1
|
Chlorogiannis DD, Aloizou AM, Mavridis T, Sänger JA, Chlorogiannis A, Madouros N, Papanagiotou P. Evolving frontiers: endovascular strategies for the treatment of delayed cerebral ischemia. Rev Neurosci 2024; 35:463-472. [PMID: 38278624 DOI: 10.1515/revneuro-2023-0148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 12/21/2023] [Indexed: 01/28/2024]
Abstract
Cerebral vasospasm and delayed cerebral ischemia represent a very challenging aspect of cerebrovascular pathophysiology, most commonly subarachnoid hemorrhage, with significantly high mortality if left untreated. Considerable advances have been made in medical treatment and prompt diagnosis, while newer endovascular modalities have recently been proposed for cases of resistant cerebral vasospasm. However, there is still paucity of data regarding which and whether a single endovascular technique is non inferior to the pharmacological standard of care. In this review, we aim to summarize the current funds of knowledge concerning cerebral vasospasm and the emerging role of the endovascular techniques for its treatment.
Collapse
Affiliation(s)
- David-Dimitris Chlorogiannis
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Society of Junior Doctors, 15123 Athens, Greece
| | - Athina-Maria Aloizou
- Department of Neurology, St. Josef-Hospital, Ruhr Universität Bochum, 44791 Bochum, Germany
| | - Theodoros Mavridis
- 1st Department of Neurology, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
- Department of Neurology, Tallaght University Hospital (TUH)/The Adelaide and Meath Hospital Dublin, Incorporating the National Children's Hospital (AMNCH), Dublin D24 NR0A, Ireland
| | | | - Anargyros Chlorogiannis
- Department of Health Economics, Policy and Management, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Nikolaos Madouros
- Hull University Teaching Hospitals NHS Trust, Hull HU3 2JZ, UK
- Society of Junior Doctors, 15123 Athens, Greece
| | - Panagiotis Papanagiotou
- First Department of Radiology, School of Medicine, National & Kapodistrian University of Athens, Areteion Hospital, 115 28 Athens, Greece
- Department of Diagnostic and Interventional Neuroradiology, Hospital Bremen-Mitte/Bremen-Ost, 28205 Bremen, Germany
| |
Collapse
|
2
|
Mittal AM, Nowicki KW, Mantena R, Cao C, Rochlin EK, Dembinski R, Lang MJ, Gross BA, Friedlander RM. Advances in biomarkers for vasospasm - Towards a future blood-based diagnostic test. World Neurosurg X 2024; 22:100343. [PMID: 38487683 PMCID: PMC10937316 DOI: 10.1016/j.wnsx.2024.100343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 02/21/2024] [Indexed: 03/17/2024] Open
Abstract
Objective Cerebral vasospasm and the resultant delayed cerebral infarction is a significant source of mortality following aneurysmal SAH. Vasospasm is currently detected using invasive or expensive imaging at regular intervals in patients following SAH, thus posing a risk of complications following the procedure and financial burden on these patients. Currently, there is no blood-based test to detect vasospasm. Methods PubMed, Web of Science, and Embase databases were systematically searched to retrieve studies related to cerebral vasospasm, aneurysm rupture, and biomarkers. The study search dated from 1997 to 2022. Data from eligible studies was extracted and then summarized. Results Out of the 632 citations screened, only 217 abstracts were selected for further review. Out of those, only 59 full text articles met eligibility and another 13 were excluded. Conclusions We summarize the current literature on the mechanism of cerebral vasospasm and delayed cerebral ischemia, specifically studies relating to inflammation, and provide a rationale and commentary on a hypothetical future bloodbased test to detect vasospasm. Efforts should be focused on clinical-translational approaches to create such a test to improve treatment timing and prediction of vasospasm to reduce the incidence of delayed cerebral infarction.
Collapse
Affiliation(s)
- Aditya M. Mittal
- University of Pittsburgh Medical Center, Department of Neurosurgery, Pittsburgh, PA, USA
| | | | - Rohit Mantena
- University of Pittsburgh Medical Center, Department of Neurosurgery, Pittsburgh, PA, USA
| | - Catherine Cao
- University of Pittsburgh Medical Center, Department of Neurosurgery, Pittsburgh, PA, USA
| | - Emma K. Rochlin
- Loyola University Stritch School of Medicine, Maywood, IL, USA
| | - Robert Dembinski
- University of Pittsburgh Medical Center, Department of Neurosurgery, Pittsburgh, PA, USA
| | - Michael J. Lang
- University of Pittsburgh Medical Center, Department of Neurosurgery, Pittsburgh, PA, USA
| | - Bradley A. Gross
- University of Pittsburgh Medical Center, Department of Neurosurgery, Pittsburgh, PA, USA
| | - Robert M. Friedlander
- University of Pittsburgh Medical Center, Department of Neurosurgery, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Bazzigaluppi P, Mester J, Joo IL, Weisspapir I, Dorr A, Koletar MM, Beckett TL, Khosravani H, Carlen P, Stefanovic B. Frequency selective neuronal modulation triggers spreading depolarizations in the rat endothelin-1 model of stroke. J Cereb Blood Flow Metab 2021; 41:2756-2768. [PMID: 33969731 PMCID: PMC8504421 DOI: 10.1177/0271678x211013656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Ischemia is one of the most common causes of acquired brain injury. Central to its noxious sequelae are spreading depolarizations (SDs), waves of persistent depolarizations which start at the location of the flow obstruction and expand outwards leading to excitotoxic damage. The majority of acute stage of stroke studies to date have focused on the phenomenology of SDs and their association with brain damage. In the current work, we investigated the role of peri-injection zone pyramidal neurons in triggering SDs by optogenetic stimulation in an endothelin-1 rat model of focal ischemia. Our concurrent two photon fluorescence microscopy data and local field potential recordings indicated that a ≥ 60% drop in cortical arteriolar red blood cell velocity was associated with SDs at the ET-1 injection site. SDs were also observed in the peri-injection zone, which subsequently exhibited elevated neuronal activity in the low-frequency bands. Critically, SDs were triggered by low- but not high-frequency optogenetic stimulation of peri-injection zone pyramidal neurons. Our findings depict a complex etiology of SDs post focal ischemia and reveal that effects of neuronal modulation exhibit spectral and spatial selectivity.
Collapse
Affiliation(s)
- Paolo Bazzigaluppi
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
- Paolo Bazzigaluppi, Sunnybrook Research Institute, 2075 Bayview Ave., S646, Toronto, ON M4N 3M5, Canada.
| | - James Mester
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Illsung L Joo
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
| | - Iliya Weisspapir
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
| | - Adrienne Dorr
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
| | | | - Tina L Beckett
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
| | - Houman Khosravani
- Division of Neurology and Interdepartmental Division of Critical Care, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
- Interdepartmental Division of Critical Care, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Peter Carlen
- Krembil Research Institute, University of Toronto, Toronto, ON, Canada
| | - Bojana Stefanovic
- Sunnybrook Research Institute, Physical Sciences, Toronto, ON, Canada
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Hosmann A, Milivojev N, Dumitrescu S, Reinprecht A, Weidinger A, Kozlov AV. Cerebral nitric oxide and mitochondrial function in patients suffering aneurysmal subarachnoid hemorrhage-a translational approach. Acta Neurochir (Wien) 2021; 163:139-149. [PMID: 32839865 PMCID: PMC7778629 DOI: 10.1007/s00701-020-04536-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/11/2020] [Indexed: 01/15/2023]
Abstract
Background Cerebral ischemia and neuroinflammation following aneurysmal subarachnoid hemorrhage (SAH) are major contributors to poor neurological outcome. Our study set out to investigate in an exploratory approach the interaction between NO and energy metabolism following SAH as both hypoxia and inflammation are known to affect nitric oxide (NO) metabolism and NO in turn affects mitochondria. Methods In seven patients under continuous multimodality neuromonitoring suffering poor-grade aneurysmal SAH, cerebral metabolism and NO levels (determined as a sum of nitrite plus nitrate) were determined in cerebral microdialysate for 14 days following SAH. In additional ex vivo experiments, rat cortex homogenate was subjected to the NO concentrations determined in SAH patients to test whether these NO concentrations impair mitochondrial function (determined by means of high-resolution respirometry). Results NO levels showed biphasic kinetics with drastically increased levels during the first 7 days (74.5 ± 29.9 μM) and significantly lower levels thereafter (47.5 ± 18.7 μM; p = 0.02). Only during the first 7 days, NO levels showed a strong negative correlation with brain tissue oxygen tension (r = − 0.78; p < 0.001) and a positive correlation with cerebral lactate (r = 0.79; p < 0.001), pyruvate (r = 0.68; p < 0.001), glutamate (r = 0.65; p < 0.001), as well as the lactate-pyruvate ratio (r = 0.48; p = 0.01), suggesting mitochondrial dysfunction. Ex vivo experiments confirmed that the increase in NO levels determined in patients during the acute phase is sufficient to impair mitochondrial function (p < 0.001). Mitochondrial respiration was inhibited irrespectively of whether glutamate (substrate of complex I) or succinate (substrate of complex II) was used as mitochondrial substrate suggesting the inhibition of mitochondrial complex IV. The latter was confirmed by direct determination of complex IV activity. Conclusions Exploratory analysis of our data suggests that during the acute phase of SAH, NO plays a key role in the neuronal damage impairing mitochondrial function and facilitating accumulation of mitochondrial substrate; further studies are required to understand mechanisms underlying this observation.
Collapse
|
5
|
Roles of TRP Channels in Neurological Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7289194. [PMID: 32963700 PMCID: PMC7492880 DOI: 10.1155/2020/7289194] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 07/02/2020] [Indexed: 11/17/2022]
Abstract
Transient receptor potential (TRP) proteins consist of a superfamily of cation channels that have been involved in diverse physiological processes in the brain as well as in the pathogenesis of neurological disease. TRP channels are widely expressed in the brain, including neurons and glial cells, as well as in the cerebral vascular endothelium and smooth muscle. Members of this channel superfamily show a wide variety of mechanisms ranging from ligand binding to voltage, physical, and chemical stimuli, implying the promising therapeutic potential of TRP in neurological diseases. In this review, we focus on the physiological functions of TRP channels in the brain and the pathological roles in neurological disorders to explore future potential neuroprotective strategies.
Collapse
|
6
|
The Role of Sartans in the Treatment of Stroke and Subarachnoid Hemorrhage: A Narrative Review of Preclinical and Clinical Studies. Brain Sci 2020; 10:brainsci10030153. [PMID: 32156050 PMCID: PMC7139942 DOI: 10.3390/brainsci10030153] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/02/2020] [Accepted: 03/05/2020] [Indexed: 12/30/2022] Open
Abstract
Background: Delayed cerebral vasospasm (DCVS) due to aneurysmal subarachnoid hemorrhage (aSAH) and its sequela, delayed cerebral ischemia (DCI), are associated with poor functional outcome. Endothelin-1 (ET-1) is known to play a major role in mediating cerebral vasoconstriction. Angiotensin-II-type-1-receptor antagonists such as Sartans may have a beneficial effect after aSAH by reducing DCVS due to crosstalk with the endothelin system. In this review, we discuss the role of Sartans in the treatment of stroke and their potential impact in aSAH. Methods: We conducted a literature research of the MEDLINE PubMed database in accordance with PRISMA criteria on articles published between 1980 to 2019 reviewing: "Sartans AND ischemic stroke". Of 227 studies, 64 preclinical and 19 clinical trials fulfilled the eligibility criteria. Results: There was a positive effect of Sartans on ischemic stroke in both preclinical and clinical settings (attenuating ischemic brain damage, reducing cerebral inflammation and infarct size, increasing cerebral blood flow). In addition, Sartans reduced DCVS after aSAH in animal models by diminishing the effect of ET-1 mediated vasoconstriction (including cerebral inflammation and cerebral epileptogenic activity reduction, cerebral blood flow autoregulation restoration as well as pressure-dependent cerebral vasoconstriction). Conclusion: Thus, Sartans might play a key role in the treatment of patients with aSAH.
Collapse
|
7
|
Role of endothelin‑1 and its receptors in cerebral vasospasm following subarachnoid hemorrhage. Mol Med Rep 2018; 18:5229-5236. [PMID: 30272323 DOI: 10.3892/mmr.2018.9513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 08/29/2018] [Indexed: 11/05/2022] Open
Abstract
Cerebral vasospasm (CVS) is a severe complication of subarachnoid hemorrhage (SAH), and endothelin‑1 (ET‑1) may be involved in its pathogenesis. The present study aimed to investigate the expression of ET‑1 in cerebrospinal fluid (CSF) in patients with SAH and to analyze rat arterial contractility and the expression levels of ET‑1 receptors in vitro. CSF samples were collected from 28 patients and the expression levels of ET‑1 were measured. Rat cerebral basilar arteries were isolated and incubated with hemorrhagic or clear CSF. Contractility, as well as ETA and ETB mRNA expression were measured. ET‑1 levels in CSF increased and reached a peak within the initial 5 days after SAH onset and then gradually subsided. After 12 or 24 h, the contraction of arteries incubated in hemorrhagic CSF was substantially stronger than those in clear CSF. The mRNA expression levels of endothelin receptor type A and B in arteries incubated in hemorrhagic CSF were significantly higher than those in clear CSF. ET‑1 and its receptors may be involved in the pathogenic mechanism of CVS following SAH. ET‑1 expression in CSF may be used as a marker in CVS and its receptors may provide novel therapeutic targets in CVS.
Collapse
|
8
|
Deng W, Kandhi S, Zhang B, Huang A, Koller A, Sun D. Extravascular Blood Augments Myogenic Constriction of Cerebral Arterioles: Implications for Hemorrhage-Induced Vasospasm. J Am Heart Assoc 2018; 7:JAHA.118.008623. [PMID: 29654195 PMCID: PMC6015404 DOI: 10.1161/jaha.118.008623] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Subarachnoid hemorrhage is a serious clinical condition that impairs local cerebral blood flow perfusion and consequently initiates neuronal dysfunction. Pressure‐sensitive myogenic vasomotor regulation is an important mechanism involved in the regulation of cerebral blood flow. We hypothesized that extravascular hemolyzed blood enhances arteriolar myogenic constriction, which in vivo may contribute to the reduction of local cerebral blood flow after subarachnoid hemorrhage. Methods and Results Arterioles isolated from the middle cerebral artery (MCA arterioles) of mice were cannulated in a perfusion chamber. Arteriolar diameters in response to step increases in intraluminal pressure (20–120 mm Hg) were measured in various experimental conditions. In response to increases in intraluminal pressure, all MCA arterioles exhibited myogenic vasoconstrictions. Compared with controls, the pressure‐induced constriction was significantly enhanced in arterioles (in vitro) exposed to extravascular hemolyzed blood or different concentrations of extracellular erythrocyte lysate (1%, 10%, and 20%) for different exposure durations (1–6 hours). The magnitude of enhancement was proportional to the lysate concentration and exposure duration. In in vivo experiments, 10 μL of autologous blood lysate were injected into the mouse subarachnoid space on the surface of the left MCA. Two hours later, MCA arterioles were isolated and left MCA arterioles displayed enhanced myogenic responses compared with the right MCA. The enhanced myogenic response was prevented by scavenge of superoxide in both in vitro and in vivo experiments. Conclusions Extravascular hemolyzed blood, perhaps by promoting vascular production of superoxide, augments myogenic constriction of cerebral arterioles, which plays a crucial role in the subarachnoid hemorrhage–induced cerebral ischemia.
Collapse
Affiliation(s)
- Wensheng Deng
- Department of Physiology, New York Medical College, Valhalla, NY.,Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai, China
| | - Sharath Kandhi
- Department of Physiology, New York Medical College, Valhalla, NY
| | - Bin Zhang
- Department of Physiology, New York Medical College, Valhalla, NY.,Department of GI Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - An Huang
- Department of Physiology, New York Medical College, Valhalla, NY
| | - Akos Koller
- Department of Physiology, New York Medical College, Valhalla, NY.,Institute of Natural Sciences, Sportgenetics and Sportgerontology Res. Group, University of Physical Education, Budapest, Hungary.,Department of Neurosurgery, Medical School, University of Pecs, Hungary
| | - Dong Sun
- Department of Physiology, New York Medical College, Valhalla, NY
| |
Collapse
|
9
|
Abstract
TRPC channels play important roles in neuronal death/survival in ischemic stroke, vasospasm in hemorrhagic stroke, thrombin-induced astrocyte pathological changes, and also in the initiation of stroke by affecting blood pressure and atherogenesis. TRPCs' unique channel characters and downstream pathways make them possible new targets for stroke therapy. TRPC proteins have different functions in different cell types. Considering TRPCs' extensive distribution in various tissues and cell types, drugs targeting them could induce more complicated effects. More specific agonists/antagonists and antibodies are required for future study of TRPCs as potential targets for stroke therapy.
Collapse
|
10
|
Li X, Xia X, Li X. Plasmid pLXSN-Mediated Adrenomedullin Gene Therapy for Cerebral Vasospasm Following Subarachnoid Hemorrhage in Rats. Med Sci Monit 2017; 23:3293-3302. [PMID: 28684727 PMCID: PMC5513563 DOI: 10.12659/msm.901914] [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] [Indexed: 01/21/2023] Open
Abstract
Background The aim of this study was to investigate the protective effect of ADM gene mediated by plasmid pVAX1 on cerebral vasospasm (CVS) after subarachnoid hemorrhage (SAH). Material/Methods The recombinant plasmid pVAX-ADM was successfully established, and 40 SD rats were randomly divided into normal saline, pVAX1, pVAX1-ADM low-dose, pVAX1-ADM mid-dose, and pVAX1-ADM high-dose groups. The circumference and diameter of basilar artery, diameter of middle cerebral artery and internal carotid artery, and thickness of basilar artery wall were observed. The levels of circulating endothelial cells (CEC) and levels of regional cerebral blood flow (rCBF) of the parietal cortex were detected at different time-points. The expression levels of serum ADM, ET-1, and NOS of each group and the neurological functions were compared. Results The circumference and diameter of basilar artery and the diameter of the middle cerebral artery and internal carotid artery in pVAX1-ADM groups were significantly longer than those in the saline group and pVAX1 group (P<0.05), but the thickness of the basilar artery wall in pVAX1-ADM groups was significantly lower (P<0.05), and the levels of growth or decrease were both dose-dependent (P<0.05). Compared with the saline group and pVAX1 group, the expression levels of serum ADM, NOS, and rCBF in pVAX1-ADM groups were significantly higher (P<0.05), but the levels of serum ET-1 and CEC were significantly lower (P<0.05). The scores of neurobehavioral functions of pVAX1-ADM groups were significantly lower (P<0.05), and the scores were also dose-dependent (P<0.05). Conclusions The recombinant eukaryotic expression plasmid pVAX1-ADM can significantly relieve cerebral vasospasm, increase the expression of serum ADM and NOS, and decrease the expression of serum ET-1 in a rat model of CVS; it is dose-dependent and can also improve nervous system function.
Collapse
Affiliation(s)
- Xin Li
- Department of Neurology, The Second Affiliated Hospital of Tianjin Medical University, Tianjin, China (mainland).,1st Department of Neurology, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang, China (mainland)
| | - Xiaoshuang Xia
- Department of Neurology, The Second Affiliated Hospital of Tianjin Medical University, Tianjin, China (mainland)
| | - Xin Li
- Department of Neurology, The Second Affiliated Hospital of Tianjin Medical University, Tianjin, China (mainland)
| |
Collapse
|
11
|
Etminan N, Macdonald R. Management of aneurysmal subarachnoid hemorrhage. HANDBOOK OF CLINICAL NEUROLOGY 2017; 140:195-228. [DOI: 10.1016/b978-0-444-63600-3.00012-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
12
|
Transient receptor potential channel 1/4 reduces subarachnoid hemorrhage-induced early brain injury in rats via calcineurin-mediated NMDAR and NFAT dephosphorylation. Sci Rep 2016; 6:33577. [PMID: 27641617 PMCID: PMC5027540 DOI: 10.1038/srep33577] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 08/31/2016] [Indexed: 02/06/2023] Open
Abstract
Transient receptor potential channel 1/4 (TRPC1/4) are considered to be related to subarachnoid hemorrhage (SAH)-induced cerebral vasospasm. In this study, a SAH rat model was employed to study the roles of TRPC1/4 in the early brain injury (EBI) after SAH. Primary cultured hippocampal neurons were exposed to oxyhemoglobin to mimic SAH in vitro. The protein levels of TRPC1/4 increased and peaked at 5 days after SAH in rats. Inhibition of TRPC1/4 by SKF96365 aggravated SAH-induced EBI, such as cortical cell death (by TUNEL staining) and degenerating (by FJB staining). In addition, TRPC1/4 overexpression could increase calcineurin activity, while increased calcineurin activity could promote the dephosphorylation of N-methyl-D-aspartate receptor (NMDAR). Calcineurin antagonist FK506 could weaken the neuroprotection and the dephosphorylation of NMDAR induced by TRPC1/4 overexpression. Contrarily, calcineurin agonist chlorogenic acid inhibited SAH-induced EBI, even when siRNA intervention of TRPC1/4 was performed. Moreover, calcineurin also could lead to the nuclear transfer of nuclear factor of activated T cells (NFAT), which is a transcription factor promoting the expressions of TRPC1/4. TRPC1/4 could inhibit SAH-induced EBI by supressing the phosphorylation of NMDAR via calcineurin. TRPC1/4-induced calcineurin activation also could promote the nuclear transfer of NFAT, suggesting a positive feedback regulation of TRPC1/4 expressions.
Collapse
|
13
|
Burrell C, Avalon NE, Siegel J, Pizzi M, Dutta T, Charlesworth MC, Freeman WD. Precision medicine of aneurysmal subarachnoid hemorrhage, vasospasm and delayed cerebral ischemia. Expert Rev Neurother 2016; 16:1251-1262. [PMID: 27314601 DOI: 10.1080/14737175.2016.1203257] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Precision medicine provides individualized treatment of diseases through leveraging patient-to-patient variation. Aneurysmal subarachnoid hemorrhage carries tremendous morbidity and mortality with cerebral vasospasm and delayed cerebral ischemia proving devastating and unpredictable. Lack of treatment measures for these conditions could be improved through precision medicine. Areas covered: Discussed are the pathophysiology of CV and DCI, treatment guidelines, and evidence for precision medicine used for prediction and prevention of poor outcomes following aSAH. A PubMed search was performed using keywords cerebral vasospasm or delayed cerebral ischemia and either biomarkers, precision medicine, metabolomics, proteomics, or genomics. Over 200 peer-reviewed articles were evaluated. The studies presented cover biomarkers identified as predictive markers or therapeutic targets following aSAH. Expert commentary: The biomarkers reviewed here correlate with CV, DCI, and neurologic outcomes after aSAH. Though practical use in clinical management of aSAH is not well established, using these biomarkers as predictive tools or therapeutic targets demonstrates the potential of precision medicine.
Collapse
Affiliation(s)
| | - Nicole E Avalon
- a Department of Neurology , Mayo Clinic , Jacksonville , FL , USA
| | - Jason Siegel
- a Department of Neurology , Mayo Clinic , Jacksonville , FL , USA
| | - Michael Pizzi
- a Department of Neurology , Mayo Clinic , Jacksonville , FL , USA
| | - Tumpa Dutta
- b Endocrine Research Unit , Mayo Clinic , Rochester , MN , USA
| | | | | |
Collapse
|
14
|
Munakata A, Naraoka M, Katagai T, Shimamura N, Ohkuma H. Role of Cyclooxygenase-2 in Relation to Nitric Oxide and Endothelin-1 on Pathogenesis of Cerebral Vasospasm After Subarachnoid Hemorrhage in Rabbit. Transl Stroke Res 2016; 7:220-7. [DOI: 10.1007/s12975-016-0466-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 02/28/2016] [Accepted: 03/29/2016] [Indexed: 10/22/2022]
|
15
|
Glial cell response after aneurysmal subarachnoid hemorrhage — Functional consequences and clinical implications. Biochim Biophys Acta Mol Basis Dis 2016; 1862:492-505. [DOI: 10.1016/j.bbadis.2015.10.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/12/2015] [Accepted: 10/15/2015] [Indexed: 12/17/2022]
|
16
|
"TRP inflammation" relationship in cardiovascular system. Semin Immunopathol 2015; 38:339-56. [PMID: 26482920 PMCID: PMC4851701 DOI: 10.1007/s00281-015-0536-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 10/08/2015] [Indexed: 02/07/2023]
Abstract
Despite considerable advances in the research and treatment, the precise relationship between inflammation and cardiovascular (CV) disease remains incompletely understood. Therefore, understanding the immunoinflammatory processes underlying the initiation, progression, and exacerbation of many cardiovascular diseases is of prime importance. The innate immune system has an ancient origin and is well conserved across species. Its activation occurs in response to pathogens or tissue injury. Recent studies suggest that altered ionic balance, and production of noxious gaseous mediators link to immune and inflammatory responses with altered ion channel expression and function. Among plausible candidates for this are transient receptor potential (TRP) channels that function as polymodal sensors and scaffolding proteins involved in many physiological and pathological processes. In this review, we will first focus on the relevance of TRP channel to both exogenous and endogenous factors related to innate immune response and transcription factors related to sustained inflammatory status. The emerging role of inflammasome to regulate innate immunity and its possible connection to TRP channels will also be discussed. Secondly, we will discuss about the linkage of TRP channels to inflammatory CV diseases, from a viewpoint of inflammation in a general sense which is not restricted to the innate immunity. These knowledge may serve to provide new insights into the pathogenesis of various inflammatory CV diseases and their novel therapeutic strategies.
Collapse
|
17
|
Arctigenin, a Potent Ingredient of Arctium lappa L., Induces Endothelial Nitric Oxide Synthase and Attenuates Subarachnoid Hemorrhage-Induced Vasospasm through PI3K/Akt Pathway in a Rat Model. BIOMED RESEARCH INTERNATIONAL 2015; 2015:490209. [PMID: 26539501 PMCID: PMC4619842 DOI: 10.1155/2015/490209] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 05/27/2015] [Indexed: 01/19/2023]
Abstract
Upregulation of protein kinase B (PKB, also known as Akt) is observed within the cerebral arteries of subarachnoid hemorrhage (SAH) animals. This study is of interest to examine Arctigenin, a potent antioxidant, on endothelial nitric oxide synthase (eNOS) and Akt pathways in a SAH in vitro study. Basilar arteries (BAs) were obtained to examine phosphatidylinositol-3-kinase (PI3K), phospho-PI3K, Akt, phospho-Akt (Western blot) and morphological examination. Endothelins (ETs) and eNOS evaluation (Western blot and immunostaining) were also determined. Arctigenin treatment significantly alleviates disrupted endothelial cells and tortured internal elastic layer observed in the SAH groups (p < 0.01). The reduced eNOS protein and phospho-Akt expression in the SAH groups were relieved by the treatment of Arctigenin (p < 0.01). This result confirmed that Arctigenin might exert dural effects in preventing SAH-induced vasospasm through upregulating eNOS expression via the PI3K/Akt signaling pathway and attenuate endothelins after SAH. Arctigenin shows therapeutic promise in the treatment of cerebral vasospasm following SAH.
Collapse
|
18
|
The change of spatial cognition ability in depression rat model and the possible association with down-regulated protein expression of TRPC6. Behav Brain Res 2015; 294:186-93. [PMID: 26248296 DOI: 10.1016/j.bbr.2015.07.062] [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: 04/27/2015] [Revised: 07/30/2015] [Accepted: 07/31/2015] [Indexed: 11/24/2022]
Abstract
An increasing number of researches have focused on the cognitive changes in depression patients. Here, we observed impaired cognitive ability in a rat depression model along with down-regulated expression of canonical transient receptor potential 6 (TRPC6) protein. The cognitive defect could be rescued by treatment with hyperforin, which can invoke TRPC6 activation. This study was designed as following: rats were randomly divided into control, stressed and stressed+hyperforin groups. Chronic unpredictable stress combined with isolation rearing was applied on rats for three weeks, except for control group. Morris water maze was applied to evaluate spatial cognitive ability while long-term potentiation (LTP) was recorded to test the synaptic plasticity. Results showed that both spatial cognition and synaptic plasticity were impaired in stress group while improved after hyperforin treatment in stressed+hyperforin group, meanwhile, Western blot assay showed that TRPC6 expression was decreased in stressed group. The histological data also presented the decline of dendritic length, dendritic spine density and the number of excitatory synapses in stress group while they were increased in stressed+hyperforin group. These results suggest that there is a well potential of TRPC6 to become a new target for selecting promising new candidates as antidepressants with therapeutically effect on impaired cognition.
Collapse
|
19
|
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
|
20
|
Yue Z, Xie J, Yu AS, Stock J, Du J, Yue L. Role of TRP channels in the cardiovascular system. Am J Physiol Heart Circ Physiol 2015; 308:H157-82. [PMID: 25416190 PMCID: PMC4312948 DOI: 10.1152/ajpheart.00457.2014] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 11/14/2014] [Indexed: 12/12/2022]
Abstract
The transient receptor potential (TRP) superfamily consists of a large number of nonselective cation channels with variable degree of Ca(2+)-permeability. The 28 mammalian TRP channel proteins can be grouped into six subfamilies: canonical, vanilloid, melastatin, ankyrin, polycystic, and mucolipin TRPs. The majority of these TRP channels are expressed in different cell types including both excitable and nonexcitable cells of the cardiovascular system. Unlike voltage-gated ion channels, TRP channels do not have a typical voltage sensor, but instead can sense a variety of other stimuli including pressure, shear stress, mechanical stretch, oxidative stress, lipid environment alterations, hypertrophic signals, and inflammation products. By integrating multiple stimuli and transducing their activity to downstream cellular signal pathways via Ca(2+) entry and/or membrane depolarization, TRP channels play an essential role in regulating fundamental cell functions such as contraction, relaxation, proliferation, differentiation, and cell death. With the use of targeted deletion and transgenic mouse models, recent studies have revealed that TRP channels are involved in numerous cellular functions and play an important role in the pathophysiology of many diseases in the cardiovascular system. Moreover, several TRP channels are involved in inherited diseases of the cardiovascular system. This review presents an overview of current knowledge concerning the physiological functions of TRP channels in the cardiovascular system and their contributions to cardiovascular diseases. Ultimately, TRP channels may become potential therapeutic targets for cardiovascular diseases.
Collapse
Affiliation(s)
- Zhichao Yue
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Jia Xie
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Albert S Yu
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Jonathan Stock
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Jianyang Du
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| | - Lixia Yue
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, Connecticut
| |
Collapse
|
21
|
Assenzio B, Martin EL, Stankevicius E, Civiletti F, Fontanella M, Boccaletti R, Berardino M, Mazzeo A, Ducati A, Simonsen U, Mascia L. Cerebrospinal fluid from patients with subarachnoid haemorrhage and vasospasm enhances endothelin contraction in rat cerebral arteries. PLoS One 2015; 10:e0116456. [PMID: 25629621 PMCID: PMC4309584 DOI: 10.1371/journal.pone.0116456] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 12/10/2014] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Previous studies have suggested that cerebrospinal fluid from patients with subarachnoid hemorrhage (SAH) leads to pronounced vasoconstriction in isolated arteries. We hypothesized that only cerebrospinal fluid from SAH patients with vasospasm would produce an enhanced contractile response to endothelin-1 in rat cerebral arteries, involving both endothelin ETA and ETB receptors. METHODS Intact rat basilar arteries were incubated for 24 hours with cerebrospinal fluid from 1) SAH patients with vasospasm, 2) SAH patients without vasospasm, and 3) control patients. Arterial segments with and without endothelium were mounted in myographs and concentration-response curves for endothelin-1 were constructed in the absence and presence of selective and combined ETA and ETB receptor antagonists. Endothelin concentrations in culture medium and receptor expression were measured. RESULTS Compared to the other groups, the following was observed in arteries exposed to cerebrospinal fluid from patients with vasospasm: 1) larger contractions at lower endothelin concentrations (p<0.05); 2) the increased endothelin contraction was absent in arteries without endothelium; 3) higher levels of endothelin secretion in the culture medium (p<0.05); 4) there was expression of ETA receptors and new expression of ETB receptors was apparent; 5) reduction in the enhanced response to endothelin after ETB blockade in the low range and after ETA blockade in the high range of endothelin concentrations; 6) after combined ETA and ETB blockade a complete inhibition of endothelin contraction was observed. CONCLUSIONS Our experimental findings showed that in intact rat basilar arteries exposed to cerebrospinal fluid from patients with vasospasm endothelin contraction was enhanced in an endothelium-dependent manner and was blocked by combined ETA and ETB receptor antagonism. Therefore we suggest that combined blockade of both receptors may play a role in counteracting vasospasm in patients with SAH.
Collapse
Affiliation(s)
- Barbara Assenzio
- Department of Anesthesia and Intensive Care, Azienda Ospedaliera Città della salute e della scienza di Torino, University of Turin, Turin, Italy
| | - Erica L. Martin
- Department of Anesthesia and Intensive Care, Azienda Ospedaliera Città della salute e della scienza di Torino, University of Turin, Turin, Italy
| | - Edgaras Stankevicius
- Institute of Physiology and Pharmacology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Federica Civiletti
- Department of Anesthesia and Intensive Care, Azienda Ospedaliera Città della salute e della scienza di Torino, University of Turin, Turin, Italy
| | - Marco Fontanella
- Division of Neurosurgery, Department of Neuroscience, Azienda Ospedaliera Città della salute e della scienza di Torino, University of Turin, Turin, Italy
| | - Riccardo Boccaletti
- Division of Neurosurgery, Department of Neuroscience, Azienda Ospedaliera Città della salute e della scienza di Torino, University of Turin, Turin, Italy
| | - Maurizio Berardino
- Department of Anesthesia and Intensive Care, Azienda Ospedaliera Città della salute e della scienza di Torino, University of Turin, Turin, Italy
| | - AnnaTeresa Mazzeo
- Department of Anesthesia and Intensive Care, Azienda Ospedaliera Città della salute e della scienza di Torino, University of Turin, Turin, Italy
| | - Alessandro Ducati
- Division of Neurosurgery, Department of Neuroscience, Azienda Ospedaliera Città della salute e della scienza di Torino, University of Turin, Turin, Italy
| | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, University of Aarhus, Aarhus, Denmark
| | - Luciana Mascia
- Department of Anesthesia and Intensive Care, Azienda Ospedaliera Città della salute e della scienza di Torino, University of Turin, Turin, Italy
- * E-mail:
| |
Collapse
|
22
|
Pathological mechanisms underlying aneurysmal subarachnoid haemorrhage and vasospasmElsevier Ltd. J Clin Neurosci 2015; 22:1-5. [DOI: 10.1016/j.jocn.2014.05.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 04/27/2014] [Accepted: 05/04/2014] [Indexed: 11/18/2022]
|
23
|
Zeng C, Tian F, Xiao B. TRPC Channels: Prominent Candidates of Underlying Mechanism in Neuropsychiatric Diseases. Mol Neurobiol 2014; 53:631-647. [DOI: 10.1007/s12035-014-9004-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 11/13/2014] [Indexed: 10/24/2022]
|
24
|
Compeer MG, Janssen GMJ, De Mey JGR. Endothelin-1 and endothelin-2 initiate and maintain contractile responses by different mechanisms in rat mesenteric and cerebral arteries. Br J Pharmacol 2014; 170:1199-209. [PMID: 23941276 DOI: 10.1111/bph.12332] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 07/29/2013] [Accepted: 08/01/2013] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND AND PURPOSE Endothelin (ET)-1 and ET-2 cause potent long-lasting vasoconstrictions by tight binding to smooth muscle ETA receptors. We tested the hypotheses that different mechanisms mediate initiation and maintenance of arterial contractile responses to ET-1 and ET-2 and that this differs among vascular beds. EXPERIMENTAL APPROACH Segments of rat mesenteric resistance artery (MRA) and basilar artery (BA) were studied in wire myographs with and without functional antagonists. KEY RESULTS Sensitivity and maximum of MRA contractile responses to ET-1 were not, or only moderately, reduced by stimulation of soluble GC, AC or K(+) -channels and by an inhibitor of receptor-operated ion channels. However, each of these reduced maintenance of ET-1 effects and relaxed ET-1-induced contractions in MRA. A calcium channel antagonist did not alter sensitivity, maximum and maintenance of ET-1 effects, but relaxed ET-1-induced contractions in MRA. A PLC inhibitor prevented contractile responses to ET-1 and ET-2 in MRA and BA, and relaxed ET-1- and ET-2-induced responses in MRA and ET-1 effects in BA. A Rho-kinase inhibitor did not modify sensitivity, maximum and maintenance of responses to both peptides in both arteries but relaxed ET-2, but not ET-1, effects in MRA and ET-1 effects in BA. CONCLUSIONS AND IMPLICATIONS PLC played a key role in arterial contractile responses to ETs, but ET-1 and ET-2 initiated and maintained vasoconstriction through different mechanisms, and these differed between MRA and BA. Selective functional antagonism may be considered for agonist- and vascular bed selective pharmacotherapy of ET-related diseases.
Collapse
Affiliation(s)
- M G Compeer
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | | | | |
Collapse
|
25
|
Zeng C, Zhou P, Jiang T, Yuan C, Ma Y, Feng L, Liu R, Tang W, Long X, Xiao B, Tian F. Upregulation and Diverse Roles of TRPC3 and TRPC6 in Synaptic Reorganization of the Mossy Fiber Pathway in Temporal Lobe Epilepsy. Mol Neurobiol 2014; 52:562-72. [PMID: 25213992 DOI: 10.1007/s12035-014-8871-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 08/15/2014] [Indexed: 11/28/2022]
Abstract
Temporal lobe epilepsy (TLE) is the most common form of intractable epilepsy and is always accompanied with hippocampal sclerosis. The molecular mechanism of this pathological phenomenon has been extensively explored, yet remains unclear. Previous studies suggest that ion channels, especially calcium channels, might play important roles. Transient receptor potential canonical channel (TRPC) is a novel cation channel dominantly permeable to Ca(2+) and widely expressed in the human brain. We measured the expression of two subtypes of TRPC channels, TRPC3 and TRPC6, in temporal lobe epileptic foci excised from patients with intractable epilepsy and in hippocampus of mice with pilocarpine-induced status epilepticus (SE), an animal model of TLE. Cortical TRPC3 and TRPC6 protein expressions were significantly higher in TLE patients compared with those in controls. Expression of TRPC3 and TRPC6 protein also increased significantly in the CA3 region of the hippocampus of SE mice. Inhibition of TRPC3 by intracerebroventricular injection of anti-TRPC3 antibody prevented aberrant-sprouted mossy fiber collaterals in the CA3 region, while inhibition of TRPC6 by anti-TRPC6 antibody reduced dendritic arborization and spine density of CA3 pyramidal neurons. Our results indicate that TRPC3 and TRPC6 participate diversely in synaptic reorganization in the mossy fiber pathway in TLE.
Collapse
Affiliation(s)
- Chang Zeng
- Health Management Center, Central South University, Changsha, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Sung HH, Choo SH, Ko M, Kang SJ, Chae MR, Kam SC, Han DH, So I, Lee SW. Increased expression of TRPC4 channels associated with erectile dysfunction in diabetes. Andrology 2014; 2:550-8. [PMID: 24782410 DOI: 10.1111/j.2047-2927.2014.00214.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 03/09/2014] [Accepted: 03/15/2014] [Indexed: 11/27/2022]
Abstract
In recent reports, an association between altered TRPC channel function and the development of various diabetic complications has drawn the attention of many investigators. The aim of this study was to investigate the expression of TRPC4 channels of corpus smooth muscle (CSM) cells in diabetes, and to evaluate the association between erectile dysfunction (ED) and altered TRPC4 channel function. The expression of TRPC4 in the penile tissue of human, normal and diabetic rat was investigated using RT-PCR, western blotting and immunohistochemistry (IHC). In vivo gene transfer of dominant negative (DN) TRPC4 into the CSM of rat was conducted. In vivo pelvic nerve stimulation was performed to measure erectile function. Expression of TRPC1, TRPC3, TRPC4 and TRPC6 in human and rat CSM tissues was confirmed by RT-PCR, western blot and IHC. In the diabetic rat, the expression levels of mRNA and protein of the TRPC4, and TRPC6 were significantly increased compared to control rats (p < 0.05). The change in TRPC4 expression in the diabetic rats was higher than those of the other TRPC subunits (p < 0.05). The IHC showed that only TRPC4 expression had a higher intensity in the diabetes compared to normal rats (p < 0.05). Gene transfection with TRPC4(DN) into the diabetic rats restored erectile function to levels similar to that of normal controls. Gene expression of TRPC4(DN) in CSM tissue was confirmed by RT-PCR 2 weeks after transfection. This study demonstrated that TRPC4 channel expression increased in the penile CSM cells of diabetic rats. The down-regulation of TRPC4 with DN form restored erectile function in the diabetic rats. The alteration of TRPC4 channel is one of pathophysiology of ED and could be a target for drug development for ED.
Collapse
Affiliation(s)
- H H Sung
- Department of Urology, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Shi X, Fu Y, Liao D, Chen Y, Liu J. Alterations of voltage-dependent calcium channel currents in basilar artery smooth muscle cells at early stage of subarachnoid hemorrhage in a rabbit model. PLoS One 2014; 9:e84129. [PMID: 24392110 PMCID: PMC3879272 DOI: 10.1371/journal.pone.0084129] [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: 09/09/2013] [Accepted: 11/12/2013] [Indexed: 02/05/2023] Open
Abstract
Objective To investigate the changes in the currents of voltage-dependent calcium channels (VDCCs) in smooth muscle cells of basilar artery in a rabbit model of subarachnoid hemorrhage (SAH). Methods New Zealand white rabbits were randomly divided into five groups: sham (C), normal (N), 24 hours (S1), 48 hours (S2) and 72 hours (S3) after SAH. Non-heparinized autologous arterial blood (1ml/kg) was injected into the cisterna magna to create SAH after intravenous anesthesia, and 1 ml/kg of saline was injected into cisterna magna in the sham group. Rabbits in group N received no injections. Basilar artery in S1, S2, S3 group were isolated at 24, 48, 72 hours after SAH. Basilar artery in group C was isolated at 72 hours after physiological saline injection. Basilar artery smooth muscle cells were isolated for all groups. Whole-cell patch-clamp technique was utilized to record cell membrane capacitance and VDCCs currents. The VDCCs antagonist nifedipine was added to the bath solution to block the Ca++ channels currents. Results There were no significant differences in the number of cells isolated, the cell size and membrane capacitance among all the five groups. VDCC currents in the S1–S3 groups had higher amplitudes than those in control and sham groups. The significant change of current amplitude was observed at 72 hours after SAH, which was higher than those of 24 and 48 hours. The VDCCs were shown to expression in human artery smooth muscle cells. Conclusions The changes of activation characteristics and voltage-current relationship at 72 hours after SAH might be an important event which leads to a series of molecular events in the microenvironment of the basilar artery smooth muscle cells. This may be the key time point for potential therapeutic intervention against subarachnoid hemorrhage.
Collapse
MESH Headings
- Animals
- Basilar Artery/drug effects
- Basilar Artery/metabolism
- Basilar Artery/physiopathology
- Blood Pressure
- Body Weight
- Brain/blood supply
- Brain/metabolism
- Calcium Channels/metabolism
- Calcium Channels, L-Type/metabolism
- Cells, Cultured
- Disease Models, Animal
- Evoked Potentials
- Heart Rate
- Humans
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Nifedipine/pharmacology
- Patch-Clamp Techniques
- Rabbits
- Subarachnoid Hemorrhage/metabolism
- Subarachnoid Hemorrhage/physiopathology
Collapse
Affiliation(s)
- Xianqing Shi
- Intensive Care Unit, Guizhou Province People's Hospital, Guiyang, China
| | - Yongjian Fu
- Intensive Care Unit, Guizhou Province People's Hospital, Guiyang, China
| | - Daqing Liao
- Laboratory of Anesthesia and Critical Medicine, West China hospital, Sichuan University, Chengdou, China
| | - Yanfang Chen
- Laboratory of Anesthesia and Critical Medicine, West China hospital, Sichuan University, Chengdou, China
| | - Jin Liu
- Laboratory of Anesthesia and Critical Medicine, West China hospital, Sichuan University, Chengdou, China
- * E-mail:
| |
Collapse
|
28
|
Abstract
TRPC4 proteins comprise six transmembrane domains, a putative pore-forming region, and an intracellularly located amino- and carboxy-terminus. Among eleven splice variants identified so far, TRPC4α and TRPC4β are the most abundantly expressed and functionally characterized. TRPC4 is expressed in various organs and cell types including the soma and dendrites of numerous types of neurons; the cardiovascular system including endothelial, smooth muscle, and cardiac cells; myometrial and skeletal muscle cells; kidney; and immune cells such as mast cells. Both recombinant and native TRPC4-containing channels differ tremendously in their permeability and other biophysical properties, pharmacological modulation, and mode of activation depending on the cellular environment. They vary from inwardly rectifying store-operated channels with a high Ca(2+) selectivity to non-store-operated channels predominantly carrying Na(+) and activated by Gαq- and/or Gαi-coupled receptors with a complex U-shaped current-voltage relationship. Thus, individual TRPC4-containing channels contribute to agonist-induced Ca(2+) entry directly or indirectly via depolarization and activation of voltage-gated Ca(2+) channels. The differences in channel properties may arise from variations in the composition of the channel complexes, in the specific regulatory pathways in the corresponding cell system, and/or in the expression pattern of interaction partners which comprise other TRPC proteins to form heteromultimeric channels. Additional interaction partners of TRPC4 that can mediate the activity of TRPC4-containing channels include (1) scaffolding proteins (e.g., NHERF) that may mediate interactions with signaling molecules in or in close vicinity to the plasma membrane such as Gα proteins or phospholipase C and with the cytoskeleton, (2) proteins in specific membrane microdomains (e.g., caveolin-1), or (3) proteins on cellular organelles (e.g., Stim1). The diversity of TRPC4-containing channels hampers the development of specific agonists or antagonists, but recently, ML204 was identified as a blocker of both recombinant and endogenous TRPC4-containing channels with an IC50 in the lower micromolar range that lacks activity on most voltage-gated channels and other TRPs except TRPC5 and TRPC3. Lanthanides are specific activators of heterologously expressed TRPC4- and TRPC5-containing channels but can block individual native TRPC4-containing channels. The biological relevance of TRPC4-containing channels was demonstrated by knockdown of TRPC4 expression in numerous native systems including gene expression, cell differentiation and proliferation, formation of myotubes, and axonal regeneration. Studies of TRPC4 single and TRPC compound knockout mice uncovered their role for the regulation of vascular tone, endothelial permeability, gastrointestinal contractility and motility, neurotransmitter release, and social exploratory behavior as well as for excitotoxicity and epileptogenesis. Recently, a single-nucleotide polymorphism (SNP) in the Trpc4 gene was associated with a reduced risk for experience of myocardial infarction.
Collapse
Affiliation(s)
- Marc Freichel
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany,
| | | | | |
Collapse
|
29
|
Shimamura N, Ohkuma H. Phenotypic transformation of smooth muscle in vasospasm after aneurysmal subarachnoid hemorrhage. Transl Stroke Res 2013; 5:357-64. [PMID: 24323729 DOI: 10.1007/s12975-013-0310-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/24/2013] [Accepted: 11/07/2013] [Indexed: 12/14/2022]
Abstract
Differentiated smooth muscle cells (SMC) control vasoconstriction and vasodilation, but they can undergo transformation, proliferate, secret cytokines, and migrate into the subendotherial layer with adverse consequences. In this review, we discuss the phenotypic transformation of SMC in cerebral vasospasm after subarachnoid hemorrhage. Phenotypic transformation starts with an insult as caused by aneurysm rupture: Elevation of intracranial and blood pressure, secretion of norepinephrine, and mechanical force on an artery are factors that can cause aneurysm. The phenotypic transformation of SMC is accelerated by inflammation, thrombin, and growth factors. A wide variety of cytokines (e.g., interleukin (IL)-1β, IL-33, matrix metalloproteinases, nitric oxidase synthases, endothelins, thromboxane A2, mitogen-activated protein kinase, platelet-derived vascular growth factors, and vascular endothelial factor) all play roles in cerebral vasospasm (CVS). We summarize the correlations between various factors and the phenotypic transformation of SMC. A new target of this study is the transient receptor potential channel in CVS. Statin together with fasdil prevents phenotypic transformation of SMC in an animal model. Clazosentan prevents CVS and improves outcome in aneurysmal subarachnoid hemorrhage in a dose-dependent manner. Clinical trials of cilostazol for the prevention of phenotypic transformation of SMC have been reported, along with requisite experimental evidence. To conquer CVS in its complexity, we will ultimately need to elucidate its general, underlying mechanism.
Collapse
Affiliation(s)
- Norihito Shimamura
- Department of Neurosurgery, Hirosaki University School of Medicine, 5-Zaihuchou, Hirosaki, Aomori Prefecture, 036-8562, Japan,
| | | |
Collapse
|
30
|
Toth P, Csiszar A, Tucsek Z, Sosnowska D, Gautam T, Koller A, Schwartzman ML, Sonntag WE, Ungvari Z. Role of 20-HETE, TRPC channels, and BKCa in dysregulation of pressure-induced Ca2+ signaling and myogenic constriction of cerebral arteries in aged hypertensive mice. Am J Physiol Heart Circ Physiol 2013; 305:H1698-708. [PMID: 24097425 DOI: 10.1152/ajpheart.00377.2013] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hypertension in the elderly substantially increases the risk of stroke and vascular cognitive impairment in part due to an impaired functional adaptation of aged cerebral arteries to high blood pressure. To elucidate the mechanisms underlying impaired autoregulatory protection in aging, hypertension was induced in young (3 mo) and aged (24 mo) C57BL/6 mice by chronic infusion of angiotensin II and pressure-induced changes in smooth muscle cell (SMC) intracellular Ca(2+) concentration ([Ca(2+)]i) and myogenic constriction of middle cerebral arteries (MCA) were assessed. In MCAs from young hypertensive mice, pressure-induced increases in vascular SMC [Ca(2+)]i and myogenic tone were increased, and these adaptive responses were inhibited by the cytochrome P-450 ω-hydroxylase inhibitor HET0016 and the transient receptor potential (TRP) channel blocker SKF96365. Administration of 20- hydroxyeicosatetraenoic acid (HETE) increased SMC [Ca(2+)]i and constricted MCAs, and these responses were inhibited by SKF96365. MCAs from aged hypertensive mice did not show adaptive increases in pressure-induced calcium signal and myogenic tone and responses to HET0016 and SKF96365 were blunted. Inhibition of large-conductance Ca(2+)-activated K(+) (BK) channels by iberiotoxin enhanced SMC [Ca(2+)]i and myogenic constriction in MCAs of young normotensive animals, whereas it was without effect in MCAs of young hypertensive mice. Iberiotoxin did not restore myogenic adaptation in MCAs of aged hypertensive mice. Thus functional maladaptation of aged cerebral arteries to hypertension is due to the dysregulation of pressure-induced 20-HETE and TRP channel-mediated SMC calcium signaling, whereas overactivation of BK channels is unlikely to play a role in this phenomenon.
Collapse
Affiliation(s)
- Peter Toth
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Endothelial control of vasodilation: integration of myoendothelial microdomain signalling and modulation by epoxyeicosatrienoic acids. Pflugers Arch 2013; 466:389-405. [PMID: 23748495 DOI: 10.1007/s00424-013-1303-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 05/24/2013] [Accepted: 05/26/2013] [Indexed: 12/17/2022]
Abstract
Endothelium-derived epoxyeicosatrienoic acids (EETs) are fatty acid epoxides that play an important role in the control of vascular tone in selected coronary, renal, carotid, cerebral and skeletal muscle arteries. Vasodilation due to endothelium-dependent smooth muscle hyperpolarization (EDH) has been suggested to involve EETs as a transferable endothelium-derived hyperpolarizing factor. However, this activity may also be due to EETs interacting with the components of other primary EDH-mediated vasodilator mechanisms. Indeed, the transfer of hyperpolarization initiated in the endothelium to the adjacent smooth muscle via gap junction connexins occurs separately or synergistically with the release of K(+) ions at discrete myoendothelial microdomain signalling sites. The net effects of such activity are smooth muscle hyperpolarization, closure of voltage-dependent Ca(2+) channels, phospholipase C deactivation and vasodilation. The spatially localized and key components of the microdomain signalling complex are the inositol 1,4,5-trisphosphate receptor-mediated endoplasmic reticulum Ca(2+) store, Ca(2+)-activated K(+) (KCa), transient receptor potential (TRP) and inward-rectifying K(+) channels, gap junctions and the smooth muscle Na(+)/K(+)-ATPase. Of these, TRP channels and connexins are key endothelial effector targets modulated by EETs. In an integrated manner, endogenous EETs enhance extracellular Ca(2+) influx (thereby amplifying and prolonging KCa-mediated endothelial hyperpolarization) and also facilitate the conduction of this hyperpolarization to spatially remote vessel regions. The contribution of EETs and the receptor and channel subtypes involved in EDH-related microdomain signalling, as a candidate for a universal EDH-mediated vasodilator mechanism, vary with vascular bed, species, development and disease and thus represent potentially selective targets for modulating specific artery function.
Collapse
|
32
|
Shen J, Pan JW, Fan ZX, Xiong XX, Zhan RY. Dissociation of vasospasm-related morbidity and outcomes in patients with aneurysmal subarachnoid hemorrhage treated with clazosentan: a meta-analysis of randomized controlled trials. J Neurosurg 2013; 119:180-9. [PMID: 23641823 DOI: 10.3171/2013.3.jns121436] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Clazosentan therapy after aneurysmal subarachnoid hemorrhage (SAH) has been found to be effective in reducing the incidence of vasospasm in randomized controlled trials. However, while vasospasm-related morbidity, including delayed ischemic neurological deficits (DINDs) and delayed cerebral infarctions, was consistently decreased, statistical significance was not demonstrated and outcomes were not affected by clazosentan treatment. The objective of this meta-analysis was to determine whether clazosentan treatment after aneurysmal SAH significantly reduced the incidence of DINDs and delayed cerebral infarctions and improved outcomes. METHODS All randomized controlled trials investigating the effect of clazosentan were retrieved via searches with sensitive and specific terms. Six variables were abstracted after the assessment of the methodological quality of the trials. Analyses were performed following the method guidelines of the Cochrane Back Review Group. RESULTS Four randomized, placebo-controlled trials met eligibility criteria, enrolling a total of 2181 patients. The meta-analysis demonstrated a significant decrease in the incidence of DINDs (relative risk [RR] 0.76 [95% CI 0.62-0.92]) and delayed cerebral infarction (RR 0.79 [95% CI 0.63-1.00]) in patients treated with clazosentan after aneurysmal SAH. However, this treatment regimen was not shown to outcomes including functional outcomes measured by Glasgow Outcome Scale-Extended (RR 1.12 [95% CI 0.96-1.30]) or mortality (RR 1.02 [95%CI 0.70-1.49]). Adverse events, including pulmonary complications, anemia, and hypotension, were all significantly increased in patients who received clazosentan therapy. CONCLUSIONS The results of the present meta-analysis show that treatment with clazosentan after aneurysmal SAH significantly reduced the incidence of the vasospasm-related DINDs and delayed cerebral infarctions, but did not improve poor neurological outcomes in patients with aneurysmal SAH. Further study is required to elucidate the dissociation between vasospasm-related morbidity and outcomes.
Collapse
Affiliation(s)
- Jian Shen
- Department of Neurosurgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou Zhejiang Province, People's Republic of China
| | | | | | | | | |
Collapse
|
33
|
Song JN, Yan WT, An JY, Hao GS, Guo XY, Zhang M, Li Y, Li DD, Sun P. Potential contribution of SOCC to cerebral vasospasm after experimental subarachnoid hemorrhage in rats. Brain Res 2013; 1517:93-103. [PMID: 23542055 DOI: 10.1016/j.brainres.2013.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Revised: 01/02/2013] [Accepted: 01/04/2013] [Indexed: 11/27/2022]
Abstract
Cerebral vasospasm (CVS) is the most treatable component of subarachnoid hemorrhage (SAH), which can be reduced by endothelin receptor antagonists. Endothelin-evoked vasospasm is considered to be mediated by Ca(2+) influx in the smooth muscle through voltage-dependent Ca(2+) channel (VDCC) and nonselective cation channels (NSCC). Because VDCC antagonists such as nimodipine have been shown to be relatively less effective than the endothelin receptor antagonists, it is assumed that NSCC maybe a more important component in mediating Ca(2+) influx during CVS. In this study, we used the basilar arteries from a "two-hemorrhage" rat model of SAH to investigate expressions of transient receptor potential channel 1 (TRPC1), transient receptor potential channel 3 (TRPC3) and stromal interaction molecule 1 (STIM1), which are considered as the promising candidates constituting NSCC. To investigate the possible role of NSCC in phenotypic switching, we performed immunohistochemical staining to examine expressions of SMα-actin and PCNA, markers of smooth muscle phenotypic switching. We found that the basilar arteries exhibited vasospasm after SAH and that vasospasm became more severe on days 5 and 7 after SAH. Elevated mRNA and protein expressions of TRPC1 and STIM1 were detected after SAH and peaked on days 5 and 7, which was in a parallel time course to the development of cerebral vasospasm. The mRNA and protein expressions of TRPC3 were not changed in the SAH group when compared with those in the control. Results of immunohistochemical staining with anti-PCNA and anti-SMα-actin antibodies also showed enhanced expression of PCNA and disappearance of SMα-actin from day 1 to day 7. Taken together, the above results supported a novel mechanism that the components of store-operated calcium channels, TRPC1 and STIM1 mediated the Ca(2+) influx and phenotypic switching in smooth muscle cells, which promoted the development of vasospasm after SAH. TRPC3, which is a component of receptor-operated calcium channels, was not involved in the above-mentioned mechanism.
Collapse
Affiliation(s)
- Jin-Ning Song
- Department of Neurosurgery, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sun L, Zhang W, Wang X, Song J, Li M. Inhibition of protein kinase C signal reduces ET receptor expression and basilar vasospasm after subarachnoid hemorrhage in rats. J Integr Neurosci 2012; 11:439-51. [DOI: 10.1142/s0219635212500288] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
35
|
Inversion of neurovascular coupling by subarachnoid blood depends on large-conductance Ca2+-activated K+ (BK) channels. Proc Natl Acad Sci U S A 2012; 109:E1387-95. [PMID: 22547803 DOI: 10.1073/pnas.1121359109] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The cellular events that cause ischemic neurological damage following aneurysmal subarachnoid hemorrhage (SAH) have remained elusive. We report that subarachnoid blood profoundly impacts communication within the neurovascular unit-neurons, astrocytes, and arterioles-causing inversion of neurovascular coupling. Elevation of astrocytic endfoot Ca(2+) to ∼400 nM by neuronal stimulation or to ∼300 nM by Ca(2+) uncaging dilated parenchymal arterioles in control brain slices but caused vasoconstriction in post-SAH brain slices. Inhibition of K(+) efflux via astrocytic endfoot large-conductance Ca(2+)-activated K(+) (BK) channels prevented both neurally evoked vasodilation (control) and vasoconstriction (SAH). Consistent with the dual vasodilator/vasoconstrictor action of extracellular K(+) ([K(+)](o)), [K(+)](o) <10 mM dilated and [K(+)](o) >20 mM constricted isolated brain cortex parenchymal arterioles with or without SAH. Notably, elevation of external K(+) to 10 mM caused vasodilation in brain slices from control animals but caused a modest constriction in brain slices from SAH model rats; this latter effect was reversed by BK channel inhibition, which restored K(+)-induced dilations. Importantly, the amplitude of spontaneous astrocytic Ca(2+) oscillations was increased after SAH, with peak Ca(2+) reaching ∼490 nM. Our data support a model in which SAH increases the amplitude of spontaneous astrocytic Ca(2+) oscillations sufficiently to activate endfoot BK channels and elevate [K(+)](o) in the restricted perivascular space. Abnormally elevated basal [K(+)](o) combined with further K(+) efflux stimulated by neuronal activity elevates [K(+)](o) above the dilation/constriction threshold, switching the polarity of arteriolar responses to vasoconstriction. Inversion of neurovascular coupling may contribute to the decreased cerebral blood flow and development of neurological deficits that commonly follow SAH.
Collapse
|
36
|
Macdonald RL. Site-Specific, Sustained-Release Drug Delivery for Subarachnoid Hemorrhage. Transl Stroke Res 2012. [DOI: 10.1007/978-1-4419-9530-8_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
37
|
Komotar RJ, Starke RM, Connolly ES. The Effect of Endothelin Receptor Antagonists on Vasospasm Following Aneurysmal Subarachnoid Hemorrhage. Neurosurgery 2011; 69:N13-4. [DOI: 10.1227/01.neu.0000407918.14115.e5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
38
|
Shi J, Ju M, Abramowitz J, Large WA, Birnbaumer L, Albert AP. TRPC1 proteins confer PKC and phosphoinositol activation on native heteromeric TRPC1/C5 channels in vascular smooth muscle: comparative study of wild-type and TRPC1-/- mice. FASEB J 2011; 26:409-19. [PMID: 21968068 DOI: 10.1096/fj.11-185611] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Ca(2+)-permeable cation channels consisting of canonical transient receptor potential 1 (TRPC1) proteins mediate Ca(2+) influx pathways in vascular smooth muscle cells (VSMCs), which regulate physiological and pathological functions. We investigated properties conferred by TRPC1 proteins to native single TRPC channels in acutely isolated mesenteric artery VSMCs from wild-type (WT) and TRPC1-deficient (TRPC1(-/-)) mice using patch-clamp techniques. In WT VSMCs, the intracellular Ca(2+) store-depleting agents cyclopiazonic acid (CPA) and 1,2-bis-(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA-AM) both evoked channel currents, which had unitary conductances of ∼2 pS. In TRPC1(-/-) VSMCs, CPA-induced channel currents had 3 subconductance states of 14, 32, and 53 pS. Passive depletion of intracellular Ca(2+) stores activated whole-cell cation currents in WT but not TRPC1(-/-) VSMCs. Differential blocking actions of anti-TRPC antibodies and coimmunoprecipitation studies revealed that CPA induced heteromeric TRPC1/C5 channels in WT VSMCs and TRPC5 channels in TRPC1(-/-) VSMCs. CPA-evoked TRPC1/C5 channel activity was prevented by the protein kinase C (PKC) inhibitor chelerythrine. In addition, the PKC activator phorbol 12,13-dibutyrate (PDBu), a PKC catalytic subunit, and phosphatidylinositol-4,5-bisphosphate (PIP(2)) and phosphatidylinositol-3,4,5-trisphosphate (PIP(3)) activated TRPC1/C5 channel activity, which was prevented by chelerythrine. In contrast, CPA-evoked TRPC5 channel activity was potentiated by chelerythrine, and inhibited by PDBu, PIP(2), and PIP(3). TRPC5 channels in TRPC1(-/-) VSMCs were activated by increasing intracellular Ca(2+) concentrations ([Ca(2+)](i)), whereas increasing [Ca(2+)](i) had no effect in WT VSMCs. We conclude that agents that deplete intracellular Ca(2+) stores activate native heteromeric TRPC1/C5 channels in VSMCs, and that TRPC1 subunits are important in determining unitary conductance and conferring channel activation by PKC, PIP(2), and PIP(3).
Collapse
Affiliation(s)
- Jian Shi
- Division of Biomedical Sciences, Cranmer St. George's, University of London, London, UK
| | | | | | | | | | | |
Collapse
|
39
|
Thilo F, Suess O, Liu Y, Tepel M. Decreased Expression of Transient Receptor Potential Channels in Cerebral Vascular Tissue from Patients After Hypertensive Intracerebral Hemorrhage. Clin Exp Hypertens 2011; 33:533-7. [DOI: 10.3109/10641963.2011.561903] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
40
|
Synthesis and in vitro evaluation of ambrisentan analogues as potential endothelin receptor antagonists. Bioorg Med Chem Lett 2011; 21:3894-7. [DOI: 10.1016/j.bmcl.2011.05.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 04/18/2011] [Accepted: 05/10/2011] [Indexed: 01/01/2023]
|
41
|
Chang CZ, Wu SC, Kwan AL, Lin CL, Hwang SL. 6-Mercaptopurine reverses experimental vasospasm and alleviates the production of endothelins in NO-independent mechanism-a laboratory study. Acta Neurochir (Wien) 2011; 153:939-49. [PMID: 21170559 DOI: 10.1007/s00701-010-0865-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Accepted: 10/29/2010] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Increased endothelin-1 (ET-1) production and diminished nitric oxide synthase (NOS) bioavailability has been observed in aneurysmal subarachnoid hemorrhage (SAH). The authors previously found that 6-mercaptopurine (6-mp) is effective in preventing and reversing arterial narrowing in a rodent SAH model. This present study is of interest to examine the effect of 6-mp on ET-1/endothelial nitric oxide synthase (eNOS) in this animal model. METHODS A rodent double hemorrhage SAH model was employed. Animals were randomly assigned to six groups (sham, SAH only, vehicle, 0.5, 1.0 and 2 mg kg(-1) day(-1) 6-mp treatment). Monoclonal CD45 immunostaining was utilized to evaluate monocytes and microglia. The level of pro-inflammatory cytokines, such as IL-1, IL-6 and TNF-α(RT-PCR), and ET-1 (ELISA) was measured. The basilar arteries (BAs) were harvested and sliced, and their cross-sectional areas were determined. Radiolabeled NOS assay kit was applied to detect eNOS. RESULTS Morphologically, convolution of internal elastic lamina, endothelial cells distortion, and necrotic smooth muscle were prevalently present in the basilar artery of SAH groups, which was absent in the 1 and 2 mg kg(-1) day(-1) 6-mp plus SAH group or the healthy controls. Significant vasospasm was noted in the vehicle group (lumen patency, 54.6%, p ≤ 0.01 compared with the sham group), but it was less prominent in the 2 mg kg(-1) day(-1) 6-mp treatment group (lumen patency, 87.6%, p < 0.05). In addition, administration with 2 mg kg(-1) day(-1) 6-mp reduced cytokine levels by 11%, 47%, and 34% for IL-1, IL-6, and TNF-α, respectively, and increased ET-1 levels were found in all the animals subject to SAH (SAH only, SAH plus vehicle, SAH plus 0.5 and 1.0 mg kg(-1) day(-1) 6-mp) except in the 2 mg kg(-1) day(-1) 6-mp SAH group, when compared with the healthy controls (no SAH). Meanwhile, treatment with 6-mp did not induce the levels of expressed eNOS in BAs in the 6-mp groups (0.5, 1.0, and 2 mg kg(-1) day(-1) 6-mp plus SAH) when compared with that in the SAH groups (p > 0.1). CONCLUSION In summary, treatment with 6-mp decreased the release of pro-inflammatory cytokines and diminished experimental vasospasm. This study offered first evidence that 6-mp dose-dependently reduces the level of ET-1 in a NO-independent mechanism, which corresponds to its antivasospastic effect in the condition of chronic vasospasm.
Collapse
MESH Headings
- Aneurysm, Ruptured/pathology
- Aneurysm, Ruptured/physiopathology
- Animals
- Chemotaxis/drug effects
- Connective Tissue/drug effects
- Connective Tissue/pathology
- Cytokines/metabolism
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Endothelin-1/metabolism
- Immunosuppressive Agents/pharmacology
- Inflammation Mediators/metabolism
- Intracranial Aneurysm/pathology
- Intracranial Aneurysm/physiopathology
- Male
- Mercaptopurine/pharmacology
- Microglia/drug effects
- Microglia/pathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Nitric Oxide/physiology
- Nitric Oxide Synthase/metabolism
- Rats
- Rats, Sprague-Dawley
- Subarachnoid Hemorrhage/pathology
- Subarachnoid Hemorrhage/physiopathology
- Vasospasm, Intracranial/pathology
- Vasospasm, Intracranial/physiopathology
Collapse
Affiliation(s)
- Chih-Zen Chang
- Department of Surgery, College of Medicine, Kaohsiung Medical University, Taiwan, Republic of China.
| | | | | | | | | |
Collapse
|
42
|
Jorks D, Major S, Oliveira-Ferreira AI, Kleeberg J, Dreier JP. Endothelin-1(1-31) induces spreading depolarization in rats. ACTA NEUROCHIRURGICA. SUPPLEMENT 2011; 110:111-7. [PMID: 21116925 DOI: 10.1007/978-3-7091-0353-1_20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
BACKGROUND The vasoconstrictor endothelin-1(1-21) (ET-1) seems to induce cerebral vasospasm after aneurismal subarachnoid hemorrhage (aSAH). Moreover, ET-1 causes spreading depolarization (SD) via vasoconstriction/ischemia. ET-1(1-31) is an alternate metabolic intermediate in the generation of ET-1. Our aim was to investigate whether endothelin-1(1-31) causes SD in a similar fashion to ET-1. METHOD Increasing concentrations of either ET-1, ET-1(1-31) or vehicle were brain topically applied in 29 rats. Each concentration was superfused for one hour while regional cerebral blood flow (rCBF) and direct current electrocorticogram (DC-ECoG) were recorded. FINDINGS In response to the highest concentration of 10(-6) M, all animals of both ET groups developed typical SD. At concentrations below 10(-6) M only ET-1 induced SD (n=14 of 19 rats). Thus, the efficacy of ET-1(1-31) to induce SD was significantly lower (P<0.001, two-tailed Fisher's Exact Test). CONCLUSIONS Our findings suggest that ET-1(1-31) less potently induces SD compared to ET-1 which implicates that it is a less potent vasoconstrictor. Speculatively, it could be interesting to shift the metabolic pathway towards the alternate intermediate ET-1(1-31) after aSAH as an alternative strategy to ETA receptor inhibition. This could decrease ET-induced vasoconstriction and SD generation while a potentially beneficial basal ETA receptor activation is maintained.
Collapse
Affiliation(s)
- D Jorks
- Department of Experimental Neurology, Charité University Medicine Berlin, Berlin, Germany
| | | | | | | | | |
Collapse
|
43
|
Abstract
Despite advances in aneurysm ablation and the initial management of patients presenting with aneurysmal subarachnoid hemorrhage, delayed cerebral ischemia remains a significant source of morbidity. Traditionally, delayed cerebral ischemia was thought to be a result of vasospasm of the proximal intracranial vessels, and clinical trials have relied largely on radiographic evidence of vasospasm as a surrogate for functional outcome. However, a number of trials have demonstrated a dissociation between angiographic vasospasm and outcome, and more recent data suggest that other mechanisms of injury, such as microvascular dysfunction and complex neuronal-glial interactions, may influence the development of delayed ischemic deficit after aneurysmal subarachnoid hemorrhage. Our evolving understanding of the pathophysiology of delayed cerebral ischemia may offer the opportunity to test new therapeutic strategies in this area and improve clinical trial design.
Collapse
Affiliation(s)
- Daniel T Laskowitz
- Departments of Medicine Neurology, Duke University, Durham NC 27710, USA.
| | | |
Collapse
|
44
|
Oliveira-Ferreira AI, Milakara D, Alam M, Jorks D, Major S, Hartings JA, Lückl J, Martus P, Graf R, Dohmen C, Bohner G, Woitzik J, Dreier JP. Experimental and preliminary clinical evidence of an ischemic zone with prolonged negative DC shifts surrounded by a normally perfused tissue belt with persistent electrocorticographic depression. J Cereb Blood Flow Metab 2010; 30:1504-19. [PMID: 20332797 PMCID: PMC2949249 DOI: 10.1038/jcbfm.2010.40] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In human cortex it has been suggested that the tissue at risk is indicated by clusters of spreading depolarizations (SDs) with persistent depression of high-frequency electrocorticographic (ECoG) activity. We here characterized this zone in the ET-1 model in rats using direct current (DC)-ECoG recordings. Topical application of the vasoconstrictor endothelin-1 (ET-1) induces focal ischemia in a concentration-dependent manner restricted to a region exposed by a cranial window, while a healthy cortex can be studied at a second naïve window. SDs originate in the ET-1-exposed cortex and invade the surrounding tissue. Necrosis is restricted to the ET-1-exposed cortex. In this study, we discovered that persistent depression occurred in both ET-1-exposed and surrounding cortex during SD clusters. However, the ET-1-exposed cortex showed longer-lasting negative DC shifts and limited high-frequency ECoG recovery after the cluster. DC-ECoG recordings of SD clusters with persistent depression from patients with aneurysmal subarachnoid hemorrhage were then analyzed for comparison. Limited ECoG recovery was associated with significantly longer-lasting negative DC shifts in a similar manner to the experimental model. These preliminary results suggest that the ischemic zone in rat and human cortex is surrounded by a normally perfused belt with persistently reduced synaptic activity during the acute injury phase.
Collapse
|
45
|
Gonzalez-Cobos JC, Trebak M. TRPC channels in smooth muscle cells. Front Biosci (Landmark Ed) 2010; 15:1023-39. [PMID: 20515740 DOI: 10.2741/3660] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Transient receptor potential canonical (TRPC) proteins constitute a family of seven (TRPC1-7) nonselective cation channels within the wider TRP superfamily. TRPC1, TRPC3, TRPC4, TRPC5 and TRPC6 channels are expressed in vascular smooth muscle cells from human vessels of all calibers and in smooth muscle from organs such as the uterus and the gastrointestinal tract. TRPC channels have recently emerged as important players in the control of smooth muscle function. This review will focus on the retrospective analysis of studies proposing contributions of TRPC channels to native calcium entry pathways in smooth muscle and to physiological and pathophysiological responses with emphasis on the vascular system.
Collapse
|
46
|
Wölfle SE, Navarro-Gonzalez MF, Grayson TH, Stricker C, Hill CE. Involvement of nonselective cation channels in the depolarisation initiating vasomotion. Clin Exp Pharmacol Physiol 2010. [DOI: 10.1111/j.1440-1681.2010.05350.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
47
|
Abstract
TRP (transient receptor potential) channels play important roles in the regulation of normal and pathological cellular function. In the vasculature, TRP channels are present both in ECs (endothelial cells) and vascular SMCs (smooth muscle cells) and contribute to vasomotor control mechanisms in most vascular beds. Vascular TRP channels are activated by various stimuli, such as mechanical perturbation, receptor activation and dietary molecules. Some of the specific roles of these channels in normal and impaired vascular function have emerged in recent years and include participation in vascular signalling processes, such as neurotransmission, hormonal signalling, NO production, myogenic tone and autoregulation of blood flow, thermoregulation, responses to oxidative stress and cellular proliferative activity. Current research is aimed at understanding the interactions of TRP channels with other vascular proteins and signalling mechanisms. These studies should reveal new targets for pharmacological therapy of vascular diseases, such as hypertension, ischaemia and vasospasm, and vascular proliferative states.
Collapse
|
48
|
Mechanosensitive channels in striated muscle and the cardiovascular system: not quite a stretch anymore. J Cardiovasc Pharmacol 2009; 54:116-22. [PMID: 19597371 DOI: 10.1097/fjc.0b013e3181aa233f] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Stretch-activated or mechanosensitive channels transduce mechanical forces into ion fluxes across the cell membrane. These channels have been implicated in several aspects of cardiovascular physiology including regulation of blood pressure, vasoreactivity, and cardiac arrhythmias, as well as the adverse remodeling associated with cardiac hypertrophy and heart failure. This review discusses mechanosensitive channels in skeletal muscle and the cardiovascular system and their role in disease pathogenesis. We describe the regulation of gating of mechanosensitive channels including direct mechanisms and indirect activation by signaling pathways, as well as the influence on activation of these channels by the underlying cytoskeleton and scaffolding proteins. We then focus on the role of transient receptor potential channels, several of which have been implicated as mechanosensitive channels, in the pathogenesis of adverse cardiac remodeling and as potential therapeutic targets in the treatment of heart failure.
Collapse
|
49
|
Vecchione C, Frati A, Di Pardo A, Cifelli G, Carnevale D, Gentile MT, Carangi R, Landolfi A, Carullo P, Bettarini U, Antenucci G, Mascio G, Busceti CL, Notte A, Maffei A, Cantore GP, Lembo G. Tumor Necrosis Factor-α Mediates Hemolysis-Induced Vasoconstriction and the Cerebral Vasospasm Evoked by Subarachnoid Hemorrhage. Hypertension 2009; 54:150-6. [DOI: 10.1161/hypertensionaha.108.128124] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Carmine Vecchione
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Alessandro Frati
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Alba Di Pardo
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Giuseppe Cifelli
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Daniela Carnevale
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Maria Teresa Gentile
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Rosa Carangi
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Alessandro Landolfi
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Pierluigi Carullo
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Umberto Bettarini
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Giovanna Antenucci
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Giada Mascio
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Carla Letizia Busceti
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Antonella Notte
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Angelo Maffei
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Gian Paolo Cantore
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| | - Giuseppe Lembo
- From the Departments of Angiocardioneurology (C.V., A.D.P., G.C., M.T.G., R.C., A.L., P.C., U.B., G.A., G.M., C.L.B., A.N., A.M., G.L.) and Neurosurgery (A.F., G.P.C.), IRCCS Neuromed, Pozzilli (IS), Italy; Department of Cell Biology and Neurosciences (D.C.), Istituto Superiore di Sanità, Rome, Italy; and Department of Experimental Medicine (G.L.), Sapienza University, Rome, Italy
| |
Collapse
|
50
|
Abramowitz J, Birnbaumer L. Physiology and pathophysiology of canonical transient receptor potential channels. FASEB J 2009; 23:297-328. [PMID: 18940894 PMCID: PMC2630793 DOI: 10.1096/fj.08-119495] [Citation(s) in RCA: 253] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Accepted: 09/25/2008] [Indexed: 11/11/2022]
Abstract
The existence of a mammalian family of TRPC ion channels, direct homologues of TRP, the visual transduction channel of flies, was discovered during 1995-1996 as a consequence of research into the mechanism by which the stimulation of the receptor-Gq-phospholipase Cbeta signaling pathway leads to sustained increases in intracellular calcium. Mammalian TRPs, TRPCs, turned out to be nonselective, calcium-permeable cation channels, which cause both a collapse of the cell's membrane potential and entry of calcium. The family comprises 7 members and is widely expressed. Many cells and tissues express between 3 and 4 of the 7 TRPCs. Despite their recent discovery, a wealth of information has accumulated, showing that TRPCs have widespread roles in almost all cells studied, including cells from excitable and nonexcitable tissues, such as the nervous and cardiovascular systems, the kidney and the liver, and cells from endothelia, epithelia, and the bone marrow compartment. Disruption of TRPC function is at the root of some familial diseases. More often, TRPCs are contributing risk factors in complex diseases. The present article reviews what has been uncovered about physiological roles of mammalian TRPC channels since the time of their discovery. This analysis reveals TRPCs as major and unsuspected gates of Ca(2+) entry that contribute, depending on context, to activation of transcription factors, apoptosis, vascular contractility, platelet activation, and cardiac hypertrophy, as well as to normal and abnormal cell proliferation. TRPCs emerge as targets for a thus far nonexistent field of pharmacological intervention that may ameliorate complex diseases.
Collapse
Affiliation(s)
- Joel Abramowitz
- Transmembrane Signaling Group, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| | | |
Collapse
|