1
|
Li S, Hong L, Yang W, Liu X, Zhang Y, Ling Y, He Z, Wang X, Yue Y, Dong Q, Wang F, Cheng X. The benefit of favorable venous outflow profile is mediated through reduced microvascular dysfunction in acute ischemic stroke. Eur Stroke J 2024; 9:432-440. [PMID: 38291622 PMCID: PMC11318418 DOI: 10.1177/23969873231224573] [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: 11/01/2023] [Accepted: 12/18/2023] [Indexed: 02/01/2024] Open
Abstract
INTRODUCTIONS Venous outflow (VO) is emerging as a marker of microvascular integrity in acute ischemic stroke. Using hemorrhagic transformation (HT) and infarct growth as mediators, we tested whether a favorable VO profile benefited functional outcome by reducing consequences of microvascular dysfunction. PATIENTS AND METHODS Patients receiving thrombectomy in three comprehensive stroke centers due to acute anterior circulation occlusion were included. VO was assessed semi-quantitatively by the opacification of ipsilateral vein of Labbé, Trolard and superficial middle cerebral vein. HT was graded on follow-up CT. Infarct growth volume (IGV) was the difference of final infarct volume and baseline core volume. The association of VO and functional independence (90-day modified Rankin Scale ⩽ 2) was examined by logistic regression. Mediation analysis was performed among VO, HT or IGV, and functional outcome in patients with or without recanalization, respectively. RESULTS In 242 patients analyzed, VO was strongly correlated with functional independence and VO ⩾ 4 was defined favorable. In 175 patients recanalized, favorable VO was associated with a reduced risk of HT (OR = 0.82, 95% CI 0.71-0.95, p = 0.008), which accounted for 13.1% of the association between VO and favorable outcome. In 67 patients without recanalization, favorable VO was associated with decreased IGV (β = -0.07, 95% CI -0.11 to -0.02, p = 0.007). The association of favorable VO and functional independence was no longer significant (aOR = 4.84, 95% CI 0.87-38.87, p = 0.089) after including IGV in the model, suggesting a complete mediation. DISCUSSION AND CONCLUSION In patients with acute anterior large vessel occlusion, the clinical benefit of VO may be mediated through reduced microvascular dysfunction.
Collapse
Affiliation(s)
- Siyuan Li
- Department of Neurology, National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Lan Hong
- Department of Neurology, National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenhao Yang
- Department of Neurology, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinyu Liu
- Department of Neurology, National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Yiran Zhang
- Department of Neurology, National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Yifeng Ling
- Department of Neurology, National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhijiao He
- Department of Neurology, National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinru Wang
- Department of Neurology, National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Yunhua Yue
- Department of Neurology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qiang Dong
- Department of Neurology, National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| | - Feng Wang
- Department of Neurology, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Cheng
- Department of Neurology, National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Denorme F, Armstrong ND, Stoller ML, Portier I, Tugolukova EA, Tanner RM, Montenont E, Bhatlekar S, Cody M, Rustad JL, Ajanel A, Tolley ND, Murray DC, Boyle JL, Nieman MT, McKenzie SE, Yost CC, Lange LA, Cushman M, Irvin MR, Bray PF, Campbell RA. The predominant PAR4 variant in individuals of African ancestry worsens murine and human stroke outcomes. J Clin Invest 2023; 133:e169608. [PMID: 37471144 PMCID: PMC10503801 DOI: 10.1172/jci169608] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 07/18/2023] [Indexed: 07/22/2023] Open
Abstract
Protease-activated receptor 4 (PAR4) (gene F2RL3) harbors a functional dimorphism, rs773902 A/G (encoding Thr120/Ala120, respectively) and is associated with greater platelet aggregation. The A allele frequency is more common in Black individuals, and Black individuals have a higher incidence of ischemic stroke than White individuals. However, it is not known whether the A allele is responsible for worse stroke outcomes. To directly test the in vivo effect of this variant on stroke, we generated mice in which F2rl3 was replaced by F2RL3, thereby expressing human PAR4 (hPAR4) with either Thr120 or Ala120. Compared with hPAR4 Ala120 mice, hPAR4 Thr120 mice had worse stroke outcomes, mediated in part by enhanced platelet activation and platelet-neutrophil interactions. Analyses of 7,620 Black subjects with 487 incident ischemic strokes demonstrated the AA genotype was a risk for incident ischemic stroke and worse functional outcomes. In humanized mice, ticagrelor with or without aspirin improved stroke outcomes in hPAR4 Ala120 mice, but not in hPAR4 Thr120 mice. P selectin blockade improved stroke outcomes and reduced platelet-neutrophil interactions in hPAR4 Thr120 mice. Our results may explain some of the racial disparity in stroke and support the need for studies of nonstandard antiplatelet therapies for patients expressing PAR4 Thr120.
Collapse
Affiliation(s)
- Frederik Denorme
- Program in Molecular Medicine and
- Department of Neurology, Division of Vascular Neurology, University of Utah, Salt Lake City, Utah, USA
| | - Nicole D. Armstrong
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | | | | - Rikki M. Tanner
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | | | | | | | | | | | | | - Julie L. Boyle
- Bioinformatics Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Marvin T. Nieman
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Steven E. McKenzie
- Department of Medicine, The Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Christian Con Yost
- Program in Molecular Medicine and
- Department of Pediatrics, Division of Neonatology, University of Utah, Salt Lake City, Utah, USA
| | - Leslie A. Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mary Cushman
- Department of Medicine, Larner College of Medicine at the University of Vermont, Burlington, Vermont, USA
| | - Marguerite R. Irvin
- Department of Neurology, Division of Vascular Neurology, University of Utah, Salt Lake City, Utah, USA
| | - Paul F. Bray
- Program in Molecular Medicine and
- Department of Internal Medicine, Division of Hematology and Hematologic Malignancies, and
| | - Robert A. Campbell
- Program in Molecular Medicine and
- Department of Internal Medicine, Division of Hematology and Hematologic Malignancies, and
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
3
|
Chou ML, Babamale AO, Walker TL, Cognasse F, Blum D, Burnouf T. Blood-brain crosstalk: the roles of neutrophils, platelets, and neutrophil extracellular traps in neuropathologies. Trends Neurosci 2023; 46:764-779. [PMID: 37500363 DOI: 10.1016/j.tins.2023.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/17/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023]
Abstract
Systemic inflammation, neurovascular dysfunction, and coagulopathy often occur concurrently in neuropathologies. Neutrophils and platelets have crucial synergistic roles in thromboinflammation and are increasingly suspected as effector cells contributing to the pathogenesis of neuroinflammatory diseases. In this review, we summarize the roles of platelet-neutrophil interactions in triggering complex pathophysiological events affecting the brain that may lead to the disruption of brain barriers, infiltration of toxic factors into the parenchyma, and amplification of neuroinflammation through the formation of neutrophil extracellular traps (NETs). We highlight the clinical significance of thromboinflammation in neurological disorders and examine the contributions of damage-associated molecular patterns (DAMPs) derived from platelets and neutrophils. These DAMPs originate from both infectious and non-infectious risk factors and contribute to the activation of inflammasomes during brain disorders. Finally, we identify knowledge gaps in the molecular mechanisms underlying neurodegenerative disease pathogenesis and emphasize the potential of interventions targeting platelets and neutrophils to treat neuroinflammatory diseases.
Collapse
Affiliation(s)
- Ming-Li Chou
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City 23561, Taiwan; INSERM UMRS 938, Centre de Recherche Saint-Antoine, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, Paris 75012, France
| | - Abdulkareem Olarewaju Babamale
- Taiwan International Graduate Program in Molecular Medicine, Academia Sinica, Taipei 11266, Taiwan; Department of Zoology, Faculty of Life Sciences, University of Ilorin, Ilorin 240003, Nigeria
| | - Tara L Walker
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Fabrice Cognasse
- Etablissement Français du Sang Auvergne-Rhône-Alpes, 42023 Saint-Étienne, France; University Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 Sainbiose, 42023 Saint-Etienne, France
| | - David Blum
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, F-59000 Lille, France; Alzheimer & Tauopathies, LabEx DISTALZ, LiCEND, Lille F-59000, France; NeuroTMULille International Laboratory, University of Lille, F-59000 Lille, France
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City 23561, Taiwan; International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City 23561, Taiwan; NeuroTMULille International Laboratory, Taipei Medical University, Taipei 10031, Taiwan; Neuroscience Research Center, Taipei Medical University, Taipei 11031, Taiwan; Brain and Consciousness Research Centre, Taipei Medical University Shuang Ho Hospital, New Taipei City 23561, Taiwan.
| |
Collapse
|
4
|
Pluta R, Miziak B, Czuczwar SJ. Post-Ischemic Permeability of the Blood-Brain Barrier to Amyloid and Platelets as a Factor in the Maturation of Alzheimer's Disease-Type Brain Neurodegeneration. Int J Mol Sci 2023; 24:10739. [PMID: 37445917 DOI: 10.3390/ijms241310739] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/13/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
The aim of this review is to present evidence of the impact of ischemic changes in the blood-brain barrier on the maturation of post-ischemic brain neurodegeneration with features of Alzheimer's disease. Understanding the processes involved in the permeability of the post-ischemic blood-brain barrier during recirculation will provide clinically relevant knowledge regarding the neuropathological changes that ultimately lead to dementia of the Alzheimer's disease type. In this review, we try to distinguish between primary and secondary neuropathological processes during and after ischemia. Therefore, we can observe two hit stages that contribute to Alzheimer's disease development. The onset of ischemic brain pathology includes primary ischemic neuronal damage and death followed by the ischemic injury of the blood-brain barrier with serum leakage of amyloid into the brain tissue, leading to increased ischemic neuronal susceptibility to amyloid neurotoxicity, culminating in the formation of amyloid plaques and ending in full-blown dementia of the Alzheimer's disease type.
Collapse
Affiliation(s)
- Ryszard Pluta
- Department of Pathophysiology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Barbara Miziak
- Department of Pathophysiology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Stanisław J Czuczwar
- Department of Pathophysiology, Medical University of Lublin, 20-059 Lublin, Poland
| |
Collapse
|
5
|
Dev P, Ekhlak M, Dash D, Pathak A. Platelet function suggests cardioembolic aetiology in cryptogenic stroke. Sci Rep 2023; 13:7615. [PMID: 37165007 PMCID: PMC10172292 DOI: 10.1038/s41598-023-32143-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 03/23/2023] [Indexed: 05/12/2023] Open
Abstract
Platelet-monocyte (PMA) and platelet-neutrophil aggregations (PNA) play critical roles in the evolution of acute ischemic stroke (AIS). The present study investigates the mechanistic basis of platelet responsiveness in cryptogenic stroke compared with cardioembolic stroke. Platelet from 16 subjects, each from cryptogenic and cardioembolic stroke groups and 18 age-matched healthy controls were subjected to different investigations. Compared to healthy controls, platelet-monocyte and platelet-neutrophil interactions were significantly elevated in cryptogenic (2.7 and 2.1 times) and cardioembolic stroke (3.9 and 2.4 times). P-selectin expression on platelet surface was 1.89 and 2.59 times higher in cryptogenic and cardioembolic strokes, respectively, compared to healthy control. Cell population with [Ca2+i] in either stroke group was significantly outnumbered (by 83% and 72%, respectively, in cryptogenic and cardioembolic stroke) in comparison to healthy controls. Noteworthy, TEG experiment revealed that the cryptogenic stroke exhibited significant decline in Reaction Time (R) and amplitude of 20 mm (K) (by 32% and 33%, respectively) while thrombin burst (α-angle) was augmented by 12%, which reflected substantial boost in thrombus formation in cryptogenic stroke. Although TEG analysis reveals a state of hypercoagulability in patients with cryptogenic stroke. However, platelets from both stroke subtypes switch to a 'hyperactive' phenotype.
Collapse
Affiliation(s)
- Priya Dev
- Department of Neurology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Mohammad Ekhlak
- Department of Biochemistry, Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Debabrata Dash
- Department of Biochemistry, Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India.
| | - Abhishek Pathak
- Department of Neurology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India.
| |
Collapse
|
6
|
Yu M, Xiao G, Han L, Peng L, Wang H, He S, Lyu M, Zhu Y. QiShen YiQi and its components attenuate acute thromboembolic stroke and carotid thrombosis by inhibition of CD62P/PSGL-1-mediated platelet-leukocyte aggregate formation. Biomed Pharmacother 2023; 160:114323. [PMID: 36738500 DOI: 10.1016/j.biopha.2023.114323] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND QiShen YiQi (QSYQ) dropping pill, a component-based Chinese medicine consisting of benefiting Qi (YQ) and activating blood (HX) components, has been reported to exert a beneficial effect on cerebral ischemia-induced stroke. However, its efficacy and pharmacological mechanism on acute thromboembolic stroke is not clear. PURPOSE This study is to explore the preventative effect and pharmacological mechanism of QSYQ and its YQ/HX components on the formation of platelet-leukocyte aggregation (PLA) in acute thromboembolic stroke. STUDY DESIGN AND METHODS In vivo thromboembolic stroke model and FeCl3-induced carotid arterial occlusion models were used. Immunohistochemistry, Western blot, RT-qPCR, and flow cytometry experiments were performed to reveal the pharmacological mechanisms of QSYQ and its YQ/HX components. RESULTS In thromboembolic stroke rats, QSYQ significantly attenuated infarct area, improved neurological recovery, reduced PLA formation, and inhibited P-selection (CD62P)/ P-selectin glycoprotein ligand-1 (PSGL-1) expressions. The YQ component preferentially down-regulated PSGL-1 expression in leukocyte, while the HX component preferentially down-regulated CD62P expression in platelet. In carotid arterial thrombosis mice, QSYQ and its YQ/HX components inhibited thrombus formation, prolonged vessel occlusion time, reduced circulating leukocytes and P-selectin expression. PLA formation and platelet/leukocyte adhesion to endothelial cell were also inhibited by QSYQ and its YQ/HX components in vitro. CONCLUSION QSYQ and YQ/HX components attenuated thromboembolic stroke and carotid thrombosis by decreasing PLA formation via inhibiting CD62P/PSGL-1 expressions. This study shed a new light on the prevention of thromboembolic stroke.
Collapse
Affiliation(s)
- Mingxing Yu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China
| | - Guangxu Xiao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China
| | - Linhong Han
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China
| | - Li Peng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China
| | - Huanyi Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China
| | - Shuang He
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China
| | - Ming Lyu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China.
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Beihua South Road, JingHai District, Tianjin 301617, China.
| |
Collapse
|
7
|
Lubega J, Kim TO, Airewele G, Grimes A, Bulsara S, Peckham E, Wanless SR, Haq H, Elyanu P, Musoke P, Lumumba M, Kekitiinwa A, Matshaba M, Scheurer M, Despotovic J. Risk factors and prognostic significance of platelet count abnormalities in children with HIV infection on antiretroviral therapy. AIDS 2023; 37:413-421. [PMID: 36129118 PMCID: PMC9877116 DOI: 10.1097/qad.0000000000003387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVES To establish the incidence, risk factors and correlation with survival of thrombocytopenia and thrombocytosis (T/T) among children with HIV infection (CWH). DESIGN A retrospective nested case control study of patients 0-18 years in five Baylor International Pediatric AIDS Initiative (BIPAI) centers in sub-Sahara Africa, 2004-2014. METHODS Clinical and laboratory variables including complete blood counts (CBC) were extracted from the BIPAI electronic medical record system. Incident cases of T/T were identified and frequency-matched on follow-up time with controls with normal platelets. We calculated the prevalence and incidence density of T/T and used conditional logistic regression to evaluate their association with selected clinical variables. We constructed Kaplan-Meier curves and a Cox proportional hazards model to evaluate the impact of T/T on survival. RESULTS Two thousand, one hundred and nine children were sampled. The incidence density of thrombocytopenia was 1 per 57.9 (95% confidence interval [CI] 50.3-66.8) CWH-years. Thrombocytopenia was higher in children with WHO Stage III/IV, lower in children on zidovudine, and had no association with use of lamivudine or nevirapine, CD4 + suppression, age, and nutrition status. Thrombocytopenia was independently associated with 2.2-fold higher mortality (95% CI 1.62-3.08). The incidence density of thrombocytosis was 1 per 11.4 (95% CI 10.7-12.1) CWH-years. Thrombocytosis was associated with higher CD4 + cell count, younger age, and use of lamivudine or nevirapine, and did not impact survival. CONCLUSIONS Platelet count is a clinically valuable biomarker of HIV clinical progression and mortality. Laboratory studies are necessary to elucidate the mechanisms of T/T.
Collapse
Affiliation(s)
- Joseph Lubega
- Baylor College of Medicine, Houston, Texas, 77030, United States
| | - Taylor O Kim
- Baylor College of Medicine, Houston, Texas, 77030, United States
| | | | - Amanda Grimes
- Baylor College of Medicine, Houston, Texas, 77030, United States
| | - Shaun Bulsara
- Baylor College of Medicine, Houston, Texas, 77030, United States
| | - Erin Peckham
- Baylor College of Medicine, Houston, Texas, 77030, United States
| | | | - Heather Haq
- Baylor College of Medicine, Houston, Texas, 77030, United States
| | | | | | | | | | | | - Michael Scheurer
- Baylor College of Medicine, Houston, Texas, 77030, United States
| | - Jenny Despotovic
- Baylor College of Medicine, Houston, Texas, 77030, United States
| |
Collapse
|
8
|
Thapa K, Shivam K, Khan H, Kaur A, Dua K, Singh S, Singh TG. Emerging Targets for Modulation of Immune Response and Inflammation in Stroke. Neurochem Res 2023; 48:1663-1690. [PMID: 36763312 DOI: 10.1007/s11064-023-03875-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 02/11/2023]
Abstract
The inflammatory and immunological responses play a significant role after stroke. The innate immune activation stimulated by microglia during stroke results in the migration of macrophages and lymphocytes into the brain and are responsible for tissue damage. The immune response and inflammation following stroke have no defined targets, and the intricacies of the immunological and inflammatory processes are only partially understood. Innate immune cells enter the brain and meninges during the acute phase, which can cause ischemia damage. Activation of systemic immunity is caused by danger signals sent into the bloodstream by injured brain cells, which is followed by a significant immunodepression that encourages life-threatening infections. Neuropsychiatric sequelae, a major source of post-stroke morbidity, may be induced by an adaptive immune response that is initiated by antigen presentation during the chronic period and is directed against the brain. Thus, the current review discusses the role of immune response and inflammation in stroke pathogenesis, their role in the progression of injury during the stroke, and the emerging targets for the modulation of the mechanism of immune response and inflammation that may have possible therapeutic benefits against stroke.
Collapse
Affiliation(s)
- Komal Thapa
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.,School of Pharmacy, Chitkara University, Rajpura, Himachal Pradesh, 174103, India
| | - Kumar Shivam
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia.,Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, 2007, Australia
| | - Sachin Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar - Delhi G.T. Road, Phagwara, Punjab, 144411, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
9
|
Cheng Q, Wang M, Jin R, Li G. Blocking of PI3-kinase beta protects against cerebral ischemia/reperfusion injury by reducing platelet activation and downstream microvascular thrombosis in rats. Sci Rep 2023; 13:2030. [PMID: 36739310 PMCID: PMC9899241 DOI: 10.1038/s41598-023-29235-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/01/2023] [Indexed: 02/06/2023] Open
Abstract
Phosphoinositide 3-kinase beta (PI3Kβ) plays an important role in platelet activation and thrombosis, but its role in stroke pathology remains unknown. In this study, we investigated whether inhibition of PI3Kβ protects against cerebral ischemia/reperfusion (I/R) injury by preventing circulating platelet activation and downstream microvascular thrombosis. We used a rat intraluminal filament model of transient middle cerebral artery occlusion (tMCAO) because the rapid restoration of cerebral blood flow to the ischemic area in both tMCAO and endovascular thrombectomy provides clinical relevance for this model. The results showed that TGX221, a selective PI3Kβ inhibitor, treatment immediately before the onset of reperfusion dose-dependently reduced infarct volume and improved neurological function. The protective effects were associated with blocking platelet activation and thrombotic response, thereby reducing downstream microvascular thrombosis, and maintaining reperfusion efficiency. These results suggest that PI3Kβ might be a promising target for treating downstream microvascular thrombosis induced by cerebral I/R injury and offer a novel adjunctive treatment to improve reperfusion therapy for acute ischemic stroke.
Collapse
Affiliation(s)
- Qiong Cheng
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, 17033, USA
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, 226000, China
| | - Min Wang
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, 17033, USA
| | - Rong Jin
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, 17033, USA.
| | - Guohong Li
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, 17033, USA.
| |
Collapse
|
10
|
Shi SX, Vodovoz SJ, Xiu Y, Liu N, Jiang Y, Katakam PVG, Bix G, Dumont AS, Wang X. T-Lymphocyte Interactions with the Neurovascular Unit: Implications in Intracerebral Hemorrhage. Cells 2022; 11:cells11132011. [PMID: 35805099 PMCID: PMC9266108 DOI: 10.3390/cells11132011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/15/2022] [Accepted: 06/22/2022] [Indexed: 11/21/2022] Open
Abstract
In the pathophysiology of hemorrhagic stroke, the perturbation of the neurovascular unit (NVU), a functional group of the microvascular and brain intrinsic cellular components, is implicated in the progression of secondary injury and partially informs the ultimate patient outcome. Given the broad NVU functions in maintaining healthy brain homeostasis through its maintenance of nutrients and energy substrates, partitioning central and peripheral immune components, and expulsion of protein and metabolic waste, intracerebral hemorrhage (ICH)-induced dysregulation of the NVU directly contributes to numerous destructive processes in the post-stroke sequelae. In ICH, the damaged NVU precipitates the emergence and evolution of perihematomal edema as well as the breakdown of the blood–brain barrier structural coherence and function, which are critical facets during secondary ICH injury. As a gateway to the central nervous system, the NVU is among the first components to interact with the peripheral immune cells mobilized toward the injured brain. The release of signaling molecules and direct cellular contact between NVU cells and infiltrating leukocytes is a factor in the dysregulation of NVU functions and further adds to the acute neuroinflammatory environment of the ICH brain. Thus, the interactions between the NVU and immune cells, and their reverberating consequences, are an area of increasing research interest for understanding the complex pathophysiology of post-stroke injury. This review focuses on the interactions of T-lymphocytes, a major cell of the adaptive immunity with expansive effector function, with the NVU in the context of ICH. In cataloging the relevant clinical and experimental studies highlighting the synergistic actions of T-lymphocytes and the NVU in ICH injury, this review aimed to feature emergent knowledge of T cells in the hemorrhagic brain and their diverse involvement with the neurovascular unit in this disease.
Collapse
|
11
|
Targeting vascular inflammation through emerging methods and drug carriers. Adv Drug Deliv Rev 2022; 184:114180. [PMID: 35271986 PMCID: PMC9035126 DOI: 10.1016/j.addr.2022.114180] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 02/18/2022] [Accepted: 03/04/2022] [Indexed: 12/16/2022]
Abstract
Acute inflammation is a common dangerous component of pathogenesis of many prevalent conditions with high morbidity and mortality including sepsis, thrombosis, acute respiratory distress syndrome (ARDS), COVID-19, myocardial and cerebral ischemia-reperfusion, infection, and trauma. Inflammatory changes of the vasculature and blood mediate the course and outcome of the pathology in the tissue site of insult, remote organs and systemically. Endothelial cells lining the luminal surface of the vasculature play the key regulatory functions in the body, distinct under normal vs. pathological conditions. In theory, pharmacological interventions in the endothelial cells might enable therapeutic correction of the overzealous damaging pro-inflammatory and pro-thrombotic changes in the vasculature. However, current agents and drug delivery systems (DDS) have inadequate pharmacokinetics and lack the spatiotemporal precision of vascular delivery in the context of acute inflammation. To attain this level of precision, many groups design DDS targeted to specific endothelial surface determinants. These DDS are able to provide specificity for desired tissues, organs, cells, and sub-cellular compartments needed for a particular intervention. We provide a brief overview of endothelial determinants, design of DDS targeted to these molecules, their performance in experimental models with focus on animal studies and appraisal of emerging new approaches. Particular attention is paid to challenges and perspectives of targeted therapeutics and nanomedicine for advanced management of acute inflammation.
Collapse
|
12
|
Dong W, Liu X, Liu W, Wang C, Zhao S, Wen S, Gong T, Chen W, Chen Q, Ye W, Li Z, Wang Y. Dual antiplatelet therapy improves functional recovery and inhibits inflammation after cerebral ischemia/reperfusion injury. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:283. [PMID: 35433995 PMCID: PMC9011245 DOI: 10.21037/atm-22-735] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/04/2022] [Indexed: 01/21/2023]
Abstract
Background Dual antiplatelet therapy with aspirin and clopidogrel (ASA + CPG) during the first 21 days has been shown to reduce the risk of major ischemic events in patients with transient ischemic attack (TIA) or minor stroke. However, the mechanisms underlying combination treatment with ASA + CPG in experimental ischemic stroke has not been fully elucidated. Methods Minor cerebral ischemia was induced in mice by transient distal middle cerebral artery occlusion (tdMCAO). Two doses of ASA + CPG (12 and 24 mg/kg/day) or vehicle were administered by gavage daily. Neurological behaviors were assessed using the modified Garcia scores, Rotarod test, Y maze, and open field test. Platelet function was assessed in vitro by flow cytometry and in vivo by bleeding and clotting time. The neutrophil ratio and the levels of inflammatory cytokines were measured by flow cytometry and the Meso Scale Discovery (MSD) electrochemilunimescence, respectively. Results Sensorimotor function was partially recovered with ASA + CPG treatment after ischemia. Anxiety levels and cognitive functions showed improvement in the ASA + CPG group at 12 mg/kg/day after 21 days. Both tail bleeding time and flow cytometry showed significantly decreased platelet function after ASA + CPG treatment. Notably, ASA + CPG at 12 mg/kg/day prolonged clotting time at 28 days after injury. Furthermore, the ratio of neutrophils, an indicator of inflammation, was reduced with 12 mg/kg/day ASA + CPG treatment in the bone marrow (BM) at 21 days and in the peripheral blood (PB) at 21 and 28 days after tdMCAO. Both doses of ASA + CPG decreased pro-inflammatory cytokine interleukin (IL)-6 expression 21 days after stroke. Taken together, these results demonstrated that combination treatment with ASA + CPG improved long-term neurological function after stroke and may inhibit platelet-neutrophil interaction by decreasing the concentration of pro-inflammatory cytokine, IL-6. Conclusions These findings indicate a neuroprotective effect of combination treatment with ASA + CPG for a duration of 21 days in an experimental acute minor stroke model. These findings provide further evidence that dual antiplatelet therapy may be a viable neuroprotective treatment to decrease the recurrence of stroke.
Collapse
Affiliation(s)
- Wen Dong
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiangrong Liu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenqian Liu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chunjuan Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shunying Zhao
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shaohong Wen
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ting Gong
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wentao Chen
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qingfang Chen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Weizhen Ye
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zixiao Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yongjun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Jurcau A, Simion A. Neuroinflammation in Cerebral Ischemia and Ischemia/Reperfusion Injuries: From Pathophysiology to Therapeutic Strategies. Int J Mol Sci 2021; 23:14. [PMID: 35008440 PMCID: PMC8744548 DOI: 10.3390/ijms23010014] [Citation(s) in RCA: 175] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/18/2021] [Accepted: 12/18/2021] [Indexed: 02/07/2023] Open
Abstract
Its increasing incidence has led stroke to be the second leading cause of death worldwide. Despite significant advances in recanalization strategies, patients are still at risk for ischemia/reperfusion injuries in this pathophysiology, in which neuroinflammation is significantly involved. Research has shown that in the acute phase, neuroinflammatory cascades lead to apoptosis, disruption of the blood-brain barrier, cerebral edema, and hemorrhagic transformation, while in later stages, these pathways support tissue repair and functional recovery. The present review discusses the various cell types and the mechanisms through which neuroinflammation contributes to parenchymal injury and tissue repair, as well as therapeutic attempts made in vitro, in animal experiments, and in clinical trials which target neuroinflammation, highlighting future therapeutic perspectives.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania;
- Neurology Ward, Clinical Municipal Hospital “dr. G. Curteanu” Oradea, 410154 Oradea, Romania
| | - Aurel Simion
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania;
- Neurorehabilitation Ward, Clinical Municipal Hospital “dr. G. Curteanu” Oradea, 410154 Oradea, Romania
| |
Collapse
|
14
|
Patel PM, Connolly MR, Coe TM, Calhoun A, Pollok F, Markmann JF, Burdorf L, Azimzadeh A, Madsen JC, Pierson RN. Minimizing Ischemia Reperfusion Injury in Xenotransplantation. Front Immunol 2021; 12:681504. [PMID: 34566955 PMCID: PMC8458821 DOI: 10.3389/fimmu.2021.681504] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/12/2021] [Indexed: 12/21/2022] Open
Abstract
The recent dramatic advances in preventing "initial xenograft dysfunction" in pig-to-non-human primate heart transplantation achieved by minimizing ischemia suggests that ischemia reperfusion injury (IRI) plays an important role in cardiac xenotransplantation. Here we review the molecular, cellular, and immune mechanisms that characterize IRI and associated "primary graft dysfunction" in allotransplantation and consider how they correspond with "xeno-associated" injury mechanisms. Based on this analysis, we describe potential genetic modifications as well as novel technical strategies that may minimize IRI for heart and other organ xenografts and which could facilitate safe and effective clinical xenotransplantation.
Collapse
Affiliation(s)
- Parth M. Patel
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Margaret R. Connolly
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Taylor M. Coe
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Anthony Calhoun
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Franziska Pollok
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Anesthesiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - James F. Markmann
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Transplantation, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Lars Burdorf
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Agnes Azimzadeh
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Joren C. Madsen
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Richard N. Pierson
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
15
|
Durán-Laforet V, Peña-Martínez C, García-Culebras A, Alzamora L, Moro MA, Lizasoain I. Pathophysiological and pharmacological relevance of TLR4 in peripheral immune cells after stroke. Pharmacol Ther 2021; 228:107933. [PMID: 34174279 DOI: 10.1016/j.pharmthera.2021.107933] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/12/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023]
Abstract
Stroke is a very common disease being the leading cause of death and disability worldwide. The immune response subsequent to an ischemic stroke is a crucial factor in its physiopathology and outcome. This response is not limited to the injury site. In fact, the immune response to the ischemic process mobilizes mainly circulating cells which upon activation will be recruited to the injury site. When a stroke occurs, molecules that are usually retained inside the cell bodies are released into the extracellular space by uncontrolled cell death. These molecules can bind to the Toll-like receptor 4 (TLR4) in circulating immune cells which are then activated, eliciting, although not exclusively, the inflammatory response to the stroke. In this review, we present an up-to-date summary of the role of the different peripheral immune cells in stroke as well as the role of TLR4 in the function of each cell type in ischemia. Also, we summarize the different antagonists developed against TLR4 and their potential as a pharmacological tool for stroke treatment.
Collapse
Affiliation(s)
- V Durán-Laforet
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital, 12 de Octubre (imas12), Madrid, Spain.
| | - C Peña-Martínez
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital, 12 de Octubre (imas12), Madrid, Spain
| | - A García-Culebras
- Neurovascular Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - L Alzamora
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital, 12 de Octubre (imas12), Madrid, Spain
| | - M A Moro
- Neurovascular Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - I Lizasoain
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Instituto de Investigación Hospital, 12 de Octubre (imas12), Madrid, Spain.
| |
Collapse
|
16
|
Platelets as drivers of ischemia/reperfusion injury after stroke. Blood Adv 2021; 5:1576-1584. [PMID: 33687431 DOI: 10.1182/bloodadvances.2020002888] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 02/01/2021] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke is a leading cause of morbidity and mortality worldwide and, despite reperfusion either via thrombolysis or thrombectomy, stroke patients often suffer from lifelong disabilities. These persistent neurological deficits may be improved by treating the ischemia/reperfusion (I/R) injury that occurs following ischemic stroke. There are currently no approved therapies to treat I/R injury, and thus it is imperative to find new targets to decrease the burden of ischemic stroke and related diseases. Platelets, cell fragments from megakaryocytes, are primarily known for their role in hemostasis. More recently, investigators have studied the nonhemostatic role of platelets in inflammatory pathologies, such as I/R injury after ischemic stroke. In this review, we seek to provide an overview of how I/R can lead to platelet activation and how activated platelets, in turn, can exacerbate I/R injury after stroke. We will also discuss potential mechanisms by which platelets may ameliorate I/R injury.
Collapse
|
17
|
Berchtold D, Priller J, Meisel C, Meisel A. Interaction of microglia with infiltrating immune cells in the different phases of stroke. Brain Pathol 2020; 30:1208-1218. [PMID: 33058417 PMCID: PMC8018083 DOI: 10.1111/bpa.12911] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 08/23/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022] Open
Abstract
Stroke, in association with its complications, is one of the leading causes of mortality and morbidity worldwide. Cerebral ischemia triggers an inflammatory response in the brain that is controlled by the activation of resident microglia as well as the infiltration of peripheral myeloid and lymphoid cells into the brain parenchyma. This inflammation has been shown to have both beneficial and detrimental effects on stroke outcome. The focus of this review lies on the functions of myeloid cells and their interaction with infiltrating lymphocytes in different phases of stroke. A detailed and time-specific understanding of the contribution of different immune cell subsets during the course of cerebral ischemia is crucial to specifically promote beneficial and inhibit detrimental effects of inflammation on stroke outcome.
Collapse
Affiliation(s)
- Daniel Berchtold
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Josef Priller
- Department of Neuropsychiatry and DZNE, Charité - Universitätsmedizin Berlin, Berlin, Germany.,UK DRI, University of Edinburgh, Edinburgh, UK
| | - Christian Meisel
- Institute for Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Meisel
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Neurocure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
18
|
Schrottmaier WC, Mussbacher M, Salzmann M, Assinger A. Platelet-leukocyte interplay during vascular disease. Atherosclerosis 2020; 307:109-120. [DOI: 10.1016/j.atherosclerosis.2020.04.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/08/2020] [Accepted: 04/29/2020] [Indexed: 02/06/2023]
|
19
|
He P, Talukder MAH, Gao F. Oxidative Stress and Microvessel Barrier Dysfunction. Front Physiol 2020; 11:472. [PMID: 32536875 PMCID: PMC7268512 DOI: 10.3389/fphys.2020.00472] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 04/17/2020] [Indexed: 12/11/2022] Open
Abstract
Clinical and experimental evidence indicate that increased vascular permeability contributes to many disease-associated vascular complications. Oxidative stress with increased production of reactive oxygen species (ROS) has been implicated in a wide variety of pathological conditions, including inflammation and many cardiovascular diseases. It is thus important to identify the role of ROS and their mechanistic significance in microvessel barrier dysfunction under pathological conditions. The role of specific ROS and their cross talk in pathological processes is complex. The mechanisms of ROS-induced increases in vascular permeability remain poorly understood. The sources of ROS in diseases have been extensively reviewed at enzyme levels. This review will instead focus on the underlying mechanisms of ROS release by leukocytes, the differentiate effects and signaling mechanisms of individual ROS on endothelial cells, pericytes and microvessel barrier function, as well as the interplay of reactive oxygen species, nitric oxide, and nitrogen species in ROS-mediated vascular barrier dysfunction. As a counter balance of excessive ROS, nuclear factor erythroid 2 related factor 2 (Nrf2), a redox-sensitive cell-protective transcription factor, will be highlighted as a potential therapeutic target for antioxidant defenses. The advantages and limitations of different experimental approaches used for the study of ROS-induced endothelial barrier function are also discussed. This article will outline the advances emerged mainly from in vivo and ex vivo studies and attempt to consolidate some of the opposing views in the field, and hence provide a better understanding of ROS-mediated microvessel barrier dysfunction and benefit the development of therapeutic strategies.
Collapse
Affiliation(s)
- Pingnian He
- Department of Cellular and Molecular Physiology, College of Medicine, The Pennsylvania State University, Hershey, PA, United States
| | - M A Hassan Talukder
- Department of Cellular and Molecular Physiology, College of Medicine, The Pennsylvania State University, Hershey, PA, United States
| | - Feng Gao
- Department of Cellular and Molecular Physiology, College of Medicine, The Pennsylvania State University, Hershey, PA, United States
| |
Collapse
|
20
|
Denorme F, Manne BK, Portier I, Eustes AS, Kosaka Y, Kile BT, Rondina MT, Campbell RA. Platelet necrosis mediates ischemic stroke outcome in mice. Blood 2020; 135:429-440. [PMID: 31800959 PMCID: PMC7005363 DOI: 10.1182/blood.2019002124] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 11/15/2019] [Indexed: 12/28/2022] Open
Abstract
Dysregulated platelet functions contribute to the development and progression of ischemic stroke. Utilizing mice with a platelet-specific deletion of cyclophilin D (CypD), a mediator of necrosis, we found that platelet necrosis regulates tissue damage and outcomes during ischemic stroke in vivo. Mice with loss of CypD in platelets (CypDplt-/-mice) exhibited significantly enhanced cerebral blood flow, improved neurological and motor functions, and reduced ischemic stroke infarct volume after cerebral ischemia-reperfusion injury. These effects were attributable, at least in part, to platelet-neutrophil interactions. Twenty-four hours after stroke, significantly more circulating platelet-neutrophil aggregates (PNAs) were found in CypDplt+/+ mice. Underscoring the role of platelet necrosis in PNA formation, we observed a significant number of phosphatidylserine (PS)+ platelets in PNAs in CypDplt+/+ mice. In contrast, significantly fewer platelets in PNAs were PS+ in CypDplt-/- counterparts. Accordingly, mice with CypD-deficient platelets had fewer neutrophils and PNAs recruited to their brain following stroke relative to wild-type counterparts. Neutrophil depletion in wild-type mice conferred protection from ischemic stroke to a similar degree as observed in mice with CypD-deficient platelets. Neutrophil depletion in CypDplt-/- mice did not further reduce infarct size. Transmission electron microscopy of ex vivo-formed PNAs revealed a propensity of necrotic platelets to interact with neutrophils. These results suggest that necrotic platelets interact with neutrophils to exacerbate brain injury during ischemic stroke. Because inhibiting platelet necrosis does not compromise hemostasis, targeting platelet CypD may be a potential therapeutic strategy to limit brain damage following ischemic stroke.
Collapse
Affiliation(s)
- Frederik Denorme
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Laboratory for Thrombosis Research, Katholieke Universiteit Leuven Campus Kulak Kortrijk, Belgium
| | | | - Irina Portier
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Alicia S Eustes
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Yasuhiro Kosaka
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Benjamin T Kile
- Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Matthew T Rondina
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
- George E. Wahlen Veterans Affairs Medical Centers Department of Internal Medicine and Geriatric Research Education and Clinical Center, Salt Lake City, UT; and
- Department of Pathology, University of Utah, Salt Lake City, UT
| | - Robert A Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| |
Collapse
|
21
|
Susanto O, Hickey MJ. Using imaging to study inflammatory platelet–leukocyte interactions in vivo. Platelets 2020; 31:610-617. [DOI: 10.1080/09537104.2020.1718632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Olivia Susanto
- Centre for Inflammatory Diseases, Monash Medical Centre, Monash University Department of Medicine, Clayton, Australia
| | - Michael J. Hickey
- Centre for Inflammatory Diseases, Monash Medical Centre, Monash University Department of Medicine, Clayton, Australia
| |
Collapse
|
22
|
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
|
23
|
Robicsek SA, Bhattacharya A, Rabai F, Shukla K, Doré S. Blood-Related Toxicity after Traumatic Brain Injury: Potential Targets for Neuroprotection. Mol Neurobiol 2019; 57:159-178. [PMID: 31617072 DOI: 10.1007/s12035-019-01766-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
Abstract
Emergency visits, hospitalizations, and deaths due to traumatic brain injury (TBI) have increased significantly over the past few decades. While the primary early brain trauma is highly deleterious to the brain, the secondary injury post-TBI is postulated to significantly impact mortality. The presence of blood, particularly hemoglobin, and its breakdown products and key binding proteins and receptors modulating their clearance may contribute significantly to toxicity. Heme, hemin, and iron, for example, cause membrane lipid peroxidation, generate reactive oxygen species, and sensitize cells to noxious stimuli resulting in edema, cell death, and increased morbidity and mortality. A wide range of other mechanisms such as the immune system play pivotal roles in mediating secondary injury. Effective scavenging of all of these pro-oxidant and pro-inflammatory metabolites as well as controlling maladaptive immune responses is essential for limiting toxicity and secondary injury. Hemoglobin metabolism is mediated by key molecules such as haptoglobin, heme oxygenase, hemopexin, and ferritin. Genetic variability and dysfunction affecting these pathways (e.g., haptoglobin and heme oxygenase expression) have been implicated in the difference in susceptibility of individual patients to toxicity and may be target pathways for potential therapeutic interventions in TBI. Ongoing collaborative efforts are required to decipher the complexities of blood-related toxicity in TBI with an overarching goal of providing effective treatment options to all patients with TBI.
Collapse
Affiliation(s)
- Steven A Robicsek
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA. .,Departments of Neurosurgery, Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Ayon Bhattacharya
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA.,Department of Pharmacology, KPC Medical College, West Bengal University of Health Sciences, Kolkata, West Bengal, India
| | - Ferenc Rabai
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA
| | - Krunal Shukla
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA
| | - Sylvain Doré
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease and McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Biomed Sci J493, Gainesville, FL, 32610, USA. .,Departments of Neurology, Psychiatry, Pharmaceutics and Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
24
|
Jansen MPB, Huisman A, Claessen N, Florquin S, Roelofs JJTH. Experimental thrombocytopenia does not affect acute kidney injury 24 hours after renal ischemia reperfusion in mice. Platelets 2019; 31:383-391. [PMID: 31364433 DOI: 10.1080/09537104.2019.1646899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The pathophysiology of renal ischemia/reperfusion (I/R) injury is characterized by excessive activation of inflammation and coagulation processes followed by abnormal renal tissue repair, resulting in renal injury and function loss. Platelets are important actors in these processes, however to what extent platelets contribute to the pathophysiology of renal I/R injury still needs to be elucidated. In the current study, we treated wild-type mice with a platelet depleting antibody, which caused thrombocytopenia. We then investigated the role of platelets during the pathophysiology of renal I/R by subjecting control wild-type mice with normal platelet counts and thrombocytopenic wild-type mice to renal I/R injury. Our results showed that in the early phase of renal I/R injury, thrombocytopenia 24 hours after ischemia reperfusion does not influence renal injury, neutrophil infiltration and accumulation of inflammatory chemokines (e.g. keratinocyte chemoattractant, monocyte chemoattractant protein 1, tumor necrosis factor alpha). In the recovery and regeneration phase of I/R injury, respectively 5 and 10 days post-ischemia, thrombocytopenia did also not affect the accumulation of intra-renal neutrophils and macrophages, renal injury, and renal fibrosis. Together, these results imply that lowering platelet counts do not impact the pathogenesis of I/R injury in mice.
Collapse
Affiliation(s)
- Marcel P B Jansen
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam Infection & Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Andras Huisman
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam Infection & Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Nike Claessen
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam Infection & Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Sandrine Florquin
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam Infection & Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam Infection & Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
25
|
Stoll G, Nieswandt B. Thrombo-inflammation in acute ischaemic stroke — implications for treatment. Nat Rev Neurol 2019; 15:473-481. [DOI: 10.1038/s41582-019-0221-1] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2019] [Indexed: 01/17/2023]
|
26
|
Wu T, Jia Z, Dong S, Han B, Zhang R, Liang Y, Zhang S, Sun J. Panax notoginseng Saponins Ameliorate Leukocyte Adherence and Cerebrovascular Endothelial Barrier Breakdown upon Ischemia-Reperfusion in Mice. J Vasc Res 2019; 56:1-10. [DOI: 10.1159/000494935] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/29/2018] [Indexed: 11/19/2022] Open
|
27
|
García-Culebras A, Durán-Laforet V, Peña-Martínez C, Ballesteros I, Pradillo JM, Díaz-Guzmán J, Lizasoain I, Moro MA. Myeloid cells as therapeutic targets in neuroinflammation after stroke: Specific roles of neutrophils and neutrophil-platelet interactions. J Cereb Blood Flow Metab 2018; 38:2150-2164. [PMID: 30129391 PMCID: PMC6282223 DOI: 10.1177/0271678x18795789] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Ischemic brain injury causes a local inflammatory response, involving the activation of resident brain cells such as microglia and the recruitment of infiltrating immune cells. Increasing evidence supports that plasticity of the myeloid cell lineage is determinant for the specific role of these cells on stroke outcome, from initiation and maintenance to resolution of post-ischemic inflammation. The aim of this review is to summarize some of the key characteristics of these cells and the mechanisms for their recruitment into the injured brain through interactions with platelets, endothelial cells and other leukocytes. Also, we discuss the existence of different leukocyte subsets in the ischemic tissue and, specifically, the impact of different myeloid phenotypes on stroke outcome, with special emphasis on neutrophils and their interplay with platelets. Knowledge of these cellular phenotypes and interactions may pave the way to new therapies able to promote protective immune responses and tissue repair after cerebral ischemia.
Collapse
Affiliation(s)
- Alicia García-Culebras
- 1 Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain.,2 Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,3 Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM, Madrid, Spain
| | - Violeta Durán-Laforet
- 1 Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain.,2 Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,3 Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM, Madrid, Spain
| | - Carolina Peña-Martínez
- 1 Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain.,2 Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,3 Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM, Madrid, Spain
| | - Iván Ballesteros
- 4 Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jesús M Pradillo
- 1 Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain.,2 Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,3 Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM, Madrid, Spain
| | - Jaime Díaz-Guzmán
- 2 Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,5 Servicio de Neurología, Hospital Universitario Doce de Octubre, Madrid, Spain
| | - Ignacio Lizasoain
- 1 Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain.,2 Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,3 Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM, Madrid, Spain
| | - María A Moro
- 1 Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain.,2 Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,3 Instituto Universitario de Investigación en Neuroquímica (IUIN), UCM, Madrid, Spain
| |
Collapse
|
28
|
Finsterbusch M, Schrottmaier WC, Kral-Pointner JB, Salzmann M, Assinger A. Measuring and interpreting platelet-leukocyte aggregates. Platelets 2018; 29:677-685. [PMID: 29461910 PMCID: PMC6178087 DOI: 10.1080/09537104.2018.1430358] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Platelets, besides their specialised role in haemostasis and atherothrombosis, actively modulate innate and adaptive immune responses with crucial roles in immune surveillance, inflammation and host defence during infection. An important prerequisite for platelet-mediated changes of immune functions involves direct engagement with different types of leukocytes. Indeed, increased platelet-leukocyte aggregates (PLAs) within the circulation and/or locally at the site of inflammation represent markers of many thrombo-inflammatory diseases, such as cardiovascular diseases, acute lung injury, renal and cerebral inflammation. Therefore, measurement of PLAs could provide an attractive and easily accessible prognostic and/or diagnostic tool for many diseases. To measure PLAs in different (patho-)physiological settings in human and animal models flow cytometric and microscopic approaches have been applied. These techniques represent complementary tools to study different aspects relating to the involvement of leukocyte subtypes and molecules, as well as location of PLAs within tissues, dynamics of their interactions and/or dynamic changes in leukocyte and platelet behaviour. This review summarises various approaches to measure and interpret PLAs and discusses potential experimental factors influencing platelet binding to leukocytes. Furthermore, we summarise insights gained from studies regarding the underlying mechanism of platelet-leukocyte interactions and discuss implications of these interactions in health and disease.
Collapse
Affiliation(s)
- Michaela Finsterbusch
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Waltraud C Schrottmaier
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Julia B Kral-Pointner
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Manuel Salzmann
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Alice Assinger
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| |
Collapse
|
29
|
Liu S, Wei C, Kang N, He Q, Liang J, Wang H, Chang L, Chen D, Zhang Q, Chang C, Zhang J, Ren H, Wu Y. Chinese medicine Tongxinluo capsule alleviates cerebral microcirculatory disturbances in ischemic stroke by modulating vascular endothelial function and inhibiting leukocyte-endothelial cell interactions in mice: A two-photon laser scanning microscopy stu. Microcirculation 2018; 25. [DOI: 10.1111/micc.12437] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 12/08/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Shen Liu
- Dongzhimen Hospital; Beijing University of Chinese Medicine; Beijing China
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine; Hebei Yiling Chinese Medicine Research Institute; Shijiazhuang China
| | - Cong Wei
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine; Hebei Yiling Chinese Medicine Research Institute; Shijiazhuang China
| | - Ning Kang
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine; Hebei Yiling Chinese Medicine Research Institute; Shijiazhuang China
| | - Qilong He
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine; Hebei Yiling Chinese Medicine Research Institute; Shijiazhuang China
| | - Junqing Liang
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine; Hebei Yiling Chinese Medicine Research Institute; Shijiazhuang China
| | - Hongtao Wang
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine; Hebei Yiling Chinese Medicine Research Institute; Shijiazhuang China
| | - Liping Chang
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine; Hebei Yiling Chinese Medicine Research Institute; Shijiazhuang China
| | - Daohong Chen
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine; Hebei Yiling Chinese Medicine Research Institute; Shijiazhuang China
| | - Qiuyan Zhang
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine; Hebei Yiling Chinese Medicine Research Institute; Shijiazhuang China
| | - Chengcheng Chang
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine; Hebei Yiling Chinese Medicine Research Institute; Shijiazhuang China
| | - Junfang Zhang
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine; Hebei Yiling Chinese Medicine Research Institute; Shijiazhuang China
| | - Hong Ren
- Graduate School; Hebei Medical University; Shijiazhuang China
| | - Yiling Wu
- Dongzhimen Hospital; Beijing University of Chinese Medicine; Beijing China
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine; Hebei Yiling Chinese Medicine Research Institute; Shijiazhuang China
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Interactions between neutrophils and platelets contribute to the progression of thromboinflammatory disease. However, the regulatory mechanism governing these interactions is poorly understood. The present review focuses on the crucial role of Ser/Thr protein kinase B (AKT)β-NADPH oxidase 2 (NOX2) signaling in regulating neutrophil and platelet activation and their heterotypic interactions under thromboinflammatory conditions. RECENT FINDINGS Growing evidence has shown that platelets, leukocytes, and blood coagulation need to be considered to treat thromboinflammatory disease in which inflammation and thrombosis occur concurrently. In addition to plasma proteins and intracellular signaling molecules, extracellular reactive oxygen species (ROS) produced from activated leukocytes could be an important factor in the pathophysiology of thromboinflammatory disease. Recent studies reveal that AKT2-NOX2 signaling has critical roles in Ca mobilization, ROS generation, degranulation, and control of the ligand-binding function of cell surface molecules, thereby promoting heterotypic cell-cell interactions in thromboinflammation. These findings have provided novel insights into attractive therapeutic targets for the prevention and treatment of thromboinflammatory disease. SUMMARY Recent discoveries concerning molecular mechanisms regulating neutrophil-platelet interactions have bridged some gaps in our knowledge of the complicated signaling pathways exacerbating thromboinflammatory conditions.
Collapse
|
31
|
Yang XM, Chen XH, Lu JF, Zhou CM, Han JY, Chen CH. In vivo observation of cerebral microcirculation after experimental subarachnoid hemorrhage in mice. Neural Regen Res 2018; 13:456-462. [PMID: 29623930 PMCID: PMC5900508 DOI: 10.4103/1673-5374.228728] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Acute brain injury caused by subarachnoid hemorrhage is the major cause of poor prognosis. The pathology of subarachnoid hemorrhage likely involves major morphological changes in the microcirculation. However, previous studies primarily used fixed tissue or delayed injury models. Therefore, in the present study, we used in vivo imaging to observe the dynamic changes in cerebral microcirculation after subarachnoid hemorrhage. Subarachnoid hemorrhage was induced by perforation of the bifurcation of the middle cerebral and anterior cerebral arteries in male C57/BL6 mice. The diameter of pial arterioles and venules was measured by in vivo fluorescence microscopy at different time points within 180 minutes after subarachnoid hemorrhage. Cerebral blood flow was examined and leukocyte adhesion/albumin extravasation was determined at different time points before and after subarachnoid hemorrhage. Cerebral pial microcirculation was abnormal and cerebral blood flow was reduced after subarachnoid hemorrhage. Acute vasoconstriction occurred predominantly in the arterioles instead of the venules. A progressive increase in the number of adherent leukocytes in venules and substantial albumin extravasation were observed between 10 and 180 minutes after subarachnoid hemorrhage. These results show that major changes in microcirculation occur in the early stage of subarachnoid hemorrhage. Our findings may promote the development of novel therapeutic strategies for the early treatment of subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Xiao-Mei Yang
- Department of Human Anatomy and Embryology, Peking University Health Science Center, Beijing, China
| | - Xu-Hao Chen
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jian-Fei Lu
- Department of Human Anatomy and Embryology, Peking University Health Science Center, Beijing, China
| | - Chang-Man Zhou
- Department of Human Anatomy and Embryology, Peking University Health Science Center, Beijing, China
| | - Jing-Yan Han
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
| | - Chun-Hua Chen
- Department of Human Anatomy and Embryology, Peking University Health Science Center, Beijing, China
| |
Collapse
|
32
|
Tong LS, Guo ZN, Ou YB, Yu YN, Zhang XC, Tang J, Zhang JH, Lou M. Cerebral venous collaterals: A new fort for fighting ischemic stroke? Prog Neurobiol 2017; 163-164:172-193. [PMID: 29199136 DOI: 10.1016/j.pneurobio.2017.11.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/03/2017] [Accepted: 11/28/2017] [Indexed: 12/13/2022]
Abstract
Stroke therapy has entered a new era highlighted by the use of endovascular therapy in addition to intravenous thrombolysis. However, the efficacy of current therapeutic regimens might be reduced by their associated adverse events. For example, over-reperfusion and futile recanalization may lead to large infarct, brain swelling, hemorrhagic complication and neurological deterioration. The traditional pathophysiological understanding on ischemic stroke can hardly address these occurrences. Accumulating evidence suggests that a functional cerebral venous drainage, the major blood reservoir and drainage system in brain, may be as critical as arterial infusion for stroke evolution and clinical sequelae. Further exploration of the multi-faceted function of cerebral venous system may add new implications for stroke outcome prediction and future therapeutic decision-making. In this review, we emphasize the anatomical and functional characteristics of the cerebral venous system and illustrate its necessity in facilitating the arterial infusion and maintaining the cerebral perfusion in the pathological stroke content. We then summarize the recent critical clinical studies that underscore the associations between cerebral venous collateral and outcome of ischemic stroke with advanced imaging techniques. A novel three-level venous system classification is proposed to demonstrate the distinct characteristics of venous collaterals in the setting of ischemic stroke. Finally, we discuss the current directions for assessment of cerebral venous collaterals and provide future challenges and opportunities for therapeutic strategies in the light of these new concepts.
Collapse
Affiliation(s)
- Lu-Sha Tong
- Department of Neurology, The 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China; Departments of Physiology, Loma Linda University, School of Medicine, CA, USA
| | - Zhen-Ni Guo
- Department of Neurology, The First Affiliated Hospital of Jilin University, Changchun, China; Departments of Physiology, Loma Linda University, School of Medicine, CA, USA
| | - Yi-Bo Ou
- Department of Neurosurgery, Tong-ji Hospital, Wuhan, China; Departments of Physiology, Loma Linda University, School of Medicine, CA, USA
| | - Yan-Nan Yu
- Department of Neurology, The 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Xiao-Cheng Zhang
- Department of Neurology, The 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Jiping Tang
- Department of Anesthesiology, Loma Linda University, School of Medicine, CA, USA
| | - John H Zhang
- Departments of Physiology, Loma Linda University, School of Medicine, CA, USA.
| | - Min Lou
- Department of Neurology, The 2nd Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China.
| |
Collapse
|
33
|
Xu XQ, Wu CJ, Lu SS, Gao QQ, Zu QQ, Liu XL, Shi HB, Liu S. Correlation between Intravoxel Incoherent Motion Magnetic Resonance Imaging Derived Metrics and Serum Soluble CD40 Ligand Level in an Embolic Canine Stroke Model. Korean J Radiol 2017; 18:835-843. [PMID: 28860901 PMCID: PMC5552467 DOI: 10.3348/kjr.2017.18.5.835] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/22/2017] [Indexed: 12/18/2022] Open
Abstract
Objective To determine the relationship between intravoxel incoherent motion (IVIM) imaging derived quantitative metrics and serum soluble CD40 ligand (sCD40L) level in an embolic canine stroke model. Materials and Methods A middle cerebral artery occlusion model was established in 24 beagle dogs. Experimental dogs were divided into low- and high-sCD40L group according to serum sCD40L level at 4.5 hours after establishing the model. IVIM imaging was scanned at 4.5 hours after model establishment using 10 b values ranging from 0 to 900 s/mm2. Quantitative metrics diffusion coefficient (D), pseudodiffusion coefficient (D*), and perfusion fraction (f) of ischemic lesions were calculated. Quantitative metrics of ischemic lesions were normalized by contralateral hemisphere using the following formula: normalized D = Dstroke / Dcontralateral. Differences in IVIM metrics between the low- and high-sCD40L groups were compared using t test. Pearson's correlation analyses were performed to determine the relationship between IVIM metrics and serum sCD40L level. Results The high-sCD40L group showed significantly lower f and normalized f values than the low-sCD40L group (f, p < 0.001; normalized f, p < 0.001). There was no significant difference in D*, normalized D*, D, or normalized D value between the two groups (All p > 0.05). Both f and normalized f values were negatively correlated with serum sCD40L level (f, r = −0.789, p < 0.001; normalized f, r = −0.823, p < 0.001). However, serum sCD40L level had no significant correlation with D*, normalized D*, D, or normalized D (All p > 0.05). Conclusion The f value derived from IVIM imaging was negatively correlated with serum sCD40L level. f value might serve as a potential imaging biomarker to assess the formation of microvascular thrombosis in hyperacute period of ischemic stroke.
Collapse
Affiliation(s)
- Xiao-Quan Xu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chen-Jiang Wu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Shan-Shan Lu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qian-Qian Gao
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qing-Quan Zu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xing-Long Liu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hai-Bin Shi
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Sheng Liu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
34
|
Lien LM, Lin KH, Huang LT, Tseng MF, Chiu HC, Chen RJ, Lu WJ. Licochalcone A Prevents Platelet Activation and Thrombus Formation through the Inhibition of PLCγ2-PKC, Akt, and MAPK Pathways. Int J Mol Sci 2017; 18:ijms18071500. [PMID: 28704925 PMCID: PMC5535990 DOI: 10.3390/ijms18071500] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/07/2017] [Accepted: 07/09/2017] [Indexed: 01/27/2023] Open
Abstract
Platelet activation is involved in cardiovascular diseases, such as atherosclerosis and ischemic stroke. Licochalcone A (LA), an active ingredient of licorice, exhibits multiple biological activities such as anti-oxidation and anti-inflammation. However, its role in platelet activation remains unclear. Therefore, the study investigated the antiplatelet mechanism of LA. Our data revealed that LA (2-10 μM) concentration dependently inhibited platelet aggregation induced by collagen, but not thrombin and U46619. LA markedly attenuated collagen-stimulated ATP release, P-selectin secretion, calcium mobilization, and GPIIbIIIa activation, but did not interfere with the collagen binding to platelets. Moreover, LA significantly reduced the activation of PLCγ2, PKC, Akt and MAPKs. Thus, LA attenuates platelet activation, possibly by inhibiting collagen receptor downstream signaling but not by blocking the collagen receptors. In addition, LA prevented adenosine diphosphate (ADP)-induced acute pulmonary thrombosis, fluorescein sodium-induced platelet thrombus formation, and middle cerebral artery occlusion/reperfusion-induced brain injury in mice, but did not affect normal hemostasis. This study demonstrated that LA effectively reduced platelet activation and thrombus formation, in part, through the inhibition of PLCγ2-PKC, Akt, and MAPK pathways, without the side effect of bleeding. These findings also indicate that LA may provide a safe and alternative therapeutic approach for preventing thromboembolic disorders such as stroke.
Collapse
Affiliation(s)
- Li-Ming Lien
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Neurology, Shin Kong Wu Ho Su Memorial Hospital, Taipei 111, Taiwan.
| | - Kuan-Hung Lin
- Department of Pharmacology and Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 110, Taiwan.
- Central Laboratory, Shin Kong Wu Ho Su Memorial Hospital, Taipei 111, Taiwan.
| | - Li-Ting Huang
- Department of Medical Research and Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan.
| | - Mei-Fang Tseng
- Department of Medical Research and Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan.
| | - Hou-Chang Chiu
- Department of Neurology, Shin Kong Wu Ho Su Memorial Hospital, Taipei 111, Taiwan.
- College of Medicine, Fu-Jen Catholic University, Taipei 242, Taiwan.
| | - Ray-Jade Chen
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Medical Research and Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan.
| | - Wan-Jung Lu
- Department of Pharmacology and Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 110, Taiwan.
- Department of Medical Research and Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan.
- Graduate Institute of Metabolism and Obesity Sciences, College of Public Health and Nutrition, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
35
|
Wang Y, Yang J, Du G, Ma D, Zhou L. Neuroprotective effects respond to cerebral ischemia without susceptibility to HB-tumorigenesis in VHL heterozygous knockout mice. Mol Carcinog 2017; 56:2342-2351. [PMID: 28574654 DOI: 10.1002/mc.22688] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 05/04/2017] [Accepted: 06/01/2017] [Indexed: 12/18/2022]
Abstract
The von Hippel-Lindau (VHL) tumor suppressor gene plays a prominent role in the development of hemangioblastomas (HBs) within specific regions of the human' central nervous system (CNS). Alterations in VHL gene are rarely observed in the more common features of human VHL-related tumors in animal models, and VHL heterozygous knockout (VHL+/-) mice do not develop HBs. We tested whether VHL heterozygous knockout mice exhibited genetic predisposition to the development of HBs and conferred a selective advantage involving growth of blood vessels to its carrier. No differences were observed between wild-type and VHL+/- mice in development ad reproduction. The heterozygous VHL+/- mice did not develop higher genetic susceptibility to CNS-HBs over their lifetime. Furthermore, this recessive VHL gene heterozygosity is relatively stable. Interestingly, we found these heterozygous VHL+/- mice gained an advantage conferring to angiogenic ability in a particular environment, compared with wild-type mice. The heterozygous VHL+/- mice obviously enhanced hypoxia inducible factor-1 (HIF)-dependent and Twist1 angiogenic mechanism in response to acute cerebral ischemia, resulting in decreased cerebral tissue damage and neuroprotective response through neovascularization. Our findings provide evidence of partial loss function of VHL as a novel precise therapeutic target in acute cerebral ischemia.
Collapse
Affiliation(s)
- Ying Wang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jingyun Yang
- Rush Alzheimer's Disease Center & Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois
| | - Guhong Du
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Dexuan Ma
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Liangfu Zhou
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
36
|
Xu S, Zhong A, Ma H, Li D, Hu Y, Xu Y, Zhang J. Neuroprotective effect of salvianolic acid B against cerebral ischemic injury in rats via the CD40/NF-κB pathway associated with suppression of platelets activation and neuroinflammation. Brain Res 2017; 1661:37-48. [DOI: 10.1016/j.brainres.2017.02.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 02/07/2017] [Accepted: 02/12/2017] [Indexed: 11/16/2022]
|
37
|
Shukla V, Shakya AK, Perez-Pinzon MA, Dave KR. Cerebral ischemic damage in diabetes: an inflammatory perspective. J Neuroinflammation 2017; 14:21. [PMID: 28115020 PMCID: PMC5260103 DOI: 10.1186/s12974-016-0774-5] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 12/07/2016] [Indexed: 12/16/2022] Open
Abstract
Stroke is one of the leading causes of death worldwide. A strong inflammatory response characterized by activation and release of cytokines, chemokines, adhesion molecules, and proteolytic enzymes contributes to brain damage following stroke. Stroke outcomes are worse among diabetics, resulting in increased mortality and disabilities. Diabetes involves chronic inflammation manifested by reactive oxygen species generation, expression of proinflammatory cytokines, and activation/expression of other inflammatory mediators. It appears that increased proinflammatory processes due to diabetes are further accelerated after cerebral ischemia, leading to increased ischemic damage. Hypoglycemia is an intrinsic side effect owing to glucose-lowering therapy in diabetics, and is known to induce proinflammatory changes as well as exacerbate cerebral damage in experimental stroke. Here, we present a review of available literature on the contribution of neuroinflammation to increased cerebral ischemic damage in diabetics. We also describe the role of hypoglycemia in neuroinflammation and cerebral ischemic damage in diabetics. Understanding the role of neuroinflammatory mechanisms in worsening stroke outcome in diabetics may help limit ischemic brain injury and improve clinical outcomes.
Collapse
Affiliation(s)
- Vibha Shukla
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, Miami, FL, 33136, USA.,Department of Neurology (D4-5), University of Miami Miller School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA
| | - Akhalesh Kumar Shakya
- Present address: Department of Microbiology and Immunology, and Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130, USA
| | - Miguel A Perez-Pinzon
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, Miami, FL, 33136, USA.,Department of Neurology (D4-5), University of Miami Miller School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Neuroscience Program, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Kunjan R Dave
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, Miami, FL, 33136, USA. .,Department of Neurology (D4-5), University of Miami Miller School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA. .,Neuroscience Program, University of Miami School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
38
|
Bhat SA, Goel R, Shukla R, Hanif K. Platelet CD40L induces activation of astrocytes and microglia in hypertension. Brain Behav Immun 2017; 59:173-189. [PMID: 27658543 DOI: 10.1016/j.bbi.2016.09.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 09/16/2016] [Accepted: 09/17/2016] [Indexed: 12/20/2022] Open
Abstract
Studies have demonstrated separately that hypertension is associated with platelet activation in the periphery (resulting in accumulation and localized inflammatory response) and glial activation in the brain. We investigated the contribution of platelets in brain inflammation, particularly glial activation in vitro and in a rat model of hypertension. We found that HTN increased the expression of adhesion molecules like JAM-1, ICAM-1, and VCAM-1 on brain endothelium and resulted in the deposition of platelets in the brain. Platelet deposition in hypertensive rats was associated with augmented CD40 and CD40L and activation of astrocytes (GFAP expression) and microglia (Iba-1 expression) in the brain. Platelets isolated from hypertensive rats had significantly higher sCD40L levels and induced more prominent glial activation than platelets from normotensive rats. Activation of platelets with ADP induced sCD40L release and activation of astrocytes and microglia. Moreover, CD40L induced glial (astrocytes and microglia) activation, NFкB and MAPK inflammatory signaling, culminating in neuroinflammation and neuronal injury (increased apoptotic cells). Importantly, injection of ADP-activated platelets into normotensive rats strongly induced activation of astrocytes and microglia and increased plasma sCD40L levels compared with control platelets. On the contrary, inhibition of platelet activation by Clopidogrel or disruption of CD40 signaling prevented astrocyte and microglial activation and provided neuroprotection in both in vivo and in vitro conditions. Thus, we have identified platelet CD40L as a key inflammatory molecule for the induction of astrocyte and microglia activation, the major contributors to inflammation-mediated injury in the brain.
Collapse
Affiliation(s)
- Shahnawaz Ali Bhat
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, U.P., India
| | - Ruby Goel
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, U.P., India
| | - Rakesh Shukla
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, U.P., India
| | - Kashif Hanif
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, U.P., India; National Institute of Pharmaceutical Education and Research, Rae Bareli, India.
| |
Collapse
|
39
|
The pathophysiological role of astrocytic endothelin-1. Prog Neurobiol 2016; 144:88-102. [DOI: 10.1016/j.pneurobio.2016.04.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 03/23/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022]
|
40
|
Fukuoka T, Hayashi T, Hirayama M, Maruyama H, Mogi M, Horiuchi M, Takao M, Tanahashi N. Platelet-endothelial cell interaction in brain microvessels of angiotensin II type-2 receptor knockout mice following transient bilateral common carotid artery occlusion. J Thromb Thrombolysis 2016; 40:401-5. [PMID: 26231766 DOI: 10.1007/s11239-015-1254-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The purpose of this study was to investigate the behavior of platelets (rolling and adhesion) in cerebral microvessels of angiotensin II type-2 receptor-knockout (AT2RKO) mice after transient bilateral carotid artery occlusion using intravital fluorescence microscopy. Twenty AT2RKO mice, consisting of 11 mice in the sham group and 9 mice in the ischemia reperfusion group (reperfusion after 15 min of bilateral, total carotid artery occlusion) were used in this study. The hole traversed the bone and dura mater, but arachnoid, pia mater, and cerebral parenchyma were preserved. Platelets were harvested from donor mice and stained using carboxyfluorescein diacetate succinimidyl ester. The number of platelets showing rolling and adhesion to pial vessels in AT2 deficient mice at 3 and 6 h after cerebral ischemia reperfusion was significantly higher than that in the sham group (P < 0.05). In addition, AT2 receptor has an inhibitory role in platelet rolling and adhesion after cerebral ischemia reperfusion.
Collapse
Affiliation(s)
- Takuya Fukuoka
- Department of Neurology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan.
| | - Takeshi Hayashi
- Department of Neurology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Makiko Hirayama
- Department of Neurology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Hajime Maruyama
- Department of Neurology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Masaki Mogi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Ehime, Japan
| | - Masatsugu Horiuchi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Ehime, Japan
| | - Masaki Takao
- Department of Neurology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Norio Tanahashi
- Department of Neurology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan
| |
Collapse
|
41
|
Dopheide JF, Rubrech J, Trumpp A, Geissler P, Zeller GC, Bock K, Dünschede F, Trinh TT, Dorweiler B, Münzel T, Radsak MP, Espinola-Klein C. Leukocyte-platelet aggregates-a phenotypic characterization of different stages of peripheral arterial disease. Platelets 2016; 27:658-667. [PMID: 27352829 DOI: 10.3109/09537104.2016.1153619] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The formation of monocyte-platelet aggregates and neutrophil-platelet aggregates (MPA and NPA, respectively) is influenced by inflammation, but also might contribute to an exacerbation of inflammatory responses in atherosclerotic plaque. The purpose of this study was to analyze MPA and NPA proportions in regard to different stages of peripheral arterial disease (PAD). Forty-five patients with intermittent claudication (IC) (3 groups: Rutherford (R)-1, R-2, and R-3; each n = 15), 20 patients with critical limb ischemia (CLI) (Rutherford 5 (40%) and 6 (60%)), and 20 healthy controls were studied. Analyses of monocyte (Mon) subpopulations (CD14++CD16- (classical) Mon1, CD14++CD16+ (intermediate) Mon2, CD14+CD16++ (non-classical) Mon3), MPA, and NPA was performed from whole blood by flow cytometry. Controls showed an increased proportion of the Mon1 subpopulation (p < 0.001), whereas CLI patients showed a significant increase of the Mon2 subpopulation compared to controls, R-1, or R-2 patients (p < 0.0001). For the Mon3 subpopulation, CLI and R-3 patients showed an increased proportion (p < 0.05). MPA formation with the proinflammatory Mon2 and Mon3 subpopulations was increased in CLI patients (both p < 0.01). Similarly, NPA was significantly increased in CLI patients (p < 0.05). Serological markers of inflammation and procoagulation (fibrinogen [r = 0.459, p < 0.001], soluble triggering receptor expressed on myeloid cells (sTREM-1) [r = 0.237, p < 0.05] and P-Selectin [r = 0.225, p < 0.05]) correlated directly with MPA formation on the Mon2 subpopulation. We found an association of inflammatory and procoagulatory markers with increased formation of MPA on the Mon2 subpopulation. Since R-3 patients also had significantly increased MPA, one can speculate that the inflammatory burden might promote an aggravation of the disease.
Collapse
Affiliation(s)
- Jörn F Dopheide
- a Department of Internal Medicine II , University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| | - Jennifer Rubrech
- a Department of Internal Medicine II , University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| | - Amelie Trumpp
- a Department of Internal Medicine II , University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| | - Philip Geissler
- a Department of Internal Medicine II , University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| | - Geraldine C Zeller
- b Department of Internal Medicine I , University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| | - Karsten Bock
- a Department of Internal Medicine II , University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| | - Friedrich Dünschede
- c Department of Cardiothoracic and Vascular Surgery, Division of Vascular Surgery , University Medical Center, Johannes-Gutenberg University , Mainz , Germany
| | - Tran Tong Trinh
- c Department of Cardiothoracic and Vascular Surgery, Division of Vascular Surgery , University Medical Center, Johannes-Gutenberg University , Mainz , Germany
| | - Bernhard Dorweiler
- c Department of Cardiothoracic and Vascular Surgery, Division of Vascular Surgery , University Medical Center, Johannes-Gutenberg University , Mainz , Germany
| | - Thomas Münzel
- a Department of Internal Medicine II , University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| | - Markus P Radsak
- d Department of Internal Medicine III , University Medical Center, Johannes Gutenberg-University , Mainz , Germany.,e Institute for Immunology, University Medical Center, Johannes Gutenberg University , Mainz , Germany
| | - Christine Espinola-Klein
- a Department of Internal Medicine II , University Medical Center, Johannes Gutenberg-University , Mainz , Germany
| |
Collapse
|
42
|
Schmidt EP, Kuebler WM, Lee WL, Downey GP. Adhesion Molecules: Master Controllers of the Circulatory System. Compr Physiol 2016; 6:945-73. [PMID: 27065171 DOI: 10.1002/cphy.c150020] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This manuscript will review our current understanding of cellular adhesion molecules (CAMs) relevant to the circulatory system, their physiological role in control of vascular homeostasis, innate and adaptive immune responses, and their importance in pathophysiological (disease) processes such as acute lung injury, atherosclerosis, and pulmonary hypertension. This is a complex and rapidly changing area of research that is incompletely understood. By design, we will begin with a brief overview of the structure and classification of the major groups of adhesion molecules and their physiological functions including cellular adhesion and signaling. The role of specific CAMs in the process of platelet aggregation and hemostasis and leukocyte adhesion and transendothelial migration will be reviewed as examples of the complex and cooperative interplay between CAMs during physiological and pathophysiological processes. The role of the endothelial glycocalyx and the glycobiology of this complex system related to inflammatory states such as sepsis will be reviewed. We will then focus on the role of adhesion molecules in the pathogenesis of specific disease processes involving the lungs and cardiovascular system. The potential of targeting adhesion molecules in the treatment of immune and inflammatory diseases will be highlighted in the relevant sections throughout the manuscript.
Collapse
Affiliation(s)
- Eric P Schmidt
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Wolfgang M Kuebler
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Departments of Surgery and Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Warren L Lee
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Division of Respirology and the Interdepartmental Division of Critical Care Medicine, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Gregory P Downey
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Departments of Medicine, Pediatrics, and Biomedical Research, National Jewish Health, Denver, Colorado, USA
- Departments of Medicine, and Immunology and Microbiology, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
43
|
Zhao Y, Schwartz EA, Palmer GM, Zennadi R. MEK1/2 inhibitors reverse acute vascular occlusion in mouse models of sickle cell disease. FASEB J 2015; 30:1171-86. [PMID: 26631480 DOI: 10.1096/fj.15-278481] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 11/09/2015] [Indexed: 12/17/2022]
Abstract
In sickle cell disease (SCD), treatment of recurrent vasoocclusive episodes, leading to pain crises and organ damage, is still a therapeutic challenge. Vasoocclusion is caused primarily by adherence of homozygous for hemoglobin S (SS) red blood cells (SSRBCs) and leukocytes to the endothelium. We tested the therapeutic benefits of MEK1/2 inhibitors in reversing vasoocclusion in nude and humanized SCD mouse models of acute vasoocclusive episodes using intravital microscopy. Administration of 0.2, 0.3, 1, or 2 mg/kg MEK1/2 inhibitor to TNF-α-pretreated nude mice before human SSRBC infusion inhibited SSRBC adhesion in inflamed vessels, prevented the progression of vasoocclusion, and reduced SSRBC organ sequestration. By use of a more clinically relevant protocol, 0.3 or 1 mg/kg MEK1/2 inhibitor given to TNF-α-pretreated nude mice after human SSRBC infusion and onset of vasoocclusion reversed SSRBC adhesion and vasoocclusion and restored blood flow. In SCD mice, 0.025, 0.05, or 0.1 mg/kg MEK1/2 inhibitor also reversed leukocyte and erythrocyte adhesion after the inflammatory trigger of vasoocclusion and improved microcirculatory blood flow. Cell adhesion was reversed by shedding of endothelial E-selectin, P-selectin, and αvβ3 integrin, and leukocyte CD44 and β2 integrin. Thus, MEK1/2 inhibitors, by targeting the adhesive function of SSRBCs and leukocytes, could represent a valuable therapeutic intervention for acute sickle cell vasoocclusive crises.
Collapse
Affiliation(s)
- Yulin Zhao
- *Division of Hematology and Departments of Medicine and Radiation Oncology, Duke University, Durham, North Carolina, USA
| | - Evan A Schwartz
- *Division of Hematology and Departments of Medicine and Radiation Oncology, Duke University, Durham, North Carolina, USA
| | - Gregory M Palmer
- *Division of Hematology and Departments of Medicine and Radiation Oncology, Duke University, Durham, North Carolina, USA
| | - Rahima Zennadi
- *Division of Hematology and Departments of Medicine and Radiation Oncology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
44
|
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
|
45
|
Carbone F, Teixeira PC, Braunersreuther V, Mach F, Vuilleumier N, Montecucco F. Pathophysiology and Treatments of Oxidative Injury in Ischemic Stroke: Focus on the Phagocytic NADPH Oxidase 2. Antioxid Redox Signal 2015; 23:460-89. [PMID: 24635113 PMCID: PMC4545676 DOI: 10.1089/ars.2013.5778] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
SIGNIFICANCE Phagocytes play a key role in promoting the oxidative stress after ischemic stroke occurrence. The phagocytic NADPH oxidase (NOX) 2 is a membrane-bound enzyme complex involved in the antimicrobial respiratory burst and free radical production in these cells. RECENT ADVANCES Different oxidants have been shown to induce opposite effects on neuronal homeostasis after a stroke. However, several experimental models support the detrimental effects of NOX activity (especially the phagocytic isoform) on brain recovery after stroke. Therapeutic strategies selectively targeting the neurotoxic ROS and increasing neuroprotective oxidants have recently produced promising results. CRITICAL ISSUES NOX2 might promote carotid plaque rupture and stroke occurrence. In addition, NOX2-derived reactive oxygen species (ROS) released by resident and recruited phagocytes enhance cerebral ischemic injury, activating the inflammatory apoptotic pathways. The aim of this review is to update evidence on phagocyte-related oxidative stress, focusing on the role of NOX2 as a potential therapeutic target to reduce ROS-related cerebral injury after stroke. FUTURE DIRECTIONS Radical scavenger compounds (such as Ebselen and Edaravone) are under clinical investigation as a therapeutic approach against stroke. On the other hand, NOX inhibition might represent a promising strategy to prevent the stroke-related injury. Although selective NOX inhibitors are not yet available, nonselective compounds (such as apocynin and fasudil) provided encouraging results in preclinical studies. Whereas additional studies are needed to better evaluate this therapeutic potential in human beings, the development of specific NOX inhibitors (such as monoclonal antibodies, small-molecule inhibitors, or aptamers) might further improve brain recovery after stroke.
Collapse
Affiliation(s)
- Federico Carbone
- 1 Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva , Geneva, Switzerland .,2 Department of Internal Medicine, University of Genoa School of Medicine , IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Priscila Camillo Teixeira
- 3 Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals , Geneva, Switzerland
| | - Vincent Braunersreuther
- 1 Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva , Geneva, Switzerland
| | - François Mach
- 1 Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva , Geneva, Switzerland
| | - Nicolas Vuilleumier
- 3 Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals , Geneva, Switzerland
| | - Fabrizio Montecucco
- 1 Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva , Geneva, Switzerland .,2 Department of Internal Medicine, University of Genoa School of Medicine , IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy .,3 Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals , Geneva, Switzerland
| |
Collapse
|
46
|
Cuartero MI, Ballesteros I, Lizasoain I, Moro MA. Complexity of the cell-cell interactions in the innate immune response after cerebral ischemia. Brain Res 2015; 1623:53-62. [PMID: 25956207 DOI: 10.1016/j.brainres.2015.04.047] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/16/2015] [Accepted: 04/17/2015] [Indexed: 12/30/2022]
Abstract
In response to brain ischemia a cascade of signals leads to the activation of the brain innate immune system and to the recruitment of blood borne derived cells to the ischemic tissue. These processes have been increasingly shown to play a role on stroke pathogenesis. Here, we discuss the key features of resident microglia and different leukocyte subsets implicated in cerebral ischemia with special emphasis of neutrophils, monocytes and microglia. We focus on how leukocytes are recruited to injured brain through a complex interplay between endothelial cells, platelets and leukocytes and describe different strategies used to inhibit their recruitment. Finally, we discuss the possible existence of different leukocyte subsets in the ischemic tissue and the repercussion of different myeloid phenotypes on stroke outcome. The knowledge of the nature of these heterogeneous cell-cell interactions may open new lines of investigation on new therapies to promote protective immune responses and tissue repair after cerebral ischemia or to block harmful responses. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- María I Cuartero
- Unidad de Investigación Neurovascular, Depto. Farmacología, Facultad de Medicina, Universidad Complutense and Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Iván Ballesteros
- Unidad de Investigación Neurovascular, Depto. Farmacología, Facultad de Medicina, Universidad Complutense and Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Ignacio Lizasoain
- Unidad de Investigación Neurovascular, Depto. Farmacología, Facultad de Medicina, Universidad Complutense and Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - María A Moro
- Unidad de Investigación Neurovascular, Depto. Farmacología, Facultad de Medicina, Universidad Complutense and Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.
| |
Collapse
|
47
|
Molnar T, Peterfalvi A, Szereday L, Pusch G, Szapary L, Komoly S, Bogar L, Illes Z. Deficient leucocyte antisedimentation is related to post-stroke infections and outcome. J Clin Pathol 2015; 61:1209-13. [PMID: 18955576 DOI: 10.1136/jcp.2008.059840] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Patients with stroke are more susceptible to infections, suggesting possible deficiencies of early immune responses, particularly of leucocytes. AIMS To serially examine leucocyte antisedimentation rate (LAR), a simple test to detect activation of leucocytes, and correlate it with S100beta, procalcitonin and outcome in patients with acute ischaemic events. METHODS Venous blood samples were taken from 61 healthy volunteers and 49 patients with acute ischaemic events (acute ischaemic stroke (AIS), n = 38; transient ischaemic attack (TIA), n = 11) within 6 hours, at 24 and 72 hours after onset of symptoms. RESULTS LAR was significantly higher in acute ischaemic events compared to controls within 6 hours after onset of stroke regardless of post-stroke infections. In addition, the increase of LAR was delayed and attenuated in TIA in contrast to AIS. A deficiency in early increase of LAR was associated with post-stroke infections and a poor outcome, measured by the Glasgow Outcome Scale in AIS. There was a positive correlation between LAR and S100beta at 72 hours after the onset of ischaemic stroke. Increased levels of S100beta at 24 and 72 hours after stroke were associated with poor outcome. CONCLUSIONS An early activation of leucocytes indicated by an increase of LAR is characteristic of acute ischaemic cerebrovascular events. A delayed and ameliorated leucocyte activation represented by LAR is characteristic of TIA in contrast to stroke. Deficient early activation predisposes to post-stroke infections related to poor outcome. In addition, the extent of tissue injury correlates with the magnitude of innate immune responses.
Collapse
Affiliation(s)
- T Molnar
- Department of Anaesthesiology and Intensive Therapy, University of Pecs, Pecs, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Chen S, Chen Y, Xu L, Matei N, Tang J, Feng H, Zhang J. Venous system in acute brain injury: Mechanisms of pathophysiological change and function. Exp Neurol 2015; 272:4-10. [PMID: 25783658 DOI: 10.1016/j.expneurol.2015.03.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 03/09/2015] [Indexed: 01/31/2023]
Abstract
Cerebral vascular injury is a major component of acute brain injury. Currently, neuroprotective strategies primarily focus on the recanalization of cerebral arteries and capillaries, and the protection of insulted neurons. Hitherto, the role of vein drainage in the pathophysiology of acute brain injury has been overlooked, due to an under appreciation of the magnitude of the impact of veins in circulation. In this review, we summarize the changes in the vein morphology and functions that are known, or likely to occur related to acute brain injury, and aim to advance the therapeutic management of acute brain injury by shifting the focus from reperfusion to another term: recirculation. Recent progress in the neurobiological understanding of the vascular neural network has demonstrated that cerebral venous systems are able to respond to acute brain injury by regulating the blood flow disharmony following brain edema, blood brain barrier disruption, ischemia, and hemorrhage. With the evidence presented in this review, future clinical management of acutely brain injured patients will expand to include the recirculation concept, establishing a harmony between arterial and venous systems, in addition to the established recanalization and reperfusion strategies.
Collapse
Affiliation(s)
- Sheng Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Liang Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Nathanael Matei
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, USA
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - JohnH Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, USA
| |
Collapse
|
49
|
Benakis C, Garcia-Bonilla L, Iadecola C, Anrather J. The role of microglia and myeloid immune cells in acute cerebral ischemia. Front Cell Neurosci 2015; 8:461. [PMID: 25642168 PMCID: PMC4294142 DOI: 10.3389/fncel.2014.00461] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 12/18/2014] [Indexed: 01/15/2023] Open
Abstract
The immune response to acute cerebral ischemia is a major contributor to stroke pathobiology. The inflammatory response is characterized by the participation of brain resident cells and peripheral leukocytes. Microglia in the brain and monocytes/neutrophils in the periphery have a prominent role in initiating, sustaining and resolving post-ischemic inflammation. In this review we aim to summarize recent literature concerning the origins, fate and role of microglia, monocytes and neutrophils in models of cerebral ischemia and to discuss their relevance for human stroke.
Collapse
Affiliation(s)
- Corinne Benakis
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College New York, NY, USA
| | - Lidia Garcia-Bonilla
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College New York, NY, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College New York, NY, USA
| |
Collapse
|
50
|
Gros A, Ollivier V, Ho-Tin-Noé B. Platelets in inflammation: regulation of leukocyte activities and vascular repair. Front Immunol 2015; 5:678. [PMID: 25610439 PMCID: PMC4285099 DOI: 10.3389/fimmu.2014.00678] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/16/2014] [Indexed: 12/29/2022] Open
Abstract
There is now a large body of evidence that platelets are central actors of inflammatory reactions. Indeed, platelets play a significant role in a variety of inflammatory diseases. These diseases include conditions as varied as atherosclerosis, arthritis, dermatitis, glomerulonephritis, or acute lung injury. In this context, one can note that inflammation is a convenient but imprecise catch-all term that is used to cover a wide range of situations. Therefore, when discussing the role of platelets in inflammation, it is important to clearly define the pathophysiological context and the exact stage of the reaction. Inflammatory reactions are indeed multistep processes that can be either acute or chronic, and their sequence can vary greatly depending on the situation and organ concerned. Here, we focus on how platelets contribute to inflammatory reactions involving recruitment of neutrophils and/or macrophages. Specifically, we review past and recent data showing that platelets intervene at various stages of these reactions to regulate parameters such as endothelial permeability, the recruitment of neutrophils and macrophages and their effector functions, as well as inflammatory bleeding. The mechanisms underlying these various modulating effect of platelets are also discussed.
Collapse
Affiliation(s)
- Angèle Gros
- Université Paris Diderot, Sorbonne Paris Cité , Paris , France ; Unit 1148, Laboratory for Vascular Translational Science, INSERM , Paris , France
| | - Véronique Ollivier
- Université Paris Diderot, Sorbonne Paris Cité , Paris , France ; Unit 1148, Laboratory for Vascular Translational Science, INSERM , Paris , France
| | - Benoît Ho-Tin-Noé
- Université Paris Diderot, Sorbonne Paris Cité , Paris , France ; Unit 1148, Laboratory for Vascular Translational Science, INSERM , Paris , France
| |
Collapse
|