Univ Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France (J.-P.D., S.L., V.S., J.G.-V., M.C., L.L., P.A., N.A., M.J.-P., J.-B.M., B.H.-T.-N., M.M.); Division of Neurology, Stroke Center, Foch Hospital, University Versailles Saint-Quentin en Yvelines, Paris, France (B.L.); Departments of Neurology and Stroke Center (P.A.) and Hematology (N.A.), AP-HP, Bichat Hospital, Paris, France; and Department of Neurology and Stroke Center, AP-HP, Lariboisière Hospital, DHU Neurovasc, Paris, France (M.M.)
| | - Mikael Mazighi
- From the Univ Paris Diderot, Sorbonne Paris Cite, Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France (J.-P.D., S.L., V.S., J.G.-V., M.C., L.L., P.A., N.A., M.J.-P., J.-B.M., B.H.-T.-N., M.M.); Division of Neurology, Stroke Center, Foch Hospital, University Versailles Saint-Quentin en Yvelines, Paris, France (B.L.); Departments of Neurology and Stroke Center (P.A.) and Hematology (N.A.), AP-HP, Bichat Hospital, Paris, France; and Department of Neurology and Stroke Center, AP-HP, Lariboisière Hospital, DHU Neurovasc, Paris, France (M.M.)
| |
Collapse
|
27
|
Dalkara T, Alarcon-Martinez L. Cerebral microvascular pericytes and neurogliovascular signaling in health and disease. Brain Res 2015; 1623:3-17. [DOI: 10.1016/j.brainres.2015.03.047] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 03/10/2015] [Accepted: 03/27/2015] [Indexed: 02/07/2023]
|
28
|
Hale SL, Kloner RA. Dabigatran treatment: effects on infarct size and the no-reflow phenomenon in a model of acute myocardial ischemia/reperfusion. J Thromb Thrombolysis 2015; 39:50-4. [PMID: 25017632 DOI: 10.1007/s11239-014-1098-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The no-reflow phenomenon occurs when an epicardial coronary artery is reopened following myocardial infarction, but portions of the intramural microvasculature fail to reperfuse. One potential mechanism for this is the presence of fibrin tactoids. In addition, some recent studies have suggested that dabigatran treatment may be associated with increased incidence of myocardial infarction. Our aim was to investigate the effect on myocardial infarct size and no-reflow in an acute model of ischemia/reperfusion. Anesthetized, open-chest rabbits were randomly assigned to receive dabigatran (Dab, 0.5 mg/kg bolus + infusion, 0.15 mg/kg/h, IV, n = 11) or vehicle (Veh, n = 11) 15 m before a 30-m coronary artery occlusion and during 2.5 h of the 3 h reperfusion procedure. At the end of the reperfusion period, infarct size (% risk zone) and no-reflow defect were measured. The ischemic risk zone (% of left ventricle) was similar in groups, 24 % in Dab and 25 % in Veh. Necrosis was neither reduced nor increased by Dab treatment; expressed as a percentage of the risk region, infarct size was 30 ± 4 % in Dab and 28 ± 5 % in Veh, p = 0.76. The extent of no-reflow was comparable, expressed either as a percent of the risk region (19 ± 3 %, Dab and 18 ± 3 %, Veh) or as a percent of the necrotic zone (67 ± 8 % Dab and 65 ± 10 % Veh). Dab treatment had no effect on heart rate or blood pressure. Dabigatran treatment did not prevent or ameliorate the no-reflow phenomenon, suggesting that fibrin does not play a major role in the development of microvascular obstruction. Dabigatran did not exacerbate myocardial infarct size.
Collapse
Affiliation(s)
- Sharon L Hale
- The Heart Institute of Good Samaritan Hospital, 1225 Wilshire Blvd, Los Angeles, CA, 90017, USA,
| | | |
Collapse
|
29
|
Jiang Y, Brynskikh AM, S-Manickam D, Kabanov AV. SOD1 nanozyme salvages ischemic brain by locally protecting cerebral vasculature. J Control Release 2015; 213:36-44. [PMID: 26093094 PMCID: PMC4684498 DOI: 10.1016/j.jconrel.2015.06.021] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 06/15/2015] [Indexed: 11/30/2022]
Abstract
Copper/zinc superoxide dismutase (CuZnSOD; SOD1) is widely considered as a potential therapeutic candidate for pathologies involving oxidative stress, but its application has been greatly hindered by delivery issues. In our previous study, nanoformulated SOD1 (cl-nanozyme) was shown to decrease infarct volume and improve sensorimotor functions after a single intravenous (IV) injection in a rat middle cerebral artery occlusion (MCAO) model of ischemia/reperfusion (I/R) injury (stroke). However, it remained unclear how cl-nanozyme was able to deliver SOD1 to the brain and exert therapeutic efficacy. The present study aims to answer this question by exploring micro-distribution pattern of cl-nanozyme in the rat brain after stroke. Immunohistochemistry studies demonstrated cl-nanozyme co-localization with fibrin along damaged arteries and capillaries in the ischemic hemisphere. We further found that cl-nanozyme can be cross-linked into thrombi formed after I/R injury in the brain, and this effect is independent of animal species (rat/mouse) used for modeling I/R injury. This work is also the first report reinforcing therapeutic potential of cl-nanozyme in a well-characterized mouse MCAO model of I/R injury.
Collapse
Affiliation(s)
- Yuhang Jiang
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Anna M Brynskikh
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Devika S-Manickam
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Alexander V Kabanov
- Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Laboratory for Chemical Design of Bionanomaterials, Faculty of Chemistry, M.V. Lomonosov Moscow State University, Moscow 117234, Russia.
| |
Collapse
|
30
|
Li TT, Fan ML, Hou SX, Li XY, Barry DM, Jin H, Luo SY, Kong F, Lau LF, Dai XR, Zhang GH, Zhou LL. A novel snake venom-derived GPIb antagonist, anfibatide, protects mice from acute experimental ischaemic stroke and reperfusion injury. Br J Pharmacol 2015; 172:3904-16. [PMID: 25917571 DOI: 10.1111/bph.13178] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 04/02/2015] [Accepted: 04/21/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Ischaemic stroke is a serious disease with limited therapy options. Glycoprotein (GP)Ib binding to von Willebrand factor (vWF) exposed at vascular injury initiates platelet adhesion and contributes to platelet aggregation. GPIb has been suggested as an effective target for antithrombotic therapy in stroke. Anfibatide is a GPIb antagonist derived from snake venom and we investigated its protective effect on experimental brain ischaemia in mice. EXPERIMENTAL APPROACH Focal cerebral ischaemia was induced by 90 min of transient middle cerebral artery occlusion (MCAO). These mice were then treated with anfibatide (4, 2, 1 μg·kg(-1) ), injected i.v., after 90 min of MCAO, followed by 1 h of reperfusion. Tirofiban, a GPIIb/IIIα antagonist, was used as a positive control. KEY RESULTS Twenty-four hours after MCAO, anfibatide-treated mice showed significantly improved ischaemic lesions in a dose-dependent manner. The mice had smaller infarct volumes, less severe neurological deficits and histopathology of cerebrum tissues compared with the untreated MCAO mice. Moreover, anfibatide decreased the amount of GPIbα, vWF and accumulation of fibrin(ogen) in the vasculature of the ischaemic hemisphere. Tirofiban had similar effects on infarct size and fibrin(ogen) deposition compared with the MCAO group. Importantly, the anfibatide-treated mice showed a lower incidence of intracerebral haemorrhage and shorter tail bleeding time compared with the tirofiban-treated mice. CONCLUSIONS AND IMPLICATIONS Our data indicate anfibatide is a safe GPIb antagonist that exerts a protective effect on cerebral ischaemia and reperfusion injury. Anfibatide is a promising candidate that could be beneficial for the treatment of ischaemic stroke.
Collapse
Affiliation(s)
- Ting-Ting Li
- Department of Pharmacology, School of Basic Medical Science, Anhui Medical University, Hefei, Anhui, China
| | - Man-Li Fan
- Department of Pharmacology, School of Basic Medical Science, Anhui Medical University, Hefei, Anhui, China
| | - Shi-Xiang Hou
- Department of Pharmacology, School of Basic Medical Science, Anhui Medical University, Hefei, Anhui, China.,Department of Pharmacy, Xuancheng People's Hospital, Xuancheng, China
| | - Xiao-Yi Li
- Zhaoke Pharmaceutical Co. Ltd, Hefei, Anhui, China
| | - Devin M Barry
- Department of Anesthesiology, School of Medicine, Washington University, St. Louis, MO, USA
| | - Hui Jin
- Department of Pharmaceutical Chemistry, Jiangsu Changjiang Pharmaceutical Co. Ltd, Shanghai, China
| | - Sheng-Yong Luo
- Department of Pharmacology, Anhui Academy of Medical Sciences, Hefei, Anhui, China
| | - Feng Kong
- Department of Pharmacology, School of Basic Medical Science, Anhui Medical University, Hefei, Anhui, China
| | - Lit-Fui Lau
- Zhaoke Pharmaceutical Co. Ltd, Hefei, Anhui, China
| | | | - Guo-Hui Zhang
- Department of Pharmacy, Xuancheng People's Hospital, Xuancheng, China.,Zhaoke Pharmaceutical Co. Ltd, Hefei, Anhui, China
| | - Lan-Lan Zhou
- Department of Pharmacology, School of Basic Medical Science, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
31
|
Zhang L, Zhang RL, Jiang Q, Ding G, Chopp M, Zhang ZG. Focal embolic cerebral ischemia in the rat. Nat Protoc 2015; 10:539-47. [PMID: 25741989 DOI: 10.1038/nprot.2015.036] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Animal models of focal cerebral ischemia are well accepted for investigating the pathogenesis and potential treatment strategies for human stroke. Occlusion of the middle cerebral artery (MCA) with an endovascular filament is a widely used model to induce focal cerebral ischemia. However, this model is not amenable to thrombolytic therapies. As thrombolysis with recombinant tissue plasminogen activator (rtPA) is a standard of care within 4.5 h of human stroke onset, suitable animal models that mimic cellular and molecular mechanisms of thrombosis and thrombolysis of stroke are required. By occluding the MCA with a fibrin-rich allogeneic clot, we previously developed an embolic model of MCA occlusion in the rat, which recapitulates the key components of thrombotic development and of thrombolytic therapy of rtPA observed from human ischemic stroke. Here we describe in detail the surgical procedures of our model, including preparing emboli from rat donors. These procedures can be typically completed within ∼30 min, and they are highly adaptable to other strains of rats, as well as mice, in both sexes. Thus, this model provides a powerful tool for translational stroke research.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Rui Lan Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Quan Jiang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Guangliang Ding
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Michael Chopp
- 1] Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA. [2] Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| |
Collapse
|
32
|
Innovative thrombolytic strategy using a heterodimer diabody against TAFI and PAI-1 in mouse models of thrombosis and stroke. Blood 2014; 125:1325-32. [PMID: 25540192 DOI: 10.1182/blood-2014-07-588319] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Circulating thrombin-activatable fibrinolysis inhibitor (TAFI) and plasminogen activator inhibitor-1 (PAI-1) are causal factors for thrombolytic failure. Therefore, we evaluated an antibody-engineered bispecific inhibitor against TAFI and PAI-1 (heterodimer diabody, Db-TCK26D6x33H1F7) in several mouse models of thrombosis and stroke. Prophylactic administration of the diabody (0.8 mg/kg) in a thromboplastin-induced model of thromboembolism led to decreased lung fibrin deposition. In a model of cerebral ischemia and reperfusion, diabody administration (0.8 mg/kg, 1 hour postocclusion) led to a mitigated cerebral injury with a 2.3-fold reduced lesion and improved functional outcomes. In a mouse model of thrombin-induced middle cerebral artery occlusion, the efficacy of the diabody was compared to the standard thrombolytic treatment with recombinant tissue-type plasminogen activator (tPA). Early administration of diabody (0.8 mg/kg) caused a twofold decrease in brain lesion size, whereas that of tPA (10 mg/kg) had a much smaller effect. Delayed administration of diabody or tPA had no effect on lesion size, whereas the combined administration of diabody with tPA caused a 1.7-fold decrease in lesion size. In contrast to tPA, the diabody did not increase accumulative bleeding. In conclusion, administration of a bispecific inhibitor against TAFI and PAI-1 results in a prominent profibrinolytic effect in mice without increased bleeding.
Collapse
|
33
|
Reed GL, Houng AK, Wang D. Microvascular thrombosis, fibrinolysis, ischemic injury, and death after cerebral thromboembolism are affected by levels of circulating α2-antiplasmin. Arterioscler Thromb Vasc Biol 2014; 34:2586-93. [PMID: 25256235 PMCID: PMC4239309 DOI: 10.1161/atvbaha.114.304530] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 09/09/2014] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Ischemic stroke is primarily attributable to thrombotic vascular occlusion. Elevated α2-antiplasmin (a2AP) levels correlate with increased stroke risk, but whether a2AP contributes to the pathogenesis of stroke is unknown. We examined how a2AP affects thrombosis, ischemic brain injury, and survival after experimental cerebral thromboembolism. APPROACH AND RESULTS We evaluated the effects of a2AP on stroke outcomes in mice with increased, normal, or no circulating a2AP, as well as in mice given an a2AP-inactivating antibody. Higher a2AP levels were correlated with greater ischemic brain injury (rs=0.88, P<0.001), brain swelling (rs=0.82, P<0.001), and reduced middle cerebral artery thrombus dissolution (rs=-0.93, P<0.001). In contrast, a2AP deficiency enhanced thrombus dissolution, increased cerebral blood flow, reduced brain infarction, and decreased brain swelling. By comparison to tissue plasminogen activator (TPA), a2AP inactivation hours after thromboembolism still reduced brain infarction (P<0.001) and hemorrhage (P<0.05). Microvascular thrombosis, a process that enhances brain ischemia, was markedly reduced in a2AP-deficient or a2AP-inactivated mice compared with TPA-treated mice or mice with increased a2AP levels (all P<0.001). Matrix metalloproteinase-9 expression, which contributes to acute brain injury, was profoundly decreased in a2AP-deficient or a2AP-inactivated mice versus TPA-treated mice or mice with increased a2AP levels (all P<0.001). a2AP inactivation markedly reduced stroke mortality versus TPA (P<0.0001). CONCLUSIONS a2AP has profound, dose-related effects on ischemic brain injury, swelling, hemorrhage, and survival after cerebral thromboembolism. By comparison to TPA, the protective effects of a2AP deficiency or inactivation seem to be mediated through reductions in microvascular thrombosis and matrix metalloproteinase-9 expression.
Collapse
Affiliation(s)
- Guy L Reed
- From the Department of Medicine, University of Tennessee Health Sciences Center, Memphis.
| | - Aiilyan K Houng
- From the Department of Medicine, University of Tennessee Health Sciences Center, Memphis
| | - Dong Wang
- From the Department of Medicine, University of Tennessee Health Sciences Center, Memphis
| |
Collapse
|
34
|
Ding G, Zhang Z, Chopp M, Li L, Zhang L, Li Q, Wei M, Jiang Q. MRI evaluation of BBB disruption after adjuvant AcSDKP treatment of stroke with tPA in rat. Neuroscience 2014; 271:1-8. [PMID: 24769225 DOI: 10.1016/j.neuroscience.2014.04.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 04/15/2014] [Accepted: 04/17/2014] [Indexed: 12/24/2022]
Abstract
The primary limitation of thrombolytic treatment of ischemic stroke with tissue plasminogen activator (tPA) is the hemorrhagic risk. We tested AcSDKP (N-acetyl-seryl-aspartyl-lysyl-proline), as an auxiliary therapeutic agent, to reduce blood-brain barrier (BBB) disruption in a combination tPA thrombolytic treatment of stroke. Wistar rats subjected to embolic stroke were randomly assigned to either the tPA monotherapy group (n=9) or combination of tPA and AcSDKP treatment group (n=9) initiated at 4 h after ischemia. Magnetic resonance imaging (MRI) measurements were performed before and after the treatments. Immunohistochemical staining and measurements were performed to confirm MRI findings. Longitudinal MRI permeability measurements with gadolinium-diethylenetriamine penta-acetic acid (Gd-DTPA) demonstrated that combination treatment of acute embolic stroke with AcSDKP and tPA significantly reduced BBB leakage, compared to tPA monotherapy, at 3 and 6 days (18.3±9.8 mm3 vs. 65.0±21.0 mm3, p<0.001) after the onset of stroke, although BBB leakage was comparable between the two groups prior to the treatments (6.8±4.4 mm3 vs. 4.3±3.3 mm3, p>0.18). The substantial reduction of BBB leakage observed in the combination treatment group was closely associated with reduced ischemic lesions measured by T2 maps (113.6±24.9 mm3 vs. 188.1±60.8 mm3, p<0.04 at 6 days). Histopathological analysis of the same population of rats showed that the combination treatment significantly reduced parenchymal fibrin deposition (0.063±0.059 mm2 vs. 0.172±0.103 mm2, p<0.03) and infarct volume (146.7±35.9 mm3 vs. 199.3±60.4 mm3, p<0.05) compared to the tPA monotherapy at 6days after stroke. MRI provides biological insight into the therapeutic benefit of combination treatment of stroke with tPA and AcSDKP 4h after onset, and demonstrates significantly improved cerebrovascular integrity with neuroprotective effects compared with tPA monotherapy.
Collapse
Affiliation(s)
- G Ding
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA
| | - Z Zhang
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA
| | - M Chopp
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA; Department of Physics, Oakland University, Rochester, MI 48309, USA
| | - L Li
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA
| | - L Zhang
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA
| | - Q Li
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA
| | - M Wei
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA
| | - Q Jiang
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA.
| |
Collapse
|
35
|
Song Y, Huang Z, Xu J, Ren D, Wang Y, Zheng X, Shen Y, Wang L, Gao H, Hou J, Pang Z, Qian J, Ge J. Multimodal SPION-CREKA peptide based agents for molecular imaging of microthrombus in a rat myocardial ischemia-reperfusion model. Biomaterials 2014; 35:2961-70. [PMID: 24393265 DOI: 10.1016/j.biomaterials.2013.12.038] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 12/13/2013] [Indexed: 02/09/2023]
Abstract
Microthrombosis plays a key role in many cardiovascular diseases. Although it is not difficult to localize thrombus within large or middle-sized vessels, the noninvasive diagnostic regimen for the detection of microthrombus remains scarce. Here we developed a nanoagent by conjucting superparamagnetic iron-oxide nanoparticle with fluorophore and a targeting element, CREKA, a peptide with special affinity for fibrin. In a rat model of myocardial ischemia-reperfusion (MI/R), the multimodal nanoagents were readily and selectively accumulated within microthrombosis, which was detectable by both magnetic resonance and optical imaging modalities. The fibrin-targeted nanoagent could be expected to have utility not only in molecular imaging of fibrin, understanding the mechanisms of microcirculation disorders, but also in targeted therapy with fibrinolytic agents.
Collapse
Affiliation(s)
- Yanan Song
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai 200032, China
| | - Zheyong Huang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai 200032, China
| | - Jianfeng Xu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai 200032, China
| | - Daoyuan Ren
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai 200032, China
| | - Yu Wang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai 201203, China
| | - Xinde Zheng
- Department of Radiology, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai 200032, China
| | - Yunli Shen
- Department of Cardiology, Shanghai East Hospital, Tongji University, 150 Jimo Road, Shanghai 200120, China
| | - Lili Wang
- Department of Radiology, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai 200032, China
| | - Hongxiang Gao
- Department of Laboratory, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai 200032, China
| | - Jiayun Hou
- Biomedical Research Center, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai 200032, China
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai 201203, China.
| | - Juying Qian
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai 200032, China.
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai 200032, China; Institute of Biomedical Science, Fudan University, 180 Feng Lin Road, Shanghai 200032, China.
| |
Collapse
|
36
|
Dalkara T, Arsava EM. Can restoring incomplete microcirculatory reperfusion improve stroke outcome after thrombolysis? J Cereb Blood Flow Metab 2012; 32:2091-9. [PMID: 23047270 PMCID: PMC3519416 DOI: 10.1038/jcbfm.2012.139] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 08/31/2012] [Accepted: 09/03/2012] [Indexed: 12/27/2022]
Abstract
Substantial experimental data and recent clinical evidence suggesting that tissue reperfusion is a better predictor of outcome after thrombolysis than recanalization necessitate that patency of microcirculation after recanalization should be reevaluated. If indeed microcirculatory blood flow cannot be sufficiently reinstituted despite complete recanalization as commonly observed in coronary circulation, it may be one of the factors contributing to low efficacy of thrombolysis in stroke. Although microvascular no-reflow is considered an irreversible process that prevents tissue recovery from injury, emerging evidence suggests that it might be reversed with pharmacological agents administered early during recanalization. Therefore, therapeutic approaches aiming at reducing microvascular obstructions may improve success rate of recanalization therapies. Importantly, promoting oxygen delivery to the tissue, where entrapped erythrocytes cannot circulate in capillaries, with ongoing serum flow may improve survival of the underreperfused tissue. Altogether, these developments bring about the exciting possibility that benefit of reperfusion therapies can be further improved by restoring microcirculatory function because survival in the penumbra critically depends on adequate blood supply. Here, we review the available evidence suggesting presence of an 'incomplete microcirculatory reperfusion' (IMR) after focal cerebral ischemia and discuss potential means that may help investigate IMR in stroke patients after recanalization therapies despite technical limitations.
Collapse
Affiliation(s)
- Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.
| | | |
Collapse
|
37
|
Gursoy-Ozdemir Y, Yemisci M, Dalkara T. Microvascular protection is essential for successful neuroprotection in stroke. J Neurochem 2012; 123 Suppl 2:2-11. [DOI: 10.1111/j.1471-4159.2012.07938.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Yasemin Gursoy-Ozdemir
- Department of Neurology, Institute of Neurological Sciences and Psychiatry; Hacettepe University; Ankara; Turkey
| | - Muge Yemisci
- Department of Neurology, Institute of Neurological Sciences and Psychiatry; Hacettepe University; Ankara; Turkey
| | - Turgay Dalkara
- Department of Neurology, Institute of Neurological Sciences and Psychiatry; Hacettepe University; Ankara; Turkey
| |
Collapse
|
38
|
Baeten KM, Akassoglou K. Extracellular matrix and matrix receptors in blood-brain barrier formation and stroke. Dev Neurobiol 2012; 71:1018-39. [PMID: 21780303 DOI: 10.1002/dneu.20954] [Citation(s) in RCA: 276] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The blood-brain barrier (BBB) is formed primarily to protect the brain microenvironment from the influx of plasma components, which may disturb neuronal functions. The BBB is a functional unit that consists mainly of specialized endothelial cells (ECs) lining the cerebral blood vessels, astrocytes, and pericytes. The BBB is a dynamic structure that is altered in neurologic diseases, such as stroke. ECs and astrocytes secrete extracellular matrix (ECM) proteins to generate and maintain the basement membranes (BMs). ECM receptors, such as integrins and dystroglycan, are also expressed at the brain microvasculature and mediate the connections between cellular and matrix components in physiology and disease. ECM proteins and receptors elicit diverse molecular signals that allow cell adaptation to environmental changes and regulate growth and cell motility. The composition of the ECM is altered upon BBB disruption and directly affects the progression of neurologic disease. The purpose of this review is to discuss the dynamic changes of ECM composition and integrin receptor expression that control BBB functions in physiology and pathology.
Collapse
Affiliation(s)
- Kim M Baeten
- Gladstone Institute of Neurological Disease, University of California, San Francisco, California 94158, USA
| | | |
Collapse
|
39
|
Zhang L, Zhang ZG, Chopp M. The neurovascular unit and combination treatment strategies for stroke. Trends Pharmacol Sci 2012; 33:415-22. [PMID: 22595494 DOI: 10.1016/j.tips.2012.04.006] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 04/06/2012] [Accepted: 04/18/2012] [Indexed: 01/01/2023]
Abstract
Tissue plasminogen activator (tPA) administered within 4.5h of symptom onset restores cerebral blood flow (CBF) and promotes neurological recovery of stroke patients. However, the narrow therapeutic time window and the risk of intracerebral hemorrhage after tPA treatment pose major hurdles to its clinical usage. In light of the failures of neuroprotective therapies in clinical trials, emerging concepts suggest that neuroprotection alone without restoration of tissue perfusion and vascular integrity may not be adequate for treatment of acute stroke. Here we review evidence of the use of adjuvant pharmacological agents to extend the therapeutic window for tPA via targeting the neurovascular unit and the underlying mechanisms of the combination therapy in experimental stroke.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA.
| | | | | |
Collapse
|
40
|
Muthaian R, Minhas G, Anand A. Pathophysiology of stroke and stroke-induced retinal ischemia: emerging role of stem cells. J Cell Physiol 2012; 227:1269-79. [PMID: 21989824 DOI: 10.1002/jcp.23048] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The current review focuses on pathophysiology, animal models and molecular analysis of stroke and retinal ischemia, and the role of stem cells in recovery of these disease conditions. Research findings associated with ischemic stroke and retinal ischemia have been discussed, and efforts towards prevention and limiting the recurrence of ischemic diseases, as well as emerging treatment possibilities with endothelial progenitor cells (EPCs) in ischemic diseases, are presented. Although most neurological diseases are still not completely understood and reliable treatment is lacking, animal models provide a major step in validating novel therapies. Stem cell approaches constitute an emerging form of cell-based therapy to treat ischemic diseases since it is an attractive source for regenerative therapy in the ischemic diseases. In this review, we highlight the advantages and limitations of this approach with a focus on key observations from preclinical animal studies and clinical trials. Further research, especially on treatment with EPCs is warranted.
Collapse
Affiliation(s)
- Rupadevi Muthaian
- Department of Neurology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | | |
Collapse
|
41
|
Zhang Y, Fan S, Yao Y, Ding J, Wang Y, Zhao Z, Liao L, Li P, Zang F, Teng GJ. In vivo near-infrared imaging of fibrin deposition in thromboembolic stroke in mice. PLoS One 2012; 7:e30262. [PMID: 22272319 PMCID: PMC3260250 DOI: 10.1371/journal.pone.0030262] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 12/12/2011] [Indexed: 12/11/2022] Open
Abstract
Objectives Thrombus and secondary thrombosis plays a key role in stroke. Recent molecular imaging provides in vivo imaging of activated factor XIII (FXIIIa), an important mediator of thrombosis or fibrinolytic resistance. The present study was to investigate the fibrin deposition in a thromboembolic stroke mice model by FXIIIa–targeted near-infrared fluorescence (NIRF) imaging. Materials and Methods The experimental protocol was approved by our institutional animal use committee. Seventy-six C57B/6J mice were subjected to thromboembolic middle cerebral artery occlusion or sham operation. Mice were either intravenously injected with the FXIIIa-targeted probe or control probe. In vivo and ex vivo NIRF imaging were performed thereafter. Probe distribution was assessed with fluorescence microscopy by spectral imaging and quantification system. MR scans were performed to measure lesion volumes in vivo, which were correlated with histology after animal euthanasia. Results In vivo significant higher fluorescence intensity over the ischemia-affected hemisphere, compared to the contralateral side, was detected in mice that received FXIIIa-targeted probe, but not in the controlled mice. Significantly NIRF signals showed time-dependent processes from 8 to 96 hours after injection of FXIIIa-targeted probes. Ex vivo NIRF image showed an intense fluorescence within the ischemic territory only in mice injected with FXIIIa-targeted probe. The fluorescence microscopy demonstrated distribution of FXIIIa-targeted probe in the ischemic region and nearby micro-vessels, and FXIIIa-targeted probe signals showed good overlap with immune-fluorescent fibrin staining images. There was a significant correlation between total targeted signal from in vivo or ex vivo NIRF images and lesion volume. Conclusion Non-invasive detection of fibrin deposition in ischemic mouse brain using NIRF imaging is feasible and this technique may provide an in vivo experimental tool in studying the role of fibrin in stroke.
Collapse
Affiliation(s)
- Yi Zhang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Shufeng Fan
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Yuyu Yao
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Jie Ding
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Yu Wang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Zhen Zhao
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Lei Liao
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Peicheng Li
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Fengchao Zang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Gao-Jun Teng
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
- * E-mail:
| |
Collapse
|
42
|
Vascular Pathology as a Potential Therapeutic Target in SCI. Transl Stroke Res 2011; 2:556-74. [PMID: 24323683 DOI: 10.1007/s12975-011-0128-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 10/21/2011] [Accepted: 10/25/2011] [Indexed: 10/15/2022]
Abstract
Acute traumatic spinal cord injury (SCI) is characterized by a progressive secondary degeneration which exacerbates the loss of penumbral tissue and neurological function. Here, we first provide an overview of the known pathophysiological mechanisms involving injured microvasculature and molecular regulators that contribute to the loss and dysfunction of existing and new blood vessels. We also highlight the differences between traumatic and ischemic injuries which may yield clues as to the more devastating nature of traumatic injuries, possibly involving toxicity associated with hemorrhage. We also discuss known species differences with implications for choosing models, their relevance and utility to translate new treatments towards the clinic. Throughout this review, we highlight the potential opportunities and proof-of-concept experimental studies for targeting therapies to endothelial cell-specific responses. Lastly, we comment on the need for vascular mechanisms to be included in drug development and non-invasive diagnostics such as serum and cerebrospinal fluid biomarkers and imaging of spinal cord pathology.
Collapse
|
43
|
Bosomtwi A, Chopp M, Zhang L, Zhang ZG, Lu M, Jiang Q. Mean microvessel segment length and radius after embolic stroke: Comparison of magnetic resonance imaging (MRI) and laser scanning confocal microscopy (LSCM). Brain Res 2011; 1381:217-27. [PMID: 21237138 DOI: 10.1016/j.brainres.2011.01.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Revised: 01/05/2011] [Accepted: 01/06/2011] [Indexed: 10/18/2022]
Abstract
We offer a new means of noninvasively assessing mean microvessel segment length and radius after ischemic stroke. This method involves measuring changes in T₂ and T₂⁎ after injecting an intravascular superparamagnetic iron oxide contrast agent and result was verified using laser scanning confocal microscopy (LSCM) of both normal brain tissue and the ischemic recovery region. Embolic stroke was induced in 8 male Wistar rats and magnetic resonance imaging (MRI) performed 1 day and 6 weeks later. On MRI taken at 6 weeks, MRI of the recovery region revealed a significant increase in mean vessel size index (VSI) (5.75 ± 0.54 vs 4.81 ± 0.3 9μm; p < 0.001) and decrease in mean segment length (MSL) (16.61 ± 2.33 vs 26.52 ± 3.20 μm; p < 0.001) compared to the normal contralateral hemisphere, comparable with published values. There was also a significant correlation between MSL and VSI measured by MRI vs LSCM in the recovery region and normal contralateral hemisphere (p < 0.001). Our data suggest that a) morphological changes in the microvasculature can be measured noninvasively using MRI, and b) both MRI and LSCM give comparable information about both of these important parameters.
Collapse
Affiliation(s)
- Asamoah Bosomtwi
- Imaging Center, Yerkes National Primate Center, Emory University, Atlanta, GA, USA
| | | | | | | | | | | |
Collapse
|
44
|
Biphasic regulation of tissue plasminogen activator activity in ischemic rat brain and in cultured neural cells: essential role of astrocyte-derived plasminogen activator inhibitor-1. Neurochem Int 2010; 58:423-33. [PMID: 21193004 DOI: 10.1016/j.neuint.2010.12.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 12/21/2010] [Accepted: 12/21/2010] [Indexed: 11/20/2022]
Abstract
In brain, the serine protease tissue plasminogen activator (tPA) and its endogenous inhibitor plasminogen activator inhibitor-1 (PAI-1) have been implicated in the regulation of various neurophysiological and pathological responses. In this study, we investigated the differential role of neurons and astrocytes in the regulation of tPA/PAI-1 activity in ischemic brain. The activity of tPA peaked transiently and then decreased in cortex and striatum along with delayed induction of PAI-1 in the inflammatory stage after MCAO/reperfusion injury. In cultured primary cells, glutamate stimulation increased tPA activity in neurons but not in other cells such as microglia and astrocytes. With LPS stimulation, a model of neuroinflammatory insults, robust PAI-1 induction was observed in astrocytes but not in neurons and microglia. The upregulation of PAI-1 by LPS in astrocytes was also verified by RT-PCR analysis as well as PAI-1 promoter reporter assay. Lastly, we checked the effects of hypoxia on tPA/PAI-1 activity. Hypoxia increased tPA release from neurons without effects on microglia, while the activity of tPA in astrocyte was decreased consistent with increased PAI-1 activity in astrocyte. Taken together, the results from the present study suggest that neurons are the major source of tPA and that the glutamate-induced stimulated release is mainly governed by neurons in the acute phase. In contrast, the massive up-regulation of PAI-1 in astrocytes during subchronic and chronic inflammatory conditions, leads to decreased tPA activity in the later stages of MCAO. Differential regulation of tPA and PAI-1 in neurons, astrocytes and microglia suggest more attention is required to understand the role of local tPA activity in the vicinity of individual cell types.
Collapse
|
45
|
Jia L, Chopp M, Zhang L, Lu M, Zhang Z. Erythropoietin in combination of tissue plasminogen activator exacerbates brain hemorrhage when treatment is initiated 6 hours after stroke. Stroke 2010; 41:2071-6. [PMID: 20671252 DOI: 10.1161/strokeaha.110.586198] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND PURPOSE Erythropoietin (EPO), a hematopoietic cytokine, exerts neuroprotective effects in experimental stroke. In the present study, we investigated the effect of recombinant human EPO (rhEPO) in combination with tissue plasminogen activator (tPA) on embolic stroke. METHODS Rats subjected to embolic middle cerebral artery occlusion (MCAO) were treated with rhEPO (5000 U/kg) in combination with tPA (10 mg/kg) at 2 or 6 hours after MCAO. Control groups consisted of ischemic rats treated with rhEPO (5000 U/kg) alone, tPA (10 mg/kg) alone, or saline at 2 or 6 hours after MCAO. RESULTS The combination therapy of rhEPO and tPA initiated 6 hours after MCAO did not reduce the ischemic lesion volume and significantly (P<0.05) increased the incidence of brain hemorrhage measured by frequency of gross hemorrhage and a quantitative spectrophotometric hemoglobin assay compared with rats treated with rhEPO alone and tPA alone. However, when the combination therapy was initiated 2 hours after MCAO, the treatment significantly (P<0.05) reduced the lesion volume and did not substantially increase the incidence of hemorrhagic transformation compared with saline-treated rats. Immunostaining analysis revealed that the combination therapy of rhEPO and tPA at 6 hours significantly (P<0.05) increased matrix metalloproteinase-9, NF-kappaB, and interleukin-1 receptor-associated kinase-1 immunoreactive cerebral vessels compared with rats treated with rhEPO alone and saline. CONCLUSIONS EPO exacerbates tPA-induced brain hemorrhage without reduction of ischemic brain damage when administered 6 hours after stroke in a rat model of embolic MCAO and that matrix metalloproteinase-9, NF-kappaB, and interleukin-1 receptor-associated kinase-1 upregulated by the delayed combination therapy may contribute to augmentation of brain hemorrhage.
Collapse
Affiliation(s)
- Longfei Jia
- Department of Neurology, Henry Ford Hospital, Detroit, Mich 48202, USA
| | | | | | | | | |
Collapse
|
46
|
Zhang L, Zhang ZG, Buller B, Jiang J, Jiang Y, Zhao D, Liu X, Morris D, Chopp M. Combination treatment with VELCADE and low-dose tissue plasminogen activator provides potent neuroprotection in aged rats after embolic focal ischemia. Stroke 2010; 41:1001-7. [PMID: 20203318 DOI: 10.1161/strokeaha.109.577288] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Treatment with a selective proteasome inhibitor, VELCADE, in combination with tissue plasminogen activator (tPA) extended the therapeutic window to 6 hours in young rats after stroke. However, stroke is a major cause of death and disability in the elderly. The present study investigated the effect of VELCADE in combination with a low-dose tPA on aged rats after embolic stroke. METHODS Male Wistar rats at the age of 18 to 20 months were treated with VELCADE (0.2 mg/kg) alone, a low-dose tPA (5 mg/kg) alone, combination of VELCADE and tPA, or saline 2 hours after embolic middle cerebral artery occlusion. To test the contribution of endothelial nitric oxide synthase to VELCADE-mediated neuroprotection, endothelial nitric oxide synthase knockout and wild-type mice were treated with VELCADE (0.5 mg/kg) 2 hours after embolic stroke. RESULTS Treatment with VELCADE significantly reduced infarct volume, whereas tPA alone did not reduce infarct volume and aggravated blood-brain barrier disruption in aged rats compared with saline-treated rats. However, the combination treatment significantly enhanced the reduction of infarct volume, which was associated with an increase in endothelial nitric oxide synthase activity compared with saline-treated rats. Additionally, the combination treatment promoted thrombolysis and did not increase the incidence of hemorrhage transformation. VELCADE significantly reduced lesion volume in wild-type mice but failed to significantly reduce lesion volume in endothelial nitric oxide synthase knockout mice. CONCLUSIONS Treatment with VELCADE exerts a neuroprotective effect in aged rats after stroke. The combination of VELCADE with the low-dose tPA further amplifies the neuroprotective effect. Endothelial nitric oxide synthase at least partly contributes to VELCADE-mediated neuroprotection after stroke.
Collapse
Affiliation(s)
- Li Zhang
- Henry Ford Health System, Department of Neurology, 2799 West Grand Boulevard, Detroit, MI 48202, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Kloek JJ, Levi M, Heger M, van der Loos CM, Gouma DJ, van Gulik TM. Cholestasis enhances liver ischemia/reperfusion-induced coagulation activation in rats. Hepatol Res 2010; 40:204-15. [PMID: 19737317 DOI: 10.1111/j.1872-034x.2009.00579.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AIM Cholestasis is associated with increased morbidity and mortality in patients undergoing major liver surgery. An additional risk is induced when vascular inflow occlusion is applied giving rise to liver ischemia/reperfusion (I/R) injury. The role of the coagulation system in this type of injury is elusive. The aim of the current study was to assess activation of coagulation following hepatic I/R injury in cholestatic rats. METHODS Male Wistar rats were randomized into two groups and subjected to bile duct ligation (BDL) or sham laparotomy. After 7 days, both groups underwent 30 min partial liver ischemia. Animals were sacrificed before ischemia or after 6 h, 24 h, and 48 h reperfusion. RESULTS Plasma AST and ALT levels were higher after I/R in cholestatic rats (P < 0.05). Hepatic necrosis, liver wet/dry ratio and neutrophil influx were increased in the BDL group up to 48 h reperfusion (P < 0.05). Liver synthetic function was decreased in the BDL group as reflected by prolonged prothrombin time after 6 h and 24 h reperfusion (P < 0.05). I/R in cholestatic rats resulted in a 12-fold vs. 7-fold (P < 0.01) increase in markers for thrombin generation and a 6-fold vs. 2-fold (P < 0.01) increase in fibrin degradation products (BDL vs. control, respectively). In addition, the cholestatic rats exhibited significantly decreased levels of antithrombin (AT) III and increased levels of the fibrinolytic inhibitor plasminogen activator inhibitor (PAI-1) during reperfusion. CONCLUSIONS Cholestasis significantly enhances I/R-induced hepatic damage and inflammation that concurs with an increased activation of coagulation and fibrinolysis.
Collapse
Affiliation(s)
- Jaap J Kloek
- Departments of Surgery, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | | | |
Collapse
|
48
|
Sezer M, Çimen A, Aslanger E, Elitok A, Umman B, Buğra Z, Yormaz E, Türkmen C, Adalet I, Nişanci Y, Umman S. Effect of Intracoronary Streptokinase Administered Immediately After Primary Percutaneous Coronary Intervention on Long-Term Left Ventricular Infarct Size, Volumes, and Function. J Am Coll Cardiol 2009; 54:1065-71. [DOI: 10.1016/j.jacc.2009.04.083] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Revised: 04/20/2009] [Accepted: 04/20/2009] [Indexed: 01/06/2023]
|
49
|
Zhang X, Zheng X, Jiang F, Zhang ZG, Katakowski M, Chopp M. Dual-color fluorescence imaging in a nude mouse orthotopic glioma model. J Neurosci Methods 2009; 181:178-85. [PMID: 19447136 DOI: 10.1016/j.jneumeth.2009.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 04/30/2009] [Accepted: 05/03/2009] [Indexed: 11/30/2022]
Abstract
We sought to establish a new orthotopic glioma model of nude mice by transfer of DsRed2, a red fluorescent protein gene, to malignant glioma cells and to perfuse the tissue with fluorescein isothiocyanate (FITC) dextran in vivo, which would permit the concurrent detection of brain tumor invasion and angiogenesis in vivo by florescence microscopy. 9L or U87 malignant glioma cells with DsRed2 expression were intracerebrally injected into the nude mice. FITC-dextran was administered intravenously to the mice bearing DsRed2-9L or DsRed2-U87 cells immediately before they were sacrificed at 10 days or 15 days after the implantation, respectively. Coronal vibratome sections were examined using 2D and 3D fluorescence microscopy and the results were compared with those examined by routine hematoxylin and eosin (H & E) staining. Angiogenesis induced by glioma was confirmed by two-dimensional and three-dimensional imaging analysis. DsRed2 fluorescence clearly demarcated the primary tumor margins and readily allowed for the visualization of local invasion at the single-cell level in the brain adjacent to tumor. We found that a few tumor cells migrated from the tumor mass along the aberrant microvasculature, but did not extend out of the angiogenic areas. However, locally invasive foci were very difficult to detect by H & E staining. We demonstrated, for the first time, that abnormal vascular structure and glioma cells can be visualized concurrently by fluorescence microscopy. This method is superior to H & E staining for the detection and study of physiologically relevant patterns of brain tumor invasion and angiogenesis in vivo.
Collapse
Affiliation(s)
- Xuepeng Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA
| | | | | | | | | | | |
Collapse
|
50
|
Transcriptomic screening of microvascular endothelial cells implicates novel molecular regulators of vascular dysfunction after spinal cord injury. J Cereb Blood Flow Metab 2008; 28:1771-85. [PMID: 18612314 PMCID: PMC2756605 DOI: 10.1038/jcbfm.2008.76] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Microvascular dysfunction is a critical pathology that underlies the evolution of secondary injury mechanisms after traumatic spinal cord injury (SCI). However, little is known of the molecular regulation of endothelial cell (EC) plasticity observed acutely after injury. One reason for this is the relative lack of methods to quickly and efficiently obtain highly enriched spinal microvascular ECs for high-throughput molecular and biochemical analyses. Adult C57Bl/6 mice received an intravenous injection of fluorescein isothiocyanate (FITC)-conjugated Lycopersicon esculentum lectin, and FITC-lectin-bound spinal microvessels were greatly enriched by fluorescence-activated cell sorter (FACS) purification. This technique allows for rapid (<1.5 h postmortem) isolation of spinal cord microvascular ECs (smvECs). The results from cell counting, reverse-transcription polymerase chain reaction (RT-PCR), and western blot analyses show a high degree of EC enrichment at mRNA and protein levels. Furthermore, a focused EC biology microarray analysis identified multiple mRNAs dramatically increased in the EC compartment 24 h after SCI, which is a time point associated with the pathologic loss of spinal vasculature. These included thrombospondin-1, CCL5/RANTES, and urokinase plasminogen activator, suggesting they may represent targets for therapeutic intervention. Furthermore, these novel methodologic approaches will likely facilitate the discovery of molecular regulators of endothelial dysfunction in a variety of central nervous system (CNS) disorders including stroke and other neurodegenerative diseases having a vascular component.
Collapse
|