1
|
Erdling A, Johansson SE, Radziwon‐Balicka A, Ansar S, Edvinsson L. Changes in P2Y 6 receptor-mediated vasoreactivity following focal and global ischemia. Physiol Rep 2022; 10:e15283. [PMID: 35466569 PMCID: PMC9035753 DOI: 10.14814/phy2.15283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 06/14/2023] Open
Abstract
Ischemia, both in the form of focal thromboembolic stroke and following subarachnoid hemorrhage (SAH), causes upregulation of vasoconstrictive receptor systems within the cerebral vasculature. Descriptions regarding changes in purinergic signaling following ischemia are lacking, especially when the importance of purinergic signaling in regulating vascular tone is taken into consideration. This prompted us to evaluate changes in P2Y6 -mediated vasomotor reactivity in two different stroke models in rat. We used wire myography to measure changes in cerebral vasoreactivity to the P2Y6 agonist UDP-β-S following either experimental SAH or transient middle cerebral artery occlusion. Changes in receptor localization or receptor expression were evaluated using immunohistochemistry and quantitative flow cytometry. Transient middle cerebral artery occlusion caused an increase in Emax when compared to sham (233.6 [206.1-258.5]% vs. 161.1 [147.1-242.6]%, p = 0.0365). No such change was seen following SAH. Both stroke models were associated with increased levels of P2Y6 receptor expression in the vascular smooth muscle cells (90.94 [86.99-99.15]% and 93.79 [89.96-96.39]% vs. 80.31 [70.80-80.86]%, p = 0.021) and p = 0.039 respectively. There was no change in receptor localization in either of the stroke models. Based on these findings, we conclude that focal ischemic stroke increases vascular sensitivity to UDP-β-S by upregulating P2Y6 receptors on vascular smooth muscle cells while experimental SAH did not induce changes in vasoreactivity in spite of increased P2Y6 receptor expression.
Collapse
Affiliation(s)
- André Erdling
- Department of Clinical SciencesDivision of Experimental Vascular ResearchLund UniversityLundSweden
- Department of Cardiothoracic Surgery, Anesthesiology and Intensive CareSkane University HospitalLundSweden
- Applied Neurovascular ResearchDepartment of Clinical SciencesLund UniversityLundSweden
| | - Sara Ellinor Johansson
- Department of Clinical Experimental ResearchGlostrup Research InstituteRigshospitalet‐GlostrupGlostrupDenmark
| | - Aneta Radziwon‐Balicka
- Department of Clinical Experimental ResearchGlostrup Research InstituteRigshospitalet‐GlostrupGlostrupDenmark
| | - Saema Ansar
- Applied Neurovascular ResearchDepartment of Clinical SciencesLund UniversityLundSweden
| | - Lars Edvinsson
- Department of Clinical SciencesDivision of Experimental Vascular ResearchLund UniversityLundSweden
- Department of Clinical Experimental ResearchGlostrup Research InstituteRigshospitalet‐GlostrupGlostrupDenmark
| |
Collapse
|
2
|
Nussbaum ES, Mikoff N, Paranjape GS. Cognitive deficits among patients surviving aneurysmal subarachnoid hemorrhage. A contemporary systematic review. Br J Neurosurg 2020; 35:384-401. [PMID: 33345644 DOI: 10.1080/02688697.2020.1859462] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) is associated with high rates of morbidity, including neurological and cognitive deficits that may be difficult to identify and quantify. This review provides an update on the cognitive deficits that may result from spontaneous aneurysmal SAH (aSAH) and identifies factors that may help predict and manage these deficits at discharge and thereafter. MATERIALS AND METHODS We conducted a systematic review of PubMed and Google Scholar to identify studies published between 2010 and 2019 that assessed cognitive deficits at discharge and during follow-up in patients with aSAH. Full-text articles were assessed for information regarding cognitive testing and factors that may be associated with functional outcomes in this population. RESULTS We reviewed 65 studies published since 2010 that described the cognitive deficits associated with non-traumatic aSAH. Such deficits may impact functional outcomes, quality of life, and return to work and may result in cognitive impairments, such as memory difficulties, speech problems, and psychiatric disorders. CONCLUSIONS Patients with aSAH, even those that appear normal at the time of hospital discharge, may harbor cognitive deficits that are difficult to detect, yet can interfere with daily functioning. Further research is needed to provide additional information and to identify stronger correlations to be used in the identification, treatment, and amelioration of long-term cognitive deficits in aSAH patients, including those who are discharged with good clinical outcomes scores.
Collapse
Affiliation(s)
- Eric S Nussbaum
- Department of Neurosurgery, National Brain Aneurysm & Tumor Center, United Hospital, Twin Cities, MN, USA
| | | | | |
Collapse
|
3
|
Lan S, Zhou L, Wang Y, Fang L, Yang L, Zheng S, Zhou X, Tang B, Duan J, Wu X, Yang C, Hong T. miRNA Profiling of Circulating Small Extracellular Vesicles From Subarachnoid Hemorrhage Rats Using Next-Generation Sequencing. Front Cell Neurosci 2020; 14:242. [PMID: 32903819 PMCID: PMC7439219 DOI: 10.3389/fncel.2020.00242] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/13/2020] [Indexed: 12/31/2022] Open
Abstract
Background Extracellular vesicles (EVs) are produced during abnormal and normal physiological conditions. Understanding the expression profile of microRNA (miRNA) in plasma-derived small extracellular vesicles (sEVs) and their roles in subarachnoid hemorrhage (SAH) that cause cerebral vasospasm (CVS) is imperative. Methods Sprague Dawley rats (250–300 g) were allocated to sham or SAH groups established using endovascular perforation method. miRNA expression profiles of plasma sEVs in both groups (each n = 4) were evaluated using next-generation sequencing (NGS). Results There were 142 microRNAs (miRNAs) significantly expressed differently between the two groups, of which 73 were up-regulated while 69 were down-regulated in SAH sEVs compared with those of sham (p < 0.05; fold change ≥ 2). The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and Gene Ontology (GO) analyses of differently expressed (DE) miRNAs revealed signaling pathways and target genes (TGs) in the SAH group. rno-miR-185-5p, rno-miR-103-3p, rno-miR-15b-3p, rno-miR-93-5p, and rno-miR-98-5p were the top five most up-regulated sEVs miRNAs. Conclusion Our results suggest that miRNA can be selectively packaged into sEVs under SAH, and this could help develop potential targets for the prevention, diagnosis, and treatment of CVS after this condition.
Collapse
Affiliation(s)
- Shihai Lan
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lin Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yimei Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Linchun Fang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Le Yang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Suyue Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - XinHui Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bin Tang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jian Duan
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiao Wu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chengxing Yang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tao Hong
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
4
|
Zaric M, Drakulic D, Dragic M, Gusevac Stojanovic I, Mitrovic N, Grkovic I, Martinovic J. Molecular Alterations and Effects of Acute Dehydroepiandrosterone Treatment Following Brief Bilateral Common Carotid Artery Occlusion: Relevance to Transient Ischemic Attack. Neuroscience 2019; 410:128-139. [PMID: 31095985 DOI: 10.1016/j.neuroscience.2019.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/01/2019] [Accepted: 05/05/2019] [Indexed: 10/26/2022]
Abstract
Transient ischemic attack (TIA) represents brief neurological dysfunction of vascular origin without detectable infarction. Despite major clinical relevance characterization of post-TIA molecular changes using appropriate experimental model is lacking and no therapeutic agent has been established yet. Neurosteroid dehydroepiandrosterone (DHEA) arose as one of the candidates for cerebral ischemia treatment but its effects on TIA-like condition remain unknown. Seeking an animal model applicable for investigation of molecular alterations in mild ischemic conditions such as TIA, 15-min bilateral common carotid artery occlusion with 24-h reperfusion was performed to induce ischemia/ reperfusion (I/R) injury in adult male Wistar rats. Additionally, effects of 4-h post-operative DHEA treatment (20 mg/kg) were investigated in physiological and I/R conditions in hippocampus (HIP) and prefrontal cortex (PFC). The study revealed absence of sensorimotor deficits, cerebral infarcts and neurodegeneration along with preserved HIP and PFC overall neuronal morphology and unaltered malondialdehyde and reduced glutathione level following I/R and/or DHEA treatment. I/R induced nitric oxide burst in HIP and PFC was accompanied with increased neuronal nitric oxide synthase protein level exclusively in HIP. DHEA had no effects in physiological conditions, while increase of Bax/Bcl2 ratio and dissipation of mitochondrial membrane potential in treated I/R group suggested DHEA-mediated exacerbation of post-ischemic changes that might lead to pro-apoptotic events in HIP. Interestingly, DHEA restored I/R-induced NO to the control level in PFC. Obtained results indicated that I/R may serve as an appropriate model for investigation of molecular changes and treatment outcome following mild ischemic conditions such as TIA.
Collapse
Affiliation(s)
- Marina Zaric
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11351 Belgrade, Republic of Serbia.
| | - Dunja Drakulic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11351 Belgrade, Republic of Serbia
| | - Milorad Dragic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11351 Belgrade, Republic of Serbia; Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Studentski trg 3, 11001 Belgrade, Republic of Serbia
| | - Ivana Gusevac Stojanovic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11351 Belgrade, Republic of Serbia
| | - Natasa Mitrovic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11351 Belgrade, Republic of Serbia
| | - Ivana Grkovic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11351 Belgrade, Republic of Serbia
| | - Jelena Martinovic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11351 Belgrade, Republic of Serbia
| |
Collapse
|
5
|
Skovsted GF, Tveden-Nyborg P, Lykkesfeldt J. Expression of endothelin type B receptors in uterine artery smooth muscle cells from pregnant Guinea pigs. Placenta 2019; 77:8-15. [PMID: 30827357 DOI: 10.1016/j.placenta.2019.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 01/15/2019] [Accepted: 01/19/2019] [Indexed: 10/27/2022]
Abstract
INTRODUCTION It is well established that upregulation of endothelin type B (ETB) receptors in vascular smooth muscle cells plays a role in pathophysiological artery remodeling as response to ischemia and atherosclerosis. This study aimed to investigate the ETB receptors function and localization under normal physiological remodeling. Specifically, in the guinea pig uterine arteries during pregnancy. METHODS Uterine artery contractility was assessed with sarafotoxin 6c and endothelin-1 in wire-myography in uterine arteries from non-pregnant and pregnant guinea pigs at gestational day 37 ± 5. Localization of ETB receptors, proliferation marker Ki-67, and SMC differentiation marker SM22α in uterine arteries were investigated with immunohistochemistry. RESULTS Uterine arteries from pregnant guinea pigs showed significantly increased ETB receptor-mediated vasoconstriction compared to uterine arteries from non-pregnant and to coronary arteries from pregnant guinea pigs (p < 0.001), suggesting that ETB-receptor upregulation in uterine artery SMCs is a normal physiological mechanism taking place during remodeling. Furthermore, uterine arteries from pregnant guinea pigs showed enhanced expression of ETB receptors, high density of Ki-67 positive SMCs and sparse SM22α staining in SMCs localized in the outer layer of the vessel wall. DISCUSSION Our results suggest that ETB receptors are expressed in dedifferentiated proliferating SMCs of uterine arteries in pregnant guinea pigs. This study provides novel insight into the function and expression of ETB receptors in uterine vascular remodeling during pregnancy.
Collapse
Affiliation(s)
- Gry Freja Skovsted
- University of Copenhagen, Faculty of Health & Medical Sciences, Experimental Pharmacology and Toxicology, Section of Experimental Animal Models, Department of Veterinary and Animal Sciences, Ridebanevej 9 - 1st Floor, DK-1870, Frederiksberg C, Denmark.
| | - Pernille Tveden-Nyborg
- University of Copenhagen, Faculty of Health & Medical Sciences, Experimental Pharmacology and Toxicology, Section of Experimental Animal Models, Department of Veterinary and Animal Sciences, Ridebanevej 9 - 1st Floor, DK-1870, Frederiksberg C, Denmark
| | - Jens Lykkesfeldt
- University of Copenhagen, Faculty of Health & Medical Sciences, Experimental Pharmacology and Toxicology, Section of Experimental Animal Models, Department of Veterinary and Animal Sciences, Ridebanevej 9 - 1st Floor, DK-1870, Frederiksberg C, Denmark
| |
Collapse
|
6
|
Blixt FW, Haanes KA, Ohlsson L, Dreisig K, Fedulov V, Warfvinge K, Edvinsson L. MEK/ERK/1/2 sensitive vascular changes coincide with retinal functional deficit, following transient ophthalmic artery occlusion. Exp Eye Res 2018; 179:142-149. [PMID: 30439349 DOI: 10.1016/j.exer.2018.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/22/2018] [Accepted: 11/05/2018] [Indexed: 11/27/2022]
Abstract
Retinal ischemia remains a major cause of blindness in the world with few acute treatments available. Recent emphasis on retinal vasculature and the ophthalmic artery's vascular properties after ischemia has shown an increase in vasoconstrictive functionality, as previously observed in cerebral arteries following stroke. Specifically, endothelin-1 (ET-1) receptor-mediated vasoconstriction regulated by the MEK/ERK1/2 pathway. In this study, the ophthalmic artery of rats was occluded for 2 h with the middle cerebral artery occlusion model. MEK/ERK1/2 inhibitor U0126 was administered at 0, 6, and 24 h following reperfusion and the functional properties of the ophthalmic artery were evaluated at 48 h post reperfusion. Additionally, retinal function was evaluated at day 1, 4, and 7 after reperfusion. Occlusion of the ophthalmic artery led to a significant increase of endothelin-1 mediated vasoconstriction which can be attenuated by U0126 treatment, most evident at higher ET-1 concentrations of 10-7 M (Emax151.0 ± 22.0% of 60 mM K+), vs non-treated ischemic arteries Emax 212.1 ± 14.7% of 60 mM K+). Retinal function also deteriorated following ischemia and was improved with treatment with a-wave amplitudes of 725 ± 36 μV in control, 560 ± 21 μV in non-treated, and 668 ± 73 μV in U0126 treated at 2 log cd*s/m2 luminance in the acute stages (1 days post-ischemia). Full spontaneous retinal recovery was observed at day 7 regardless of treatment. In conclusion, this is the first study to show a beneficial in vivo effect of U0126 on vascular contractility following ischemia in the ophthalmic artery. Coupled with the knowledge obtained from cerebral vasculature, these results point towards a novel therapeutic approach following ischemia-related injuries to the eye.
Collapse
Affiliation(s)
- Frank W Blixt
- Department of Clinical Sciences, Division of Experimental Vascular Research, Lund University, Lund, Sweden.
| | - Kristian Agmund Haanes
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet, Glostrup, Denmark
| | - Lena Ohlsson
- Department of Clinical Sciences, Division of Experimental Vascular Research, Lund University, Lund, Sweden
| | - Karin Dreisig
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet, Glostrup, Denmark
| | - Vadim Fedulov
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet, Glostrup, Denmark
| | - Karin Warfvinge
- Department of Clinical Sciences, Division of Experimental Vascular Research, Lund University, Lund, Sweden; Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet, Glostrup, Denmark
| | - Lars Edvinsson
- Department of Clinical Sciences, Division of Experimental Vascular Research, Lund University, Lund, Sweden; Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet, Glostrup, Denmark
| |
Collapse
|
7
|
Blixt FW, Haanes KA, Ohlsson L, Tolstrup Christiansen A, Warfvinge K, Edvinsson L. Increased endothelin-1-mediated vasoconstriction after organ culture in rat and pig ocular arteries can be suppressed with MEK/ERK1/2 inhibitors. Acta Ophthalmol 2018; 96:e619-e625. [PMID: 29369532 DOI: 10.1111/aos.13651] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 10/23/2017] [Indexed: 01/19/2023]
Abstract
PURPOSE Even though retinal vascular changes following ischaemia have been poorly understood, the upregulation of vasoconstrictive endothelin-1 (ET-1) receptors (ETA /ETB ) following global cerebral ischaemia has been described. The aim of this study was to investigate whether or not the MEK/ERK1/2 pathway is involved in the observed upregulation and whether specific MEK/ERK1/2 inhibitors U0126 and trametinib can prevent it. METHODS The aim was also to localize ETA and ETB receptors using immunohistochemistry in both fresh rat ophthalmic arteries and after 24-hr organ culture and study the receptors functionally using myography. Pig retinal arteries also underwent 24-hr organ culture to validate similar responses across species and the retinal vasculature. RESULTS Results showed that following organ culture there is a significant increase in ET-1-mediated vasoconstriction, in particular via the ETB receptor. Furthermore, immunohistochemistry revealed a clear increase in pERK in the smooth muscle cells of rat ophthalmic artery. U0126 and trametinib were successful in attenuating the functional vasoconstriction in both rat and pig, as well as restoring immunofluorescence of pERK to fresh levels and counteracting ETB expression in the smooth muscle cells of the rat ophthalmic artery. CONCLUSION This is the first study to show that the MEK/ERK1/2 pathway in responsible for the increase in functional vasoconstriction via ET-1 receptor in rat ophthalmic and pig retinal arteries. Furthermore, this study is the first to suggest a way of inhibiting and preventing such an increase. With these results, we suggest a novel approach in retinal ischaemia therapy.
Collapse
Affiliation(s)
- Frank W. Blixt
- Department of Clinical Sciences; Division of Experimental Vascular Research; Lund University; Lund Sweden
| | - Kristian Agmund Haanes
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
| | - Lena Ohlsson
- Department of Clinical Sciences; Division of Experimental Vascular Research; Lund University; Lund Sweden
| | | | - Karin Warfvinge
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
| | - Lars Edvinsson
- Department of Clinical Sciences; Division of Experimental Vascular Research; Lund University; Lund Sweden
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
| |
Collapse
|
8
|
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
|
9
|
Li HT, Wang J, Li SF, Cheng L, Tang WZ, Feng YG. Upregulation of microRNA‑24 causes vasospasm following subarachnoid hemorrhage by suppressing the expression of endothelial nitric oxide synthase. Mol Med Rep 2018; 18:1181-1187. [PMID: 29845232 DOI: 10.3892/mmr.2018.9050] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 01/12/2017] [Indexed: 11/06/2022] Open
Abstract
MicroRNA (miR)‑24 has been reported to associate with various diseases by acting on different signaling pathways. The present study aimed to elucidate the association between miR‑24 expression levels and vasospasm following subarachnoid hemorrhage (SAH), and its underlying mechanism. An miR online database was searched, identifying endothelial nitric oxide synthase (NOS3) as a potential target gene of miR‑24. A luciferase reporter assay performed to investigate the regulatory association between miR‑24 and NOS3 revealed that miR‑24 bound to the NOS3 3' untranslated region and inhibited NOS3 expression. Reverse transcription‑quantitative polymerase chain reaction and western blot analysis were performed to investigate the miR‑24 and NOS3 expression levels in samples from patients with SAH, and demonstrated a negative correlation between the two. In addition, miR‑24 expression levels were increased in SAH patients with vasospasm compared with those without, whereas the opposite results were observed for NOS3. Vascular smooth muscle cells (VSMCs) transfected with an miR‑24 inhibitor exhibited increased expression levels of NOS3, whereas those transfected with an miR‑24 mimic or NOS3 small interfering RNA exhibited reduced expression levels of NOS3, compared with the control. These results indicated a negative regulatory association between miR‑24 and NOS3. Downregulation of NOS3 may induce vasospasm following SAH, which may be due to the upregualtion of miR‑24 in VSMCs.
Collapse
Affiliation(s)
- Huan-Ting Li
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Jing Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Shi-Fang Li
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Lei Cheng
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Wan-Zhong Tang
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yu-Gong Feng
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
10
|
Spray S, Johansson SE, Radziwon-Balicka A, Haanes KA, Warfvinge K, Povlsen GK, Kelly PAT, Edvinsson L. Enhanced contractility of intraparenchymal arterioles after global cerebral ischaemia in rat - new insights into the development of delayed cerebral hypoperfusion. Acta Physiol (Oxf) 2017; 220:417-431. [PMID: 27864916 DOI: 10.1111/apha.12834] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/11/2016] [Accepted: 11/15/2016] [Indexed: 12/13/2022]
Abstract
AIM Delayed cerebral hypoperfusion is a secondary complication found in the days after transient global cerebral ischaemia that worsens the ischaemic damage inflicted by the initial transient episode of global cerebral ischaemia. A recent study demonstrated increased cerebral vasoconstriction in the large arteries on the brain surface (pial arteries) after global cerebral ischaemia. However, smaller arterioles inside the brain (parenchymal arterioles) are equally important in the regulation of cerebral blood flow and yet their pathophysiology after global cerebral ischaemia is largely unknown. Therefore, we investigated whether increased contractility occurs in the intraparenchymal arterioles. METHODS Global cerebral ischaemia was induced in male Wistar rats by bilateral common carotid occlusion for 15 min combined with hypovolaemia. Regional cerebral blood flow was determined by quantitative autoradiography. Intraparenchymal arterioles were isolated and pressurized, and concentration-response curves to endothelin-1 with and without the endothelin B receptor-selective antagonist BQ788 was generated. Endothelin B receptor expression was investigated by quantitative flow cytometry and immunohistochemistry. RESULTS We observed increased endothelin-1-mediated contractility of parenchymal arterioles correlating with reduced cerebral blood flow of the cortex, hippocampus and caudate nucleus 48 h after global cerebral ischaemia. The increased endothelin-1-mediated contractility was abolished by BQ788, and the vascular smooth muscle cell-specific expression of endothelin B receptors was significantly increased after global cerebral ischaemia. CONCLUSION Increased endothelin-1-mediated contractility and expression of endothelin B receptors in the intraparenchymal vasculature contributes to the development of delayed cerebral hypoperfusion after global cerebral ischaemia in combination with vascular changes of the pial vasculature.
Collapse
Affiliation(s)
- S. Spray
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
| | - S. E. Johansson
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
| | - A. Radziwon-Balicka
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
| | - K. A. Haanes
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
| | - K. Warfvinge
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
- Division of Experimental Vascular Research; Department of Clinical Sciences; Lund University Hospital; Lund Sweden
| | - G. K. Povlsen
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
| | - P. A. T. Kelly
- Centre for Cognitive and Neural System; University of Edinburgh; Edinburgh UK
| | - L. Edvinsson
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
- Division of Experimental Vascular Research; Department of Clinical Sciences; Lund University Hospital; Lund Sweden
| |
Collapse
|
11
|
Spray S, Johansson SE, Edwards AVG, Larsen MR, Radziwon-Balicka A, Povlsen GK, Edvinsson L. Alterations in the Cerebral Microvascular Proteome Expression Profile After Transient Global Cerebral Ischemia in Rat. J Mol Neurosci 2016; 61:396-411. [DOI: 10.1007/s12031-016-0875-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 11/29/2016] [Indexed: 11/29/2022]
|
12
|
Schachtschneider KM, Liu Y, Rund LA, Madsen O, Johnson RW, Groenen MAM, Schook LB. Impact of neonatal iron deficiency on hippocampal DNA methylation and gene transcription in a porcine biomedical model of cognitive development. BMC Genomics 2016; 17:856. [PMID: 27809765 PMCID: PMC5094146 DOI: 10.1186/s12864-016-3216-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 10/26/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Iron deficiency is a common childhood micronutrient deficiency that results in altered hippocampal function and cognitive disorders. However, little is known about the mechanisms through which neonatal iron deficiency results in long lasting alterations in hippocampal gene expression and function. DNA methylation is an epigenetic mark involved in gene regulation and altered by environmental factors. In this study, hippocampal DNA methylation and gene expression were assessed via reduced representation bisulfite sequencing and RNA-seq on samples from a previous study reporting reduced hippocampal-based learning and memory in a porcine biomedical model of neonatal iron deficiency. RESULTS In total 192 differentially expressed genes (DEGs) were identified between the iron deficient and control groups. GO term and pathway enrichment analysis identified DEGs associated with hypoxia, angiogenesis, increased blood brain barrier (BBB) permeability, and altered neurodevelopment and function. Of particular interest are genes previously implicated in cognitive deficits and behavioral disorders in humans and mice, including HTR2A, HTR2C, PAK3, PRSS12, and NETO1. Altered genome-wide DNA methylation was observed across 0.5 million CpG and 2.4 million non-CpG sites. In total 853 differentially methylated (DM) CpG and 99 DM non-CpG sites were identified between groups. Samples clustered by group when comparing DM non-CpG sites, suggesting high conservation of non-CpG methylation in response to neonatal environment. In total 12 DM sites were associated with 9 DEGs, including genes involved in angiogenesis, neurodevelopment, and neuronal function. CONCLUSIONS Neonatal iron deficiency leads to altered hippocampal DNA methylation and gene regulation involved in hypoxia, angiogenesis, increased BBB permeability, and altered neurodevelopment and function. Together, these results provide new insights into the mechanisms through which neonatal iron deficiency results in long lasting reductions in cognitive development in humans.
Collapse
Affiliation(s)
- Kyle M. Schachtschneider
- Department of Animal Sciences, University of Illinois, 1201 W Gregory Drive, Urbana, IL 61801 USA
- Animal Breeding and Genomics Centre, Wageningen University, P.O. Box 338, Wageningen, 6700AH The Netherlands
| | - Yingkai Liu
- Department of Animal Sciences, University of Illinois, 1201 W Gregory Drive, Urbana, IL 61801 USA
- College of Animal Science and Technology, Sichuan Agricultural University, Wenjiang, Huimin Road #221, Chengdu, 610000 China
| | - Laurie A. Rund
- Department of Animal Sciences, University of Illinois, 1201 W Gregory Drive, Urbana, IL 61801 USA
| | - Ole Madsen
- Animal Breeding and Genomics Centre, Wageningen University, P.O. Box 338, Wageningen, 6700AH The Netherlands
| | - Rodney W. Johnson
- Department of Animal Sciences, University of Illinois, 1201 W Gregory Drive, Urbana, IL 61801 USA
| | - Martien A. M. Groenen
- Animal Breeding and Genomics Centre, Wageningen University, P.O. Box 338, Wageningen, 6700AH The Netherlands
| | - Lawrence B. Schook
- Department of Animal Sciences, University of Illinois, 1201 W Gregory Drive, Urbana, IL 61801 USA
- Institute for Genomic Biology, University of Illinois, 1206 W Gregory Drive, Urbana, IL 61801 USA
| |
Collapse
|
13
|
Povlsen GK, Longden TA, Bonev AD, Hill-Eubanks DC, Nelson MT. Uncoupling of neurovascular communication after transient global cerebral ischemia is caused by impaired parenchymal smooth muscle Kir channel function. J Cereb Blood Flow Metab 2016; 36:1195-201. [PMID: 27052838 PMCID: PMC4929704 DOI: 10.1177/0271678x16638350] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/18/2016] [Accepted: 01/18/2016] [Indexed: 12/28/2022]
Abstract
Transient global cerebral ischemia is often followed by delayed disturbances of cerebral blood flow, contributing to neuronal injury. The pathophysiological processes underlying such disturbances are incompletely understood. Here, using an established model of transient global cerebral ischemia, we identify dramatically impaired neurovascular coupling following ischemia. This impairment results from the loss of functional inward rectifier potassium (KIR) channels in the smooth muscle of parenchymal arterioles. Therapeutic strategies aimed at protecting or restoring cerebrovascular KIR channel function may therefore improve outcomes following ischemia.
Collapse
Affiliation(s)
- Gro Klitgaard Povlsen
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington, Vermont, USA Department of Clinical Experimental Research, Glostrup Research Institute, Glostrup University Hospital, Glostrup, Denmark
| | - Thomas A Longden
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Adrian D Bonev
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington, Vermont, USA
| | | | - Mark T Nelson
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington, Vermont, USA Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
14
|
Blixt FW, Johansson SE, Johnson L, Haanes KA, Warfvinge K, Edvinsson L. Enhanced Endothelin-1 Mediated Vasoconstriction of the Ophthalmic Artery May Exacerbate Retinal Damage after Transient Global Cerebral Ischemia in Rat. PLoS One 2016; 11:e0157669. [PMID: 27322388 PMCID: PMC4913955 DOI: 10.1371/journal.pone.0157669] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 06/02/2016] [Indexed: 11/18/2022] Open
Abstract
Cerebral vasculature is often the target of stroke studies. However, the vasculature supplying the eye might also be affected by ischemia. The aim of the present study was to investigate if the transient global cerebral ischemia (GCI) enhances vascular effect of endothelin-1 (ET-1) and 5-hydroxytryptamine/serotonin (5-HT) on the ophthalmic artery in rats, leading to delayed retinal damage. This was preformed using myography on the ophthalmic artery, coupled with immunohistochemistry and electroretinogram (ERG) to assess the ischemic consequences on the retina. Results showed a significant increase of ET-1 mediated vasoconstriction at 48 hours post ischemia. The retina did not exhibit any morphological changes throughout the study. However, we found an increase of GFAP and vimentin expression at 72 hours and 7 days after ischemia, indicating Müller cell mediated gliosis. ERG revealed significantly decreased function at 72 hours, but recovered almost completely after 7 days. In conclusion, we propose that the increased contractile response via ET-1 receptors in the ophthalmic artery after 48 hours may elicit negative retinal consequences due to a second ischemic period. This may exacerbate retinal damage after ischemia as illustrated by the decreased retinal function and Müller cell activation. The ophthalmic artery and ET-1 mediated vasoconstriction may be a valid and novel therapeutic target after longer periods of ischemic insults.
Collapse
Affiliation(s)
- Frank W. Blixt
- Department of Clinical Sciences, Division of Experimental Vascular Research, Lund University, Lund, Sweden
- * E-mail:
| | - Sara Ellinor Johansson
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet, Glostrup, Denmark
| | - Leif Johnson
- Department of Ophthalmology, Glostrup Research Institute, Rigshospitalet, Glostrup, Denmark
| | - Kristian Agmund Haanes
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet, Glostrup, Denmark
| | - Karin Warfvinge
- Department of Clinical Sciences, Division of Experimental Vascular Research, Lund University, Lund, Sweden
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet, Glostrup, Denmark
| | - Lars Edvinsson
- Department of Clinical Sciences, Division of Experimental Vascular Research, Lund University, Lund, Sweden
- Department of Clinical Experimental Research, Glostrup Research Institute, Rigshospitalet, Glostrup, Denmark
| |
Collapse
|
15
|
Davenport AP, Hyndman KA, Dhaun N, Southan C, Kohan DE, Pollock JS, Pollock DM, Webb DJ, Maguire JJ. Endothelin. Pharmacol Rev 2016; 68:357-418. [PMID: 26956245 PMCID: PMC4815360 DOI: 10.1124/pr.115.011833] [Citation(s) in RCA: 556] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The endothelins comprise three structurally similar 21-amino acid peptides. Endothelin-1 and -2 activate two G-protein coupled receptors, ETA and ETB, with equal affinity, whereas endothelin-3 has a lower affinity for the ETA subtype. Genes encoding the peptides are present only among vertebrates. The ligand-receptor signaling pathway is a vertebrate innovation and may reflect the evolution of endothelin-1 as the most potent vasoconstrictor in the human cardiovascular system with remarkably long lasting action. Highly selective peptide ETA and ETB antagonists and ETB agonists together with radiolabeled analogs have accurately delineated endothelin pharmacology in humans and animal models, although surprisingly no ETA agonist has been discovered. ET antagonists (bosentan, ambrisentan) have revolutionized the treatment of pulmonary arterial hypertension, with the next generation of antagonists exhibiting improved efficacy (macitentan). Clinical trials continue to explore new applications, particularly in renal failure and for reducing proteinuria in diabetic nephropathy. Translational studies suggest a potential benefit of ETB agonists in chemotherapy and neuroprotection. However, demonstrating clinical efficacy of combined inhibitors of the endothelin converting enzyme and neutral endopeptidase has proved elusive. Over 28 genetic modifications have been made to the ET system in mice through global or cell-specific knockouts, knock ins, or alterations in gene expression of endothelin ligands or their target receptors. These studies have identified key roles for the endothelin isoforms and new therapeutic targets in development, fluid-electrolyte homeostasis, and cardiovascular and neuronal function. For the future, novel pharmacological strategies are emerging via small molecule epigenetic modulators, biologicals such as ETB monoclonal antibodies and the potential of signaling pathway biased agonists and antagonists.
Collapse
Affiliation(s)
- Anthony P Davenport
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| | - Kelly A Hyndman
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| | - Neeraj Dhaun
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| | - Christopher Southan
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| | - Donald E Kohan
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| | - Jennifer S Pollock
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| | - David M Pollock
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| | - David J Webb
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| | - Janet J Maguire
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom (A.P.D., J.J.M.); IUPHAR/BPS Guide to PHARMACOLOGY, Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, Edinburgh, United Kingdom (C.S.); Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah (D.E.K.); Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama (K.A.H., J.S.P., D.M.P.); and Department of Renal Medicine, Royal Infirmary of Edinburgh (N.D.) and University/British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute (D.J.W.N.D.), Edinburgh, Scotland, United Kingdom
| |
Collapse
|
16
|
Johansson SE, Andersen XEDR, Hansen RH, Povlsen GK, Edvinsson L. Cerebrovascular endothelin-1 hyper-reactivity is associated with transient receptor potential canonical channels 1 and 6 activation and delayed cerebral hypoperfusion after forebrain ischaemia in rats. Acta Physiol (Oxf) 2015; 214:376-89. [PMID: 25939574 DOI: 10.1111/apha.12519] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 01/19/2015] [Accepted: 04/29/2015] [Indexed: 12/17/2022]
Abstract
AIM In this study, we aimed to investigate whether changes in cerebrovascular voltage-dependent calcium channels and non-selective cation channels contribute to the enhanced endothelin-1-mediated vasoconstriction in the delayed hypoperfusion phase after experimental transient forebrain ischaemia. METHODS Experimental forebrain ischaemia was induced in Wistar male rats by a two-vessel occlusion model, and the cerebral blood flow was measured by magnetic resonance imaging two days after reperfusion. In vitro vasoreactivity studies, immunofluorescence and quantitative PCR were performed on cerebral arteries from ischaemic or sham-operated rats to evaluate changes in vascular voltage-dependent calcium channels, transient receptor potential canonical channels as well as endothelin-1 receptor function and expression. RESULTS The expression of transient receptor potential canonical channels 1 and 6 in the vascular smooth muscle cells was enhanced and correlated with decreased cerebral blood flow two days after forebrain ischaemia. Furthermore, under conditions when voltage-dependent calcium channels were inhibited, endothelin-1-induced cerebrovascular contraction was enhanced and this enhancement was presumably mediated by Ca(2+) influx via upregulated transient receptor potential canonical channels 1 and 6. CONCLUSIONS Our data demonstrates that endothelin-1-mediated influx of extracellular Ca(2+) activates transient receptor potential canonical channels 1 and 6 in cerebral vascular smooth muscle cells. This seems to have an important role in the enhanced cerebral vasoconstriction in the delayed post-ischaemic hypoperfusion phase after experimental forebrain ischaemia.
Collapse
Affiliation(s)
- S. E. Johansson
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
| | - X. E. D. R. Andersen
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
| | - R. H. Hansen
- Research Group; Department of Radiology; Copenhagen University Hospital; Herlev Denmark
| | - G. K. Povlsen
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
| | - L. Edvinsson
- Department of Clinical Experimental Research; Glostrup Research Institute; Rigshospitalet; Glostrup Denmark
- Division of Experimental Vascular Research; Department of Clinical Sciences; Lund University; Lund Sweden
| |
Collapse
|
17
|
Lu XZ, Bi XY, He X, Zhao M, Xu M, Yu XJ, Zhao ZH, Zang WJ. Activation of M3 cholinoceptors attenuates vascular injury after ischaemia/reperfusion by inhibiting the Ca2+/calmodulin-dependent protein kinase II pathway. Br J Pharmacol 2015; 172:5619-33. [PMID: 25953628 DOI: 10.1111/bph.13183] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 03/24/2015] [Accepted: 04/20/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE The activation of M3 cholinoceptors (M3 receptors) by choline reduces cardiovascular risk, but it is unclear whether these receptors can regulate ischaemia/reperfusion (I/R)-induced vascular injury. Thus, the primary goal of the present study was to explore the effects of choline on the function of mesenteric arteries following I/R, with a major focus on Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) regulation. EXPERIMENTAL APPROACH Rats were given choline (10 mg · kg(-1), i.v.) and then the superior mesenteric artery was occluded for 60 min (ischaemia), followed by 90 min of reperfusion. The M3 receptor antagonist, 4-diphenylacetoxy-N-methylpiperidine methiodide (4-DAMP), was injected (0.12 μg · kg(-1), i.v.) 5 min prior to choline treatment. Vascular function was examined in rings of mesenteric arteries isolated after the reperfusion procedure. Vascular superoxide anion production, CaMKII and the levels of Ca(2+)-cycling proteins were also assessed. KEY RESULTS Choline treatment attenuated I/R-induced vascular dysfunction, blocked elevations in the levels of reactive oxygen species (ROS) and decreased the up-regulated expression of oxidised CaMKII and phosphorylated CaMKII. In addition, choline reversed the abnormal expression of Ca(2+)-cycling proteins, including Na(+)Ca(2+) exchanger, inositol 1,4,5-trisphosphate receptor, sarcoplasmic reticulum Ca(2+)-ATPase and phospholamban. All of these cholinergic effects of choline were abolished by 4-DAMP. CONCLUSIONS AND IMPLICATIONS Our data suggest that inhibition of the ROS-mediated CaMKII pathway and modulation of Ca(2+)-cycling proteins may be novel mechanisms underlying choline-induced vascular protection. These results represent a significant addition to the understanding of the pharmacological roles of M3 receptors in the vasculature, providing a new therapeutic strategy for I/R-induced vascular injury.
Collapse
Affiliation(s)
- Xing-Zhu Lu
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xue-Yuan Bi
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xi He
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Ming Zhao
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Man Xu
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xiao-Jiang Yu
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Zheng-Hang Zhao
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Wei-Jin Zang
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
18
|
Durgan DJ, Crossland RF, Lloyd EE, Phillips SC, Bryan RM. Increased cerebrovascular sensitivity to endothelin-1 in a rat model of obstructive sleep apnea: a role for endothelin receptor B. J Cereb Blood Flow Metab 2015; 35:402-11. [PMID: 25425077 PMCID: PMC4348382 DOI: 10.1038/jcbfm.2014.214] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/30/2014] [Accepted: 11/05/2014] [Indexed: 11/09/2022]
Abstract
Obstructive sleep apnea (OSA) is associated with cerebrovascular diseases. However, little is known regarding the effects of OSA on the cerebrovascular wall. We tested the hypothesis that OSA augments endothelin-1 (ET-1) constrictions of cerebral arteries. Repeated apneas (30 or 60 per hour) were produced in rats during the sleep cycle (8 hours) by remotely inflating a balloon implanted in the trachea. Four weeks of apneas produced a 23-fold increase in ET-1 sensitivity in isolated and pressurized posterior cerebral arteries (PCAs) compared with PCAs from sham-operated rats (EC50=10(-9.2) mol/L versus 10(-10.6) mol/L; P<0.001). This increased sensitivity was abolished by the ET-B receptor antagonist, BQ-788. Constrictions to the ET-B receptor agonist, IRL-1620, were greater in PCAs from rats after 2 or 4 weeks of apneas compared with that from sham-operated rats (P=0.013). Increased IRL-1620 constrictions in PCAs from OSA rats were normalized with the transient receptor potential channel (TRPC) blocker, SKF96365, or the Rho kinase (ROCK) inhibitor, Y27632. These data show that OSA increases the sensitivity of PCAs to ET-1 through enhanced ET-B activity, and enhanced activity of TRPCs and ROCK. We conclude that enhanced ET-1 signaling is part of a pathologic mechanism associated with adverse cerebrovascular outcomes of OSA.
Collapse
Affiliation(s)
- David J Durgan
- Department of Anesthesiology, Baylor College of Medicine, Houston, Texas, USA
| | - Randy F Crossland
- 1] Department of Anesthesiology, Baylor College of Medicine, Houston, Texas, USA [2] Department of Molecular Physiology and Biophysics (Graduate Program in Cardiovascular Sciences), Baylor College of Medicine, Houston, Texas, USA
| | - Eric E Lloyd
- Department of Anesthesiology, Baylor College of Medicine, Houston, Texas, USA
| | - Sharon C Phillips
- Department of Anesthesiology, Baylor College of Medicine, Houston, Texas, USA
| | - Robert M Bryan
- 1] Department of Anesthesiology, Baylor College of Medicine, Houston, Texas, USA [2] Department of Molecular Physiology and Biophysics (Graduate Program in Cardiovascular Sciences), Baylor College of Medicine, Houston, Texas, USA [3] Department of Medicine (Cardiovascular Sciences), Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
19
|
Regulation of microRNAs miR-30a and miR-143 in cerebral vasculature after experimental subarachnoid hemorrhage in rats. BMC Genomics 2015; 16:119. [PMID: 25766280 PMCID: PMC4342208 DOI: 10.1186/s12864-015-1341-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 02/12/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND microRNAs (miRNAs) are important regulators of translation and have been implicated in the pathogenesis of a number of cardiovascular diseases, including stroke, and suggested as possible prognostic biomarkers. Our aim was to identify miRNAs that are differentially regulated in cerebral arteries after subarachnoid hemorrhage (SAH), using a rat injection model of SAH and a qPCR-based screen of 728 rat miRNAs. Additionally, serum was analyzed for a possible spill-over to the circulation of regulated miRNAs from the vessel walls. RESULTS We identified 482 different miRNAs expressed in cerebral arteries post-SAH. Two miRNAs, miR-30a and miR-143, were significantly upregulated in cerebral arteries after SAH when compared to sham-operated animals. However, none of these exhibited significantly altered serum levels after SAH versus post-sham surgery. The most robust upregulation was seen for miR-143, which has several predicted targets and is a strong regulator of vascular morphology. We hypothesize that miR-30a and miR-143 may play a role in the vascular wall changes seen after SAH. CONCLUSIONS We report that miR-30a and miR-143 in the cerebral arteries show significant changes over time after SAH, but do not differ from sham-operated rats at 24 h post-SAH. Although this finding suggests interesting novel possible mechanisms involved in post-SAH cerebrovascular changes, the lack of regulation of these miRNAs in serum excludes their use as blood-borne biomarkers for cerebrovascular changes following SAH.
Collapse
|
20
|
Regulatory mechanism of endothelin receptor B in the cerebral arteries after focal cerebral ischemia. PLoS One 2014; 9:e113624. [PMID: 25479176 PMCID: PMC4257531 DOI: 10.1371/journal.pone.0113624] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 10/29/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Increased expression of endothelin receptor type B (ETBR), a vasoactive receptor, has recently been implied in the reduced cerebral blood flow and exacerbated neuronal damage after ischemia-reperfusion (I/R). The study explores the regulatory mechanisms of ETBR to identify drug targets to restore normal cerebral artery contractile function as part of successful neuroprotective therapy. METHODS We have employed in vitro methods on human and rat cerebral arteries to study the regulatory mechanisms and the efficacy of target selective inhibitor, Mithramycin A (MitA), to block the ETBR mediated contractile properties. Later, middle cerebral artery occluded (MCAO) rats were used to substantiate the observations. Quantative PCR, immunohistochemistry, western blot and wire myograph methods were employed to study the expression and contractile properties of cerebral arteries. RESULTS Increased expression of specificity protein (Sp1) was observed in human and rat cerebral arteries after organ culture, strongly correlating with the ETBR upregulation. Similar observations were made in MCAO rats. Treatment with MitA, a Sp1 specific inhibitor, significantly downregulated the ETBR mRNA and protein levels. It also significantly reduced the ETBR mediated cerebrovascular contractility. Detailed analysis indicated that ERK1/2 mediated phosphorylation of Sp1 might be essential for ETBR transcription. CONCLUSION Transcription factor Sp1 regulates the ETBR mediated vasoconstriction in focal cerebral ischemia via MEK-ERK signaling, which is also conserved in humans. The results show that MitA can effectively be used to block ETBR mediated vasoconstriction as a supplement to an existing ischemic stroke therapy.
Collapse
|
21
|
Johansson SE, Larsen SS, Povlsen GK, Edvinsson L. Early MEK1/2 inhibition after global cerebral ischemia in rats reduces brain damage and improves outcome by preventing delayed vasoconstrictor receptor upregulation. PLoS One 2014; 9:e92417. [PMID: 24642693 PMCID: PMC3958517 DOI: 10.1371/journal.pone.0092417] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 02/21/2014] [Indexed: 11/18/2022] Open
Abstract
Background Global cerebral ischemia following cardiac arrest is associated with increased cerebral vasoconstriction and decreased cerebral blood flow, contributing to delayed neuronal cell death and neurological detriments in affected patients. We hypothesize that upregulation of contractile ETB and 5-HT1B receptors, previously demonstrated in cerebral arteries after experimental global ischemia, are a key mechanism behind insufficient perfusion of the post-ischemic brain, proposing blockade of this receptor upregulation as a novel target for prevention of cerebral hypoperfusion and delayed neuronal cell death after global cerebral ischemia. The aim was to characterize the time-course of receptor upregulation and associated neuronal damage after global ischemia and investigate whether treatment with the MEK1/2 inhibitor U0126 can prevent cerebrovascular receptor upregulation and thereby improve functional outcome after global cerebral ischemia. Incomplete global cerebral ischemia was induced in Wistar rats and the time-course of enhanced contractile responses and the effect of U0126 in cerebral arteries were studied by wire myography and the neuronal cell death by TUNEL. The expression of ETB and 5-HT1B receptors was determined by immunofluorescence. Results Enhanced vasoconstriction peaked in fore- and midbrain arteries 3 days after ischemia. Neuronal cell death appeared initially in the hippocampus 3 days after ischemia and gradually increased until 7 days post-ischemia. Treatment with U0126 normalised cerebrovascular ETB and 5-HT1B receptor expression and contractile function, reduced hippocampal cell death and improved survival rate compared to vehicle treated animals. Conclusions Excessive cerebrovascular expression of contractile ETB and 5-HT1B receptors is a delayed response to global cerebral ischemia peaking 3 days after the insult, which likely contributes to the development of delayed neuronal damage. The enhanced cerebrovascular contractility can be prevented by treatment with the MEK1/2 inhibitor U0126, diminishes neuronal damage and improves survival rate, suggesting MEK1/2 inhibition as a novel strategy for early treatment of neurological consequences following global cerebral ischemia.
Collapse
Affiliation(s)
- Sara Ellinor Johansson
- Department of Clinical Experimental Research, Glostrup Research Institute, Glostrup Hospital, Glostrup, Denmark
- * E-mail:
| | - Stine Schmidt Larsen
- Department of Clinical Experimental Research, Glostrup Research Institute, Glostrup Hospital, Glostrup, Denmark
| | - Gro Klitgaard Povlsen
- Department of Clinical Experimental Research, Glostrup Research Institute, Glostrup Hospital, Glostrup, Denmark
| | - Lars Edvinsson
- Department of Clinical Experimental Research, Glostrup Research Institute, Glostrup Hospital, Glostrup, Denmark
- Division of Experimental Vascular Research, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
22
|
Jalewa J, Joshi A, McGinnity TM, Prasad G, Wong-Lin K, Hölscher C. Neural circuit interactions between the dorsal raphe nucleus and the lateral hypothalamus: an experimental and computational study. PLoS One 2014; 9:e88003. [PMID: 24516577 PMCID: PMC3916338 DOI: 10.1371/journal.pone.0088003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 01/02/2014] [Indexed: 12/22/2022] Open
Abstract
Orexinergic/hypocretinergic (Ox) neurotransmission plays an important role in regulating sleep, as well as in anxiety and depression, for which the serotonergic (5-HT) system is also involved in. However, little is known regarding the direct and indirect interactions between 5-HT in the dorsal raphe nucleus (DRN) and Ox neurons in the lateral hypothalamus (LHA). In this study, we report the additional presence of 5-HT1BR, 5-HT2AR, 5-HT2CR and fast ligand-gated 5-HT3AR subtypes on the Ox neurons of transgenic Ox-enhanced green fluorescent protein (Ox-EGFP) and wild type C57Bl/6 mice using single and double immunofluorescence (IF) staining, respectively, and quantify the colocalization for each 5-HT receptor subtype. We further reveal the presence of 5-HT3AR and 5-HT1AR on GABAergic neurons in LHA. We also identify NMDAR1, OX1R and OX2R on Ox neurons, but none on adjacent GABAergic neurons. This suggests a one-way relationship between LHA's GABAergic and Ox neurons, wherein GABAergic neurons exerts an inhibitory effect on Ox neurons under partial DRN's 5-HT control. We also show that Ox axonal projections receive glutamatergic (PSD-95 immunopositive) and GABAergic (Gephyrin immunopositive) inputs in the DRN. We consider these and other available findings into our computational model to explore possible effects of neural circuit connection types and timescales on the DRN-LHA system's dynamics. We find that if the connections from 5-HT to LHA's GABAergic neurons are weakly excitatory or inhibitory, the network exhibits slow oscillations; not observed when the connection is strongly excitatory. Furthermore, if Ox directly excites 5-HT neurons at a fast timescale, phasic Ox activation can lead to an increase in 5-HT activity; no significant effect with slower timescale. Overall, our experimental and computational approaches provide insights towards a more complete understanding of the complex relationship between 5-HT in the DRN and Ox in the LHA.
Collapse
Affiliation(s)
- Jaishree Jalewa
- School of Biomedical Sciences, University of Ulster, Coleraine, Northern Ireland, United Kingdom
| | - Alok Joshi
- Intelligent Systems Research Centre, University of Ulster, Magee Campus, Londonderry, Northern Ireland, United Kingdom
| | - T. Martin McGinnity
- Intelligent Systems Research Centre, University of Ulster, Magee Campus, Londonderry, Northern Ireland, United Kingdom
| | - Girijesh Prasad
- Intelligent Systems Research Centre, University of Ulster, Magee Campus, Londonderry, Northern Ireland, United Kingdom
| | - KongFatt Wong-Lin
- Intelligent Systems Research Centre, University of Ulster, Magee Campus, Londonderry, Northern Ireland, United Kingdom
- * E-mail: (CH); (KW)
| | - Christian Hölscher
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
- * E-mail: (CH); (KW)
| |
Collapse
|
23
|
Rasmussen MNP, Hornbak M, Larsen SS, Sheykhzade M, Edvinsson L. Permanent distal occlusion of middle cerebral artery in rat causes local increased ETB, 5-HT₁B and AT₁ receptor-mediated contractility downstream of occlusion. J Vasc Res 2013; 50:396-409. [PMID: 23988741 DOI: 10.1159/000354242] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 07/05/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS In response to experimental stroke, a characteristic functional and expressional upregulation of contractile G-protein-coupled receptors has been uncovered in the affected cerebral vasculature; however, the mechanism initiating this phenomenon remains unknown. METHODS Using a model of permanent distal occlusion of rat middle cerebral arteries, we investigated whether there was a regional difference in receptor-mediated contractility of segments located upstream and downstream of the occlusion site. The contractile response to endothelin, angiotensin and 5-hydroxytryptamine receptor stimulation was studied by sensitive wire myograph. RESULTS Only downstream segments exhibited an augmented contractile response to stimulation with each of the three ligands, with the response towards sarafotoxin 6c being especially augmented compared to sham, upstream and contralateral controls. This functional increase did not seem to relate to ischemic tissue damage, inflammatory cell infiltration or the element of reperfusion. Interestingly, immunohistochemistry did not show any difference in the level of immunoreactivity towards endothelin B (ETB) receptors between groups. CONCLUSION Single artery occlusion without significant visible infarct resulted in locally increased ETB, angiotensin type 1 and 5-hydroxytryptamine 1B receptor-mediated contractile responses only in segments located downstream of the occlusion site. This suggests lack of wall stress as an initiating trigger leading to regulation of contractile response after cerebral stroke.
Collapse
Affiliation(s)
- Marianne N P Rasmussen
- Department of Clinical Experimental Research, Glostrup Research Institute, Glostrup University Hospital, Glostrup, Denmark
| | | | | | | | | |
Collapse
|
24
|
Marín-Prida J, Pavón-Fuentes N, Llópiz-Arzuaga A, Fernández-Massó JR, Delgado-Roche L, Mendoza-Marí Y, Santana SP, Cruz-Ramírez A, Valenzuela-Silva C, Nazábal-Gálvez M, Cintado-Benítez A, Pardo-Andreu GL, Polentarutti N, Riva F, Pentón-Arias E, Pentón-Rol G. Phycocyanobilin promotes PC12 cell survival and modulates immune and inflammatory genes and oxidative stress markers in acute cerebral hypoperfusion in rats. Toxicol Appl Pharmacol 2013; 272:49-60. [PMID: 23732081 DOI: 10.1016/j.taap.2013.05.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 05/17/2013] [Accepted: 05/21/2013] [Indexed: 01/23/2023]
Abstract
Since the inflammatory response and oxidative stress are involved in the stroke cascade, we evaluated here the effects of Phycocyanobilin (PCB, the C-Phycocyanin linked tetrapyrrole) on PC12 cell survival, the gene expression and the oxidative status of hypoperfused rat brain. After the permanent bilateral common carotid arteries occlusion (BCCAo), the animals were treated with saline or PCB, taking samples 24h post-surgery. Global gene expression was analyzed with GeneChip Rat Gene ST 1.1 from Affymetrix; the expression of particular genes was assessed by the Fast SYBR Green RT-PCR Master Mix and Bioplex methods; and redox markers (MDA, PP, CAT, SOD) were evaluated spectrophotometrically. The PCB treatment prevented the H2O2 and glutamate induced PC12 cell injury assessed by the MTT assay, and modulated 190 genes (93 up- and 97 down-regulated) associated to several immunological and inflammatory processes in BCCAo rats. Furthermore, PCB positively modulated 19 genes mostly related to a detrimental pro-inflammatory environment and counteracted the oxidative imbalance in the treated BCCAo animals. Our results support the view of an effective influence of PCB on major inflammatory mediators in acute cerebral hypoperfusion. These results suggest that PCB has a potential to be a treatment for ischemic stroke for which further studies are needed.
Collapse
Affiliation(s)
- Javier Marín-Prida
- Centre for Research and Biological Evaluations (CEIEB), Institute of Pharmacy and Food, University of Havana, Ave. 23 e/ 214 y 222, La Lisa, PO Box: 430, Havana, Cuba
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Frydrychowski AF, Winklewski PJ, Szarmach A, Halena G, Bandurski T. Near-infrared transillumination back scattering sounding--new method to assess brain microcirculation in patients with chronic carotid artery stenosis. PLoS One 2013; 8:e61936. [PMID: 23613977 PMCID: PMC3629110 DOI: 10.1371/journal.pone.0061936] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 03/15/2013] [Indexed: 11/18/2022] Open
Abstract
Purpose The purpose of the study was to assess the responses of pial artery pulsation (cc-TQ) and subarachnoid width (sas-TQ) to acetazolamide challenge in patients with chronic carotid artery stenosis and relate these responses to changes in peak systolic velocity (PSV), cerebral blood flow (CBF), cerebral blood volume (CBV), mean transit time (MTT) and time to peak response (TTP). Methods Fifteen patients with carotid artery stenosis ≥90% on the ipsilateral side and <50% on the contralateral side were enrolled into the study. PSV was assessed using colour-coded duplex sonography, CBF, CBV, MTT and TTP with perfusion computed tomography, cc-TQ and sas-TQ with near-infrared transillumination/backscattering sounding (NIR-T/BSS). Results Based on the ipsilateral/contralateral cc-TQ ratio after acetazolamide challenge two groups of patients were distinguished: the first group with a ratio ≥1 and the second with a ratio <1. In the second group increases in CBF and CBV after the acetazolamide test were significantly higher in both hemispheres (ipsilateral: +33.0%±8.1% vs. +15.3%±4.4% and +26.3%±6.6% vs. +14.3%±5.1%; contralateral: +26.8%±7.0% vs. +17.6%±5.6% and +20.0%±7.3% vs. +10.0%±3.7%, respectively), cc-TQ was significantly higher only on the ipsilateral side (+37.3%±9.3% vs. +26.6%±8.6%) and the decrease in sas-TQ was less pronounced on the ipsilateral side (−0.7%±1.5% vs. −10.2%±1.5%), in comparison with the first group. The changes in sas-TQ following the acetazolamide test were consistent with the changes in TTP. Conclusions The ipsilateral/contralateral cc-TQ ratio following acetazolamide challenge may be used to distinguish patient groups characterized by different haemodynamic parameters. Further research on a larger group of patients is warranted.
Collapse
Affiliation(s)
| | - Pawel J. Winklewski
- Institute of Human Physiology, Medical University of Gdansk, Gdansk, Poland
- * E-mail:
| | | | - Grzegorz Halena
- Department of Cardiovascular Surgery, Medical University of Gdansk, Gdansk, Poland
| | - Tomasz Bandurski
- Department of Nuclear Medicine and Radiological Informatics, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
26
|
Povlsen GK, Johansson SE, Larsen CC, Samraj AK, Edvinsson L. Early events triggering delayed vasoconstrictor receptor upregulation and cerebral ischemia after subarachnoid hemorrhage. BMC Neurosci 2013; 14:34. [PMID: 23496889 PMCID: PMC3618233 DOI: 10.1186/1471-2202-14-34] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 03/07/2013] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Upregulation of vasoconstrictor receptors in cerebral arteries, including endothelin B (ETB) and 5-hydroxytryptamine 1B (5-HT(1B)) receptors, has been suggested to contribute to delayed cerebral ischemia, a feared complication after subarachnoid hemorrhage (SAH). This receptor upregulation has been shown to be mediated by intracellular signalling via the mitogen activated protein kinase kinase (MEK1/2)--extracellular regulated kinase 1/2 (ERK1/2) pathway. However, it is not known what event(s) that trigger MEK-ERK1/2 activation and vasoconstrictor receptor upregulation after SAH.We hypothesise that the drop in cerebral blood flow (CBF) and wall tension experienced by cerebral arteries in acute SAH is a key triggering event. We here investigate the importance of the duration of this acute CBF drop in a rat SAH model in which a fixed amount of blood is injected into the prechiasmatic cistern either at a high rate resulting in a short acute CBF drop or at a slower rate resulting in a prolonged acute CBF drop. RESULTS We demonstrate that the duration of the acute CBF drop is determining for a) degree of early ERK1/2 activation in cerebral arteries, b) delayed upregulation of vasoconstrictor receptors in cerebral arteries and c) delayed CBF reduction, neurological deficits and mortality. Moreover, treatment with an inhibitor of MEK-ERK1/2 signalling during an early time window from 6 to 24 h after SAH was sufficient to completely prevent delayed vasoconstrictor receptor upregulation and improve neurological outcome several days after the SAH. CONCLUSIONS Our findings suggest a series of events where 1) the acute CBF drop triggers early MEK-ERK1/2 activation, which 2) triggers the transcriptional upregulation of vasoconstrictor receptors in cerebral arteries during the following days, where 3) the resulting enhanced cerebrovascular contractility contribute to delayed cerebral ischemia.
Collapse
Affiliation(s)
- Gro Klitgaard Povlsen
- Department of Clinical Experimental Research, Glostrup Research Institute, Glostrup University Hospital, Nordre Ringvej 69, Glostrup, DK 2600, Denmark
| | - Sara Ellinor Johansson
- Department of Clinical Experimental Research, Glostrup Research Institute, Glostrup University Hospital, Nordre Ringvej 69, Glostrup, DK 2600, Denmark
| | | | - Ajoy Kumar Samraj
- Department of Clinical Experimental Research, Glostrup Research Institute, Glostrup University Hospital, Nordre Ringvej 69, Glostrup, DK 2600, Denmark
| | - Lars Edvinsson
- Department of Clinical Experimental Research, Glostrup Research Institute, Glostrup University Hospital, Nordre Ringvej 69, Glostrup, DK 2600, Denmark
| |
Collapse
|
27
|
Early brain injury: a common mechanism in subarachnoid hemorrhage and global cerebral ischemia. Stroke Res Treat 2013; 2013:394036. [PMID: 23533958 PMCID: PMC3603523 DOI: 10.1155/2013/394036] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 01/27/2013] [Indexed: 12/19/2022] Open
Abstract
Early brain injury (EBI) has become an area of extreme interest in the recent years and seems to be a common denominator in the pathophysiology of global transient ischemia and subarachnoid hemorrhage (SAH). In this paper, we highlight the importance of cerebral hypoperfusion and other mechanisms that occur in tandem in both pathologies and underline their possible roles in triggering brain injury after hemorrhagic or ischemic strokes.
Collapse
|
28
|
Ansar S, Eftekhari S, Waldsee R, Nilsson E, Nilsson O, Säveland H, Edvinsson L. MAPK signaling pathway regulates cerebrovascular receptor expression in human cerebral arteries. BMC Neurosci 2013; 14:12. [PMID: 23343134 PMCID: PMC3663811 DOI: 10.1186/1471-2202-14-12] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 12/26/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cerebral ischemia results in enhanced expression of contractile cerebrovascular receptors, such as endothelin type B (ET(B)), 5-hydroxytryptamine type 1B (5-HT(1B)), angiotensin II type 1 (AT(1)) and thromboxane (TP) receptors in the cerebral arteries within the ischemic area. The receptor upregulation occurs via activation of the mitogen-activated protein kinases (MAPK) pathway. Previous studies have shown that inhibitors of the MAPK pathway diminished the ischemic area and contractile cerebrovascular receptors after experimental cerebral ischemia. The aim of this study was to examine if the upregulation of contractile cerebrovascular receptors after 48 h of organ culture of human cerebral arteries involves MAPK pathways and if it can be prevented by a MEK1/2 inhibitor. Human cerebral arteries were obtained from patients undergoing intracranial tumor surgery. The vessels were divided into ring segments and incubated for 48 h in the presence or absence of the specific MEK1/2 inhibitor U0126. The vessels were then examined by using in vitro pharmacological methods and protein immunohistochemistry. RESULTS After organ culture of the cerebral arteries the contractile responses to endothelin (ET)-1, angiotensin (Ang) II and thromboxane (TP) were enhanced in comparison with fresh human arteries. However, 5-carboxamidotryptamine (5-CT) induced decreased contractile responses after organ culture as compared to fresh arteries. Incubation with U0126 diminished the maximum contraction elicited by application of ET-1, Ang II and U46619 in human cerebral arteries. In addition, the MEK1/2 inhibitor decreased the contractile response to 5-CT. Immunohistochemistry revealed that organ culture resulted in increased expression of endothelin ET(A), endothelin ET(B) angiotensin AT(2), 5-hydroxytryptamine 5-HT(1B) and thromboxane A2 receptors, and elevated levels of activated pERK1/2, all localized to the smooth muscle cells of the cerebral arteries. Co-incubation with U0126 normalized these proteins. CONCLUSION The study demonstrated that there is a clear association between human cerebrovascular receptor upregulation via transcription involving activation of the MAPK pathway after organ culture. Inhibition of the MAPK pathways attenuated the vasoconstriction mediated by ET, AT and TP receptors in human cerebral arteries and the enhanced expression of their receptors. The results indicate that MAPK inhibition might be a novel target for treatment of cerebrovascular disorders.
Collapse
Affiliation(s)
- Saema Ansar
- Department of Clinical Sciences, Division of Experimental Vascular Research, Lund University, Lund, Sweden.
| | | | | | | | | | | | | |
Collapse
|
29
|
Kamp MA, Dibué M, Schneider T, Steiger HJ, Hänggi D. Calcium and potassium channels in experimental subarachnoid hemorrhage and transient global ischemia. Stroke Res Treat 2012; 2012:382146. [PMID: 23251831 PMCID: PMC3518967 DOI: 10.1155/2012/382146] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Accepted: 10/27/2012] [Indexed: 11/23/2022] Open
Abstract
Healthy cerebrovascular myocytes express members of several different ion channel families which regulate resting membrane potential, vascular diameter, and vascular tone and are involved in cerebral autoregulation. In animal models, in response to subarachnoid blood, a dynamic transition of ion channel expression and function is initiated, with acute and long-term effects differing from each other. Initial hypoperfusion after exposure of cerebral vessels to oxyhemoglobin correlates with a suppression of voltage-gated potassium channel activity, whereas delayed cerebral vasospasm involves changes in other potassium channel and voltage-gated calcium channels expression and function. Furthermore, expression patterns and function of ion channels appear to differ between main and small peripheral vessels, which may be key in understanding mechanisms behind subarachnoid hemorrhage-induced vasospasm. Here, changes in calcium and potassium channel expression and function in animal models of subarachnoid hemorrhage and transient global ischemia are systematically reviewed and their clinical significance discussed.
Collapse
Affiliation(s)
- Marcel A. Kamp
- Department for Neurosurgery, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany
- Institute for Neurophysiology, University of Cologne, Robert-Koch-Straße 39, 50931 Cologne, Germany
| | - Maxine Dibué
- Department for Neurosurgery, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany
- Institute for Neurophysiology, University of Cologne, Robert-Koch-Straße 39, 50931 Cologne, Germany
- Center of Molecular Medicine, Cologne, Germany
| | - Toni Schneider
- Institute for Neurophysiology, University of Cologne, Robert-Koch-Straße 39, 50931 Cologne, Germany
- Center of Molecular Medicine, Cologne, Germany
| | - Hans-Jakob Steiger
- Department for Neurosurgery, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Daniel Hänggi
- Department for Neurosurgery, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany
| |
Collapse
|