1
|
Sharma HS, Feng L, Muresanu DF, Castellani RJ, Sharma A. Neuroprotective effects of a potent bradykinin B2 receptor antagonist HOE-140 on microvascular permeability, blood flow disturbances, edema formation, cell injury and nitric oxide synthase upregulation following trauma to the spinal cord. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 146:103-152. [DOI: 10.1016/bs.irn.2019.06.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
2
|
Enhancement of bradykinin-induced relaxation by focal brain ischemia in the rat middle cerebral artery: Receptor expression upregulation and activation of multiple pathways. PLoS One 2018; 13:e0198553. [PMID: 29912902 PMCID: PMC6005516 DOI: 10.1371/journal.pone.0198553] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 05/21/2018] [Indexed: 01/06/2023] Open
Abstract
Focal brain ischemia markedly affects cerebrovascular reactivity. So far, these changes have mainly been related to alterations in the level of smooth muscle cell function while alterations of the endothelial lining have not yet been studied in detail. We have, therefore, investigated the effects of ischemia/reperfusion injury on bradykinin (BK)-induced relaxation since BK is an important mediator of tissue inflammation and affects vascular function in an endothelium-dependent manner. Focal brain ischemia was induced in rats by endovascular filament occlusion (2h) of the middle cerebral artery (MCA). After 22h reperfusion, both MCAs were harvested and the response to BK studied in organ bath experiments. Expression of the BK receptor subtypes 1 and 2 (B1, B2) was determined by real-time semi-quantitative RT-qPCR methodology, and whole mount immunofluorescence staining was performed to show the B2 receptor protein expression. In control animals, BK did not induce significant vasomotor effects despite a functionally intact endothelium and robust expression of B2 mRNA. After ischemia/reperfusion injury, BK induced a concentration-related sustained relaxation in all arteries studied, more pronounced in the ipsilateral than in the contralateral MCA. The B2 mRNA was significantly upregulated and the B1 mRNA displayed de novo expression, again more pronounced ipsi- than contralaterally. Endothelial cells displaying B2 receptor immunofluorescence were observed scattered or clustered in previously occluded MCAs. Relaxation to BK was mediated by B2 receptor activation, abolished after endothelium denudation, and largely diminished by blocking nitric oxide (NO) release or soluble guanylyl cyclase activity. Relaxation to BK was partially inhibited by charybdotoxin (ChTx), but not apamin or iberiotoxin suggesting activation of an endothelium-dependent hyperpolarization pathway. When the NO-cGMP pathway was blocked, BK induced a transient relaxation which was suppressed by ChTx. After ischemia/reperfusion injury BK elicits endothelium-dependent relaxation which was not detectable in control MCAs. This gain of function is mediated by B2 receptor activation and involves the release of NO and activation of an endothelium-dependent hyperpolarization. It goes along with increased B2 mRNA and protein expression, leaving the functional role of the de novo B1 receptor expression still open.
Collapse
|
3
|
Sang H, Liu L, Wang L, Qiu Z, Li M, Yu L, Zhang H, Shi R, Yu S, Guo R, Ye R, Liu X, Zhang R. Opposite roles of bradykinin B1 and B2 receptors during cerebral ischaemia-reperfusion injury in experimental diabetic rats. Eur J Neurosci 2016; 43:53-65. [PMID: 26565562 DOI: 10.1111/ejn.13133] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 10/29/2015] [Accepted: 11/06/2015] [Indexed: 02/05/2023]
Affiliation(s)
- Hongfei Sang
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Ling Liu
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Liumin Wang
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Zhongming Qiu
- Department of Neurology; The 117th Hospital of PLA; Xihu District Hangzhou Zhejiang Province China
| | - Min Li
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Linjie Yu
- Nanjing University School of Medicine; Nanjing China
| | - Hao Zhang
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Ruifeng Shi
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Shuhong Yu
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Ruibing Guo
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Ruidong Ye
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Xinfeng Liu
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Renliang Zhang
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| |
Collapse
|
4
|
Lalkovičová M, Bonová P, Burda J, Danielisová V. Effect of Bradykinin Postconditioning on Ischemic and Toxic Brain Damage. Neurochem Res 2015. [PMID: 26216051 PMCID: PMC4536273 DOI: 10.1007/s11064-015-1675-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Brain damage caused by ischemia or toxic agents leads in selectively vulnerable regions to apoptosis-like delayed neuronal death and can result in irreversible damage. Selectively vulnerable neurons of the CA1 area of hippocampus are particularly sensitive to ischemic damage. We investigated the effects of bradykinin (BR) postconditioning on cerebral ischemic and toxic injury. Transient forebrain ischemia was induced by four-vessel occlusion for 10 min and toxic injury was induced by trimethyltin (TMT, 8 µg/kg i.p.). BR as a postconditioner at a dose of 150 µg/kg was applied intraperitoneally 48 h after ischemia or TMT intoxication. Experimental animals were divided into groups according to the length of survival (short—3 and 7 days, and long—28 days survival) and according to the applied ischemic or toxic injury. Glutamate concentration was lowered in both CA1 and dentate gyrus areas of hippocampus after the application of BR postconditioning in both ischemic and toxic brain damage. The number of degenerated neurons in the hippocampal CA1 region was significantly lower in BR-treated ischemic and toxic groups compared to vehicle group. The behavioral test used in our experiments confirms also the memory improvement in conditioned animals. The rats’ ability to form spatial maps and learn was preserved, which is visible from our Barnes maze results. By using the methods of delayed postconditioning is possible to stimulate the endogenous protective mechanisms of the organism and induce the neuroprotective effect. In this study we demonstrated that BR postconditioning, if applied before the onset of irreversible neurodegenerative changes, induced neuroprotection against ischemic or toxic injury.
Collapse
Affiliation(s)
- Mária Lalkovičová
- Department of Neurochemistry, Institute of Neurobiology, Slovak Academy of Sciences, Šoltésovej 4-6, 04001, Kosice, Slovak Republic,
| | | | | | | |
Collapse
|
5
|
Ay I, Napadow V, Ay H. Electrical stimulation of the vagus nerve dermatome in the external ear is protective in rat cerebral ischemia. Brain Stimul 2014; 8:7-12. [PMID: 25312600 DOI: 10.1016/j.brs.2014.09.009] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 09/11/2014] [Accepted: 09/17/2014] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Although cervical vagus nerve stimulation is effective for reducing infarct volume in rats, it is not feasible for acute human stroke as it requires surgical incision of the neck. We hypothesized that stimulation of the dermatome in the external ear innervated by the vagus nerve (auricular vagus nerve stimulation; aVNS) reduces infarct volume after transient focal ischemia in rats. METHODS Animals were randomized to active aVNS or sham stimulation. For aVNS, electrical stimulation of the left cavum concha (1 h duration) using percutaneous needles was initiated 30 min after induction of ischemia. Behavioral and tissue outcome were measured 24 h after induction of ischemia. In a separate experimental dataset, c-Fos immunohistochemistry was performed to identify the brain regions activated after the stimulation. RESULTS Stimulation of the left cavum concha resulted in bilateral c-Fos staining in the nuclei tractus solitarii and the loci coerulei in all animals. There was no c-Fos staining in any part of the brainstem in sham control animals. The mean infarct volume (SD) as calculated by indirect method was 44.20 ± 7.58% in controls and 31.65 ± 9.67% in treated animals (P < 0.0001). The effect of aVNS on tissue outcome was associated with better neurological scores at 24 h after ischemia (P < 0.0001). CONCLUSIONS Electric stimulation of the vagus nerve dermatome in the external ear activates brainstem afferent vagal nuclei and reduces infarct volume in rats. This finding has potential to facilitate the development of treatments that leverage the brain's endogenous neuroprotective pathways at the setting of acute ischemic stroke.
Collapse
Affiliation(s)
- Ilknur Ay
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital East, 149 13th Street, Charlestown, MA 02129, USA.
| | - Vitaly Napadow
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital East, 149 13th Street, Charlestown, MA 02129, USA; Department of Biomedical Engineering, Kyunghee University, Yongin, South Korea
| | - Hakan Ay
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital East, 149 13th Street, Charlestown, MA 02129, USA
| |
Collapse
|
6
|
Albert-Weißenberger C, Sirén AL, Kleinschnitz C. Ischemic stroke and traumatic brain injury: the role of the kallikrein-kinin system. Prog Neurobiol 2012; 101-102:65-82. [PMID: 23274649 DOI: 10.1016/j.pneurobio.2012.11.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 11/15/2012] [Accepted: 11/20/2012] [Indexed: 12/16/2022]
Abstract
Acute ischemic stroke and traumatic brain injury are a major cause of mortality and morbidity. Due to the paucity of therapies, there is a pressing clinical demand for new treatment options. Successful therapeutic strategies for these conditions must target multiple pathophysiological mechanisms occurring at different stages of brain injury. In this respect, the kallikrein-kinin system is an ideal target linking key pathological hallmarks of ischemic and traumatic brain damage such as edema formation, inflammation, and thrombosis. In particular, the kinin receptors, plasma kallikrein, and coagulation factor XIIa are highly attractive candidates for pharmacological development, as kinin receptor antagonists or inhibitors of plasma kallikrein and coagulation factor XIIa are neuroprotective in animal models of stroke and traumatic brain injury. Nevertheless, conflicting preclinical evaluation as well as limited and inconclusive data from clinical trials suggest caution when transferring observations made in animals into the human situation. This review summarizes current evidence on the pathological significance of the kallikrein-kinin system during ischemic and traumatic brain damage, with a particular focus on experimental data derived from animal models. Experimental findings are also compared with human data if available, and potential therapeutic implications are discussed.
Collapse
|
7
|
Ay I, Ay H. Ablation of the sphenopalatine ganglion does not attenuate the infarct reducing effect of vagus nerve stimulation. Auton Neurosci 2012; 174:31-5. [PMID: 23273773 DOI: 10.1016/j.autneu.2012.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 11/19/2012] [Accepted: 12/03/2012] [Indexed: 12/28/2022]
Abstract
Electrical stimulation of the cervical vagus nerve reduces infarct size by approximately 50% after cerebral ischemia in rats. The mechanism of ischemic protection by vagus nerve stimulation (VNS) is not known. In this study, we investigated whether the infarct reducing effect of VNS was mediated by activation of the parasympathetic vasodilator fibers that originate from the sphenopalatine ganglion (SPG) and innervate the anterior cerebral circulation. We examined the effects of electrical stimulation of the cervical vagus nerve in two groups of rats: one with and one without SPG ablation. Electrical stimulation was initiated 30 min after induction of ischemia, and lasted for 1h. Measurement of infarct size 24h later revealed that the volume of ischemic damage was smaller in those animals that received VNS treatment (41.32±2.07% vs. 24.19±2.62% of the contralateral hemispheric volume, n=6 in both; p<0.05). SPG ablation did not abolish this effect; the reduction in infarct volume following VNS was 58% in SPG-damaged animals, 41% in SPG-intact animals (p>0.05). In both SPG-intact and SPG-damaged animals VNS treatment resulted in better motor outcome (p<0.05 vs. corresponding controls for both). Our findings show that VNS can protect the brain against acute ischemic injury, and that this effect is not mediated by SPG projections.
Collapse
Affiliation(s)
- Ilknur Ay
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital East, Charlestown, MA 02129, USA.
| | | |
Collapse
|
8
|
Schöller K, Feiler S, Anetsberger S, Kim SW, Plesnila N. Contribution of Bradykinin Receptors to the Development of Secondary Brain Damage After Experimental Subarachnoid Hemorrhage. Neurosurgery 2011; 68:1118-23. [DOI: 10.1227/neu.0b013e31820a0024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Karsten Schöller
- Department of Neurosurgery and University of Munich Medical Center–Grosshadern, Ludwig-Maximilians University, Munich, Germany
- Institute for Surgical Research, University of Munich Medical Center–Grosshadern, Ludwig-Maximilians University, Munich, Germany
| | - Sergej Feiler
- Department of Neurosurgery and University of Munich Medical Center–Grosshadern, Ludwig-Maximilians University, Munich, Germany
- Institute for Surgical Research, University of Munich Medical Center–Grosshadern, Ludwig-Maximilians University, Munich, Germany
| | - Stephanie Anetsberger
- Institute for Surgical Research, University of Munich Medical Center–Grosshadern, Ludwig-Maximilians University, Munich, Germany
| | - Seong-Woong Kim
- Institute for Surgical Research, University of Munich Medical Center–Grosshadern, Ludwig-Maximilians University, Munich, Germany
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Nikolaus Plesnila
- Department of Neurosurgery and University of Munich Medical Center–Grosshadern, Ludwig-Maximilians University, Munich, Germany
- Institute for Surgical Research, University of Munich Medical Center–Grosshadern, Ludwig-Maximilians University, Munich, Germany
- Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
9
|
Bhaskar S, Tian F, Stoeger T, Kreyling W, de la Fuente JM, Grazú V, Borm P, Estrada G, Ntziachristos V, Razansky D. Multifunctional Nanocarriers for diagnostics, drug delivery and targeted treatment across blood-brain barrier: perspectives on tracking and neuroimaging. Part Fibre Toxicol 2010; 7:3. [PMID: 20199661 PMCID: PMC2847536 DOI: 10.1186/1743-8977-7-3] [Citation(s) in RCA: 281] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Accepted: 03/03/2010] [Indexed: 01/03/2023] Open
Abstract
Nanotechnology has brought a variety of new possibilities into biological discovery and clinical practice. In particular, nano-scaled carriers have revolutionalized drug delivery, allowing for therapeutic agents to be selectively targeted on an organ, tissue and cell specific level, also minimizing exposure of healthy tissue to drugs. In this review we discuss and analyze three issues, which are considered to be at the core of nano-scaled drug delivery systems, namely functionalization of nanocarriers, delivery to target organs and in vivo imaging. The latest developments on highly specific conjugation strategies that are used to attach biomolecules to the surface of nanoparticles (NP) are first reviewed. Besides drug carrying capabilities, the functionalization of nanocarriers also facilitate their transport to primary target organs. We highlight the leading advantage of nanocarriers, i.e. their ability to cross the blood-brain barrier (BBB), a tightly packed layer of endothelial cells surrounding the brain that prevents high-molecular weight molecules from entering the brain. The BBB has several transport molecules such as growth factors, insulin and transferrin that can potentially increase the efficiency and kinetics of brain-targeting nanocarriers. Potential treatments for common neurological disorders, such as stroke, tumours and Alzheimer's, are therefore a much sought-after application of nanomedicine. Likewise any other drug delivery system, a number of parameters need to be registered once functionalized NPs are administered, for instance their efficiency in organ-selective targeting, bioaccumulation and excretion. Finally, direct in vivo imaging of nanomaterials is an exciting recent field that can provide real-time tracking of those nanocarriers. We review a range of systems suitable for in vivo imaging and monitoring of drug delivery, with an emphasis on most recently introduced molecular imaging modalities based on optical and hybrid contrast, such as fluorescent protein tomography and multispectral optoacoustic tomography. Overall, great potential is foreseen for nanocarriers in medical diagnostics, therapeutics and molecular targeting. A proposed roadmap for ongoing and future research directions is therefore discussed in detail with emphasis on the development of novel approaches for functionalization, targeting and imaging of nano-based drug delivery systems, a cutting-edge technology poised to change the ways medicine is administered.
Collapse
Affiliation(s)
- Sonu Bhaskar
- Instituto Universitario de Nanociencia de Aragón (INA), Universidad de Zaragoza, Zaragoza, Spain
- Zaragoza University Hospital-Miguel Servet, and Instituto Aragonés de Ciencias de la Salud (I+CS), Zaragoza, Spain
| | - Furong Tian
- Comprehensive Pneumology Centre, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Neuherberg, Germany
| | - Tobias Stoeger
- Comprehensive Pneumology Centre, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Neuherberg, Germany
| | - Wolfgang Kreyling
- Comprehensive Pneumology Centre, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jesús M de la Fuente
- Instituto Universitario de Nanociencia de Aragón (INA), Universidad de Zaragoza, Zaragoza, Spain
| | - Valeria Grazú
- Instituto Universitario de Nanociencia de Aragón (INA), Universidad de Zaragoza, Zaragoza, Spain
| | - Paul Borm
- Centre of Expertise in Life Sciences, Zuyd University, Heerlen, the Netherlands
| | - Giovani Estrada
- Institute of Bioinformatics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Vasilis Ntziachristos
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, and Technische Universität München, Germany
| | - Daniel Razansky
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, and Technische Universität München, Germany
| |
Collapse
|
10
|
Kim D, Cho SH, Kim JS, Jo SH, Lee SJ, Kim KT, Choi SY. Human astrocytic bradykinin B(2) receptor modulates zymosan-induced cytokine expression in 1321N1 cells. Peptides 2010; 31:101-7. [PMID: 19854233 DOI: 10.1016/j.peptides.2009.10.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Revised: 10/14/2009] [Accepted: 10/14/2009] [Indexed: 11/17/2022]
Abstract
Bradykinin is an important modulator of the neurons and glial cells of the nervous system. Bradykinin secreted from neurons affects astrocytic functions such as neurovascular coupling and astrocytic cytokine production. In human astrocytes, however, the detailed mechanism of bradykinin-mediated modulation of astrocytic functions has not yet been fully elucidated. Here, we report the functional expression of the bradykinin B(2) receptor and its modulation of zymosan-induced cytokine expression in human astrocytoma 1321N1 cells. Bradykinin increased cytosolic [Ca(2+)] in a concentration-dependent manner, whereas [des-Arg(10)] kallidin (an agonist of the B(1) receptor) did not have this effect. Bradykinin also triggered intracellular InsP(3) production. Pretreating the cells with HOE140 (icatibant acetate, a B(2) receptor antagonist) inhibited the bradykinin-induced increase in cytosolic [Ca(2+)] and InsP(3) production. In contrast, [des-Arg(10)]HOE140 (a B(1) receptor antagonist) did not show any inhibitory effect. Bradykinin increased the zymosan-induced expression of TNF-alpha, and interleukin 1beta (IL-1beta) but did not affect the expression of interleukin 6 (IL-6) or interleukin 10 (IL-10). Interestingly, a cyclooxygenase-2 specific inhibitor blocked the bradykinin-induced effect. In contrast to the result in human glioma cells, bradykinin inhibits the zymosan-induced expression of TNF-alpha and IL-1beta in rat astrocytes, which shows a species-dependent manner. These data suggest that bradykinin B(2) receptors are expressed in human astrocytoma cells and that they modulate the expression pattern of inflammatory cytokines.
Collapse
Affiliation(s)
- Donghoon Kim
- Department of Physiology and Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
11
|
Thal SC, Sporer S, Schmid-Elsaesser R, Plesnila N, Zausinger S. Inhibition of bradykinin B2 receptors before, not after onset of experimental subarachnoid hemorrhage prevents brain edema formation and improves functional outcome. Crit Care Med 2009; 37:2228-34. [DOI: 10.1097/ccm.0b013e3181a068fc] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
12
|
Endothelin-1 induced MCAO: dose dependency of cerebral blood flow. J Neurosci Methods 2009; 179:22-8. [PMID: 19428507 DOI: 10.1016/j.jneumeth.2009.01.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Revised: 12/22/2008] [Accepted: 01/08/2009] [Indexed: 12/31/2022]
Abstract
The purpose of this study was to characterize the magnitude and duration of cerebral blood flow (CBF) reduction in the somatosensory cortical region in a rat model of middle cerebral artery occlusion (MCAO) induced by endothelin-1 (ET1) microinjection under isoflurane anesthesia. MCAO was induced by microinjection of ET1 proximal to the MCA in 41 isoflurane-anesthetized male Sprague-Dawley rats. Three doses of ET1 were studied, 60 pmol (Group 1), 150 pmol (Group 2), and 300 pmol (Group 3). CBF was monitored for 4h following injection using a laser Doppler probe stereotaxically inserted into the left somatosensory cortical region. Computed tomography perfusion imaging was used to verify the extent and duration of blood flow reduction in a subset of 12 animals. The magnitude and duration of blood flow reduction was variable (60-92% of baseline). The 300 pmol dose provided the greatest sustained decrease in blood flow. Evidence of tissue damage was obtained in cases where CBF decreased to <40% of baseline. At the doses studied, ET1-induced ischemia in the presence of isoflurane anesthesia can be used as a minimally invasive but variable model of MCAO. The model is well suited for acute imaging studies of ischemia.
Collapse
|
13
|
Chiang WC, Chien CT, Lin WW, Lin SL, Chen YM, Lai CF, Wu KD, Chao J, Tsai TJ. Early activation of bradykinin B2 receptor aggravates reactive oxygen species generation and renal damage in ischemia/reperfusion injury. Free Radic Biol Med 2006; 41:1304-14. [PMID: 17015177 DOI: 10.1016/j.freeradbiomed.2006.07.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2006] [Revised: 06/09/2006] [Accepted: 07/11/2006] [Indexed: 11/20/2022]
Abstract
The kallikrein/kinin system is beneficial in ischemia/reperfusion injury in heart, controversial in brain, but detrimental in lung, liver, and intestine. We examined the role of the kallikrein/kinin system in acute ischemia/reperfusion renal injury induced by 40 min occlusion of the renal artery followed by reperfusion. Rats were infused with tissue kallikrein protein 5 days before (pretreated group) or after (treated group) ischemia. Two days later, the pretreated group exhibited the worst renal dysfunction, followed by the treated group, then the control group. Kallikrein increased tubular necrosis and inflammatory cell infiltration with generation of more tumor necrosis factor-alpha and monocyte chemoattractant protein-1. Reactive oxygen species (ROS), malondialdehyde, and reduced/oxidized glutathione measurement revealed that the oxidative stress was augmented by kallikrein administration in both ischemic and reperfusion phases. The groups with more ROS generation also had more apoptotic renal cells. The deleterious effects of kallikrein on ischemia/reperfusion injury were reversed by cotreatment with bradykinin B2 receptor (B2R) antagonist, but not B1 receptor antagonist, and were not associated with hemodynamic changes. We conclude that early activation of B2R augmented ROS generation in ischemia/reperfusion renal injury, resulting in subsequent apoptosis, inflammation, and tissue damage. This finding suggests the potential application of B2R antagonists in acute ischemic renal disease associated with bradykinin activation.
Collapse
Affiliation(s)
- Wen-Chih Chiang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10016, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Storini C, Bergamaschini L, Gesuete R, Rossi E, Maiocchi D, De Simoni MG. Selective inhibition of plasma kallikrein protects brain from reperfusion injury. J Pharmacol Exp Ther 2006; 318:849-54. [PMID: 16705080 DOI: 10.1124/jpet.106.105064] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have studied the effect of DX-88, a selective recombinant inhibitor of human plasma kallikrein, in transient or permanent focal brain ischemia (with or without reperfusion, respectively) induced in C57BL/6 mice. Twenty-four hours after transient ischemia, DX-88 administered at the beginning of ischemia (pre) induced a dose-dependent reduction of ischemic volume that, at the dose of 30 microg/mouse, reached 49% of the volume of saline-treated mice. At the same dose, DX-88 was also able to reduce brain swelling to 32%. Mice treated with DX-88 pre had significantly lower general and focal deficit score. Fluoro-Jade staining, a marker for neuronal degeneration, showed that DX-88-treated mice had a reduction in the number of degenerating cells, compared with saline-treated mice. Seven days after transient ischemia, the DX-88 protective effect was still present. When the inhibitor was injected at the end of ischemia (post), it was still able to reduce ischemic volume, brain swelling, and neurological deficits. DX-88 efficacy was lost when the inhibitor was given 30 min after the beginning of reperfusion (1 h post) or when reperfusion was not present (permanent occlusion model). This study shows that DX-88 has a strong neuroprotective effect in the early phases of brain ischemia preventing reperfusion injury and indicates that inhibition of plasma kallikrein may be a useful tool in the strategy aimed at reducing the detrimental effects linked to reperfusion.
Collapse
Affiliation(s)
- Claudio Storini
- Department of Neuroscience, Mario Negri Institute, Milan, Italy
| | | | | | | | | | | |
Collapse
|