1
|
Yang H, You L, Wang Z, Yang L, Wang X, Wu W, Zhi H, Rong G, Sheng Y, Liu X, Liu L. Bile duct ligation elevates 5-HT levels in cerebral cortex of rats partly due to impairment of brain UGT1A6 expression and activity via ammonia accumulation. Redox Biol 2024; 69:103019. [PMID: 38163420 PMCID: PMC10794929 DOI: 10.1016/j.redox.2023.103019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/19/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024] Open
Abstract
Hepatic encephalopathy (HE) is often associated with endogenous serotonin (5-HT) disorders. However, the reason for elevated brain 5-HT levels due to liver failure remains unclear. This study aimed to investigate the mechanism by which liver failure increases brain 5-HT levels and the role in behavioral abnormalities in HE. Using bile duct ligation (BDL) rats as a HE model, we verified the elevated 5-HT levels in the cortex but not in the hippocampus and striatum, and found that this cortical 5-HT overload may be caused by BDL-mediated inhibition of UDP-glucuronosyltransferase 1A6 (UGT1A6) expression and activity in the cortex. The intraventricular injection of the UGT1A6 inhibitor diclofenac into rats demonstrated that the inhibition of brain UGT1A6 activity significantly increased cerebral 5-HT levels and induced HE-like behaviors. Co-immunofluorescence experiments demonstrated that UGT1A6 is primarily expressed in astrocytes. In vitro studies confirmed that NH4Cl activates the ROS-ERK pathway to downregulate UGT1A6 activity and expression in U251 cells, which can be reversed by the oxidative stress antagonist N-acetyl-l-cysteine and the ERK inhibitor U0126. Silencing Hepatocyte Nuclear Factor 4α (HNF4α) suppressed UGT1A6 expression whilst overexpressing HNF4α increased Ugt1a6 promotor activity. Meanwhile, both NH4Cl and the ERK activator TBHQ downregulated HNF4α and UGT1A6 expression. In the cortex of hyperammonemic rats, we also found activation of the ROS-ERK pathway, decreases in HNF4α and UGT1A6 expression, and increases in brain 5-HT content. These results prove that the ammonia-mediated ROS-ERK pathway activation inhibits HNF4α expression to downregulate UGT1A6 expression and activity, thereby increasing cerebral 5-HT content and inducing manic-like HE symptoms. This is the first study to reveal the mechanism of elevated cortical 5-HT concentration in a state of liver failure and elucidate its association with manic-like behaviors in HE.
Collapse
Affiliation(s)
- Hanyu Yang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China; Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Linjun You
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, 210009, Nanjing, China
| | - Zhongyan Wang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Lu Yang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xun Wang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Wenhan Wu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hao Zhi
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Guangmei Rong
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yun Sheng
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiaodong Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China; Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Li Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China; Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
2
|
Rolfes L, Riek-Burchardt M, Pawlitzki M, Minnerup J, Bock S, Schmidt M, Meuth SG, Gunzer M, Neumann J. Neutrophil granulocytes promote flow stagnation due to dynamic capillary stalls following experimental stroke. Brain Behav Immun 2021; 93:322-330. [PMID: 33486002 DOI: 10.1016/j.bbi.2021.01.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/01/2022] Open
Abstract
Flow stagnation of peri-ischemic capillaries due to dynamic leukocyte stalls has been described to be a contributor to ongoing penumbral injury in transient brain ischemia, but has not been investigated in permanent experimental stroke so far. Moreover, it is discussed that obstructing neutrophils are involved in this process; however, their contribution has not yet been proven. Here, we characterize the dynamics of neutrophil granulocytes in two models of permanent stroke (photothrombosis and permanent middle cerebral artery occlusion) using intravital two-photon fluorescence microscopy. Different to previous studies on LysM-eGFP+ cells we additionally apply a transgenic mouse model with tdTomato-expressing neutrophils to avoid interference from additional immune cell subsets. We identify repetitively occurring capillary stalls of varying duration promoted by neutrophils in both models of permanent cerebral ischemia, validating the suitability of our new transgenic mouse model in determining neutrophil occlusion formation in vivo. Flow cytometric analysis of peripheral blood (PB) and brain tissue from mice subjected to photothrombosis reveal an increase in the total proportion of neutrophils, with selective upregulation of endothelial adherence markers in the PB. In conclusion, the dynamic microcirculatory stall phenomenon that is described after transient ischemia followed by reperfusion also occurs after permanent small- or large-vessel stroke and is clearly attributable to neutrophils.
Collapse
Affiliation(s)
- Leoni Rolfes
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Germany.
| | | | - Marc Pawlitzki
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Germany; Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.
| | - Jens Minnerup
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Germany.
| | - Stefanie Bock
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Germany.
| | - Mariella Schmidt
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Germany.
| | - Sven G Meuth
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Germany; Department of Neurology, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Germany.
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital Essen, Germany.
| | - Jens Neumann
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
3
|
Ramaglia V, Florescu A, Zuo M, Sheikh-Mohamed S, Gommerman JL. Stromal Cell–Mediated Coordination of Immune Cell Recruitment, Retention, and Function in Brain-Adjacent Regions. THE JOURNAL OF IMMUNOLOGY 2021; 206:282-291. [DOI: 10.4049/jimmunol.2000833] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/27/2020] [Indexed: 12/15/2022]
|
4
|
Mastorakos P, McGavern D. The anatomy and immunology of vasculature in the central nervous system. Sci Immunol 2020; 4:4/37/eaav0492. [PMID: 31300479 DOI: 10.1126/sciimmunol.aav0492] [Citation(s) in RCA: 183] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 06/13/2019] [Indexed: 12/21/2022]
Abstract
Barriers between circulation and the central nervous system (CNS) play a key role in the development and modulation of CNS immune responses. Structural variations in the vasculature traversing different anatomical regions within the CNS strongly influence where and how CNS immune responses first develop. Here, we provide an overview of cerebrovascular anatomy, focusing on the blood-CNS interface and how anatomical variations influence steady-state immunology in the compartment. We then discuss how CNS vasculature is affected by and influences the development of different pathophysiological states, such as CNS autoimmune disease, cerebrovascular injury, cerebral ischemia, and infection.
Collapse
Affiliation(s)
- Panagiotis Mastorakos
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Dorian McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
5
|
Orion D, von Landenberg P, Itsekson-Hayosh Z, Schwammenthal Y, Tsabari R, Merzeliak O, Chapman J, Tanne D. Plasma myeloperoxidase levels in acute brain ischaemia and high grade carotid stenosis. Eur J Neurol 2020; 27:1604-1611. [PMID: 32335972 DOI: 10.1111/ene.14279] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 04/15/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND PURPOSE Myeloperoxidase (MPO) is an important oxidative enzyme participating in different stages of cardiovascular disease and predicts prognosis. Little is known about its role in acute cerebrovascular events and carotid plaque vulnerability. In this study, the aim was to assess plasma MPO levels in acute stroke patients and their correlation to stroke severity and stroke outcome. METHODS Plasma MPO levels were assessed in patients presenting with acute brain ischaemia within 36 h of symptom onset (n = 144, mean age 64.7 ± 11.6 years, 67% men) and in patients with moderate-to-severe carotid stenosis undergoing carotid artery stenting (n = 51, mean age 66.3 ± 8.4 years, 75% men). Patients presenting with acute brain ischaemia were assessed serially for stroke severity and disability. RESULTS Plasma MPO concentrations (ng/ml) were associated with interleukin-6 (r = 0.38, P < 0.0001) and gender (median interquartile range) of 68.6 (49.8-107.0) vs. 59.7 (42.7-85.5) in women vs. men (P = 0.02). In acute brain ischaemia, MPO concentrations were associated with non-lacunar subtype (bottom, middle and top tertiles 37.5%, 71.7% and 71.7% respectively; P = 0.001), with stroke severity (baseline National Institutes of Health Stroke Scale score > 10, bottom, middle and top tertiles 6.3%, vs. 41.7% and 31.3%, respectively; P < 0.006) as well as with stroke severity at days 1-2, days 4-5 and at discharge (P < 0.05 for all), but less with disability at discharge (modified Rankin Scale score ≥ 2, 41.7% vs. 60.4% and 58.7% for the bottom, middle and top tertiles, respectively; P = 0.096). CONCLUSIONS Amongst patients with acute brain ischaemia, plasma MPO concentrations were associated with stroke severity and non-lacunar subtype, but not with long-term functional disability.
Collapse
Affiliation(s)
- D Orion
- Department of Neurology and Sagol Neuroscience Center, Stroke Center, Sheba Medical Center, Tel Hashomer, Israel
| | - P von Landenberg
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg Universität Mainz Klinikum, Mainz, Germany
| | - Z Itsekson-Hayosh
- Department of Neurology and Sagol Neuroscience Center, Stroke Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Y Schwammenthal
- Department of Neurology and Sagol Neuroscience Center, Stroke Center, Sheba Medical Center, Tel Hashomer, Israel
| | - R Tsabari
- Department of Neurology and Sagol Neuroscience Center, Stroke Center, Sheba Medical Center, Tel Hashomer, Israel
| | - O Merzeliak
- Department of Neurology and Sagol Neuroscience Center, Stroke Center, Sheba Medical Center, Tel Hashomer, Israel
| | - J Chapman
- Department of Neurology and Sagol Neuroscience Center, Stroke Center, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - D Tanne
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Stroke and Cognition Institute, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
6
|
Nakamura A, Otani K, Shichita T. Lipid mediators and sterile inflammation in ischemic stroke. Int Immunol 2020; 32:719-725. [DOI: 10.1093/intimm/dxaa027] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/16/2020] [Indexed: 12/18/2022] Open
Abstract
Abstract
Stroke is one of the major causes of lethality and disability, yet few effective therapies have been established for ischemic stroke. Inflammation in the ischemic brain is induced by the infiltration and subsequent activation of immune cells. Loss of cerebral blood flow and ischemic brain-cell death trigger the activation of infiltrating immune cells and drastic changes in the lipid content of the ischemic brain. In particular, polyunsaturated fatty acids and their metabolites regulate cerebral post-ischemic inflammation and ischemic stroke pathologies. In this review, we discuss the relationships between the lipid mediators and cerebral post-ischemic inflammation and their relevance to possible future therapeutic strategies targeting lipid mediators for ischemic stroke.
Collapse
Affiliation(s)
- Akari Nakamura
- Stroke Renaissance Project, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa, Setagaya-ku, Tokyo, Japan
- Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kento Otani
- Stroke Renaissance Project, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa, Setagaya-ku, Tokyo, Japan
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo, Japan
| | - Takashi Shichita
- Stroke Renaissance Project, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa, Setagaya-ku, Tokyo, Japan
- Precursory Research for Innovative Medical Care (PRIME), Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| |
Collapse
|
7
|
Gad SN, Nofal S, Raafat EM, Ahmed AAE. Lixisenatide Reduced Damage in Hippocampus CA1 Neurons in a Rat Model of Cerebral Ischemia-Reperfusion Possibly Via the ERK/P38 Signaling Pathway. J Mol Neurosci 2020; 70:1026-1037. [PMID: 32040827 DOI: 10.1007/s12031-020-01497-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 02/05/2020] [Indexed: 12/11/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) is a gut-derived peptide that has various physiological actions. One of its main actions is the regulation of blood glucose level when it is elevated as it potentiates insulin release. It is also known that GLP-1 protects neurons from damage caused by neurodegenerative diseases. Lixisenatide is one of the GLP-1 analogues that has a strong affinity to the GLP-1 receptor. Experimental animal studies have shown that it holds a neuroprotective effect in Parkinson, myocardial, and cerebral ischemic disease animal models. The beneficial effect of lixisenatide on the brain after cerebral ischemia-reperfusion (I/R) is not clarified yet; thus, it needs further explanatory studies. Our research is the first to study the effect of lixisenatide on myeloperoxidase (MPO) and toll-like receptors (TLRs)/mitogen-activated protein kinase (MAPK) pathway in a rat model of cerebral I/R. Lixisenatide with 2 doses 0.7 and 7 nmol/kg was given intraperitoneal in 2 different groups for 14 days; then, the bilateral common carotid artery was occluded for 1 h followed by reperfusion for 1 h. Examination of hippocampus CA1 neurons by Nissl stain showed that the number of intact neurons was elevated in the lixisenatide-treated group related to the control group (I/R group). Lixisenatide exhibited neuroprotection action possibly via downregulation of MPO, TLR2/4, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and pP38 and upregulation of phosphorylated extracellular signal-regulated kinase (pERK1/2); thus, this study gives possible link between lixisenatide and TLR/MAPK pathway following cerebral I/R and supports the use of lixisenatide for neuroprotection against stroke.
Collapse
Affiliation(s)
- Salma N Gad
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt.
| | - Shahira Nofal
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| | - Eman M Raafat
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| | - Amany A E Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| |
Collapse
|
8
|
Amtul Z, Frías C, Randhawa J, Hill DJ, Arany EJ. The spatial cerebral damage caused by larger infarct and β-amyloid toxicity is driven by the anatomical/functional connectivity. J Comp Neurol 2020; 528:48-60. [PMID: 31265125 DOI: 10.1002/cne.24738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/04/2019] [Accepted: 06/18/2019] [Indexed: 11/07/2022]
Abstract
Large cerebral infarctions are major predictors of death and severe disability from stroke. Conversely, data concerning these types of infarctions and the affected adjacent brain circuits are scarce. It remains to be determined if the co-morbid concurrence of large infarct and β-amyloid (Aβ) toxicity can precipitate the early development of dementia. Here, we described a dose-dependent effect of a unilateral striatal injection of vasoconstrictive endothelin-1 (ET-1) along with Aβ toxicity on CNS pathogenesis; driven by the anatomical and functional networks within a brain circuit. After 21 days of treatment, a high dose (60 pmol) of ET-1 (E60) alone caused the greatest increase in neuroinflammation, mainly in the ipsilateral striatum and distant regions with synaptic links to the striatal lesion such as white matter (subcortical white matter, corpus callosum, internal capsule, anterior commissure), gray matter (globus pallidus, thalamus), and cortices (cingulate, motor, somatosensory, entorhinal). The combined E60 + Aβ treatment also extended perturbation in the contralateral hemisphere of these rats, such as increased deposition of amyloid precursor protein fragments associated with the appearance of degenerating cells and the leakage of laminin from the basement membrane across a compromised blood-brain barrier. However, the cerebral damage induced by the 6 pmol ET-1 (E6), Aβ and E6 + Aβ rats was not detrimental enough to injure the complete network. The appreciation of the causal interactions among distinct anatomical units in the brain after ischemia and Aβ toxicity will help in the design of effective and alternative therapeutics that may disassociate the synergistic or additive association between the infarcts and Aβ toxicity.
Collapse
Affiliation(s)
- Zareen Amtul
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada
| | - Carmen Frías
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada
| | - Jasmine Randhawa
- Department of Biology, University of Western Ontario, London, Ontario, Canada
| | - David J Hill
- Department of Medicine, Physiology, and Pharmacology, and Pediatrics, University of Western Ontario, London, Ontario, Canada.,Lawson Health Research Institute, London, Ontario, Canada
| | - Edith J Arany
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
9
|
Pivotal role of innate myeloid cells in cerebral post-ischemic sterile inflammation. Semin Immunopathol 2018; 40:523-538. [PMID: 30206661 DOI: 10.1007/s00281-018-0707-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/04/2018] [Indexed: 12/17/2022]
Abstract
Inflammatory responses play a multifaceted role in regulating both disability and recovery after ischemic brain injury. In the acute phase of ischemic stroke, resident microglia elicit rapid inflammatory responses by the ischemic milieu. After disruption of the blood-brain barrier, peripheral-derived neutrophils and mononuclear phagocytes infiltrate into the ischemic brain. These infiltrating myeloid cells are activated by the endogenous alarming molecules released from dying brain cells. Inflammation after ischemic stroke thus typically consists of sterile inflammation triggered by innate immunity, which exacerbates the pathologies of ischemic stroke and worsens neurological prognosis. Infiltrating immune cells sustain the post-ischemic inflammation for several days; after this period, however, these cells take on a repairing function, phagocytosing inflammatory mediators and cellular debris. This time-specific polarization of immune cells in the ischemic brain is a potential novel therapeutic target. In this review, we summarize the current understanding of the phase-dependent role of innate myeloid cells in ischemic stroke and discuss the cellular and molecular mechanisms of their inflammatory or repairing polarization from a therapeutic perspective.
Collapse
|
10
|
Enzmann G, Kargaran S, Engelhardt B. Ischemia-reperfusion injury in stroke: impact of the brain barriers and brain immune privilege on neutrophil function. Ther Adv Neurol Disord 2018; 11:1756286418794184. [PMID: 30181779 PMCID: PMC6111395 DOI: 10.1177/1756286418794184] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/29/2018] [Indexed: 02/06/2023] Open
Abstract
Reperfusion injury following ischemic stroke is a complex pathophysiological process involving numerous mechanisms ranging from the release of excitatory amino acids and ion disequilibrium to the induction of apoptosis and necrosis, to oxidative stress and inflammation. The migration of neutrophils into the brain parenchyma and release of their abundant proteases are generally considered the main cause of neuronal cell death and acute reperfusion injury following ischemic stroke. Recent findings in experimental and human stroke have challenged this view, as the majority of neutrophils were rather found to accumulate within the neurovascular unit (NVU) and the subarachnoid space (SAS) where they remain separated from the brain parenchyma by the glia limitans. The brain parenchyma is an immune-privileged site that is not readily accessible to immune cells and does not elicit stereotypic adaptive or innate immune responses. Understanding brain immune privilege requires intimate knowledge of its unique anatomy in which the brain barriers, that include the glia limitans, establish compartments that differ remarkably with regard to their accessibility to the immune system. We here propose that the brain immune privilege also extends to an ischemic insult, where the brain parenchyma does not evoke a rapid infiltration of neutrophils as observed in ischemic events in peripheral organs. Rather, neutrophil accumulation in the NVU and SAS could have a potential impact on cerebrospinal fluid (CSF) drainage from the central nervous system (CNS) and thus on edema formation and reperfusion injury after ischemic stroke. Integrating the anatomical and functional implications of the brain immune privilege with the unquestionable role of neutrophils in reperfusion injury is a prerequisite to exploit appropriate strategies for therapeutic interventions aiming to reduce neuronal cell death after ischemic stroke.
Collapse
Affiliation(s)
- Gaby Enzmann
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, Bern 3012, Switzerland
| | - Soghra Kargaran
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, Bern 3012, Switzerland
| |
Collapse
|
11
|
Targeting antioxidant enzyme expression as a therapeutic strategy for ischemic stroke. Neurochem Int 2016; 107:23-32. [PMID: 28043837 DOI: 10.1016/j.neuint.2016.12.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/05/2016] [Accepted: 12/17/2016] [Indexed: 11/24/2022]
Abstract
During ischemic stroke, neurons and glia are subjected to damage during the acute and neuroinflammatory phases of injury. Production of reactive oxygen species (ROS) from calcium dysregulation in neural cells and the invasion of activated immune cells are responsible for stroke-induced neurodegeneration. Scientists have failed thus far to identify antioxidant-based drugs that can enhance neural cell survival and improve recovery after stroke. However, several groups have demonstrated success in protecting against stroke by increasing expression of antioxidant enzymes in neural cells. These enzymes, which include but are not limited to enzymes in the glutathione peroxidase, catalase, and superoxide dismutase families, degrade ROS that otherwise damage cellular components such as DNA, proteins, and lipids. Several groups have identified cellular therapies including neural stem cells and human umbilical cord blood cells, which exert neuroprotective and oligoprotective effects through the release of pro-survival factors that activate PI3K/Akt signaling to upregulation of antioxidant enzymes. Other studies demonstrate that treatment with soluble factors released by these cells yield similar changes in enzyme expression after stroke. Treatment with the cytokine leukemia inhibitory factor increases the expression of peroxiredoxin IV and metallothionein III in glia and boosts expression of superoxide dismutase 3 in neurons. Through cell-specific upregulation of these enzymes, LIF and other Akt-inducing factors have the potential to protect multiple cell types against damage from ROS during the early and late phases of ischemic damage.
Collapse
|
12
|
Amantea D, Certo M, Petrelli F, Bagetta G. Neuroprotective Properties of a Macrolide Antibiotic in a Mouse Model of Middle Cerebral Artery Occlusion: Characterization of the Immunomodulatory Effects and Validation of the Efficacy of Intravenous Administration. Assay Drug Dev Technol 2016; 14:298-307. [PMID: 27392039 DOI: 10.1089/adt.2016.728] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Repurposing the macrolide antibiotic azithromycin has recently been suggested as a promising neuroprotective strategy for the acute treatment of ischemic stroke. Here, we aim at further characterizing the immunomodulatory properties of intraperitoneal (i.p.) administration of this drug and, more importantly, at assessing whether neuroprotection can also be achieved by the more clinically relevant intravenous (i.v.) route of administration in a mouse model of focal cerebral ischemia induced by transient (30-min) middle cerebral artery occlusion (MCAo). A single i.p. injection of azithromycin (150 mg/kg) upon reperfusion prevented ischemia-induced spleen contraction and increased the number of MAC-1-immunopositive microglia/macrophages in the ischemic hemisphere 48 h after the insult. This was paralleled by an elevation of alternatively activated phenotypes (i.e., Ym1-immunopositive M2-polarized cells) and by a reduced expression of the pro-inflammatory marker myeloperoxidase. More importantly, i.v. administration of azithromycin upon reperfusion reduced MCAo-induced infarct volume and cerebral edema to an extent comparable to that obtained via the i.p. route. Although the i.p. route is often used for research purposes, it is impractical in the clinical setting; however, i.v. administration can easily be used in ischemic stroke patients who usually have i.v. access already established on hospital admission. The neuroprotective efficacy of the clinically relevant i.v. administration of azithromycin, together with its beneficial immunomodulatory properties reported in mice subjected to transient MCAo, suggests that this macrolide antibiotic can be effectively repurposed for the acute treatment of ischemic stroke. To this end, further work is needed to validate the efficacy of azithromycin in the clinical setting.
Collapse
Affiliation(s)
- Diana Amantea
- 1 Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria , Rende (CS), Italy
| | - Michelangelo Certo
- 1 Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria , Rende (CS), Italy
| | - Francesco Petrelli
- 1 Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria , Rende (CS), Italy
| | - Giacinto Bagetta
- 1 Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria , Rende (CS), Italy .,2 University Consortium for Adaptive Disorders and Head Pain (UCADH), Section of Neuropharmacology of Normal and Pathological Neuronal Plasticity, University of Calabria , Rende, Italy
| |
Collapse
|
13
|
Herz J, Sabellek P, Lane TE, Gunzer M, Hermann DM, Doeppner TR. Role of Neutrophils in Exacerbation of Brain Injury After Focal Cerebral Ischemia in Hyperlipidemic Mice. Stroke 2015; 46:2916-25. [PMID: 26337969 DOI: 10.1161/strokeaha.115.010620] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 07/16/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE Inflammation-related comorbidities contribute to stroke-induced immune responses and brain damage. We previously showed that hyperlipidemia exacerbates ischemic brain injury, which is associated with elevated peripheral and cerebral granulocyte numbers. Herein, we evaluate the contribution of neutrophils to the exacerbation of ischemic brain injury. METHODS Wild-type mice fed with a normal chow and ApoE knockout mice fed with a high cholesterol diet were exposed to middle cerebral artery occlusion. CXCR2 was blocked using the selective antagonist SB225002 (2 mg/kg) or neutralizing CXCR2 antiserum. Neutrophils were depleted using an anti-Ly6G antibody. At 72 hours post ischemia, immunohistochemistry, flow cytometry, and real-time polymerase chain reaction were performed to determine cerebral tissue injury and immunologic changes in the blood, bone marrow, and brain. Functional outcome was assessed by accelerated rota rod and tight rope tests at 4, 7, and 14 days post ischemia. RESULTS CXCR2 antagonization reduced neurological deficits and infarct volumes that were exacerbated in hyperlipidemic ApoE-/- mice. This effect was mimicked by neutrophil depletion. Cerebral neutrophil infiltration and peripheral neutrophilia, which were increased on ischemia in hyperlipidemia, were attenuated by CXCR2 antagonization. This downscaling of neutrophil responses was associated with increased neutrophil apoptosis and reduced levels of CXCR2, inducible nitric oxide synthase, and NADPH oxidase 2 expression on bone marrow neutrophils. CONCLUSIONS Our data demonstrate a role of neutrophils in the exacerbation of ischemic brain injury induced by hyperlipidemia. Accordingly, CXCR2 blockade, which prevents neutrophil recruitment into the brain, might be an effective option for stroke treatment in patients with hyperlipidemia.
Collapse
Affiliation(s)
- Josephine Herz
- From the Department of Neurology (J.H., P.S., D.M.H., T.R.D.), Department of Pediatrics I (J.H.), and Institute of Experimental Immunology and Imaging, University Duisburg-Essen (M.G.), University Hospital Essen, Essen, Germany; and Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City (T.E.L.).
| | - Pascal Sabellek
- From the Department of Neurology (J.H., P.S., D.M.H., T.R.D.), Department of Pediatrics I (J.H.), and Institute of Experimental Immunology and Imaging, University Duisburg-Essen (M.G.), University Hospital Essen, Essen, Germany; and Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City (T.E.L.)
| | - Thomas E Lane
- From the Department of Neurology (J.H., P.S., D.M.H., T.R.D.), Department of Pediatrics I (J.H.), and Institute of Experimental Immunology and Imaging, University Duisburg-Essen (M.G.), University Hospital Essen, Essen, Germany; and Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City (T.E.L.)
| | - Matthias Gunzer
- From the Department of Neurology (J.H., P.S., D.M.H., T.R.D.), Department of Pediatrics I (J.H.), and Institute of Experimental Immunology and Imaging, University Duisburg-Essen (M.G.), University Hospital Essen, Essen, Germany; and Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City (T.E.L.)
| | - Dirk M Hermann
- From the Department of Neurology (J.H., P.S., D.M.H., T.R.D.), Department of Pediatrics I (J.H.), and Institute of Experimental Immunology and Imaging, University Duisburg-Essen (M.G.), University Hospital Essen, Essen, Germany; and Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City (T.E.L.)
| | - Thorsten R Doeppner
- From the Department of Neurology (J.H., P.S., D.M.H., T.R.D.), Department of Pediatrics I (J.H.), and Institute of Experimental Immunology and Imaging, University Duisburg-Essen (M.G.), University Hospital Essen, Essen, Germany; and Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City (T.E.L.)
| |
Collapse
|
14
|
Maysami S, Haley MJ, Gorenkova N, Krishnan S, McColl BW, Lawrence CB. Prolonged diet-induced obesity in mice modifies the inflammatory response and leads to worse outcome after stroke. J Neuroinflammation 2015; 12:140. [PMID: 26239227 PMCID: PMC4524371 DOI: 10.1186/s12974-015-0359-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 07/03/2015] [Indexed: 12/05/2022] Open
Abstract
Background Obesity increases the risk for ischaemic stroke and is associated with worse outcome clinically and experimentally. Most experimental studies have used genetic models of obesity. Here, a more clinically relevant model, diet-induced obesity, was used to study the impact of obesity over time on the outcome and inflammatory response after stroke. Methods Male C57BL/6 mice were maintained on a high-fat (60 % fat) or control (12 % fat) diet for 2, 3, 4 and 6 months when experimental stroke was induced by transient occlusion of the middle cerebral artery (MCAo) for either 20 (6-month diet) or 30 min (2-, 3-, 4- and 6-month diet). Ischaemic damage, blood–brain barrier (BBB) integrity, neutrophil number and chemokine expression in the brain were assessed at 24 h. Plasma chemokine levels (at 4 and 24 h) and neutrophil number in the liver (at 24 h) were measured. Physiological parameters (body weight and blood glucose) were measured in naïve control- and high-fat-fed mice at all time points and blood pressure at 3 and 6 months. Blood cell counts were also assessed in naïve 6-month control- and high-fat-fed mice. Results Mice fed a high-fat diet for 6 months had greater body weight, blood glucose and white and red blood cell count but no change in systolic blood pressure. After 4 and 6 months of high-fat feeding, and in the latter group with a 30-min (but not 20-min) occlusion of the MCA, obese mice had greater ischaemic brain damage. An increase in blood–brain barrier permeability, chemokine expression (CXCL-1 and CCL3), neutrophil number and microglia/macrophage cells was observed in the brains of 6-month high-fat-fed mice after 30-min MCAo. In response to stroke, chemokine (CXCL-1) expression in the plasma and liver was significantly different in obese mice (6-month high-fat fed), and a greater number of neutrophils were detected in the liver of control but not obese mice. Conclusions The detrimental effects of diet-induced obesity on stroke were therefore dependent on the severity of obesity and length of ischaemic challenge. The altered inflammatory response in obese mice may play a key role in its negative impact on stroke.
Collapse
Affiliation(s)
- Samaneh Maysami
- Faculty of Life Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.,Faculty of Medical and Human Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Michael J Haley
- Faculty of Life Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Natalia Gorenkova
- Faculty of Life Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Siddharth Krishnan
- Faculty of Life Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Barry W McColl
- The Roslin Institute and R(D)SVS, University of Edinburgh Easter Bush, Midlothian, EH25 9RG, UK
| | - Catherine B Lawrence
- Faculty of Life Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
15
|
Neumann J, Riek-Burchardt M, Herz J, Doeppner TR, König R, Hütten H, Etemire E, Männ L, Klingberg A, Fischer T, Görtler MW, Heinze HJ, Reichardt P, Schraven B, Hermann DM, Reymann KG, Gunzer M. Very-late-antigen-4 (VLA-4)-mediated brain invasion by neutrophils leads to interactions with microglia, increased ischemic injury and impaired behavior in experimental stroke. Acta Neuropathol 2015; 129:259-77. [PMID: 25391494 DOI: 10.1007/s00401-014-1355-2] [Citation(s) in RCA: 205] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 10/08/2014] [Accepted: 10/08/2014] [Indexed: 12/17/2022]
Abstract
Neuronal injury from ischemic stroke is aggravated by invading peripheral immune cells. Early infiltrates of neutrophil granulocytes and T-cells influence the outcome of stroke. So far, however, neither the timing nor the cellular dynamics of neutrophil entry, its consequences for the invaded brain area, or the relative importance of T-cells has been extensively studied in an intravital setting. Here, we have used intravital two-photon microscopy to document neutrophils and brain-resident microglia in mice after induction of experimental stroke. We demonstrated that neutrophils immediately rolled, firmly adhered, and transmigrated at sites of endothelial activation in stroke-affected brain areas. The ensuing neutrophil invasion was associated with local blood-brain barrier breakdown and infarct formation. Brain-resident microglia recognized both endothelial damage and neutrophil invasion. In a cooperative manner, they formed cytoplasmic processes to physically shield activated endothelia and trap infiltrating neutrophils. Interestingly, the systemic blockade of very-late-antigen-4 immediately and very effectively inhibited the endothelial interaction and brain entry of neutrophils. This treatment thereby strongly reduced the ischemic tissue injury and effectively protected the mice from stroke-associated behavioral impairment. Behavioral preservation was also equally well achieved with the antibody-mediated depletion of myeloid cells or specifically neutrophils. In contrast, T-cell depletion more effectively reduced the infarct volume without improving the behavioral performance. Thus, neutrophil invasion of the ischemic brain is rapid, massive, and a key mediator of functional impairment, while peripheral T-cells promote brain damage. Acutely depleting T-cells and inhibiting brain infiltration of neutrophils might, therefore, be a powerful early stroke treatment.
Collapse
|
16
|
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
|
17
|
Semple BD, Trivedi A, Gimlin K, Noble-Haeusslein LJ. Neutrophil elastase mediates acute pathogenesis and is a determinant of long-term behavioral recovery after traumatic injury to the immature brain. Neurobiol Dis 2014; 74:263-80. [PMID: 25497734 DOI: 10.1016/j.nbd.2014.12.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 11/19/2014] [Accepted: 12/01/2014] [Indexed: 12/21/2022] Open
Abstract
While neutrophil elastase (NE), released by activated neutrophils, is a key mediator of secondary pathogenesis in adult models of brain ischemia and spinal cord injury, no studies to date have examined this protease in the context of the injured immature brain, where there is notable vulnerability resulting from inadequate antioxidant reserves and prolonged exposure to infiltrating neutrophils. We thus reasoned that NE may be a key determinant of secondary pathogenesis, and as such, adversely influence long-term neurological recovery. To address this hypothesis, wild-type (WT) and NE knockout (KO) mice were subjected to a controlled cortical impact at post-natal day 21, approximating a toddler-aged child. To determine if NE is required for neutrophil infiltration into the injured brain, and whether this protease contributes to vasogenic edema, we quantified neutrophil numbers and measured water content in the brains of each of these genotypes. While leukocyte trafficking was indistinguishable between genotypes, vasogenic edema was markedly attenuated in the NE KO. To determine if early pathogenesis is dependent on NE, indices of cell death (TUNEL and activated caspase-3) were quantified across genotypes. NE KO mice showed a reduction in these markers of cell death in the injured hippocampus, which corresponded to greater preservation of neuronal integrity as well as reduced expression of heme oxygenase-1, a marker of oxidative stress. WT mice, treated with a competitive inhibitor of NE at 2, 6 and 12h post-injury, likewise showed a reduction in cell death and oxidative stress compared to vehicle-treated controls. We next examined the long-term behavioral and structural consequences of NE deficiency. NE KO mice showed an improvement in long-term spatial memory retention and amelioration of injury-induced hyperactivity. However, volumetric and stereological analyses found comparable tissue loss in the injured cortex and hippocampus independent of genotype. Further, WT mice treated acutely with the NE inhibitor showed no long-term behavioral or structural improvements. Together, these findings validate the central role of NE in both acute pathogenesis and chronic functional recovery, and support future exploration of the therapeutic window, taking into account the prolonged period of neutrophil trafficking into the injured immature brain.
Collapse
Affiliation(s)
- Bridgette D Semple
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA; Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Parkville, VIC 3000, Australia.
| | - Alpa Trivedi
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Kayleen Gimlin
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA.
| | - Linda J Noble-Haeusslein
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA; Department of Physical Therapy and Rehabilitation Sciences, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
18
|
Famakin BM. The Immune Response to Acute Focal Cerebral Ischemia and Associated Post-stroke Immunodepression: A Focused Review. Aging Dis 2014; 5:307-26. [PMID: 25276490 DOI: 10.14336/ad.2014.0500307] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 07/07/2014] [Accepted: 07/08/2014] [Indexed: 12/20/2022] Open
Abstract
It is currently well established that the immune system is activated in response to transient or focal cerebral ischemia. This acute immune activation occurs in response to damage, and injury, to components of the neurovascular unit and is mediated by the innate and adaptive arms of the immune response. The initial immune activation is rapid, occurs via the innate immune response and leads to inflammation. The inflammatory mediators produced during the innate immune response in turn lead to recruitment of inflammatory cells and the production of more inflammatory mediators that result in activation of the adaptive immune response. Under ideal conditions, this inflammation gives way to tissue repair and attempts at regeneration. However, for reasons that are just being understood, immunosuppression occurs following acute stroke leading to post-stroke immunodepression. This review focuses on the current state of knowledge regarding innate and adaptive immune activation in response to focal cerebral ischemia as well as the immunodepression that can occur following stroke. A better understanding of the intricate and complex events that take place following immune response activation, to acute cerebral ischemia, is imperative for the development of effective novel immunomodulatory therapies for the treatment of acute stroke.
Collapse
Affiliation(s)
- Bolanle M Famakin
- National Institutes of Health, National Institute of Neurological Diseases and Stroke, Stroke Branch, Branch, Bethesda, MD, 20892, USA
| |
Collapse
|
19
|
Easton AS. Neutrophils and stroke – Can neutrophils mitigate disease in the central nervous system? Int Immunopharmacol 2013; 17:1218-25. [DOI: 10.1016/j.intimp.2013.06.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 02/15/2013] [Accepted: 06/09/2013] [Indexed: 12/19/2022]
|
20
|
Dhungana H, Malm T, Denes A, Valonen P, Wojciechowski S, Magga J, Savchenko E, Humphreys N, Grencis R, Rothwell N, Koistinaho J. Aging aggravates ischemic stroke-induced brain damage in mice with chronic peripheral infection. Aging Cell 2013; 12:842-50. [PMID: 23725345 DOI: 10.1111/acel.12106] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2013] [Indexed: 01/13/2023] Open
Abstract
Ischemic stroke is confounded by conditions such as atherosclerosis, diabetes, and infection, all of which alter peripheral inflammatory processes with concomitant impact on stroke outcome. The majority of the stroke patients are elderly, but the impact of interactions between aging and inflammation on stroke remains unknown. We thus investigated the influence of age on the outcome of stroke in animals predisposed to systemic chronic infection. Th1-polarized chronic systemic infection was induced in 18-22 month and 4-month-old C57BL/6j mice by administration of Trichuris muris (gut parasite). One month after infection, mice underwent permanent middle cerebral artery occlusion and infarct size, brain gliosis, and brain and plasma cytokine profiles were analyzed. Chronic infection increased the infarct size in aged but not in young mice at 24 h. Aged, ischemic mice showed altered plasma and brain cytokine responses, while the lesion size correlated with plasma prestroke levels of RANTES. Moreover, the old, infected mice exhibited significantly increased neutrophil recruitment and upregulation of both plasma interleukin-17α and tumor necrosis factor-α levels. Neither age nor infection status alone or in combination altered the ischemia-induced brain microgliosis. Our results show that chronic peripheral infection in aged animals renders the brain more vulnerable to ischemic insults, possibly by increasing the invasion of neutrophils and altering the inflammation status in the blood and brain. Understanding the interactions between age and infections is crucial for developing a better therapeutic regimen for ischemic stroke and when modeling it as a disease of the elderly.
Collapse
Affiliation(s)
- Hiramani Dhungana
- Department of Neurobiology; A. I. Virtanen Institute for Molecular Sciences; Biocenter Kuopio; University of Eastern Finland; PO Box 1627; FI-70211; Kuopio; Finland
| | - Tarja Malm
- Department of Neurobiology; A. I. Virtanen Institute for Molecular Sciences; Biocenter Kuopio; University of Eastern Finland; PO Box 1627; FI-70211; Kuopio; Finland
| | | | - Piia Valonen
- Department of Neurobiology; A. I. Virtanen Institute for Molecular Sciences; Biocenter Kuopio; University of Eastern Finland; PO Box 1627; FI-70211; Kuopio; Finland
| | - Sara Wojciechowski
- Department of Neurobiology; A. I. Virtanen Institute for Molecular Sciences; Biocenter Kuopio; University of Eastern Finland; PO Box 1627; FI-70211; Kuopio; Finland
| | - Johanna Magga
- Department of Neurobiology; A. I. Virtanen Institute for Molecular Sciences; Biocenter Kuopio; University of Eastern Finland; PO Box 1627; FI-70211; Kuopio; Finland
| | - Ekaterina Savchenko
- Department of Neurobiology; A. I. Virtanen Institute for Molecular Sciences; Biocenter Kuopio; University of Eastern Finland; PO Box 1627; FI-70211; Kuopio; Finland
| | - Neil Humphreys
- Faculty of Life Sciences; University of Manchester; Oxford Road; Manchester; M13 9PT; UK
| | - Richard Grencis
- Faculty of Life Sciences; University of Manchester; Oxford Road; Manchester; M13 9PT; UK
| | - Nancy Rothwell
- Faculty of Life Sciences; University of Manchester; Oxford Road; Manchester; M13 9PT; UK
| | | |
Collapse
|
21
|
Enzmann G, Mysiorek C, Gorina R, Cheng YJ, Ghavampour S, Hannocks MJ, Prinz V, Dirnagl U, Endres M, Prinz M, Beschorner R, Harter PN, Mittelbronn M, Engelhardt B, Sorokin L. The neurovascular unit as a selective barrier to polymorphonuclear granulocyte (PMN) infiltration into the brain after ischemic injury. Acta Neuropathol 2013; 125:395-412. [PMID: 23269317 PMCID: PMC3578720 DOI: 10.1007/s00401-012-1076-3] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 12/14/2012] [Accepted: 12/14/2012] [Indexed: 01/05/2023]
Abstract
The migration of polymorphonuclear granulocytes (PMN) into the brain parenchyma and release of their abundant proteases are considered the main causes of neuronal cell death and reperfusion injury following ischemia. Yet, therapies targeting PMN egress have been largely ineffective. To address this discrepancy we investigated the temporo-spatial localization of PMNs early after transient ischemia in a murine transient middle cerebral artery occlusion (tMCAO) model and human stroke specimens. Using specific markers that distinguish PMN (Ly6G) from monocytes/macrophages (Ly6C) and that define the cellular and basement membrane boundaries of the neurovascular unit (NVU), histology and confocal microscopy revealed that virtually no PMNs entered the infarcted CNS parenchyma. Regardless of tMCAO duration, PMNs were mainly restricted to luminal surfaces or perivascular spaces of cerebral vessels. Vascular PMN accumulation showed no spatial correlation with increased vessel permeability, enhanced expression of endothelial cell adhesion molecules, platelet aggregation or release of neutrophil extracellular traps. Live cell imaging studies confirmed that oxygen and glucose deprivation followed by reoxygenation fail to induce PMN migration across a brain endothelial monolayer under flow conditions in vitro. The absence of PMN infiltration in infarcted brain tissues was corroborated in 25 human stroke specimens collected at early time points after infarction. Our observations identify the NVU rather than the brain parenchyma as the site of PMN action after CNS ischemia and suggest reappraisal of targets for therapies to reduce reperfusion injury after stroke.
Collapse
Affiliation(s)
- Gaby Enzmann
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012 Bern, Switzerland
| | - Caroline Mysiorek
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany
| | - Roser Gorina
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012 Bern, Switzerland
| | - Yu-Jung Cheng
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany
| | - Sharang Ghavampour
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany
| | - Melanie-Jane Hannocks
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany
| | | | - Ulrich Dirnagl
- Department of Neurology, Berlin, Germany
- Center for Stroke Research Berlin, Charité University, Berlin, Germany
| | - Matthias Endres
- Department of Neurology, Berlin, Germany
- Center for Stroke Research Berlin, Charité University, Berlin, Germany
| | - Marco Prinz
- Department of Neuropathology, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Rudi Beschorner
- Department of Neuropathology, Institute of Pathology and Neuropathology, University of Tübingen, Tübingen, Germany
| | - Patrick N. Harter
- Institute of Neurology (Edinger Institute), University of Frankfurt, Frankfurt, Germany
| | - Michel Mittelbronn
- Institute of Neurology (Edinger Institute), University of Frankfurt, Frankfurt, Germany
| | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012 Bern, Switzerland
| | - Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany
| |
Collapse
|
22
|
Watcharotayangul J, Mao L, Xu H, Vetri F, Baughman VL, Paisansathan C, Pelligrino DA. Post-ischemic vascular adhesion protein-1 inhibition provides neuroprotection in a rat temporary middle cerebral artery occlusion model. J Neurochem 2012; 123 Suppl 2:116-24. [PMID: 23050649 DOI: 10.1111/j.1471-4159.2012.07950.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
We examined the neuroprotective efficacy associated with post-ischemic vascular adhesion protein-1 (VAP-1) blockade in rats subjected to transient (1 h) middle cerebral artery occlusion (MCAo). We compared saline-treated control rats to rats treated with a highly selective VAP-1 inhibitor, LJP-1586 [Z-3-fluoro-2-(4-methoxybenzyl) allylamine hydrochloride]. Initial intraperitoneal LJP-1586 (or saline control) treatments were delayed until 6 h or 12 h reperfusion. At 72-h reperfusion, LJP-1586-treated rats displayed 51% and 33% smaller infarct volumes, relative to their controls, in the 6- and 12-h treatment groups, respectively. However, only in the 6-h treatment group was the infarct volume reduction significant (p < 0.05). On the other hand, we observed significantly improved neurologic functions in both 6- and 12-h treatment groups, versus their matched controls (p < 0.05). Also, the effect of 6-h LJP-1586 treatment on post-ischemic leukocyte trafficking in pial venules overlying the ischemic cortex was evaluated using intravital microscopy. These experiments revealed that: 1) LJP-1586 did not affect intravascular leukocyte (largely neutrophil) adhesion, at least out to 12-h reperfusion; and 2) the onset of neutrophil extravasation, which occurred between 6-8-h reperfusion in control rats, was prevented by LJP-1586-treatment. In conclusion, in rats subjected to transient MCAo, selective VAP-1 pharmacologic blockade provided neuroprotection, with a prolonged therapeutic window of 6-12-h reperfusion.
Collapse
|
23
|
Tang NY, Liu CH, Hsieh CT, Hsieh CL. The Anti-Inflammatory Effect of Paeoniflorin on Cerebral Infarction Induced by Ischemia-Reperfusion Injury in Sprague-Dawley Rats. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2012; 38:51-64. [PMID: 20128044 DOI: 10.1142/s0192415x10007786] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Paeoniflorin, a component in Paeonia lactiflora Pall, inhibits nuclear factor-κB expression in chronic hypoperfusion rat and has anti-inflammatory properties. Therefore, the aim of the present study was to investigate the effect of paeoniflorin on cerebral infarct, and the involvement of anti-inflammation. We established an animal model of cerebral infarct by occluding both the common carotid arteries and the right middle cerebral artery for 90 min, followed by reperfusion of 24 hours. The ratios of cerebral infarction area to total brain area, and neuro-deficit score were used as an index to observe the effects of paeoniflorin on cerebral infarct. ED1 (mouse anti rat CD68), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), intercellular adhesion molecular-1 (ICAM-1), myeloperoxidase (MPO) immunostaining and apoptotic cells in the cerebral infarction region also were studied. The results indicated that both pre-treatment and post-treatment with paeoniflorin reduced the ratio of cerebral infarction area; pre-treatment with paeoniflorin also reduced the neurological deficit score. The counts of ED1, IL-1β, TNF-α, ICAM-1 of microvessels and MPO immunoreactive cells and apoptotic cells were increased in the cerebral infarction region; however, these increases were reduced by Paeoniflorin pre-treatment. In conclusion, Paeoniflorin reduced cerebral infarct and neurological deficit in ischemia-reperfusion injured rats, suggesting that paeoniflorin may have a similar effect in humans and might be a suitable treatment for stroke. Paeoniflorin reduced cerebral infarct, at least in part, involves the anti-inflammatory properties.
Collapse
Affiliation(s)
- Nou-Ying Tang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chung-Hsiang Liu
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan
| | - Ching-Tou Hsieh
- Department of Internal Medicine, Jen-Ai Hospital, Taichung, Taiwan
| | - Ching-Liang Hsieh
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
- Acupuncture Research Center, China Medical University, Taichung, Taiwan
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
24
|
Mu S, Ouyang L, Liu B, Qu H, Zhu Y, Li K, Lei W. Relationship between inflammatory reaction and ischemic injury of caudate-putamen in rats: inflammatory reaction and brain ischemia. Anat Sci Int 2010; 86:86-97. [PMID: 20809266 DOI: 10.1007/s12565-010-0091-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Accepted: 06/30/2010] [Indexed: 10/19/2022]
Abstract
Inflammatory response after middle cerebral artery occlusion (MCAO) has been a focus of research recently, but the effect of inflammatory cells on ischemic neurons remains unclear. In order to study the effect of the inflammatory reaction on brain ischemic injury, we observed the morphology, number and distribution of CD3-, CD8-, ED1- and ED2-positive cells systematically in the caudate-putamen of rats in a MCAO model. The present results show that all four types of inflammatory cells first infiltrated the ischemic penumbra and then migrated into the center of the ischemic area, but the morphological changes and infiltration processes differed significantly; the infiltration of CD3- and CD8-positive cells into the ischemic area started at 3 days postischemia, and their number peaked at 1 week; however, although ED1- and ED2-positive cells were also observed at 3 days after ischemia, they reached their maximum number at 2 and 4 weeks, respectively. Moreover, ED1-and ED2-positive cells showed evident hyperplasia and hypertrophy in morphology. Our results also showed that the response of CD3-, CD8-, ED1- and ED2-positive cells in the ischemic area and the pathological changes in ischemic brain tissue could be inhibited by cyclosporine A. The results suggest that the infiltration and reaction of inflammatory cells are involved in the pathological process of ischemic brain injury.
Collapse
Affiliation(s)
- Shuhua Mu
- Department of Anatomy, Zhongshan Medical School of Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
25
|
Semple BD, Bye N, Ziebell JM, Morganti-Kossmann MC. Deficiency of the chemokine receptor CXCR2 attenuates neutrophil infiltration and cortical damage following closed head injury. Neurobiol Dis 2010; 40:394-403. [PMID: 20621186 DOI: 10.1016/j.nbd.2010.06.015] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 06/23/2010] [Accepted: 06/26/2010] [Indexed: 12/21/2022] Open
Abstract
The contribution of infiltrated neutrophils to secondary damage following traumatic brain injury remains controversial. Chemokines that regulate neutrophil migration by signaling through the CXCR2 receptor are markedly elevated by brain injury and are associated with the propagation of secondary damage. This study thus investigated the function of CXCR2 in posttraumatic inflammation and secondary degeneration by examining Cxcr2-deficient (Cxcr2(-/-)) mice over 14 days following closed head injury (CHI). We demonstrate a significant attenuation of neutrophil infiltration in Cxcr2(-/-) mice at 12 hours and 7 days after CHI, despite increased levels of CXC neutrophil-attracting chemokines in the lesioned cortex. This coincides with reduced tissue damage, neuronal loss, and cell death in Cxcr2(-/-) mice compared to wild-type controls, with heterozygotes showing intermediate responses. In contrast, blood-brain barrier permeability and functional recovery did not appear to be affected by Cxcr2 deletion. This study highlights the deleterious contribution of neutrophils to posttraumatic neurodegeneration and demonstrates the importance of CXC chemokine signaling in this process. Therefore, CXCR2 antagonistic therapeutics currently in development for other inflammatory conditions may also be of benefit in posttraumatic neuroinflammation.
Collapse
Affiliation(s)
- Bridgette D Semple
- National Trauma Research Institute, The Alfred Hospital, Melbourne, Victoria, Australia.
| | | | | | | |
Collapse
|
26
|
Tu XK, Yang WZ, Wang CH, Shi SS, Zhang YL, Chen CM, Yang YK, Jin CD, Wen S. Zileuton Reduces Inflammatory Reaction and Brain Damage Following Permanent Cerebral Ischemia in Rats. Inflammation 2010; 33:344-52. [DOI: 10.1007/s10753-010-9191-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
27
|
Farias S, Frey LC, Murphy RC, Heidenreich KA. Injury-related production of cysteinyl leukotrienes contributes to brain damage following experimental traumatic brain injury. J Neurotrauma 2010; 26:1977-86. [PMID: 19886806 DOI: 10.1089/neu.2009.0877] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The leukotrienes belong to a family of biologically active lipids derived from arachidonate that are often involved in inflammatory responses. In the central nervous system, a group of leukotrienes, known as the cysteinyl leukotrienes, is generated in brain tissue in response to a variety of acute brain injuries. Although the exact clinical significance of this excess production remains unclear, the cysteinyl leukotrienes may contribute to injury-related disruption of the brain-blood barrier and exacerbate secondary injury processes. In the present study, the formation and role of cysteinyl leukotrienes was explored in the fluid percussion injury model of traumatic brain injury in rats. The results showed that levels of the cysteinyl leukotrienes were elevated after fluid percussion injury with a maximal formation 1 hour after the injury. Neutrophils contributed to cysteinyl leukotriene formation in the injured brain hemisphere, potentially through a transcellular biosynthetic mechanism. Furthermore, pharmacological reduction of cysteinyl leukotriene formation after the injury, using MK-886, resulted in reduction of brain lesion volumes, suggesting that the cysteinyl leukotrienes play an important role in traumatic brain injury.
Collapse
Affiliation(s)
- Santiago Farias
- Department of Pharmacology, University of Colorado at Denver, Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | | | | | | |
Collapse
|
28
|
Clausen F, Hånell A, Björk M, Hillered L, Mir AK, Gram H, Marklund N. Neutralization of interleukin-1beta modifies the inflammatory response and improves histological and cognitive outcome following traumatic brain injury in mice. Eur J Neurosci 2009; 30:385-96. [PMID: 19614750 DOI: 10.1111/j.1460-9568.2009.06820.x] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Interleukin-1beta (IL-1beta) may play a central role in the inflammatory response following traumatic brain injury (TBI). We subjected 91 mice to controlled cortical impact (CCI) brain injury or sham injury. Beginning 5 min post-injury, the IL-1beta neutralizing antibody IgG2a/k (1.5 microg/mL) or control antibody was infused at a rate of 0.25 microL/h into the contralateral ventricle for up to 14 days using osmotic minipumps. Neutrophil and T-cell infiltration and microglial activation was evaluated at days 1-7 post-injury. Cognition was assessed using Morris water maze, and motor function using rotarod and cylinder tests. Lesion volume and hemispheric tissue loss were evaluated at 18 days post-injury. Using this treatment strategy, cortical and hippocampal tissue levels of IgG2a/k reached 50 ng/mL, sufficient to effectively inhibit IL-1betain vitro. IL-1beta neutralization attenuated the CCI-induced cortical and hippocampal microglial activation (P < 0.05 at post-injury days 3 and 7), and cortical infiltration of neutrophils (P < 0.05 at post-injury day 7). There was only a minimal cortical infiltration of activated T-cells, attenuated by IL-1beta neutralization (P < 0.05 at post-injury day 7). CCI induced a significant deficit in neurological motor and cognitive function, and caused a loss of hemispheric tissue (P < 0.05). In brain-injured animals, IL-1beta neutralizing treatment resulted in reduced lesion volume, hemispheric tissue loss and attenuated cognitive deficits (P < 0.05) without influencing neurological motor function. Our results indicate that IL-1beta is a central component in the post-injury inflammatory response that, in view of the observed positive neuroprotective and cognitive effects, may be a suitable pharmacological target for the treatment of TBI.
Collapse
Affiliation(s)
- Fredrik Clausen
- Department of Neuroscience, Section for Neurosurgery, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
29
|
Matharu NM, McGettrick HM, Salmon M, Kissane S, Vohra RK, Rainger G, Nash GB. Inflammatory responses of endothelial cells experiencing reduction in flow after conditioning by shear stress. J Cell Physiol 2008; 216:732-41. [DOI: 10.1002/jcp.21457] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
30
|
Small Molecule Selectin Inhibitor in Global Cerebral Ischemia and Controlled Hemorrhagic Shock. ACTA ACUST UNITED AC 2008; 65:678-84. [PMID: 18784584 DOI: 10.1097/ta.0b013e3181843f3a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
31
|
Demiralay R, Gürsan N, Erdem H. The effects of erdosteine, N-acetylcysteine, and vitamin E on nicotine-induced apoptosis of hippocampal neural cells. J Cell Biochem 2008; 104:1740-6. [DOI: 10.1002/jcb.21739] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
32
|
Microglia cells protect neurons by direct engulfment of invading neutrophil granulocytes: a new mechanism of CNS immune privilege. J Neurosci 2008; 28:5965-75. [PMID: 18524901 DOI: 10.1523/jneurosci.0060-08.2008] [Citation(s) in RCA: 205] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Microglial cells maintain the immunological integrity of the healthy brain and can exert protection from traumatic injury. During ischemic tissue damage such as stroke, peripheral immune cells acutely infiltrate the brain and may exacerbate neurodegeneration. Whether and how microglia can protect from this insult is unknown. Polymorphonuclear neutrophils (PMNs) are a prominent immunologic infiltrate of ischemic lesions in vivo. Here, we show in organotypic brain slices that externally applied invading PMNs massively enhance ischemic neurotoxicity. This, however, is counteracted by additional application of microglia. Time-lapse imaging shows that microglia exert protection by rapid engulfment of apoptotic, but, strikingly, also viable, motile PMNs in cell culture and within brain slices. PMN engulfment is mediated by integrin- and lectin-based recognition. Interference with this process using RGDS peptides and N-acetyl-glucosamine blocks engulfment of PMNs and completely abrogates the neuroprotective function of microglia. Thus, engulfment of invading PMNs by microglia may represent an entirely new mechanism of CNS immune privilege.
Collapse
|
33
|
Oh JK, Jung JW, Oh HR, Han YN, Ryu JH. Neuroprotective Effects of Ginkgo biloba extract, GBB, in the Transient Ischemic Rat Model. Biomol Ther (Seoul) 2007. [DOI: 10.4062/biomolther.2007.15.3.169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
34
|
Villa P, Triulzi S, Cavalieri B, Di Bitondo R, Bertini R, Barbera S, Bigini P, Mennini T, Gelosa P, Tremoli E, Sironi L, Ghezzi P. The interleukin-8 (IL-8/CXCL8) receptor inhibitor reparixin improves neurological deficits and reduces long-term inflammation in permanent and transient cerebral ischemia in rats. Mol Med 2007. [PMID: 17592546 DOI: 10.2119/2007-00008.villa] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Leukocyte infiltration is viewed as a pharmacological target in cerebral ischemia. We previously reported that reparixin, a CXCL8 receptor blocker that inhibits neutrophil infiltration, and related molecules can reduce infarct size in a rat model of transient middle cerebral artery occlusion (MCAO). The study aims were to compare the effects of reparixin in transient and permanent MCAO using varied treatment schedules and therapeutic windows to evaluate effects on long-term neurological deficits and late inflammatory response. Reparixin, administered for 1 to 3 days, 3.5 to 6 h after MCAO, ameliorates neurological function recovery and inhibits long-term inflammation. The infarct size reduction at 24 h, evaluated by TTC staining, is more pronounced in transient MCAO. MRI analysis identified a decrease in the progression of infarct size by reparixin that was more evident at 48 h in permanent MCAO, and was associated with a significantly improved recovery from long-term neurological deficits.
Collapse
Affiliation(s)
- Pia Villa
- Mario Negri Institute, 20157 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Villa P, Triulzi S, Cavalieri B, Di Bitondo R, Bertini R, Barbera S, Bigini P, Mennini T, Gelosa P, Tremoli E, Sironi L, Ghezzi P. The interleukin-8 (IL-8/CXCL8) receptor inhibitor reparixin improves neurological deficits and reduces long-term inflammation in permanent and transient cerebral ischemia in rats. MOLECULAR MEDICINE (CAMBRIDGE, MASS.) 2007; 13:125-33. [PMID: 17592546 PMCID: PMC1892761 DOI: 10.2119/2007–00008.villa] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Accepted: 02/05/2007] [Indexed: 01/30/2023]
Abstract
Leukocyte infiltration is viewed as a pharmacological target in cerebral ischemia. We previously reported that reparixin, a CXCL8 receptor blocker that inhibits neutrophil infiltration, and related molecules can reduce infarct size in a rat model of transient middle cerebral artery occlusion (MCAO). The study aims were to compare the effects of reparixin in transient and permanent MCAO using varied treatment schedules and therapeutic windows to evaluate effects on long-term neurological deficits and late inflammatory response. Reparixin, administered for 1 to 3 days, 3.5 to 6 h after MCAO, ameliorates neurological function recovery and inhibits long-term inflammation. The infarct size reduction at 24 h, evaluated by TTC staining, is more pronounced in transient MCAO. MRI analysis identified a decrease in the progression of infarct size by reparixin that was more evident at 48 h in permanent MCAO, and was associated with a significantly improved recovery from long-term neurological deficits.
Collapse
Affiliation(s)
- Pia Villa
- Mario Negri Institute, 20157 Milan, Italy
- CNR, Institute of Neuroscience, 20129 Milan, Italy
| | | | | | | | | | | | | | | | - Paolo Gelosa
- Department of Pharmacological Sciences, University of Milan, 20133 Milan, Italy
| | - Elena Tremoli
- Department of Pharmacological Sciences, University of Milan, 20133 Milan, Italy
- Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy
| | - Luigi Sironi
- Department of Pharmacological Sciences, University of Milan, 20133 Milan, Italy
| | - Pietro Ghezzi
- Mario Negri Institute, 20157 Milan, Italy
- Address correspondence and reprint requests to Pietro Ghezzi, Laboratory of Neuroim-munology, Mario Negri Institute, via Eritrea 62, 20157 Milan, Italy. Phone: + 39-02-39014486; Fax: + 39-02-3546277; E-mail:
| |
Collapse
|
36
|
Morkos AA, Hopper AO, Deming DD, Yellon SM, Wycliffe N, Ashwal S, Sowers LC, Peverini RL, Angeles DM. Elevated total peripheral leukocyte count may identify risk for neurological disability in asphyxiated term neonates. J Perinatol 2007; 27:365-70. [PMID: 17443199 DOI: 10.1038/sj.jp.7211750] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE The present study investigated the relationship between neurologic outcome and total circulating white blood cell (WBC) and absolute neutrophil counts (ANCs) in the first week of life in term infants with hypoxic-ischemic encephalopathy (HIE). STUDY DESIGN Long-term neurologic outcome at 18 months was measured retrospectively in 30 term neonates with HIE using the Pediatric Cerebral Performance Category Scale (PCPCS) score with outcomes dichotomized as either good or poor. We then compared white blood cell and ANC levels during the first 4 days of life and magnetic resonance imaging (MRI) obtained within the first month life between the two PCPCS groups. MRI was quantified using a validated scoring system. RESULTS Neonates with good long-term outcomes had significantly lower MRI scores (indicating lesser injury) than neonates with poor outcomes. More importantly, neonates with poor outcomes had significantly higher WBC and ANC levels as early as12 h after birth and up to 96 h after birth compared to those with good outcomes. These data suggest that elevated peripheral neutrophil counts in the first 96 h of life may signal or predict adverse long-term outcome. CONCLUSIONS Our findings suggest that elevated peripheral neutrophil counts in the first 96 h of life in term infants with HIE may contribute to abnormal neurodevelopmental outcome.
Collapse
Affiliation(s)
- A A Morkos
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
McColl BW, Rothwell NJ, Allan SM. Systemic inflammatory stimulus potentiates the acute phase and CXC chemokine responses to experimental stroke and exacerbates brain damage via interleukin-1- and neutrophil-dependent mechanisms. J Neurosci 2007; 27:4403-12. [PMID: 17442825 PMCID: PMC6672305 DOI: 10.1523/jneurosci.5376-06.2007] [Citation(s) in RCA: 271] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Systemic inflammatory stimuli, such as infection, increase the risk of stroke and are associated with poorer clinical outcome. The mechanisms underlying the impact of systemic inflammatory stimuli on stroke are not well defined. We investigated the impact of systemic inflammation on experimental stroke and potential mechanisms involved. Focal cerebral ischemia was induced by intraluminal filament occlusion of the middle cerebral artery (MCAo). Brain damage and neurological deficit 24 h after MCAo were exacerbated by systemic lipopolysaccharide (LPS) administration. This exacerbation was critically dependent on interleukin (IL)-1, because coadministration of IL-1 receptor antagonist abolished the effect of LPS on brain damage. Systemic administration of IL-1 increased ischemic damage to a similar extent as LPS and also exacerbated brain edema. IL-1 markedly potentiated circulating levels of the acute phase proteins, serum amyloid A and IL-6, and the neutrophil-selective CXC chemokines, KC and macrophage inflammatory protein-2. Neutrophil mobilization and cortical neutrophil infiltration were aggravated by IL-1 before changes in ischemic damage. Neutropenia abolished the damaging effects of systemic IL-1. These data show for the first time that an acute systemic inflammatory stimulus is detrimental to outcome after experimental stroke and highlight IL-1 as a critical mediator in this paradigm. Our data suggest IL-1-induced potentiation of neutrophil mobilization via CXC chemokine induction is a putative mechanism underlying this effect. Our results may help to explain the poorer outcome in stroke patients presenting with infection and may have implications for neurodegenerative diseases involving neurovascular alterations, such as Alzheimer's disease, in which systemic inflammation can modulate disease progression.
Collapse
Affiliation(s)
- Barry W McColl
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom.
| | | | | |
Collapse
|
38
|
Lee MY, Kuan YH, Chen HY, Chen TY, Chen ST, Huang CC, Yang IP, Hsu YS, Wu TS, Lee EJ. Intravenous administration of melatonin reduces the intracerebral cellular inflammatory response following transient focal cerebral ischemia in rats. J Pineal Res 2007; 42:297-309. [PMID: 17349029 DOI: 10.1111/j.1600-079x.2007.00420.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We have previously shown that exogenous melatonin improves the preservation of the blood-brain barrier (BBB) and neurovascular unit following cerebral ischemia-reperfusion. Recent evidence indicates that postischemic microglial activation exaggerates the damage to the BBB. Herein, we explored whether melatonin mitigates the cellular inflammatory response after transient focal cerebral ischemia for 90 min in rats. Melatonin (5 mg/kg) or vehicle was given intravenously at reperfusion onset. Immunohistochemistry and flow cytometric analysis were used to evaluate the cellular inflammatory response at 48 hr after reperfusion. Relative to controls, melatonin-treated animals did not have significantly changed systemic cellular inflammatory responses in the bloodstream (P > 0.05). Melatonin, however, significantly decreased the cellular inflammatory response by 41% (P < 0.001) in the ischemic hemisphere. Specifically, melatonin effectively decreased the extent of neutrophil emigration (Ly6G-positive/CD45-positive) and macrophage/activated microglial infiltration (CD11b-positive/CD45-positive) by 51% (P < 0.01) and 66% (P < 0.01), respectively, but did not significantly alter the population composition of T lymphocyte (CD3-positive/CD45-positive; P > 0.05). This melatonin-mediated decrease in the cellular inflammatory response was accompanied by both reduced brain infarction and improved neurobehavioral outcome by 43% (P < 0.001) and 50% (P < 0.001), respectively. Thus, intravenous administration of melatonin upon reperfusion effectively decreased the emigration of circulatory neutrophils and macrophages/monocytes into the injured brain and inhibited focal microglial activation following cerebral ischemia-reperfusion. The finding demonstrates melatonin's inhibitory ability against the cellular inflammatory response after cerebral ischemia-reperfusion, and further supports its pleuripotent neuroprotective actions suited either as a monotherapy or an add-on to the thrombolytic therapy for ischemic stroke patients.
Collapse
Affiliation(s)
- Ming-Yang Lee
- Neurophysiology Laboratory, Neurosurgical Service, Department of Surgery, National Cheng Kung University Medical Center and Medical School, Tainan, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Pustovidko A, Potselueva M, Kochegarov A, Evtodienko Y. Role of Ca2+ in activation of reactive oxygen species production in polymorphonuclear leukocytes during tumour growth in rats. LUMINESCENCE 2007; 22:199-205. [PMID: 17262735 DOI: 10.1002/bio.949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The role of Ca(2+) ions in PMA-induced generation of reactive oxygen species (ROS) by polymorphonuclear leukocytes (PMNL) was studied during Zajdela hepatoma growth in the peritoneal cavity of rats. In PMNL from control healthy animals, a manifold Ca(2+)-induced enhancement of ROS generation and its significant reduction in the presence of Ca(2+) binding agent (BAPTA-AM) were observed. In contrast, ROS generation by PMNL from tumour-carrying animals dramatically increased in Ca(2+)-free medium, being practically insensitive to the agents, which can increase or decrease intracellular Ca(2+) levels. Free cytosolic Ca(2+) ([Ca(2+)](i)) in control PMNL was found to be relatively low ( approximately 250 nmol/L), rising slowly after Ca(2+) addition and further to two-fold in the presence of Ca(2+) and ionomycin in the incubating medium. Tumour growth in animals was accompanied with a significant [Ca(2+)](i) elevation. In Ca(2+)-free medium, [Ca(2+)](i) elevation was up to 480 nmol/L in tPMNL with the additions of Ca(+) and ionomycin as well as EGTA and ionomycin being able to increase [Ca(2+)](i) to 700-900 nmol/L onward. It was concluded that a higher Ca(2+) permeability of the plasma membrane and higher Ca(2+) accumulation in intracellular pools of PMNL was developed at the advanced stages of malignant disease. These results indicate the primed state of circulating PMNL and the independence of PMA-induced ROS generation at intra- and extracellular Ca(2+) levels at the advanced stages of tumour growth in animals.
Collapse
Affiliation(s)
- A Pustovidko
- A. N. Belozersky Institute of Physicochemical Biology, Moscow State University, Moscow 119899, Russia.
| | | | | | | |
Collapse
|
40
|
Weston RM, Jones NM, Jarrott B, Callaway JK. Inflammatory cell infiltration after endothelin-1-induced cerebral ischemia: histochemical and myeloperoxidase correlation with temporal changes in brain injury. J Cereb Blood Flow Metab 2007; 27:100-14. [PMID: 16736051 DOI: 10.1038/sj.jcbfm.9600324] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Accumulation of neutrophils in brain after transient focal stroke remains controversial with some studies showing neutrophils to be deleterious, whereas others suggest neutrophils do not contribute to ischemic injury. Myeloperoxidase (MPO) has been used extensively as a marker for quantifying neutrophil accumulation, but is an indirect method and does not detect neutrophils alone. To elucidate the interaction of macrophages in the neutrophil inflammatory response, we conducted double-label immunofluorescence in brain sections at 0, 1, 2, 3, 7, and 15 days after ischemia. Each of these results was obtained from the same animal to determine correlations between neutrophil infiltration and ischemic damage. It was found that MPO activity increased up to 3 days after cerebral ischemia. Dual-staining revealed that macrophages engulf neutrophils in the brain and that this engulfment of neutrophils increased with time, with 50% of neutrophils in the brain engulfed at 3 days and approximately 85% at 15 days (N=5, P<0.05). Interestingly, at 7 days the amount of dual-staining was decreased to 20% (N=5, P<0.05). Neutrophil infiltration was positively correlated with ischemic damage in both the cortex and striatum (r(2)=0.86 and 0.80, respectively, P<0.01). The results of this study indicate that the MPO from neutrophils phagocytized by macrophages may continue to contribute to the overall MPO activity, and that previous assessments that have utilized this marker to measure neutrophil accumulation may have mis-calculated the number of neutrophils within the ischemic territory and hence their contribution to the evolution of the infarct at later time points. Thus any biphasic infiltration of neutrophils may have been masked by the accumulation of macrophages.
Collapse
Affiliation(s)
- Robert M Weston
- Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | | | | | | |
Collapse
|
41
|
Matsumoto H, Kumon Y, Watanabe H, Ohnishi T, Shudou M, Ii C, Takahashi H, Imai Y, Tanaka J. Antibodies to CD11b, CD68, and lectin label neutrophils rather than microglia in traumatic and ischemic brain lesions. J Neurosci Res 2007; 85:994-1009. [PMID: 17265469 DOI: 10.1002/jnr.21198] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Resident quiescent microglia have been thought to respond rapidly to various pathologic events in the brain by proliferating and producing many bioactive substances, including proinflammatory cytokines and nitric oxide (NO). In this study, we investigated the reaction of microglia in traumatic and ischemic lesions caused by stab wounds and the transient 90-min occlusion of middle cerebral artery in a mature rat brain. Although many Iba1(+) resident microglia underwent apoptotic degeneration in the lesion core within 24 hr after the onset of the brain insult as revealed by TUNEL staining, numerous small, round, isolectin B4(+)/CD11b(+)/CD68(+) cells were localized in the lesion core. These small, round cells with diameters of 7-9 mum and polymorph nuclei expressed neutrophil-specific elastase, alkaline phosphatase, and platelet-activating factor receptor. Accordingly, they were not activated microglia but neutrophils. Immunohistochemical staining with antibodies to inducible NO synthase (iNOS) showed that most iNOS(+) cells were neutrophils. The results from spatial and kinetic analyses using RT-PCR and immunoblotting were consistent with the immunohistochemical observations. These results suggest the necessity of reevaluating the traditional view on the roles of activated microglia in severe neuropathologic events. Note that the traditional microglial markers isolectin B4, CD11b, and CD68 are not specific for microglia, particularly in a pathologic brain.
Collapse
MESH Headings
- Alkaline Phosphatase/metabolism
- Animals
- Antibodies
- Antigens, CD/analysis
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/analysis
- Antigens, Differentiation, Myelomonocytic/immunology
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Surface/immunology
- Biomarkers/analysis
- Biomarkers/metabolism
- Brain Infarction/immunology
- Brain Infarction/pathology
- Brain Infarction/physiopathology
- Brain Injuries/immunology
- Brain Injuries/pathology
- Brain Injuries/physiopathology
- Brain Ischemia/immunology
- Brain Ischemia/pathology
- Brain Ischemia/physiopathology
- CD11 Antigens/analysis
- CD11 Antigens/immunology
- CD11 Antigens/metabolism
- Chemotaxis, Leukocyte/immunology
- Female
- Infarction, Middle Cerebral Artery/immunology
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/physiopathology
- Leukocyte Elastase/metabolism
- Male
- Microglia/immunology
- Neutrophils/immunology
- Nitric Oxide Synthase Type II/metabolism
- Plant Lectins/immunology
- Platelet Membrane Glycoproteins/metabolism
- Rats
- Rats, Wistar
- Receptors, G-Protein-Coupled/metabolism
Collapse
Affiliation(s)
- Hiroaki Matsumoto
- Department of Neurosurgery, Graduate School of Medicine, Ehime University, Ehime, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Weston RM, Jarrott B, Ishizuka Y, Callaway JK. AM-36 modulates the neutrophil inflammatory response and reduces breakdown of the blood brain barrier after endothelin-1 induced focal brain ischaemia. Br J Pharmacol 2006; 149:712-23. [PMID: 17016500 PMCID: PMC2014659 DOI: 10.1038/sj.bjp.0706918] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND AND PURPOSE Following transient focal stroke, rapid accumulation and activation of neutrophils in the ischaemic region is deleterious due to release of reactive oxygen species and myeloperoxidase (MPO). The purpose of this study was to examine whether AM-36, both a Na+ channel blocker and an antioxidant, afforded neuroprotection by modulating neutrophil accumulation into brain, following endothelin-1 (ET-1) induced middle cerebral artery occlusion (MCAo) in conscious rats. EXPERIMENTAL APPROACH AM-36 was administered at 3 and 24 h after ET-1-induced MCAo. Functional recovery was determined using grid-walking and cylinder tests. Image analysis of brain sections was used to determine infarct volume. The effect of AM-36 on neutrophil infiltration and their interaction with macrophages was examined in rats at 48 h following MCAo by both an MPO assay and double-label immunofluorescence. Blood brain barrier (BBB) breakdown was measured by the area stained by intravenous Evans Blue. KEY RESULTS AM-36 reduced functional deficits in both tests such that no difference existed from pre-ischaemic values at 48 h. Neutrophil infiltration, assessed by MPO activity, and infarct volume were significantly reduced following AM-36 administration by 54 and 60% respectively. Similarly, immunofluorescence revealed that AM-36 reduced neutrophil infiltration by approximately 50% in selected brain regions, when compared to controls, and also modulated macrophage phagocytosis of neutrophils. Breakdown of the BBB was significantly reduced by 60% following AM-36 treatment. CONCLUSIONS AND IMPLICATIONS These findings suggest that AM-36 can directly modulate the neutrophil inflammatory response and reduce BBB breakdown following MCAo.
Collapse
Affiliation(s)
- R M Weston
- Department of Pharmacology, Monash University Clayton, Australia
- Howard Florey Institute, The University of Melbourne Parkville, Australia
| | - B Jarrott
- Howard Florey Institute, The University of Melbourne Parkville, Australia
| | - Y Ishizuka
- Department of Pharmacology, Monash University Clayton, Australia
- Department of Psychiatry, Miyazaki Medical College Kihara, Kiyotake, Miyazaki, Japan
| | - J K Callaway
- Howard Florey Institute, The University of Melbourne Parkville, Australia
- Author for correspondence:
| |
Collapse
|
43
|
Margaill I, Plotkine M, Lerouet D. Antioxidant strategies in the treatment of stroke. Free Radic Biol Med 2005; 39:429-43. [PMID: 16043015 DOI: 10.1016/j.freeradbiomed.2005.05.003] [Citation(s) in RCA: 305] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2004] [Revised: 04/19/2005] [Accepted: 05/04/2005] [Indexed: 11/22/2022]
Abstract
Excessive production of free radicals is known to lead to cell injury in a variety of diseases, such as cerebral ischemia. In this review, we describe some of the numerous studies that have examined this oxidative stress and the efficiency of antioxidant strategies in focal cerebral ischemia. Besides using genetically modified mice, these strategies can be divided into three groups: (1) inhibition of free radical production, (2) scavenging of free radicals, and (3) increase of free radical degradation by using agents mimicking the enzymatic activity of endogenous antioxidants. Finally, the clinical trials that have tested or are currently testing the efficiency of antioxidants in patients suffering from stroke are reviewed. The results presented here lead us to consider that antioxidants are very promising drugs for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Isabelle Margaill
- Faculté des Sciences Pharmaceutiques et Biologiques, Laboratoire de Pharmacologie (UPRES EA 2510), Université René Descartes, 4 avenue de l'Observatoire, 75006 Paris, France.
| | | | | |
Collapse
|
44
|
Pétrault O, Ouk T, Gautier S, Laprais M, Gelé P, Bastide M, Bordet R. Pharmacological neutropenia prevents endothelial dysfunction but not smooth muscle functions impairment induced by middle cerebral artery occlusion. Br J Pharmacol 2005; 144:1051-8. [PMID: 15700030 PMCID: PMC1576087 DOI: 10.1038/sj.bjp.0706124] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
1. The polymorphonuclear neutrophils (PMN) activation and mobilization observed in acute cerebral infarction contribute to the brain tissue damage, but PMN could also be involved in postischemic functional injury of ischemied blood vessel. 2. This study was undertaken to investigate whether pharmacological neutropenia could modify the postischemic endothelial dysfunction in comparison to smooth muscle whose impairment is likely more related to reperfusion and oxidative stress. 3. A cerebral ischemia-reperfusion by endoluminal occlusion of right middle cerebral artery (MCA) was performed 4 days after intravenous administration of vinblastine or 12 h after RP-3 anti-rat neutrophils monoclonal antibody (mAb RP-3) injection into the peritoneal cavity, on male Wistar rats with 1-h ischemia then followed by 24-h reperfusion period. Brain infarct volume was measured by histomorphometric analysis and vascular endothelial and smooth muscle reactivity of MCA was analysed using Halpern myograph. 4. Neutropenia induced a neuroprotective effect as demonstrated by a significant decrease of brain infarct size. In parallel to neuroprotection, neutropenia prevented postischemic impairment of endothelium-dependent relaxing response to acetylcholine. In contrast, smooth muscle functional alterations were not prevented by neutropenia. Ischemia-reperfusion-induced myogenic tone impairment remained unchanged in vinblastine and mAb RP-3-treated rats. Postischemic Kir2.x-dependent relaxation impairment was not prevented in neutropenic conditions. The fully relaxation of smooth muscle response to sodium nitroprusside was similar in all groups. 5. Our results evidenced the dissociate prevention of pharmacologically induced neutropenia on postischemic vascular endothelial and smooth muscle impairment. The selective endothelial protection by neutropenia is parallel to a neuroprotective effect suggesting a possible relationship between the two phenomena.
Collapse
MESH Headings
- Animals
- Dose-Response Relationship, Drug
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/physiology
- Infarction, Middle Cerebral Artery/physiopathology
- Infarction, Middle Cerebral Artery/prevention & control
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Neutropenia/chemically induced
- Neutropenia/physiopathology
- Rats
- Rats, Wistar
- Vinblastine/toxicity
Collapse
Affiliation(s)
- Olivier Pétrault
- EA 1046-Laboratoire de Pharmacologie, Institut de Médecine Prédictive et de Recherche Thérapeutique, Université de Lille 2 et Centre Hospitalier Universitaire de Lille, 1 place de Verdun 59045, Lille Cedex, France
| | - Thavarak Ouk
- EA 1046-Laboratoire de Pharmacologie, Institut de Médecine Prédictive et de Recherche Thérapeutique, Université de Lille 2 et Centre Hospitalier Universitaire de Lille, 1 place de Verdun 59045, Lille Cedex, France
| | - Sophie Gautier
- EA 1046-Laboratoire de Pharmacologie, Institut de Médecine Prédictive et de Recherche Thérapeutique, Université de Lille 2 et Centre Hospitalier Universitaire de Lille, 1 place de Verdun 59045, Lille Cedex, France
| | - Maud Laprais
- EA 1046-Laboratoire de Pharmacologie, Institut de Médecine Prédictive et de Recherche Thérapeutique, Université de Lille 2 et Centre Hospitalier Universitaire de Lille, 1 place de Verdun 59045, Lille Cedex, France
| | - Patrick Gelé
- EA 1046-Laboratoire de Pharmacologie, Institut de Médecine Prédictive et de Recherche Thérapeutique, Université de Lille 2 et Centre Hospitalier Universitaire de Lille, 1 place de Verdun 59045, Lille Cedex, France
| | - Michèle Bastide
- EA 1046-Laboratoire de Pharmacologie, Institut de Médecine Prédictive et de Recherche Thérapeutique, Université de Lille 2 et Centre Hospitalier Universitaire de Lille, 1 place de Verdun 59045, Lille Cedex, France
- IUT A, Université Sciences et Techniques de Lille, Villeneuve d'Ascq, France
| | - Régis Bordet
- EA 1046-Laboratoire de Pharmacologie, Institut de Médecine Prédictive et de Recherche Thérapeutique, Université de Lille 2 et Centre Hospitalier Universitaire de Lille, 1 place de Verdun 59045, Lille Cedex, France
- Author for correspondence:
| |
Collapse
|
45
|
Ambrosini A, Louin G, Croci N, Plotkine M, Jafarian-Tehrani M. Characterization of a rat model to study acute neuroinflammation on histopathological, biochemical and functional outcomes. J Neurosci Methods 2004; 144:183-91. [PMID: 15910976 DOI: 10.1016/j.jneumeth.2004.11.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2004] [Revised: 11/01/2004] [Accepted: 11/04/2004] [Indexed: 11/22/2022]
Abstract
Neuroinflammation is one of the events occurring after acute brain injuries. The aim of the present report was to characterize a rat model to study acute neuroinflammation on the histopathological, biochemical and functional outcomes. Lipopolysaccharide (LPS), known as a strong immunostimulant, was directly injected into the hippocampus. The spatiotemporal evolution of inducible NOS (iNOS) and cell death was studied from 6 h to 7 days. A perfect time course correlation was observed between iNOS immunoreactivity and iNOS activity showing an acute, expansive and transient iNOS induction in the hippocampus with a peak at 24 h. It was associated with a marked increase in NO metabolite (NO(x)) levels, and a high level of myeloperoxidase (MPO) activity. This inflammation precedes a massive cellular loss including at least neurons and astrocytes, and a drop of constitutive NOS activity, restrictive to the ipsilateral hippocampus from 48 h after LPS injection. Moreover, sensorimotor function impairment occurred from 24 h to 7 days with a maximum at 24 h post-LPS injection. Therefore, we characterized an in vivo model of acute neuroinflammation and neurodegeneration, in relation with a neurological deficit, which may be a powerful tool for mechanistic studies and for further evaluation of the potential neuroprotective agents.
Collapse
Affiliation(s)
- Andrea Ambrosini
- Laboratoire de Pharmacologie (UPRES EA 2510), Université René Descartes, 4, avenue de l'Observatoire, 75270 Paris cedex 06, France
| | | | | | | | | |
Collapse
|
46
|
Kazmierski R, Guzik P, Ambrosius W, Ciesielska A, Moskal J, Kozubski W. Predictive value of white blood cell count on admission for in-hospital mortality in acute stroke patients. Clin Neurol Neurosurg 2004; 107:38-43. [PMID: 15567551 DOI: 10.1016/j.clineuro.2004.03.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2003] [Revised: 03/03/2004] [Accepted: 03/18/2004] [Indexed: 11/21/2022]
Abstract
OBJECTIVE In the present study, we sought to determine the predictive value of white blood cell (WBC) count measured on admission for in-hospital death in acute stroke patients. METHODS WBC count was measured automatically in 400 consecutive acute stroke patients (67.5 +/- 12.9 years old; 226 female) on admission to hospital. Patients included into the study had symptoms starting less than 12 h prior to hospitalization and no known causes of inflammation. Logistic regression adjusted for age, gender, the presence of diabetes, hypertension, atrial fibrillation, previous stroke and ischemic heart disease was used for the calculation of odds ratio (OR) with 95% confidence interval (CI) for in-hospital mortality. RESULTS Stroke patients with WBC counts in the third tertile (over 9.7 x 10(3) microL(-1)) had more than eight times (OR: 8.26; 95% CI: 3.95-17.25; P < 0.0001) increased risk of in-hospital mortality as compared with the rest of the patients. The WBC count increment of every 1000 cell/microL was associated with OR for in-hospital death of 1.27 (95% CI: 1.17-1.39; P < 0.0001). There was also a significant correlation between patients' WBC count and their clinical condition and degree of disability at the time of admission to hospital. CONCLUSION An increased WBC count within the first 12 h of onset of an ischemic stroke is a strong prognostic factor for in-hospital mortality.
Collapse
Affiliation(s)
- Radoslaw Kazmierski
- Department of Neurology, University of Medical Sciences, Przybyszewskiego 49, PL 60-355 Poznañ, Poland.
| | | | | | | | | | | |
Collapse
|